Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article in English | MEDLINE | ID: mdl-33468657

ABSTRACT

DNA damage repair genes are modifiers of disease onset in Huntington's disease (HD), but how this process intersects with associated disease pathways remains unclear. Here we evaluated the mechanistic contributions of protein inhibitor of activated STAT-1 (PIAS1) in HD mice and HD patient-derived induced pluripotent stem cells (iPSCs) and find a link between PIAS1 and DNA damage repair pathways. We show that PIAS1 is a component of the transcription-coupled repair complex, that includes the DNA damage end processing enzyme polynucleotide kinase-phosphatase (PNKP), and that PIAS1 is a SUMO E3 ligase for PNKP. Pias1 knockdown (KD) in HD mice had a normalizing effect on HD transcriptional dysregulation associated with synaptic function and disease-associated transcriptional coexpression modules enriched for DNA damage repair mechanisms as did reduction of PIAS1 in HD iPSC-derived neurons. KD also restored mutant HTT-perturbed enzymatic activity of PNKP and modulated genomic integrity of several transcriptionally normalized genes. The findings here now link SUMO modifying machinery to DNA damage repair responses and transcriptional modulation in neurodegenerative disease.


Subject(s)
DNA Repair Enzymes/genetics , DNA Repair , DNA/genetics , Huntingtin Protein/genetics , Huntington Disease/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Inhibitors of Activated STAT/genetics , Protein Processing, Post-Translational , Small Ubiquitin-Related Modifier Proteins/genetics , Animals , Cell Differentiation , DNA/metabolism , DNA Damage , DNA Repair Enzymes/metabolism , Disease Models, Animal , Female , Humans , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Huntington Disease/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Primary Cell Culture , Protein Inhibitors of Activated STAT/antagonists & inhibitors , Protein Inhibitors of Activated STAT/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Small Ubiquitin-Related Modifier Proteins/antagonists & inhibitors , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Transcription, Genetic
2.
Proc Natl Acad Sci U S A ; 116(22): 10952-10961, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31088970

ABSTRACT

Neuroinflammation is an important contributor to neuronal pathology and death in neurodegenerative diseases and neuronal injury. Therapeutic interventions blocking the activity of the inflammatory kinase IKKß, a key regulator of neuroinflammatory pathways, is protective in several animal models of neurodegenerative disease and neuronal injury. In Huntington's disease (HD), however, significant questions exist as to the impact of blocking or diminishing the activity of IKKß on HD pathology given its potential role in Huntingtin (HTT) degradation. In cell culture, IKKß phosphorylates HTT serine (S) 13 and activates HTT degradation, a process that becomes impaired with polyQ expansion. To investigate the in vivo relationship of IKKß to HTT S13 phosphorylation and HD progression, we crossed conditional tamoxifen-inducible IKKß knockout mice with R6/1 HD mice. Behavioral assays in these mice showed a significant worsening of HD pathological phenotypes. The increased behavioral pathology correlated with reduced levels of endogenous mouse full-length phospho-S13 HTT, supporting the importance of IKKß in the phosphorylation of HTT S13 in vivo. Notably, many striatal autophagy genes were up-regulated in HD vs. control mice; however, IKKß knockout partially reduced this up-regulation in HD, increased striatal neurodegeneration, and enhanced an activated microglial response. We propose that IKKß is protective in striatal neurons early in HD progression via phosphorylation of HTT S13. As IKKß is also required for up-regulation of some autophagy genes and HTT is a scaffold for selective autophagy, IKKß may influence autophagy through multiple mechanisms to maintain healthy striatal function, thereby reducing neuronal degeneration to slow HD onset.


