73 results on '"Shihabuddin LS"'
Search Results
2. Acid [beta]-glucosidase mutants linked to gaucher disease, parkinson disease, and lewy body dementia alter [alpha]-synuclein processing.
- Author
-
Cullen V, Sardi SP, Ng J, Xu YH, Sun Y, Tomlinson JJ, Kolodziej P, Kahn I, Saftig P, Woulfe J, Rochet JC, Glicksman MA, Cheng SH, Grabowski GA, Shihabuddin LS, and Schlossmacher MG
- Subjects
ANIMAL experimentation ,ANIMALS ,BIOLOGICAL models ,CELLS ,DOSE-effect relationship in pharmacology ,ENZYME-linked immunosorbent assay ,GAUCHER'S disease ,GENES ,GENETIC techniques ,GLYCOSIDASES ,IMMUNOSUPPRESSIVE agents ,LEWY body dementia ,MICE ,GENETIC mutation ,NERVE tissue proteins ,PARKINSON'S disease ,PROTEINS ,PROTEOLYTIC enzymes ,RATS ,RAPAMYCIN ,PHARMACODYNAMICS - Abstract
OBJECTIVE: Heterozygous mutations in the GBA1 gene elevate the risk of Parkinson disease and dementia with Lewy bodies; both disorders are characterized by misprocessing of [alpha]-synuclein (SNCA). A loss in lysosomal acid-[beta]-glucosidase enzyme (GCase) activity due to biallelic GBA1 mutations underlies Gaucher disease. We explored mechanisms for the gene's association with increased synucleinopathy risk. METHODS: We analyzed the effects of wild-type (WT) and several GBA mutants on SNCA in cellular and in vivo models using biochemical and immunohistochemical protocols. RESULTS: We observed that overexpression of all GBA mutants examined (N370S, L444P, D409H, D409V, E235A, and E340A) significantly raised human SNCA levels to 121 to 248% of vector control (p < 0.029) in neural MES23.5 and PC12 cells, but without altering GCase activity. Overexpression of WT GBA in neural and HEK293-SNCA cells increased GCase activity, as expected (ie, to 167% in MES-SNCA, 128% in PC12-SNCA, and 233% in HEK293-SNCA; p < 0.002), but had mixed effects on SNCA. Nevertheless, in HEK293-SNCA cells high GCase activity was associated with SNCA reduction by <=32% (p = 0.009). Inhibition of cellular GCase activity (to 8-20% of WT; p < 0.0017) did not detectably alter SNCA levels. Mutant GBA-induced SNCA accumulation could be pharmacologically reversed in D409V-expressing PC12-SNCA cells by rapamycin, an autophagy-inducer (<=40%; 10[mu]M; p < 0.02). Isofagomine, a GBA chaperone, showed a related trend. In mice expressing two D409Vgba knockin alleles without signs of Gaucher disease (residual GCase activity, >=20%), we recorded an age-dependent rise of endogenous Snca in hippocampal membranes (125% vs WT at 52 weeks; p = 0.019). In young Gaucher disease mice (V394Lgba+/+//prosaposin[ps]-null//ps-transgene), which demonstrate neurological dysfunction after age 10 weeks (GCase activity, <=10%), we recorded no significant change in endogenous Snca levels at 12 weeks of age. However, enhanced neuronal ubiquitin signals and axonal spheroid formation were already present. The latter changes were similar to those seen in three week-old cathepsin D-deficient mice. INTERPRETATION: Our results demonstrate that GBA mutants promote SNCA accumulation in a dose- and time-dependent manner, thereby identifying a biochemical link between GBA1 mutation carrier status and increased synucleinopathy risk. In cell culture models, this gain of toxic function effect can be mitigated by rapamycin. Loss in GCase activity did not immediately raise SNCA concentrations, but first led to neuronal ubiquitinopathy and axonal spheroids, a phenotype shared with other lysosomal storage disorders. ANN NEUROL 2011; [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
3. The engineered AAV2-HBKO promotes non-invasive gene delivery to large brain regions beyond ultrasound targeted sites.
- Author
-
Kofoed RH, Noseworthy K, Wu K, Sivadas S, Stanek L, Elmer B, Hynynen K, Shihabuddin LS, and Aubert I
- Abstract
Magnetic resonance imaging-guided focused ultrasound combined with microbubbles injected in the bloodstream (MRIgFUS) temporarily increases the permeability of the blood-brain barrier (BBB), which facilitates the entry of intravenously administered adeno-associated viruses (AAVs) from the blood to targeted brain areas. To date, the properties of the AAVs used for MRIgFUS delivery resulted in cell transduction limited to MRIgFUS-targeted sites. Considering future clinical applications, strategies are needed to deliver genes to multiple locations and large brain volumes while creating minimal BBB modulation. Here we combine MRIgFUS with a vector that has enhanced biodistribution following brain entry, AAV2-HBKO, to mediate broad gene delivery to targeted brain regions at levels with potential therapeutic relevance. Expression of a reporter gene was achieved in 13% and 21% of all neurons present in the striatum and thalamus, respectively, while targeting only 28% of the brain regions with MRIgFUS. Compared with AAV9, MRIgFUS-mediated delivery of AAV2-HBKO showed greater diffusion in the brain and a higher percentage of the neurons expressing the transgene. MRIgFUS AAV2-HBKO gene delivery to the brain has the potential to reach levels that are functionally and clinically relevant, and this even when using relatively low intravenous AAV dosages, compared with what is currently used in clinical trials., Competing Interests: L.S. and L.S.S. were paid employees of Sanofi when most of the work was done. B.E. is a paid employee of Sanofi., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
4. Glucosylceramide synthase inhibition reduces ganglioside GM3 accumulation, alleviates amyloid neuropathology, and stabilizes remote contextual memory in a mouse model of Alzheimer's disease.
- Author
-
Dodge JC, Tamsett TJ, Treleaven CM, Taksir TV, Piepenhagen P, Sardi SP, Cheng SH, and Shihabuddin LS
- Subjects
- Amyloid beta-Peptides, Animals, Disease Models, Animal, G(M3) Ganglioside, Glucosyltransferases, Memory, Long-Term, Mice, Mice, Transgenic, Plaque, Amyloid, Alzheimer Disease pathology
- Abstract
Background: Gangliosides are highly enriched in the brain and are critical for its normal development and function. However, in some rare neurometabolic diseases, a deficiency in lysosomal ganglioside hydrolysis is pathogenic and leads to early-onset neurodegeneration, neuroinflammation, demyelination, and dementia. Increasing evidence also suggests that more subtle ganglioside accumulation contributes to the pathogenesis of more common neurological disorders including Alzheimer's disease (AD). Notably, ganglioside GM3 levels are elevated in the brains of AD patients and in several mouse models of AD, and plasma GM3 levels positively correlate with disease severity in AD patients., Methods: Tg2576 AD model mice were fed chow formulated with a small molecule inhibitor of glucosylceramide synthase (GCSi) to determine whether reducing glycosphingolipid synthesis affected aberrant GM3 accumulation, amyloid burden, and disease manifestations in cognitive impairment. GM3 was measured with LC-MS, amyloid burden with ELISA and amyloid red staining, and memory was assessed using the contextual fear chamber test., Results: GCSi mitigated soluble Aβ42 accumulation in the brains of AD model mice when treatment was started prophylactically. Remarkably, GCSi treatment also reduced soluble Aβ42 levels and amyloid plaque burden in aged (i.e., 70 weeks old) AD mice with preexisting neuropathology. Our analysis of contextual memory in Tg2576 mice showed that impairments in remote (cortical-dependent) memory consolidation preceded deficits in short-term (hippocampal-dependent) contextual memory, which was consistent with soluble Aβ42 accumulation occurring more rapidly in the cortex of AD mice compared to the hippocampus. Notably, GCSi treatment significantly stabilized remote memory consolidation in AD mice-especially in mice with enhanced cognitive training. This finding was consistent with GCSi treatment lowering aberrant GM3 accumulation in the cortex of AD mice., Conclusions: Collectively, our results indicate that glycosphingolipids regulated by GCS are important modulators of Aβ neuropathology and that glycosphingolipid homeostasis plays a critical role in the consolidation of remote memories., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
5. Glucosylceramide in cerebrospinal fluid of patients with GBA-associated and idiopathic Parkinson's disease enrolled in PPMI.