Subject(s)
Huntington Disease , I-kappa B Kinase , Animals , Autophagy/genetics , Corpus Striatum/cytology , Corpus Striatum/pathology , Disease Models, Animal , Disease Progression , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Huntington Disease/pathology , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Male , Mice , Mice, Knockout , Microglia/cytology , Microglia/pathology , Phosphorylation/genetics
3.
Brain ; 143(1): 266-288, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31848580

ABSTRACT

Huntington's disease is associated with a reactive microglial response and consequent inflammation. To address the role of these cells in disease pathogenesis, we depleted microglia from R6/2 mice, a rapidly progressing model of Huntington's disease marked by behavioural impairment, mutant huntingtin (mHTT) accumulation, and early death, through colony-stimulating factor 1 receptor inhibition (CSF1Ri) with pexidartinib (PLX3397) for the duration of disease. Although we observed an interferon gene signature in addition to downregulated neuritogenic and synaptic gene pathways with disease, overt inflammation was not evident by microglial morphology or cytokine transcript levels in R6/2 mice. Nonetheless, CSF1Ri-induced microglial elimination reduced or prevented disease-related grip strength and object recognition deficits, mHTT accumulation, astrogliosis, and striatal volume loss, the latter of which was not associated with reductions in cell number but with the extracellular accumulation of chondroitin sulphate proteoglycans (CSPGs)-a primary component of glial scars. A concurrent loss of proteoglycan-containing perineuronal nets was also evident in R6/2 mice, and microglial elimination not only prevented this but also strikingly increased perineuronal nets in the brains of naïve littermates, suggesting a new role for microglia as homeostatic regulators of perineuronal net formation and integrity.


Subject(s)
Aminopyridines/pharmacology , Extracellular Matrix/drug effects , Huntingtin Protein/drug effects , Huntington Disease/metabolism , Microglia/drug effects , Neostriatum/drug effects , Pyrroles/pharmacology , Recognition, Psychology/drug effects , Animals , Astrocytes/drug effects , Chondroitin Sulfate Proteoglycans/drug effects , Chondroitin Sulfate Proteoglycans/metabolism , Cytokines/drug effects , Cytokines/genetics , Disease Models, Animal , Down-Regulation , Extracellular Matrix/metabolism , Hand Strength , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/pathology , Huntington Disease/physiopathology , Inflammation , Mice , Mice, Transgenic , Neostriatum/pathology , Neurites/drug effects , RNA, Messenger/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Synapses/drug effects , Transcriptome
4.
Nucleic Acids Res ; 47(21): 11284-11303, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31612951

ABSTRACT

Endocytosis is a mechanism by which cells sense their environment and internalize various nutrients, growth factors and signaling molecules. This process initiates at the plasma membrane, converges with autophagy, and terminates at the lysosome. It is well-established that cellular uptake of antisense oligonucleotides (ASOs) proceeds through the endocytic pathway; however, only a small fraction escapes endosomal trafficking while the majority are rendered inactive in the lysosome. Since these pathways converge and share common molecular machinery, it is unclear if autophagy-related trafficking participates in ASO uptake or whether modulation of autophagy affects ASO activity and localization. To address these questions, we investigated the effects of autophagy modulation on ASO activity in cells and mice. We found that enhancing autophagy through small-molecule mTOR inhibition, serum-starvation/fasting, and ketogenic diet, increased ASO-mediated target reduction in vitro and in vivo. Additionally, autophagy activation enhanced the localization of ASOs into autophagosomes without altering intracellular concentrations or trafficking to other compartments. These results support a novel role for autophagy and the autophagosome as a previously unidentified compartment that participates in and contributes to enhanced ASO activity. Further, we demonstrate non-chemical methods to enhance autophagy and subsequent ASO activity using translatable approaches such as fasting or ketogenic diet.


Subject(s)
Autophagy/physiology , Oligonucleotides, Antisense/metabolism , Animals , Autophagosomes/metabolism , Biological Transport/physiology , Cells, Cultured , Endocytosis/physiology , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Oligonucleotides, Antisense/genetics , RNA Interference , Signal Transduction
5.
Neurobiol Dis ; 109(Pt A): 127-136, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28986324