- Author
-
Huh YE, Park H, Chiang MSR, Tuncali I, Liu G, Locascio JJ, Shirvan J, Hutten SJ, Rotunno MS, Viel C, Shihabuddin LS, Wang B, Sardi SP, and Scherzer CR
- Abstract
Protein-coding variants in the GBA gene modulate susceptibility and progression in ~10% of patients with Parkinson's disease (PD). GBA encodes the β-glucocerebrosidase enzyme that hydrolyzes glucosylceramide. We hypothesized that GBA mutations will lead to glucosylceramide accumulation in cerebrospinal fluid (CSF). Glucosylceramide, ceramide, sphingomyelin, and lactosylceramide levels were measured by liquid chromatography-tandem mass spectrometry in CSF of 411 participants from the Parkinson's Progression Markers Initiative (PPMI) cohort, including early stage, de novo PD patients with abnormal dopamine transporter neuroimaging and healthy controls. Forty-four PD patients carried protein-coding GBA variants (GBA-PD) and 227 carried wild-type alleles (idiopathic PD). The glucosylceramide fraction was increased (P = 0.0001), and the sphingomyelin fraction (a downstream metabolite) was reduced (P = 0.0001) in CSF of GBA-PD patients compared to healthy controls. The ceramide fraction was unchanged, and lactosylceramide was below detection limits. We then used the ratio of glucosylceramide to sphingomyelin (the GlcCer/SM ratio) to explore whether these two sphingolipid fractions altered in GBA-PD were useful for stratifying idiopathic PD patients. Idiopathic PD patients in the top quartile of GlcCer/SM ratios at baseline showed a more rapid decline in Montreal Cognitive Assessment scores during longitudinal follow-up compared to those in the lowest quartile with a P-value of 0.036. The GlcCer/SM ratio was negatively associated with α-synuclein levels in CSF of PD patients. This study highlights glucosylceramide as a pathway biomarker for GBA-PD patients and the GlcCer/SM ratio as a potential stratification tool for clinical trials of idiopathic PD patients. Our sphingolipids data together with the clinical, imaging, omics, and genetic characterization of PPMI will contribute a useful resource for multi-modal biomarkers development., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
6. Preclinical pharmacology of glucosylceramide synthase inhibitor venglustat in a GBA-related synucleinopathy model.
- Author
-
Viel C, Clarke J, Kayatekin C, Richards AM, Chiang MSR, Park H, Wang B, Shihabuddin LS, and Sardi SP
- Subjects
- Animals, Disease Models, Animal, Hippocampus drug effects, Hippocampus metabolism, Mice, Mice, Inbred C57BL, Mutation genetics, Parkinson Disease metabolism, Carbamates pharmacology, Glucosylceramidase metabolism, Glucosylceramides metabolism, Glucosyltransferases antagonists & inhibitors, Quinuclidines pharmacology, Synucleinopathies drug therapy, Synucleinopathies metabolism
- Abstract
Mutations in GBA, the gene encoding the lysosomal enzyme glucocerebrosidase (GCase), represent the greatest genetic risk factor for developing synucleinopathies including Parkinson's disease (PD). Additionally, PD patients harboring a mutant GBA allele present with an earlier disease onset and an accelerated disease progression of both motor and non-motor symptoms. Preclinical studies in mouse models of synucleinopathy suggest that modulation of the sphingolipid metabolism pathway via inhibition of glucosylceramide synthase (GCS) using a CNS-penetrant small molecule may be a potential treatment for synucleinopathies. Here, we aim to alleviate the lipid storage burden by inhibiting the de novo synthesis of the primary glycosphingolipid substrate of GCase, glucosylceramide (GlcCer). We have previously shown that systemic GCS inhibition reduced GlcCer and glucosylsphingosine (GlcSph) accumulation, slowed α-synuclein buildup in the hippocampus, and improved cognitive deficits. Here, we studied the efficacy of a brain-penetrant clinical candidate GCS inhibitor, venglustat, in mouse models of GBA-related synucleinopathy, including a heterozygous Gba mouse model which more closely replicates the typical GBA-PD patient genotype. Collectively, these data support the rationale for modulation of GCase-related sphingolipid metabolism as a therapeutic strategy for treating GBA-related synucleinopathies., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
7. Viral alpha-synuclein knockdown prevents spreading synucleinopathy.
- Author
-
Menon S, Kofoed RH, Nabbouh F, Xhima K, Al-Fahoum Y, Langman T, Mount HTJ, Shihabuddin LS, Sardi SP, Fraser PE, Watts JC, Aubert I, and Tandon A
- Abstract
The accumulation of aggregated alpha-synuclein (α-syn) in Parkinson's disease, dementia with Lewy bodies and multiple system atrophy is thought to involve a common prion-like mechanism, whereby misfolded α-syn provides a conformational template for further accumulation of pathological α-syn. We tested whether silencing α-syn gene expression could reduce native non-aggregated α-syn substrate and thereby disrupt the propagation of pathological α-syn initiated by seeding with synucleinopathy-affected mouse brain homogenates. Unilateral intracerebral injections of adeno-associated virus serotype-1 encoding microRNA targeting the α-syn gene reduced the extent and severity of both the α-syn pathology and motor deficits. Importantly, a moderate 50% reduction in α-syn was sufficient to prevent the spread of α-syn pathology to distal brain regions. Our study combines behavioural, immunohistochemical and biochemical data that strongly support α-syn knockdown gene therapy for synucleinopathies., (© The Author(s) (2021). Published by Oxford University Press on behalf of the Guarantors of Brain.)
- Published
- 2021
- Full Text
- View/download PDF
8. Gain of toxic function by long-term AAV9-mediated SMN overexpression in the sensorimotor circuit.
- Author
-
Van Alstyne M, Tattoli I, Delestrée N, Recinos Y, Workman E, Shihabuddin LS, Zhang C, Mentis GZ, and Pellizzoni L
- Subjects
- Animals, Dependovirus, Ganglia, Spinal metabolism, Ganglia, Spinal pathology, Gene Transfer Techniques, Genetic Therapy adverse effects, Genetic Vectors, Injections, Intraventricular, Mice, Motor Disorders genetics, Motor Disorders metabolism, Motor Disorders pathology, Survival of Motor Neuron 1 Protein genetics, Motor Neurons metabolism, Motor Neurons pathology, Nerve Degeneration genetics, Nerve Degeneration metabolism, Nerve Degeneration pathology, Survival of Motor Neuron 1 Protein toxicity
- Abstract
The neurodegenerative disease spinal muscular atrophy (SMA) is caused by deficiency in the survival motor neuron (SMN) protein. Currently approved SMA treatments aim to restore SMN, but the potential for SMN expression beyond physiological levels is a unique feature of adeno-associated virus serotype 9 (AAV9)-SMN gene therapy. Here, we show that long-term AAV9-mediated SMN overexpression in mouse models induces dose-dependent, late-onset motor dysfunction associated with loss of proprioceptive synapses and neurodegeneration. Mechanistically, aggregation of overexpressed SMN in the cytoplasm of motor circuit neurons sequesters components of small nuclear ribonucleoproteins, leading to splicing dysregulation and widespread transcriptome abnormalities with prominent signatures of neuroinflammation and the innate immune response. Thus, long-term SMN overexpression interferes with RNA regulation and triggers SMA-like pathogenic events through toxic gain-of-function mechanisms. These unanticipated, SMN-dependent and neuron-specific liabilities warrant caution on the long-term safety of treating individuals with SMA with AAV9-SMN and the risks of uncontrolled protein expression by gene therapy.
- Published
- 2021
- Full Text
- View/download PDF
9. Sterol auto-oxidation adversely affects human motor neuron viability and is a neuropathological feature of amyotrophic lateral sclerosis.
- Author
-
Dodge JC, Yu J, Sardi SP, and Shihabuddin LS
- Subjects
- Amyotrophic Lateral Sclerosis metabolism, Animals, Cell Death physiology, Cells, Cultured, Disease Models, Animal, Feces chemistry, Humans, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells pathology, Mice, Mice, Transgenic, Motor Neurons metabolism, Nervous System Diseases metabolism, Spinal Cord metabolism, Sterols metabolism, Superoxide Dismutase-1 genetics, Amyotrophic Lateral Sclerosis pathology, Motor Neurons pathology, Nervous System Diseases pathology, Spinal Cord pathology, Sterols chemistry, Superoxide Dismutase-1 metabolism
- Abstract
Aberrant cholesterol homeostasis is implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS), a fatal neuromuscular disease that is due to motor neuron (MN) death. Cellular toxicity from excess cholesterol is averted when it is enzymatically oxidized to oxysterols and bile acids (BAs) to promote its removal. In contrast, the auto oxidation of excess cholesterol is often detrimental to cellular survival. Although oxidized metabolites of cholesterol are altered in the blood and CSF of ALS patients, it is unknown if increased cholesterol oxidation occurs in the SC during ALS, and if exposure to oxidized cholesterol metabolites affects human MN viability. Here, we show that in the SOD1
G93A mouse model of ALS that several oxysterols, BAs and auto oxidized sterols are increased in the lumbar SC, plasma, and feces during disease. Similar changes in cholesterol oxidation were found in the cervical SC of sporadic ALS patients. Notably, auto-oxidized sterols, but not oxysterols and BAs, were toxic to iPSC derived human MNs. Thus, increased cholesterol oxidation is a manifestation of ALS and non-regulated sterol oxidation likely contributes to MN death. Developing therapeutic approaches to restore cholesterol homeostasis in the SC may lead to a treatment for ALS.- Published
- 2021
- Full Text
- View/download PDF
10. Neutral Lipid Cacostasis Contributes to Disease Pathogenesis in Amyotrophic Lateral Sclerosis.
- Author
-
Dodge JC, Jensen EH, Yu J, Sardi SP, Bialas AR, Taksir TV, Bangari DS, and Shihabuddin LS
- Subjects
- Amyotrophic Lateral Sclerosis metabolism, Animals, Cell Death, Cholesterol Esters metabolism, Gray Matter metabolism, Humans, Lysophosphatidylcholines metabolism, Male, Mice, Mice, Transgenic, Motor Neurons pathology, Receptors, G-Protein-Coupled genetics, Receptors, Phospholipase A2 metabolism, Spinal Cord metabolism, Superoxide Dismutase-1 genetics, Triglycerides metabolism, Amyotrophic Lateral Sclerosis pathology, Lipid Metabolism
- Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disease characterized by motor neuron (MN) death. Lipid dysregulation manifests during disease; however, it is unclear whether lipid homeostasis is adversely affected in the in the spinal cord gray matter (GM), and if so, whether it is because of an aberrant increase in lipid synthesis. Moreover, it is unknown whether lipid dysregulation contributes to MN death. Here, we show that cholesterol ester (CE) and triacylglycerol levels are elevated several-fold in the spinal cord GM of male sporadic ALS patients. Interestingly, HMG-CoA reductase, the rate-limiting enzyme in cholesterol synthesis, was reduced in the spinal cord GM of ALS patients. Increased cytosolic phospholipase A2 activity and lyso-phosphatidylcholine (Lyso-PC) levels in ALS patients suggest that CE accumulation was driven by acyl group transfer from PC to cholesterol. Notably, Lyso-PC, a byproduct of CE synthesis, was toxic to human MNs in vitro Elevations in CE, triacylglycerol, and Lyso-PC were also found in the spinal cord of SOD1
G93A mice, a model of ALS. Similar to ALS patients, a compensatory downregulation of cholesterol synthesis occurred in the spinal cord of SOD1G93A mice; levels of sterol regulatory element binding protein 2, a transcriptional regulator of cholesterol synthesis, progressively declined. Remarkably, overexpressing sterol regulatory element binding protein 2 in the spinal cord of normal mice to model CE accumulation led to ALS-like lipid pathology, MN death, astrogliosis, paralysis, and reduced survival. Thus, spinal cord lipid dysregulation in ALS likely contributes to neurodegeneration and developing therapies to restore lipid homeostasis may lead to a treatment for ALS. SIGNIFICANCE STATEMENT Neurons that control muscular function progressively degenerate in patients with amyotrophic lateral sclerosis (ALS). Lipid dysregulation is a feature of ALS; however, it is unclear whether disrupted lipid homeostasis (i.e., lipid cacostasis) occurs proximal to degenerating neurons in the spinal cord, what causes it, and whether it contributes to neurodegeneration. Here we show that lipid cacostasis occurs in the spinal cord gray matter of ALS patients. Lipid accumulation was not associated with an aberrant increase in synthesis or reduced hydrolysis, as enzymatic and transcriptional regulators of lipid synthesis were downregulated during disease. Last, we demonstrated that genetic induction of lipid cacostasis in the CNS of normal mice was associated with ALS-like lipid pathology, astrogliosis, neurodegeneration, and clinical features of ALS., (Copyright © 2020 the authors.)- Published
- 2020
- Full Text
- View/download PDF
11. Cerebrospinal fluid proteomics implicates the granin family in Parkinson's disease.
- Author
-
Rotunno MS, Lane M, Zhang W, Wolf P, Oliva P, Viel C, Wills AM, Alcalay RN, Scherzer CR, Shihabuddin LS, Zhang K, and Sardi SP
- Subjects
- Aged, Biomarkers cerebrospinal fluid, Female, Humans, Male, Middle Aged, Proteomics, Tandem Mass Spectrometry, Chromogranins cerebrospinal fluid, Parkinson Disease cerebrospinal fluid
- Abstract
Parkinson's disease, the most common age-related movement disorder, is a progressive neurodegenerative disease with unclear etiology. Better understanding of the underlying disease mechanism(s) is an urgent need for the development of disease-modifying therapeutics. Limited studies have been performed in large patient cohorts to identify protein alterations in cerebrospinal fluid (CSF), a proximal site to pathology. We set out to identify disease-relevant protein changes in CSF to gain insights into the etiology of Parkinson's disease and potentially assist in disease biomarker identification. In this study, we used liquid chromatography-tandem mass spectrometry in data-independent acquisition (DIA) mode to identify Parkinson's-relevant biomarkers in cerebrospinal fluid. We quantified 341 protein groups in two independent cohorts (n = 196) and a longitudinal cohort (n = 105 samples, representing 40 patients) consisting of Parkinson's disease and healthy control samples from three different sources. A first cohort of 53 Parkinson's disease and 72 control samples was analyzed, identifying 53 proteins with significant changes (p < 0.05) in Parkinson's disease relative to healthy control. We established a biomarker signature and multiple protein ratios that differentiate Parkinson's disease from healthy controls and validated these results in an independent cohort. The second cohort included 28 Parkinson's disease and 43 control samples. Independent analysis of these samples identified 41 proteins with significant changes. Evaluation of the overlapping changes between the two cohorts identified 13 proteins with consistent and significant changes (p < 0.05). Importantly, we found the extended granin family proteins as reduced in disease, suggesting a potential common mechanism for the biological reduction in monoamine neurotransmission in Parkinson's patients. Our study identifies several novel protein changes in Parkinson's disease cerebrospinal fluid that may be exploited for understanding etiology of disease and for biomarker development.
- Published
- 2020
- Full Text
- View/download PDF
12. Viral delivery of a microRNA to Gba to the mouse central nervous system models neuronopathic Gaucher disease.
- Author
-
Jackson KL, Viel C, Clarke J, Bu J, Chan M, Wang B, Shihabuddin LS, and Sardi SP
- Subjects
- Animals, Dependovirus, Disease Models, Animal, Fibroblasts metabolism, Gait physiology, Gaucher Disease metabolism, Genetic Vectors, Glucosylceramidase metabolism, Mice, MicroRNAs metabolism, NIH 3T3 Cells, Brain metabolism, Gaucher Disease genetics, Glucosylceramidase genetics, MicroRNAs genetics, Motor Activity physiology, Spinal Cord metabolism
- Abstract
Pathological mutations in GBA, encoding lysosomal glucocerebrosidase (GCase), cause Gaucher disease (GD). GD is a multi-system disease with great phenotypic variation between individuals. It has been classified into type 1 with primarily peripheral involvement and types 2 and 3 with varying degrees of neurological involvement. GD is characterized by decreased GCase activity and subsequent accumulation of its lipid substrates, glucosylceramide and glucosylsphingosine. Current murine models of neuronopathic GD mostly replicate the severe aspects of the neurological symptoms developing rapid progression and early lethality, thus presenting a short window for therapeutic testing. In order to develop a model of chronic neuronopathic GD, we reduced GCase in the central nervous system (CNS) of a mild GD mouse model (Gba
D409V/D409V ) via intracerebroventricular administration of an adeno-associated virus encoding a microRNA to Gba (AAV-GFP-miR-Gba). GbaD409V/D409V mice have significantly reduced GCase activity and increased substrate accumulation in the CNS. Phenotypically, these mice partially recapitulate features of mild type 1 GD. Their neurological examination reveals cognitive impairment with normal motor features. Administration of AAV-GFP-miR-Gba into GbaD409V/D409V pups in the CNS caused progressive lipid substrate accumulation. Phenotypically, AAV1-GFP-miR-Gba-treated mice were indistinguishable from their littermates until 10 weeks of age, when they started developing progressive neurological impairments, including hyperactivity, abnormal gait, and head retroflexion. Importantly, these impairments can be prevented by simultaneous administration of a miR-resistant GBA, demonstrating that the pathological effects are specifically due to Gba mRNA reduction. This novel model of neuronopathic GD offers several advantages over current models including slower progression of neurological complications and an increased lifespan, which make it more amenable for therapeutic testing., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
13. Astrocyte transduction is required for rescue of behavioral phenotypes in the YAC128 mouse model with AAV-RNAi mediated HTT lowering therapeutics.