ABSTRACT

Huntington's disease (HD) is caused by a CAG repeat expansion that encodes a polyglutamine (polyQ) expansion in the HD disease protein, huntingtin (HTT). PolyQ expansion promotes misfolding and aggregation of mutant HTT (mHTT) within neurons. The cellular pathways, including ubiquitin-dependent processes, by which mHTT is regulated remain incompletely understood. Ube2W is the only ubiquitin conjugating enzyme (E2) known to ubiquitinate substrates at their amino (N)-termini, likely favoring substrates with disordered N-termini. By virtue of its N-terminal polyQ domain, HTT has an intrinsically disordered amino terminus. In studies employing immortalized cells, primary neurons and a knock-in (KI) mouse model of HD, we tested the effect of Ube2W deficiency on mHTT levels, aggregation and neurotoxicity. In cultured cells, deficiency of Ube2W activity markedly decreases mHTT aggregate formation and increases the level of soluble monomers, while reducing mHTT-induced cytotoxicity. Consistent with this result, the absence of Ube2W in HdhQ200 KI mice significantly increases levels of soluble monomeric mHTT while reducing insoluble oligomeric species. This study sheds light on the potential function of the non-canonical ubiquitin-conjugating enzyme, Ube2W, in this polyQ neurodegenerative disease.


Subject(s)
Huntingtin Protein/metabolism , Huntington Disease/enzymology , Neurons/enzymology , Ubiquitin-Conjugating Enzymes/metabolism , Animals , Corpus Striatum/enzymology , Corpus Striatum/pathology , Disease Models, Animal , Gene Knock-In Techniques , HEK293 Cells , Humans , Huntington Disease/genetics , Inclusion Bodies/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology , Primary Cell Culture , Ubiquitin-Conjugating Enzymes/genetics
6.
Proc Natl Acad Sci U S A ; 111(47): 16889-94, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25385587

ABSTRACT

Although dominant gain-of-function triplet repeat expansions in the Huntingtin (HTT) gene are the underlying cause of Huntington disease (HD), understanding the normal functions of nonmutant HTT protein has remained a challenge. We report here findings that suggest that HTT plays a significant role in selective autophagy. Loss of HTT function in Drosophila disrupts starvation-induced autophagy in larvae and conditional knockout of HTT in the mouse CNS causes characteristic cellular hallmarks of disrupted autophagy, including an accumulation of striatal p62/SQSTM1 over time. We observe that specific domains of HTT have structural similarities to yeast Atg proteins that function in selective autophagy, and in particular that the C-terminal domain of HTT shares structural similarity to yeast Atg11, an autophagic scaffold protein. To explore possible functional similarity between HTT and Atg11, we investigated whether the C-terminal domain of HTT interacts with mammalian counterparts of yeast Atg11-interacting proteins. Strikingly, this domain of HTT coimmunoprecipitates with several key Atg11 interactors, including the Atg1/Unc-51-like autophagy activating kinase 1 kinase complex, autophagic receptor proteins, and mammalian Atg8 homologs. Mutation of a phylogenetically conserved WXXL domain in a C-terminal HTT fragment reduces coprecipitation with mammalian Atg8 homolog GABARAPL1, suggesting a direct interaction. Collectively, these data support a possible central role for HTT as an Atg11-like scaffold protein. These findings have relevance to both mechanisms of disease pathogenesis and to therapeutic intervention strategies that reduce levels of both mutant and normal HTT.


Subject(s)
Autophagy , Microtubule-Associated Proteins/physiology , Animals , Animals, Genetically Modified , Drosophila , Drosophila Proteins , Huntingtin Protein , Mice , Microtubule-Associated Proteins/genetics
7.
J Undergrad Neurosci Educ ; 16(1): A95-A101, 2017.
Article in English | MEDLINE | ID: mdl-29371848

ABSTRACT

Flipped instruction using online enrichment is a popular way to enhance active learning in the laboratory setting. Graduate student teaching assistants at University of California, Irvine flipped an upper division undergraduate neurobiology and behavior lab using the new online software platform "Rocketmix." The following research study compares the impact of pre-lab online instruction (front flipping) and post-lab online instruction (back flipping) on student exam performance. We describe a novel method for unbiased categorization of exam questions by degree of difficulty. Multi-choice instruction encourages students to consider all distractors and discourages verbal cues and process of elimination techniques. Eighteen identical questions were evenly distributed across exam versions with multiple choice instruction (single answer) or a more challenging multi-choice instruction (more than one answer). Student performance on multiple choice questions were used to categorize the degree of difficulty of questions that were presented in multi-choice format. Our findings reveal that pre-lab instruction resulted in better student performance compared with post-lab instruction on questions of moderate difficulty. This effect was significant for both male and female students. Student survey data on the flipped lab format is provided, indicating that students appreciated the online instructional modules, finding them both informative and useful during lab exercises and exams.

8.
Res Sq ; 2023 Jan 27.
Article in English | MEDLINE | ID: mdl-36747781

ABSTRACT

Neurodevelopmental disorders (NDDs) are frequently associated with dendritic abnormalities in pyramidal neurons that affect arbor complexity, spine density, and synaptic communication 1,2. The underlying genetic causes are often complex, obscuring the molecular pathways that drive these disorders 3. Next-generation sequencing has identified recurrent de novo missense mutations in a handful of genes associated with NDDs, offering a unique opportunity to decipher the molecular pathways 4. One such gene is PACS1, which encodes the multi-functional trafficking protein PACS1 (or PACS-1); a single recurrent de novo missense mutation, c607C>T (PACS1R203W), causes developmental delay and intellectual disability (ID) 5,6. The processes by which PACS1R203W causes PACS1 syndrome are unknown, and there is no curative treatment. We show that PACS1R203W increases the interaction between PACS1 and the α-tubulin deacetylase HDAC6, elevating enzyme activity and appropriating control of its posttranscriptional regulation. Consequently, PACS1R203W reduces acetylation of α-tubulin and cortactin, causing the Golgi to fragment and enter developing neurites, leading to increased dendrite arborization. The dendrites, however, are beset with diminished spine density and fewer functional synapses, characteristic of ID pathology. Treatment of PACS1 syndrome mice with PACS1- or HDAC6-targeting antisense oligonucleotides restores neuronal structure and synaptic transmission, suggesting PACS1R203W/HDAC6 may be targeted for treating PACS1 syndrome neuropathology.

9.
Nat Commun ; 14(1): 6547, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37848409

ABSTRACT

PACS1 syndrome is a neurodevelopmental disorder (NDD) caused by a recurrent de novo missense mutation in PACS1 (p.Arg203Trp (PACS1R203W)). The mechanism by which PACS1R203W causes PACS1 syndrome is unknown, and no curative treatment is available. Here, we use patient cells and PACS1 syndrome mice to show that PACS1 (or PACS-1) is an HDAC6 effector and that the R203W substitution increases the PACS1/HDAC6 interaction, aberrantly potentiating deacetylase activity. Consequently, PACS1R203W reduces acetylation of α-tubulin and cortactin, causing the Golgi ribbon in hippocampal neurons and patient-derived neural progenitor cells (NPCs) to fragment and overpopulate dendrites, increasing their arborization. The dendrites, however, are beset with varicosities, diminished spine density, and fewer functional synapses, characteristic of NDDs. Treatment of PACS1 syndrome mice or patient NPCs with PACS1- or HDAC6-targeting antisense oligonucleotides, or HDAC6 inhibitors, restores neuronal structure and synaptic transmission in prefrontal cortex, suggesting that targeting PACS1R203W/HDAC6 may be an effective therapy for PACS1 syndrome.


Subject(s)
Histone Deacetylases , Tubulin , Humans , Mice , Animals , Histone Deacetylase 6/genetics , Histone Deacetylase 6/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Tubulin/metabolism , Neurons/metabolism , Protein Processing, Post-Translational , Syndrome , Acetylation , Histone Deacetylase Inhibitors/pharmacology , Vesicular Transport Proteins/genetics
10.
Nat Med ; 26(9): 1444-1451, 2020 09.
Article in English | MEDLINE | ID: mdl-32719489