- Author
-
Stanek LM, Bu J, and Shihabuddin LS
- Subjects
- Animals, Astrocytes pathology, Brain metabolism, Brain pathology, Dependovirus, Disease Models, Animal, Genetic Vectors, Huntingtin Protein genetics, Huntington Disease pathology, Mice, Mice, Transgenic, MicroRNAs, Neurons metabolism, Neurons pathology, Phenotype, Transduction, Genetic, Astrocytes metabolism, Huntingtin Protein antagonists & inhibitors, Huntington Disease metabolism
- Abstract
Huntington's disease (HD) is a fatal autosomal dominant neurodegenerative disease caused by a CAG expansion, which translates into an elongated polyglutamine (polyQ) repeat near the amino-terminus of the huntingtin protein (HTT). This results in production of a toxic mutant huntingtin protein (mHTT) that leads to neuronal dysfunction and death. Currently, no disease-modifying treatments are available; however, numerous therapeutic strategies aimed at lowering HTT levels in the brain are under development. To date, studies have not closely examined the contribution of mHTT in neurons vs astrocytes to disease pathophysiology. To better understand the role of astrocytes in HD pathophysiology and the need for cell type specific targeting of HTT lowering therapeutic strategies, AAV capsids were employed that selectively transduce neurons, or both neurons and astrocytes. These vectors carrying miRNA sequences directed against HTT were injected into the YAC128 mouse model of HD to selectively lower HTT expression in neurons alone versus neurons and astrocytes. The results suggested that HTT lowering in neurons alone was not sufficient to rescue the motor phenotype in YAC128 mice. Furthermore, HTT lowering in both cell types was required to achieve maximal functional benefit. The study suggested that astrocyte dysfunction may play a critical role in HD pathogenesis, and thus astrocytes represent an important therapeutic target., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
14. New Frontiers in Parkinson's Disease: From Genetics to the Clinic.
- Author
-
Shihabuddin LS, Brundin P, Greenamyre JT, Stephenson D, and Sardi SP
- Subjects
- Animals, Clinical Trials as Topic methods, Drug Delivery Systems methods, Humans, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 genetics, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 metabolism, Mutation drug effects, Parkinson Disease metabolism, alpha-Synuclein genetics, alpha-Synuclein metabolism, Antiparkinson Agents administration & dosage, Drug Delivery Systems trends, Mutation physiology, Parkinson Disease drug therapy, Parkinson Disease genetics
- Abstract
The greatest unmet therapeutic need in Parkinson's disease (PD) is a treatment that slows the relentless progression of the symptoms and the neurodegenerative process. This review highlights the utility of genetics to understand the pathogenic mechanisms and develop novel therapeutic approaches for PD. The focus is on strategies provided by genetic studies: notably via the reduction and clearance of α-synuclein, inhibition of LRRK2 kinase activity, and modulation of glucocerebrosidase-related substrates. In addition, the critical role of precompetitive public-private partnerships in supporting trial design optimization, overall drug development, and regulatory approvals is illustrated. With these great advances, the promise of developing transformative therapies that halt or slow disease progression is a tangible goal., (Copyright © 2018 the authors 0270-6474/18/389375-08$15.00/0.)
- Published
- 2018
- Full Text
- View/download PDF
15. Extensive Transduction and Enhanced Spread of a Modified AAV2 Capsid in the Non-human Primate CNS.
- Author
-
Naidoo J, Stanek LM, Ohno K, Trewman S, Samaranch L, Hadaczek P, O'Riordan C, Sullivan J, San Sebastian W, Bringas JR, Snieckus C, Mahmoodi A, Mahmoodi A, Forsayeth J, Bankiewicz KS, and Shihabuddin LS
- Subjects
- Animals, Axonal Transport drug effects, Brain drug effects, Brain pathology, Capsid Proteins administration & dosage, Capsid Proteins genetics, Central Nervous System drug effects, Central Nervous System pathology, Dependovirus, Disease Models, Animal, Genetic Vectors administration & dosage, Genetic Vectors genetics, Heparan Sulfate Proteoglycans administration & dosage, Heparan Sulfate Proteoglycans genetics, Humans, Infusions, Intraventricular, Motor Neurons drug effects, Neurons pathology, Primates, Spinal Cord pathology, Thalamus drug effects, Genetic Therapy, Genetic Vectors adverse effects, Neurons drug effects, Parvovirinae genetics, Spinal Cord drug effects
- Abstract
The present study was designed to characterize transduction of non-human primate brain and spinal cord with a modified adeno-associated virus serotype 2, incapable of binding to the heparan sulfate proteoglycan receptor, referred to as AAV2-HBKO. AAV2-HBKO was infused into the thalamus, intracerebroventricularly or via a combination of both intracerebroventricular and thalamic delivery. Thalamic injection of this modified vector encoding GFP resulted in widespread CNS transduction that included neurons in deep cortical layers, deep cerebellar nuclei, several subcortical regions, and motor neuron transduction in the spinal cord indicative of robust bidirectional axonal transport. Intracerebroventricular delivery similarly resulted in widespread cortical transduction, with one striking distinction that oligodendrocytes within superficial layers of the cortex were the primary cell type transduced. Robust motor neuron transduction was also observed in all levels of the spinal cord. The combination of thalamic and intracerebroventricular delivery resulted in transduction of oligodendrocytes in superficial cortical layers and neurons in deeper cortical layers. Several subcortical regions were also transduced. Our data demonstrate that AAV2-HBKO is a powerful vector for the potential treatment of a wide number of neurological disorders, and highlight that delivery route can significantly impact cellular tropism and pattern of CNS transduction., (Published by Elsevier Inc.)
- Published
- 2018
- Full Text
- View/download PDF
16. Dysregulation of Mdm2 and Mdm4 alternative splicing underlies motor neuron death in spinal muscular atrophy.
- Author
-
Van Alstyne M, Simon CM, Sardi SP, Shihabuddin LS, Mentis GZ, and Pellizzoni L
- Subjects
- Animals, Cell Death, Exons, Mice, Motor Neurons pathology, Muscular Atrophy, Spinal metabolism, Muscular Atrophy, Spinal physiopathology, NIH 3T3 Cells, Nerve Degeneration metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-mdm2 metabolism, Ribonucleoproteins, Small Nuclear biosynthesis, Tumor Suppressor Protein p53 metabolism, Alternative Splicing, Motor Neurons metabolism, Muscular Atrophy, Spinal genetics, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-mdm2 genetics
- Abstract
Ubiquitous deficiency in the survival motor neuron (SMN) protein causes death of motor neurons-a hallmark of the neurodegenerative disease spinal muscular atrophy (SMA)-through poorly understood mechanisms. Here, we show that the function of SMN in the assembly of spliceosomal small nuclear ribonucleoproteins (snRNPs) regulates alternative splicing of Mdm2 and Mdm4, two nonredundant repressors of p53. Decreased inclusion of critical Mdm2 and Mdm4 exons is most prominent in SMA motor neurons and correlates with both snRNP reduction and p53 activation in vivo. Importantly, increased skipping of Mdm2 and Mdm4 exons regulated by SMN is necessary and sufficient to synergistically elicit robust p53 activation in wild-type mice. Conversely, restoration of full-length Mdm2 and Mdm4 suppresses p53 induction and motor neuron degeneration in SMA mice. These findings reveal that loss of SMN-dependent regulation of Mdm2 and Mdm4 alternative splicing underlies p53-mediated death of motor neurons in SMA, establishing a causal link between snRNP dysfunction and neurodegeneration., (© 2018 Van Alstyne et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2018
- Full Text
- View/download PDF
17. Rationally designed AAV2 and AAVrh8R capsids provide improved transduction in the retina and brain.
- Author
-
Sullivan JA, Stanek LM, Lukason MJ, Bu J, Osmond SR, Barry EA, O'Riordan CR, Shihabuddin LS, Cheng SH, and Scaria A
- Subjects
- Animals, Brain metabolism, Capsid metabolism, Capsid Proteins genetics, Capsid Proteins metabolism, Dependovirus immunology, Gene Transfer Techniques, Genetic Vectors, HeLa Cells, Heparitin Sulfate, Humans, Mice, Mice, Inbred C57BL, Photoreceptor Cells metabolism, Retina metabolism, Transduction, Genetic methods, Genetic Therapy methods, Parvovirinae growth & development, Parvovirinae immunology
- Abstract
The successful application of adeno-associated virus (AAV) gene delivery vectors as a therapeutic paradigm will require efficient gene delivery to the appropriate cells in affected organs. In this study, we utilized a rational design approach to introduce modifications to the AAV2 and AAVrh8R capsids and the resulting variants were evaluated for transduction activity in the retina and brain. The modifications disrupted either capsid/receptor binding or altered capsid surface charge. Specifically, we mutated AAV2 amino acids R585A and R588A, which are required for binding to its receptor, heparan sulfate proteoglycans, to generate a variant referred to as AAV2-HBKO. In contrast to parental AAV2, the AAV2-HBKO vector displayed low-transduction activity following intravitreal delivery to the mouse eye; however, following its subretinal delivery, AAV2-HBKO resulted in significantly greater photoreceptor transduction. Intrastriatal delivery of AAV2-HBKO to mice facilitated widespread striatal and cortical expression, in contrast to the restricted transduction pattern of the parental AAV2 vector. Furthermore, we found that altering the surface charge on the AAVrh8R capsid by modifying the number of arginine residues on the capsid surface had a profound impact on subretinal transduction. The data further validate the potential of capsid engineering to improve AAV gene therapy vectors for clinical applications.
- Published
- 2018
- Full Text
- View/download PDF
18. Glucosylceramide synthase inhibition alleviates aberrations in synucleinopathy models.
- Author
-
Sardi SP, Viel C, Clarke J, Treleaven CM, Richards AM, Park H, Olszewski MA, Dodge JC, Marshall J, Makino E, Wang B, Sidman RL, Cheng SH, and Shihabuddin LS
- Subjects
- Animals, Disease Models, Animal, Gene Expression Regulation, Glucosyltransferases genetics, Humans, Mice, Mutation, Parkinson Disease enzymology, Parkinson Disease pathology, Protein Aggregation, Pathological drug therapy, Protein Aggregation, Pathological genetics, Protein Aggregation, Pathological pathology, Ubiquitin metabolism, tau Proteins metabolism, Carbamates pharmacology, Enzyme Inhibitors administration & dosage, Glucosyltransferases antagonists & inhibitors, Parkinson Disease drug therapy, Quinuclidines pharmacology, alpha-Synuclein genetics
- Abstract
Mutations in the glucocerebrosidase gene ( GBA ) confer a heightened risk of developing Parkinson's disease (PD) and other synucleinopathies, resulting in a lower age of onset and exacerbating disease progression. However, the precise mechanisms by which mutations in GBA increase PD risk and accelerate its progression remain unclear. Here, we investigated the merits of glucosylceramide synthase (GCS) inhibition as a potential treatment for synucleinopathies. Two murine models of synucleinopathy (a Gaucher-related synucleinopathy model, Gba
D409V/D409V and a A53T-α-synuclein overexpressing model harboring wild-type alleles of GBA , A53T-SNCA mouse model) were exposed to a brain-penetrant GCS inhibitor, GZ667161. Treatment of GbaD409V/D409V mice with the GCS inhibitor reduced levels of glucosylceramide and glucosylsphingosine in the central nervous system (CNS), demonstrating target engagement. Remarkably, treatment with GZ667161 slowed the accumulation of hippocampal aggregates of α-synuclein, ubiquitin, and tau, and improved the associated memory deficits. Similarly, prolonged treatment of A53T-SNCA mice with GZ667161 reduced membrane-associated α-synuclein in the CNS and ameliorated cognitive deficits. The data support the contention that prolonged antagonism of GCS in the CNS can affect α-synuclein processing and improve behavioral outcomes. Hence, inhibition of GCS represents a disease-modifying therapeutic strategy for GBA -related synucleinopathies and conceivably for certain forms of sporadic disease.- Published
- 2017
- Full Text
- View/download PDF
19. Glucocerebrosidase modulates cognitive and motor activities in murine models of Parkinson's disease.
- Author
-
Rockenstein E, Clarke J, Viel C, Panarello N, Treleaven CM, Kim C, Spencer B, Adame A, Park H, Dodge JC, Cheng SH, Shihabuddin LS, Masliah E, and Sardi SP
- Subjects
- Animals, Cognition drug effects, Disease Models, Animal, Dopamine, Gaucher Disease genetics, Gene Expression, Glucosylceramidase genetics, Glucosylceramidase therapeutic use, Humans, Mice, Motor Activity drug effects, Mutation, Parkinson Disease drug therapy, Parkinson Disease genetics, alpha-Synuclein cerebrospinal fluid, alpha-Synuclein metabolism, Glucosylceramidase metabolism, Glucosylceramidase physiology
- Abstract
Mutations in GBA1, the gene encoding glucocerebrosidase, are associated with an enhanced risk of developing synucleinopathies such as Parkinson's disease (PD) and dementia with Lewy bodies. A higher prevalence and increased severity of motor and non-motor symptoms is observed in PD patients harboring mutant GBA1 alleles, suggesting a link between the gene or gene product and disease development. Interestingly, PD patients without mutations in GBA1 also exhibit lower levels of glucocerebrosidase activity in the central nervous system (CNS), implicating this lysosomal enzyme in disease pathogenesis. Here, we investigated whether modulation of glucocerebrosidase activity in murine models of synucleinopathy (expressing wild type Gba1) affected α-synuclein accumulation and behavioral phenotypes. Partial inhibition of glucocerebrosidase activity in PrP-A53T-SNCA mice using the covalent inhibitor conduritol-B-epoxide induced a profound increase in soluble α-synuclein in the CNS and exacerbated cognitive and motor deficits. Conversely, augmenting glucocerebrosidase activity in the Thy1-SNCA mouse model of PD delayed the progression of synucleinopathy. Adeno-associated virus-mediated expression of glucocerebrosidase in the Thy1-SNCA mouse striatum led to decrease in the levels of the proteinase K-resistant fraction of α-synuclein, amelioration of behavioral aberrations and protection from loss of striatal dopaminergic markers. These data indicate that increasing glucocerebrosidase activity can influence α-synuclein homeostasis, thereby reducing the progression of synucleinopathies. This study provides robust in vivo evidence that augmentation of CNS glucocerebrosidase activity is a potential therapeutic strategy for PD, regardless of the mutation status of GBA1., (© The Author 2016. Published by Oxford University Press.)
- Published
- 2016
- Full Text
- View/download PDF
20. Widespread AAV1- and AAV2-mediated transgene expression in the nonhuman primate brain: implications for Huntington's disease.
- Author
-
Hadaczek P, Stanek L, Ciesielska A, Sudhakar V, Samaranch L, Pivirotto P, Bringas J, O'Riordan C, Mastis B, San Sebastian W, Forsayeth J, Cheng SH, Bankiewicz KS, and Shihabuddin LS
- Abstract
Huntington's disease (HD) is caused by a toxic gain-of-function associated with the expression of the mutant huntingtin (htt) protein. Therefore, the use of RNA interference to inhibit Htt expression could represent a disease-modifying therapy. The potential of two recombinant adeno-associated viral vectors (AAV), AAV1 and AAV2, to transduce the cortico-striatal tissues that are predominantly affected in HD was explored. Green fluorescent protein was used as a reporter in each vector to show that both serotypes were broadly distributed in medium spiny neurons in the striatum and cortico-striatal neurons after infusion into the putamen and caudate nucleus of nonhuman primates (NHP), with AAV1-directed expression being slightly more robust than AAV2-driven expression. This study suggests that both serotypes are capable of targeting neurons that degenerate in HD, and it sets the stage for the advanced preclinical evaluation of an RNAi-based therapy for this disease.
- Published
- 2016
- Full Text
- View/download PDF
21. No evidence for substrate accumulation in Parkinson brains with GBA mutations.
- Author
-
Gegg ME, Sweet L, Wang BH, Shihabuddin LS, Sardi SP, and Schapira AH
- Subjects
- Cerebellum pathology, Humans, Mutation, Putamen pathology, Cerebellum metabolism, Glucosylceramidase metabolism, Putamen metabolism, Tissue Banks, beta-Glucosidase genetics
- Abstract
Background: To establish whether Parkinson's disease (PD) brains previously described to have decreased glucocerebrosidase activity exhibit accumulation of the lysosomal enzyme's substrate, glucosylceramide, or other changes in lipid composition., Methods: Lipidomic analyses and cholesterol measurements were performed on the putamen (n = 5-7) and cerebellum (n = 7-14) of controls, Parkinson's disease brains with heterozygote GBA1 mutations (PD+GBA), or sporadic PD., Results: Total glucosylceramide levels were unchanged in both PD+GBA and sporadic PD brains when compared with controls. No changes in glucosylsphingosine (deacetylated glucosylceramide), sphingomyelin, gangliosides (GM2, GM3), or total cholesterol were observed in either putamen or cerebellum., Conclusions: This study did not demonstrate glucocerebrosidase substrate accumulation in PD brains with heterozygote GBA1 mutations in areas of the brain with low α-synuclein pathology., (© 2015 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.)