ABSTRACT

CLN3 Batten disease is an autosomal recessive, neurodegenerative, lysosomal storage disease caused by mutations in CLN3, which encodes a lysosomal membrane protein1-3. There are no disease-modifying treatments for this disease that affects up to 1 in 25,000 births, has an onset of symptoms in early childhood and typically is fatal by 20-30 years of life4-7. Most patients with CLN3 Batten have a deletion encompassing exons 7 and 8 (CLN3∆ex7/8), creating a reading frameshift7,8. Here we demonstrate that mice with this deletion can be effectively treated using an antisense oligonucleotide (ASO) that induces exon skipping to restore the open reading frame. A single treatment of neonatal mice with an exon 5-targeted ASO-induced robust exon skipping for more than a year, improved motor coordination, reduced histopathology in Cln3∆ex7/8 mice and increased survival in a new mouse model of the disease. ASOs also induced exon skipping in cell lines derived from patients with CLN3 Batten disease. Our findings demonstrate the utility of ASO-based reading-frame correction as an approach to treat CLN3 Batten disease and broaden the therapeutic landscape for ASOs in the treatment of other diseases using a similar strategy.


Subject(s)
Membrane Glycoproteins/genetics , Molecular Chaperones/genetics , Neuronal Ceroid-Lipofuscinoses/drug therapy , Neuronal Ceroid-Lipofuscinoses/genetics , Oligonucleotides, Antisense/therapeutic use , Animals , Cell Line , Codon, Nonsense/genetics , Disease Models, Animal , Frameshift Mutation/genetics , Humans , Mice
11.
JCI Insight ; 4(12)2019 06 20.
Article in English | MEDLINE | ID: mdl-31217349

ABSTRACT

African Americans develop end-stage renal disease at a higher rate compared with European Americans due to 2 polymorphisms (G1 and G2 risk variants) in the apolipoprotein L1 (APOL1) gene common in people of African ancestry. Although this compelling genetic evidence provides an exciting opportunity for personalized medicine in chronic kidney disease, drug discovery efforts have been greatly hindered by the fact that APOL1 expression is lacking in rodents. Here, we describe a potentially novel physiologically relevant genomic mouse model of APOL1-associated renal disease that expresses human APOL1 from the endogenous human promoter, resulting in expression in similar tissues and at similar relative levels as humans. While naive APOL1-transgenic mice did not exhibit a renal disease phenotype, administration of IFN-γ was sufficient to robustly induce proteinuria only in APOL1 G1 mice, despite inducing kidney APOL1 expression in both G0 and G1 mice, serving as a clinically relevant "second hit." Treatment of APOL1 G1 mice with IONIS-APOL1Rx, an antisense oligonucleotide (ASO) targeting APOL1 mRNA, prior to IFN-γ challenge robustly and dose-dependently inhibited kidney and liver APOL1 expression and protected against IFN-γ-induced proteinuria, indicating that the disease-relevant cell types are sensitive to ASO treatment. Therefore, IONIS-APOL1Rx may be an effective therapeutic for APOL1 nephropathies and warrants further development.


Subject(s)
Apolipoprotein L1/genetics , Interferon-gamma , Oligonucleotides, Antisense/therapeutic use , Proteinuria/drug therapy , Proteinuria/etiology , Animals , Cell Line , Female , Humans , Mice , Mice, Transgenic
12.
J Vis Exp ; (132)2018 02 27.
Article in English | MEDLINE | ID: mdl-29553509

ABSTRACT

The accumulation of misfolded proteins is central to pathology in Huntington's disease (HD) and many other neurodegenerative disorders. Specifically, a key pathological feature of HD is the aberrant accumulation of mutant HTT (mHTT) protein into high molecular weight complexes and intracellular inclusion bodies composed of fragments and other proteins. Conventional methods to measure and understand the contributions of various forms of mHTT-containing aggregates include fluorescence microscopy, western blot analysis, and filter trap assays. However, most of these methods are conformation specific, and therefore may not resolve the full state of mHTT protein flux due to the complex nature of aggregate solubility and resolution. For the identification of aggregated mHTT and various modified forms and complexes, separation and solubilization of the cellular aggregates and fragments is mandatory. Here we describe a method to isolate and visualize soluble mHTT, monomers, oligomers, fragments, and an insoluble high molecular weight (HMW) accumulated mHTT species. HMW mHTT tracks with disease progression, corresponds with mouse behavior readouts, and has been beneficially modulated by certain therapeutic interventions1. This approach can be used with mouse brain, peripheral tissues, and cell culture but may be adapted to other model systems or disease contexts.


Subject(s)
Dose Fractionation, Radiation , Huntingtin Protein/genetics , Huntington Disease/drug therapy , Animals , Humans , Huntingtin Protein/metabolism , Mice , Models, Biological
13.
J Huntingtons Dis ; 7(4): 321-335, 2018.
Article in English | MEDLINE | ID: mdl-30452420

ABSTRACT

BACKGROUND: Biochemical analysis of mutant huntingtin (mHTT) aggregation species in HD mice is a common measure to track disease. A longitudinal and systematic study of how tissue processing affects detection of conformers has not yet been reported. Understanding the homeostatic flux of mHTT over time and under different processing conditions would aid in interpretation of pre-clinical assessments of disease interventions. OBJECTIVE: Provide a systematic evaluation of tissue lysis methods and molecular and biochemical assays in parallel with behavioral readouts in R6/2 mice to establish a baseline for HTT exon1 protein accumulation. METHODS: Established biochemical methods were used to process tissue from R6/2 mice of specific ages following behavior tasks. Aggregation states and accumulation of mHTT exon 1 protein were evaluated using multiple break and assay methods to determine potential conformational flux assay specificity in detection of mHTT species, and tissue specificity of conformers. RESULTS: Detection of mHTT exon 1 protein species varied based on biochemical processing and analysis providing a baseline for subsequent studies in R6/2 mice. Insoluble, high molecular weight species of mHTT exon 1 protein increased and tracked with onset of behavioral impairments in R6/2 mice using multiple assay methods. CONCLUSIONS: Conformational flux from soluble monomer to high molecular weight, insoluble species of mHTT exon 1 protein was generally consistent for multiple assay methods throughout R6/2 disease progression; however, the results support the use of multiple biochemical techniques to detect mHTT exon 1 protein species for preclinical assessments in HD mouse models expressing mHTT exon 1 protein.


Subject(s)
Brain/metabolism , Huntingtin Protein/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Disease Models, Animal , Electrophoresis, Agar Gel , Electrophoresis, Polyacrylamide Gel , Exons , Humans , Huntingtin Protein/genetics , Huntington Disease/metabolism , Longitudinal Studies , Mice , Mice, Inbred Strains , Mice, Transgenic , Specimen Handling
14.
J Huntingtons Dis ; 7(2): 137-150, 2018.
Article in English | MEDLINE | ID: mdl-29843246

ABSTRACT

BACKGROUND: Huntington's disease (HD) is a progressive neurodegenerative disorder associated with aging, caused by an expanded polyglutamine (polyQ) repeat within the Huntingtin (HTT) protein. In HD, degeneration of the striatum and atrophy of the cortex are observed while cerebellum is less affected. OBJECTIVE: To test the hypothesis that HTT protein levels decline with age, which together with HTT mutation could influence disease progression. METHODS: Using whole brain cell lysates, a unique method of SDS-PAGE and western analysis was used to quantitate HTT protein, which resolves as a monomer and as a high molecular weight species that is modulated by the presence of transglutaminase 2. HTT levels were measured in striatum, cortex and cerebellum in congenic homozygous Q140 and HdhQ150 knock-in mice and WT littermate controls. RESULTS: Mutant HTT in both homozygous knock-in HD mouse models and WT HTT in control striatal and cortical tissues significantly declined in a progressive manner over time. Levels of mutant HTT in HD cerebellum remained high during aging. CONCLUSIONS: A general decline in mutant HTT levels in striatum and cortex is observed that may contribute to disease progression in homozygous knock-in HD mouse models through reduction of HTT function. In cerebellum, sustained levels of mutant HTT with aging may be protective to this tissue which is less overtly affected in HD.