- Published
- 2015
- Full Text
- View/download PDF
22. Glycosphingolipids are modulators of disease pathogenesis in amyotrophic lateral sclerosis.
- Author
-
Dodge JC, Treleaven CM, Pacheco J, Cooper S, Bao C, Abraham M, Cromwell M, Sardi SP, Chuang WL, Sidman RL, Cheng SH, and Shihabuddin LS
- Subjects
- Amyotrophic Lateral Sclerosis enzymology, Animals, Disease Models, Animal, Disease Progression, G(M3) Ganglioside administration & dosage, Glucosyltransferases antagonists & inhibitors, Humans, Injections, Intraventricular, Male, Mice, Mice, Transgenic, Spinal Cord physiopathology, Superoxide Dismutase genetics, Superoxide Dismutase metabolism, Amyotrophic Lateral Sclerosis physiopathology, Glycosphingolipids physiology
- Abstract
Recent genetic evidence suggests that aberrant glycosphingolipid metabolism plays an important role in several neuromuscular diseases including hereditary spastic paraplegia, hereditary sensory neuropathy type 1, and non-5q spinal muscular atrophy. Here, we investigated whether altered glycosphingolipid metabolism is a modulator of disease course in amyotrophic lateral sclerosis (ALS). Levels of ceramide, glucosylceramide, galactocerebroside, lactosylceramide, globotriaosylceramide, and the gangliosides GM3 and GM1 were significantly elevated in spinal cords of ALS patients. Moreover, enzyme activities (glucocerebrosidase-1, glucocerebrosidase-2, hexosaminidase, galactosylceramidase, α-galactosidase, and β-galactosidase) mediating glycosphingolipid hydrolysis were also elevated up to threefold. Increased ceramide, glucosylceramide, GM3, and hexosaminidase activity were also found in SOD1(G93A) mice, a familial model of ALS. Inhibition of glucosylceramide synthesis accelerated disease course in SOD1(G93A) mice, whereas infusion of exogenous GM3 significantly slowed the onset of paralysis and increased survival. Our results suggest that glycosphingolipids are likely important participants in pathogenesis of ALS and merit further analysis as potential drug targets.
- Published
- 2015
- Full Text
- View/download PDF
23. Partial rescue of some features of Huntington Disease in the genetic absence of caspase-6 in YAC128 mice.
- Author
-
Wong BKY, Ehrnhoefer DE, Graham RK, Martin DDO, Ladha S, Uribe V, Stanek LM, Franciosi S, Qiu X, Deng Y, Kovalik V, Zhang W, Pouladi MA, Shihabuddin LS, and Hayden MR
- Subjects
- Animals, Body Weight, Brain-Derived Neurotrophic Factor metabolism, Caspase 6 genetics, Corpus Striatum metabolism, Depression metabolism, Disease Models, Animal, Insulin-Like Growth Factor I metabolism, Mice, Mice, Transgenic, Motor Activity, Caspase 6 metabolism, Huntington Disease enzymology, Serotonin Plasma Membrane Transport Proteins metabolism
- Abstract
Huntington Disease (HD) is a progressive neurodegenerative disease caused by an elongated CAG repeat in the huntingtin (HTT) gene that encodes a polyglutamine tract in the HTT protein. Proteolysis of the mutant HTT protein (mHTT) has been detected in human and murine HD brains and is implicated in the pathogenesis of HD. Of particular importance is the site at amino acid (aa) 586 that contains a caspase-6 (Casp6) recognition motif. Activation of Casp6 occurs presymptomatically in human HD patients and the inhibition of mHTT proteolysis at aa586 in the YAC128 mouse model results in the full rescue of HD-like phenotypes. Surprisingly, Casp6 ablation in two different HD mouse models did not completely prevent the generation of this fragment, and therapeutic benefits were limited, questioning the role of Casp6 in the disease. We have evaluated the impact of the loss of Casp6 in the YAC128 mouse model of HD. Levels of the mHTT-586 fragment are reduced but not absent in the absence of Casp6 and we identify caspase 8 as an alternate enzyme that can generate this fragment. In vivo, the ablation of Casp6 results in a partial rescue of body weight gain, normalized IGF-1 levels, a reversal of the depression-like phenotype and decreased HTT levels. In the YAC128/Casp6-/- striatum there is a concomitant reduction in p62 levels, a marker of autophagic activity, suggesting increased autophagic clearance. These results implicate the HTT-586 fragment as a key contributor to certain features of HD, irrespective of the enzyme involved in its generation., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
24. Gaucher-related synucleinopathies: the examination of sporadic neurodegeneration from a rare (disease) angle.
- Author
-
Sardi SP, Cheng SH, and Shihabuddin LS
- Subjects
- Gaucher Disease genetics, Glucosylceramidase genetics, Humans, Mutation genetics, Gaucher Disease complications, Gaucher Disease metabolism, Neurodegenerative Diseases etiology, alpha-Synuclein metabolism
- Abstract
Gaucher disease, the most common lysosomal storage disease, is caused by a recessively inherited deficiency in glucocerebrosidase and subsequent accumulation of toxic lipid substrates. Heterozygous mutations in the lysosomal glucocerebrosidase gene (GBA1) have recently been recognized as the highest genetic risk factor for the development of α-synuclein aggregation disorders ("synucleinopathies"), including Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Despite the wealth of experimental, clinical and genetic evidence that supports the association between mutant genotypes and synucleinopathy risk, the precise mechanisms by which GBA1 mutations lead to PD and DLB remain unclear. Decreased glucocerebrosidase activity has been demonstrated to promote α-synuclein misprocessing. Furthermore, aberrant α-synuclein species have been reported to downregulate glucocerebrosidase activity, which further contributes to disease progression. In this review, we summarize the recent findings that highlight the complexity of this pathogenetic link and how several pathways that connect glucocerebrosidase insufficiency with α-synuclein misprocessing have emerged as potential therapeutic targets. From a translational perspective, we discuss how various therapeutic approaches to lysosomal dysfunction have been explored for the treatment of GBA1-related synucleinopathies, and potentially, for non-GBA1-associated neurodegenerative diseases. In summary, the link between GBA1 and synucleinopathies has become the paradigm of how the study of a rare lysosomal disease can transform the understanding of the etiopathology, and hopefully the treatment, of a more prevalent and multifactorial disorder., (Copyright © 2015 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
25. Translational fidelity of intrathecal delivery of self-complementary AAV9-survival motor neuron 1 for spinal muscular atrophy.
- Author
-
Passini MA, Bu J, Richards AM, Treleaven CM, Sullivan JA, O'Riordan CR, Scaria A, Kells AP, Samaranch L, San Sebastian W, Federici T, Fiandaca MS, Boulis NM, Bankiewicz KS, Shihabuddin LS, and Cheng SH
- Subjects
- Animals, Genetic Vectors genetics, Genetic Vectors metabolism, Mice, Mice, Knockout, Motor Neurons metabolism, Motor Neurons pathology, Muscular Atrophy, Spinal genetics, Muscular Atrophy, Spinal metabolism, Muscular Atrophy, Spinal pathology, Spinal Cord metabolism, Spinal Cord pathology, Swine, Dependovirus, Genetic Vectors pharmacology, Injections, Spinal, Muscular Atrophy, Spinal therapy, Protein Biosynthesis, Survival of Motor Neuron 1 Protein biosynthesis, Survival of Motor Neuron 1 Protein genetics
- Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disease caused by mutations in survival motor neuron 1 (SMN1). Previously, we showed that central nervous system (CNS) delivery of an adeno-associated viral (AAV) vector encoding SMN1 produced significant improvements in survival in a mouse model of SMA. Here, we performed a dose-response study in SMA mice to determine the levels of SMN in the spinal cord necessary for efficacy, and measured the efficiency of motor neuron transduction in the spinal cord after intrathecal delivery in pigs and nonhuman primates (NHPs). CNS injections of 5e10, 1e10, and 1e9 genome copies (gc) of self-complementary AAV9 (scAAV9)-hSMN1 into SMA mice extended their survival from 17 to 153, 70, and 18 days, respectively. Spinal cords treated with 5e10, 1e10, and 1e9 gc showed that 70-170%, 30-100%, and 10-20% of wild-type levels of SMN were attained, respectively. Furthermore, detectable SMN expression in a minimum of 30% motor neurons correlated with efficacy. A comprehensive analysis showed that intrathecal delivery of 2.5e13 gc of scAAV9-GFP transduced 25-75% of the spinal cord motor neurons in NHPs. Thus, the extent of gene expression in motor neurons necessary to confer efficacy in SMA mice could be obtained in large-animal models, justifying the continual development of gene therapy for SMA.
- Published
- 2014
- Full Text
- View/download PDF
26. iPSC-derived neurons from GBA1-associated Parkinson's disease patients show autophagic defects and impaired calcium homeostasis.