Subject(s)
Corpus Striatum/metabolism , Disease Progression , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Aging , Animals , Cerebellum/metabolism , Cerebral Cortex/metabolism , Disease Models, Animal , Female , Gene Knock-In Techniques , Homozygote , Huntingtin Protein/genetics , Male , Mice, Inbred C57BL , Mutant Proteins/genetics , Mutant Proteins/metabolism
15.
Stem Cell Reports ; 10(1): 58-72, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29233555

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder with no disease-modifying treatment. Expansion of the glutamine-encoding repeat in the Huntingtin (HTT) gene causes broad effects that are a challenge for single treatment strategies. Strategies based on human stem cells offer a promising option. We evaluated efficacy of transplanting a good manufacturing practice (GMP)-grade human embryonic stem cell-derived neural stem cell (hNSC) line into striatum of HD modeled mice. In HD fragment model R6/2 mice, transplants improve motor deficits, rescue synaptic alterations, and are contacted by nerve terminals from mouse cells. Furthermore, implanted hNSCs are electrophysiologically active. hNSCs also improved motor and late-stage cognitive impairment in a second HD model, Q140 knockin mice. Disease-modifying activity is suggested by the reduction of aberrant accumulation of mutant HTT protein and expression of brain-derived neurotrophic factor (BDNF) in both models. These findings hold promise for future development of stem cell-based therapies.


Subject(s)
Cognition , Huntington Disease/therapy , Motor Activity , Neural Stem Cells/transplantation , Recovery of Function , Animals , Cell Line , Disease Models, Animal , Heterografts , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/pathology , Humans , Huntington Disease/metabolism , Huntington Disease/pathology , Huntington Disease/physiopathology , Mice , Neural Stem Cells/metabolism , Neural Stem Cells/pathology
17.
Stem Cells Transl Med ; 6(6): 1477-1490, 2017 06.
Article in English | MEDLINE | ID: mdl-28225193

ABSTRACT

Synucleinopathies are a group of neurodegenerative disorders sharing the common feature of misfolding and accumulation of the presynaptic protein α-synuclein (α-syn) into insoluble aggregates. Within this diverse group, Dementia with Lewy Bodies (DLB) is characterized by the aberrant accumulation of α-syn in cortical, hippocampal, and brainstem neurons, resulting in multiple cellular stressors that particularly impair dopamine and glutamate neurotransmission and related motor and cognitive function. Recent studies show that murine neural stem cell (NSC) transplantation can improve cognitive or motor function in transgenic models of Alzheimer's and Huntington's disease, and DLB. However, examination of clinically relevant human NSCs in these models is hindered by the challenges of xenotransplantation and the confounding effects of immunosuppressant drugs on pathology and behavior. To address this challenge, we developed an immune-deficient transgenic model of DLB that lacks T-, B-, and NK-cells, yet exhibits progressive accumulation of human α-syn (h-α-syn)-laden inclusions and cognitive and motor impairments. We demonstrate that clinically relevant human neural progenitor cells (line CNS10-hNPCs) survive, migrate extensively and begin to differentiate preferentially into astrocytes following striatal transplantation into this DLB model. Critically, grafted CNS10-hNPCs rescue both cognitive and motor deficits after 1 and 3 months and, furthermore, restore striatal dopamine and glutamate systems. These behavioral and neurochemical benefits are likely achieved by reducing α-syn oligomers. Collectively, these results using a new model of DLB demonstrate that hNPC transplantation can impact a broad array of disease mechanisms and phenotypes and suggest a cellular therapeutic strategy that should be pursued. Stem Cells Translational Medicine 2017;6:1477-1490.


Subject(s)
Lewy Body Disease/therapy , Neural Stem Cells/transplantation , Stem Cell Transplantation/methods , alpha-Synuclein/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cells, Cultured , Humans , Memory , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis
18.
Neuron ; 94(1): 93-107.e6, 2017 Apr 05.
Article in English | MEDLINE | ID: mdl-28384479