- Author
-
Schöndorf DC, Aureli M, McAllister FE, Hindley CJ, Mayer F, Schmid B, Sardi SP, Valsecchi M, Hoffmann S, Schwarz LK, Hedrich U, Berg D, Shihabuddin LS, Hu J, Pruszak J, Gygi SP, Sonnino S, Gasser T, and Deleidi M
- Subjects
- Cell Differentiation, Glycoside Hydrolases metabolism, Humans, Induced Pluripotent Stem Cells enzymology, Neurons enzymology, Parkinson Disease immunology, Parkinson Disease metabolism, Autophagy, Calcium metabolism, Glucosylceramidase genetics, Homeostasis, Induced Pluripotent Stem Cells pathology, Neurons pathology, Parkinson Disease pathology
- Abstract
Mutations in the acid β-glucocerebrosidase (GBA1) gene, responsible for the lysosomal storage disorder Gaucher's disease (GD), are the strongest genetic risk factor for Parkinson's disease (PD) known to date. Here we generate induced pluripotent stem cells from subjects with GD and PD harbouring GBA1 mutations, and differentiate them into midbrain dopaminergic neurons followed by enrichment using fluorescence-activated cell sorting. Neurons show a reduction in glucocerebrosidase activity and protein levels, increase in glucosylceramide and α-synuclein levels as well as autophagic and lysosomal defects. Quantitative proteomic profiling reveals an increase of the neuronal calcium-binding protein 2 (NECAB2) in diseased neurons. Mutant neurons show a dysregulation of calcium homeostasis and increased vulnerability to stress responses involving elevation of cytosolic calcium. Importantly, correction of the mutations rescues such pathological phenotypes. These findings provide evidence for a link between GBA1 mutations and complex changes in the autophagic/lysosomal system and intracellular calcium homeostasis, which underlie vulnerability to neurodegeneration.
- Published
- 2014
- Full Text
- View/download PDF
27. Silencing mutant huntingtin by adeno-associated virus-mediated RNA interference ameliorates disease manifestations in the YAC128 mouse model of Huntington's disease.
- Author
-
Stanek LM, Sardi SP, Mastis B, Richards AR, Treleaven CM, Taksir T, Misra K, Cheng SH, and Shihabuddin LS
- Subjects
- Animals, Behavior, Animal, Disease Models, Animal, HEK293 Cells, Humans, Huntingtin Protein, Mice, Mice, Transgenic, MicroRNAs metabolism, Neostriatum metabolism, Neostriatum pathology, RNA, Messenger genetics, RNA, Messenger metabolism, Transduction, Genetic, Dependovirus metabolism, Huntington Disease genetics, Huntington Disease pathology, Mutant Proteins genetics, Nerve Tissue Proteins genetics, RNA Interference
- Abstract
Huntington's disease (HD) is a fatal autosomal dominant neurodegenerative disease caused by an increase in the number of polyglutamine residues in the huntingtin (Htt) protein. With the identification of the underlying basis of HD, therapies are being developed that reduce expression of the causative mutant Htt. RNA interference (RNAi) that seeks to selectively reduce the expression of such disease-causing agents is emerging as a potential therapeutic strategy for this and similar disorders. This study examines the merits of administering a recombinant adeno-associated viral (AAV) vector designed to deliver small interfering RNA (siRNA) that targets the degradation of the Htt transcript. The aim was to lower Htt levels and to correct the behavioral, biochemical, and neuropathological deficits shown to be associated with the YAC128 mouse model of HD. Our data demonstrate that AAV-mediated RNAi is effective at transducing greater than 80% of the cells in the striatum and partially reducing the levels (~40%) of both wild-type and mutant Htt in this region. Concomitant with these reductions are significant improvements in behavioral deficits, reduction of striatal Htt aggregates, and partial correction of the aberrant striatal transcriptional profile observed in YAC128 mice. Importantly, a partial reduction of both the mutant and wild-type Htt levels is not associated with any notable overt neurotoxicity. Collectively, these results support the continued development of AAV-mediated RNAi as a therapeutic strategy for HD.
- Published
- 2014
- Full Text
- View/download PDF
28. Metabolic signatures of amyotrophic lateral sclerosis reveal insights into disease pathogenesis.
- Author
-
Dodge JC, Treleaven CM, Fidler JA, Tamsett TJ, Bao C, Searles M, Taksir TV, Misra K, Sidman RL, Cheng SH, and Shihabuddin LS
- Subjects
- Acidosis etiology, Acidosis genetics, Acidosis metabolism, Amyotrophic Lateral Sclerosis etiology, Amyotrophic Lateral Sclerosis genetics, Animals, Disease Models, Animal, Disease Progression, Glycogen metabolism, Humans, Mice, Mice, Transgenic, Mutation, Superoxide Dismutase genetics, Superoxide Dismutase metabolism, Amyotrophic Lateral Sclerosis metabolism
- Abstract
Metabolic dysfunction is an important modulator of disease course in amyotrophic lateral sclerosis (ALS). We report here that a familial mouse model (transgenic mice over-expressing the G93A mutation of the Cu/Zn superoxide dismutase 1 gene) of ALS enters a progressive state of acidosis that is associated with several metabolic (hormonal) alternations that favor lipolysis. Extensive investigation of the major determinants of H(+) concentration (i.e., the strong ion difference and the strong ion gap) suggests that acidosis is also due in part to the presence of an unknown anion. Consistent with a compensatory response to avert pathological acidosis, ALS mice harbor increased accumulation of glycogen in CNS and visceral tissues. The altered glycogen is associated with fluctuations in lysosomal and neutral α-glucosidase activities. Disease-related changes in glycogen, glucose, and α-glucosidase activity are also found in spinal cord tissue samples of autopsied patients with ALS. Collectively, these data provide insights into the pathogenesis of ALS as well as potential targets for drug development.
- Published
- 2013
- Full Text
- View/download PDF
29. Augmenting CNS glucocerebrosidase activity as a therapeutic strategy for parkinsonism and other Gaucher-related synucleinopathies.
- Author
-
Sardi SP, Clarke J, Viel C, Chan M, Tamsett TJ, Treleaven CM, Bu J, Sweet L, Passini MA, Dodge JC, Yu WH, Sidman RL, Cheng SH, and Shihabuddin LS
- Subjects
- Animals, Brain pathology, Brain physiopathology, Dependovirus metabolism, Disease Models, Animal, Gaucher Disease pathology, Gaucher Disease physiopathology, Glucosylceramidase administration & dosage, Glucosylceramidase genetics, Glucosylceramidase therapeutic use, Hippocampus metabolism, Hippocampus pathology, Hippocampus physiopathology, Humans, Memory, Mice, Mice, Transgenic, Parkinsonian Disorders physiopathology, Protein Structure, Quaternary, Psychosine analogs & derivatives, Psychosine metabolism, alpha-Synuclein genetics, tau Proteins chemistry, tau Proteins metabolism, Brain enzymology, Gaucher Disease drug therapy, Gaucher Disease enzymology, Glucosylceramidase metabolism, Parkinsonian Disorders drug therapy, Parkinsonian Disorders enzymology, alpha-Synuclein metabolism
- Abstract
Mutations of GBA1, the gene encoding glucocerebrosidase, represent a common genetic risk factor for developing the synucleinopathies Parkinson disease (PD) and dementia with Lewy bodies. PD patients with or without GBA1 mutations also exhibit lower enzymatic levels of glucocerebrosidase in the central nervous system (CNS), suggesting a possible link between the enzyme and the development of the disease. Previously, we have shown that early treatment with glucocerebrosidase can modulate α-synuclein aggregation in a presymptomatic mouse model of Gaucher-related synucleinopathy (Gba1(D409V/D409V)) and ameliorate the associated cognitive deficit. To probe this link further, we have now evaluated the efficacy of augmenting glucocerebrosidase activity in the CNS of symptomatic Gba1(D409V/D409V) mice and in a transgenic mouse model overexpressing A53T α-synuclein. Adeno-associated virus-mediated expression of glucocerebrosidase in the CNS of symptomatic Gba1(D409V/D409V) mice completely corrected the aberrant accumulation of the toxic lipid glucosylsphingosine and reduced the levels of ubiquitin, tau, and proteinase K-resistant α-synuclein aggregates. Importantly, hippocampal expression of glucocerebrosidase in Gba1(D409V/D409V) mice (starting at 4 or 12 mo of age) also reversed their cognitive impairment when examined using a novel object recognition test. Correspondingly, overexpression of glucocerebrosidase in the CNS of A53T α-synuclein mice reduced the levels of soluble α-synuclein, suggesting that increasing the glycosidase activity can modulate α-synuclein processing and may modulate the progression of α-synucleinopathies. Hence, increasing glucocerebrosidase activity in the CNS represents a potential therapeutic strategy for GBA1-related and non-GBA1-associated synucleinopathies, including PD.
- Published
- 2013
- Full Text
- View/download PDF
30. Antisense oligonucleotide-mediated correction of transcriptional dysregulation is correlated with behavioral benefits in the YAC128 mouse model of Huntington's disease.
- Author
-
Stanek LM, Yang W, Angus S, Sardi PS, Hayden MR, Hung GH, Bennett CF, Cheng SH, and Shihabuddin LS
- Subjects
- Animals, Disease Models, Animal, Dopamine and cAMP-Regulated Phosphoprotein 32 drug effects, Dopamine and cAMP-Regulated Phosphoprotein 32 genetics, Enkephalins drug effects, Enkephalins genetics, Huntingtin Protein, Hypoxanthine Phosphoribosyltransferase drug effects, Hypoxanthine Phosphoribosyltransferase genetics, Infusions, Intraventricular, Mice, Neostriatum metabolism, Nerve Tissue Proteins metabolism, Nuclear Proteins metabolism, Real-Time Polymerase Chain Reaction, Receptor, Cannabinoid, CB1 drug effects, Receptor, Cannabinoid, CB1 genetics, Receptors, Dopamine D1 drug effects, Receptors, Dopamine D1 genetics, Receptors, Dopamine D2 drug effects, Receptors, Dopamine D2 genetics, Behavior, Animal drug effects, Gene Expression Regulation drug effects, Huntington Disease genetics, Motor Skills drug effects, Neostriatum drug effects, Nerve Tissue Proteins drug effects, Nuclear Proteins drug effects, Oligonucleotides, Antisense pharmacology
- Abstract
Background: Huntington's disease (HD) is a neurological disorder caused by mutations in the huntingtin (HTT) gene, the product of which leads to selective and progressive neuronal cell death in the striatum and cortex. Transcriptional dysregulation has emerged as a core pathologic feature in the CNS of human and animal models of HD. It is still unclear whether perturbations in gene expression are a consequence of the disease or importantly, contribute to the pathogenesis of HD., Objective: To examine if transcriptional dysregulation can be ameliorated with antisense oligonucleotides that reduce levels of mutant Htt and provide therapeutic benefit in the YAC128 mouse model of HD., Methods: Quantitative real-time PCR analysis was used to evaluate dysregulation of a subset of striatal genes in the YAC128 mouse model. Transcripts were then evaluated following ICV delivery of antisense oligonucleotides (ASO). Rota rod and Porsolt swim tests were used to evaluate phenotypic deficits in these mice following ASO treatment., Results: Transcriptional dysregulation was detected in the YAC128 mouse model and appears to progress with age. ICV delivery of ASOs directed against mutant Htt resulted in reduction in mutant Htt levels and amelioration in behavioral deficits in the YAC128 mouse model. These improvements were correlated with improvements in the levels of several dysregulated striatal transcripts., Conclusions: The role of transcriptional dysregulation in the pathogenesis of Huntington's disease is not well understood, however, a wealth of evidence now strongly suggests that changes in transcriptional signatures are a prominent feature in the brains of both HD patients and animal models of the disease. Our study is the first to show that a therapeutic agent capable of improving an HD disease phenotype is concomitantly correlated with normalization of a subset of dysregulated striatal transcripts. Our data suggests that correction of these disease-altered transcripts may underlie, at least in part, the therapeutic efficacy shown associated with ASO-mediated correction of HD phenotypes and may provide a novel set of early biomarkers for evaluating future therapeutic concepts for HD.
- Published
- 2013
- Full Text
- View/download PDF
31. Merits of combination cortical, subcortical, and cerebellar injections for the treatment of Niemann-Pick disease type A.
- Author
-
Bu J, Ashe KM, Bringas J, Marshall J, Dodge JC, Cabrera-Salazar MA, Forsayeth J, Schuchman EH, Bankiewicz KS, Cheng SH, Shihabuddin LS, and Passini MA
- Subjects
- Animals, Brain enzymology, Dependovirus genetics, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, Genetic Vectors genetics, Injections, Macaca fascicularis, Male, Mice, Mice, Knockout, Niemann-Pick Disease, Type A pathology, Primates metabolism, Sphingomyelin Phosphodiesterase genetics, Sphingomyelin Phosphodiesterase metabolism, Genetic Therapy, Niemann-Pick Disease, Type A therapy, Sphingomyelin Phosphodiesterase deficiency
- Abstract
Niemann-Pick disease Type A (NPA) is a neuronopathic lysosomal storage disease (LSD) caused by the loss of acid sphingomyelinase (ASM). The goals of the current study are to ascertain the levels of human ASM that are efficacious in ASM knockout (ASMKO) mice, and determine whether these levels can be attained in non-human primates (NHPs) using a multiple parenchymal injection strategy. Intracranial injections of different doses of AAV1-hASM in ASMKO mice demonstrated that only a small amount of enzyme (<0.5 mg hASM/g tissue) was sufficient to increase survival, and that increasing the amount of hASM did not enhance this survival benefit until a new threshold level of >10 mg hASM/g tissue was reached. In monkeys, injection of 12 tracts of AAV1-hASM resulted in efficacious levels of enzyme in broad regions of the brain that was aided, in part, by axonal transport of adeno-associated virus (AAV) and movement through the perivascular space. This study demonstrates that a combination cortical, subcortical, and cerebellar injection protocol could provide therapeutic levels of hASM to regions of the NHP brain that are highly affected in NPA patients. The information from this study might help design new AAV-mediated enzyme replacement protocols for NPA and other neuronopathic LSDs in future clinical trials.
- Published
- 2012
- Full Text
- View/download PDF
32. Gene transfer to the CNS is efficacious in immune-primed mice harboring physiologically relevant titers of anti-AAV antibodies.
- Author
-
Treleaven CM, Tamsett TJ, Bu J, Fidler JA, Sardi SP, Hurlbut GD, Woodworth LA, Cheng SH, Passini MA, Shihabuddin LS, and Dodge JC
- Subjects
- Adult, Animals, Antibodies, Viral metabolism, Biomarkers metabolism, Brain metabolism, Dependovirus immunology, Disease Models, Animal, Gene Transfer Techniques, Genetic Vectors, Humans, Immunization, Mice, Niemann-Pick Disease, Type A genetics, Niemann-Pick Disease, Type A immunology, Niemann-Pick Disease, Type A metabolism, Transgenes, Antibodies, Viral immunology, Brain immunology, Dependovirus genetics, Genetic Therapy methods, Niemann-Pick Disease, Type A therapy
- Abstract
Central nervous system (CNS)-directed gene therapy with recombinant adeno-associated virus (AAV) vectors has been used effectively to slow disease course in mouse models of several neurodegenerative diseases. However, these vectors were typically tested in mice without prior exposure to the virus, an immunological scenario unlikely to be duplicated in human patients. Here, we examined the impact of pre-existing immunity on AAV-mediated gene delivery to the CNS of normal and diseased mice. Antibody levels in brain tissue were determined to be 0.6% of the levels found in systemic circulation. As expected, transgene expression in brains of mice with relatively high serum antibody titers was reduced by 59-95%. However, transduction activity was unaffected in mice that harbored more clinically relevant antibody levels. Moreover, we also showed that markers of neuroinflammation (GFAP, Iba1, and CD3) and histopathology (hematoxylin and eosin (H&E)) were not enhanced in immune-primed mice (regardless of pre-existing antibody levels). Importantly, we also demonstrated in a mouse model of Niemann Pick Type A (NPA) disease that pre-existing immunity did not preclude either gene transfer to the CNS or alleviation of disease-associated neuropathology. These findings support the continued development of AAV-based therapies for the treatment of neurological disorders.
- Published
- 2012
- Full Text
- View/download PDF
33. Sustained therapeutic reversal of Huntington's disease by transient repression of huntingtin synthesis.
- Author
-
Kordasiewicz HB, Stanek LM, Wancewicz EV, Mazur C, McAlonis MM, Pytel KA, Artates JW, Weiss A, Cheng SH, Shihabuddin LS, Hung G, Bennett CF, and Cleveland DW
- Subjects
- Animals, Corpus Striatum metabolism, Corpus Striatum pathology, Disease Models, Animal, Disease Progression, Huntingtin Protein, Huntington Disease genetics, Huntington Disease pathology, Infusions, Spinal, Macaca mulatta, Mice, Nerve Tissue Proteins biosynthesis, Nerve Tissue Proteins genetics, Neurons metabolism, Neurons pathology, Nuclear Proteins biosynthesis, Nuclear Proteins genetics, Oligodeoxyribonucleotides, Antisense administration & dosage, Time, Treatment Outcome, Huntington Disease therapy, Nerve Tissue Proteins antagonists & inhibitors, Nuclear Proteins antagonists & inhibitors, Oligodeoxyribonucleotides, Antisense therapeutic use
- Abstract
The primary cause of Huntington's disease (HD) is expression of huntingtin with a polyglutamine expansion. Despite an absence of consensus on the mechanism(s) of toxicity, diminishing the synthesis of mutant huntingtin will abate toxicity if delivered to the key affected cells. With antisense oligonucleotides (ASOs) that catalyze RNase H-mediated degradation of huntingtin mRNA, we demonstrate that transient infusion into the cerebrospinal fluid of symptomatic HD mouse models