ABSTRACT

Huntington's disease (HD) is caused by an expanded CAG repeat in the Huntingtin (HTT) gene. The mechanism(s) by which mutant HTT (mHTT) causes disease is unclear. Nucleocytoplasmic transport, the trafficking of macromolecules between the nucleus and cytoplasm, is tightly regulated by nuclear pore complexes (NPCs) made up of nucleoporins (NUPs). Previous studies offered clues that mHTT may disrupt nucleocytoplasmic transport and a mutation of an NUP can cause HD-like pathology. Therefore, we evaluated the NPC and nucleocytoplasmic transport in multiple models of HD, including mouse and fly models, neurons transfected with mHTT, HD iPSC-derived neurons, and human HD brain regions. These studies revealed severe mislocalization and aggregation of NUPs and defective nucleocytoplasmic transport. HD repeat-associated non-ATG (RAN) translation proteins also disrupted nucleocytoplasmic transport. Additionally, overexpression of NUPs and treatment with drugs that prevent aberrant NUP biology also mitigated this transport defect and neurotoxicity, providing future novel therapy targets.


Subject(s)
Active Transport, Cell Nucleus/genetics , Huntingtin Protein/genetics , Huntington Disease/genetics , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore/metabolism , Adult , Animals , Disease Models, Animal , Drosophila , Drosophila Proteins , Female , Humans , Induced Pluripotent Stem Cells , Male , Mice , Middle Aged , Mutation , Young Adult
19.
Neuron ; 90(3): 507-20, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27146268

ABSTRACT

The disruption of protein quality control networks is central to pathology in Huntington's disease (HD) and other neurodegenerative disorders. The aberrant accumulation of insoluble high-molecular-weight protein complexes containing the Huntingtin (HTT) protein and SUMOylated protein corresponds to disease manifestation. We previously identified an HTT-selective E3 SUMO ligase, PIAS1, that regulates HTT accumulation and SUMO modification in cells. Here we investigated whether PIAS1 modulation in neurons alters HD-associated phenotypes in vivo. Instrastriatal injection of a PIAS1-directed miRNA significantly improved behavioral phenotypes in rapidly progressing mutant HTT (mHTT) fragment R6/2 mice. PIAS1 reduction prevented the accumulation of mHTT and SUMO- and ubiquitin-modified proteins, increased synaptophysin levels, and normalized key inflammatory markers. In contrast, PIAS1 overexpression exacerbated mHTT-associated phenotypes and aberrant protein accumulation. These results confirm the association between aberrant accumulation of expanded polyglutamine-dependent insoluble protein species and pathogenesis, and they link phenotypic benefit to reduction of these species through PIAS1 modulation.


Subject(s)
Brain/metabolism , Huntington Disease/genetics , Mutation/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , Protein Inhibitors of Activated STAT/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Animals , Disease Models, Animal , Humans , Huntingtin Protein/genetics , Mice , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Phenotype
20.
Cell Rep ; 4(2): 362-75, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23871671

ABSTRACT

A key feature in Huntington disease (HD) is the accumulation of mutant Huntingtin (HTT) protein, which may be regulated by posttranslational modifications. Here, we define the primary sites of SUMO modification in the amino-terminal domain of HTT, show modification downstream of this domain, and demonstrate that HTT is modified by the stress-inducible SUMO-2. A systematic study of E3 SUMO ligases demonstrates that PIAS1 is an E3 SUMO ligase for both HTT SUMO-1 and SUMO-2 modification and that reduction of dPIAS in a mutant HTT Drosophila model is protective. SUMO-2 modification regulates accumulation of insoluble HTT in HeLa cells in a manner that mimics proteasome inhibition and can be modulated by overexpression and acute knockdown of PIAS1. Finally, the accumulation of SUMO-2-modified proteins in the insoluble fraction of HD postmortem striata implicates SUMO-2 modification in the age-related pathogenic accumulation of mutant HTT and other cellular proteins that occurs during HD progression.


Subject(s)
Huntington Disease/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Inhibitors of Activated STAT/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Catalytic Domain , Drosophila , Female , HeLa Cells , Humans , Huntingtin Protein , Huntington Disease/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Molecular Sequence Data , Mutation , Protein Inhibitors of Activated STAT/genetics , Protein Processing, Post-Translational , Small Ubiquitin-Related Modifier Proteins/genetics , Transfection , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL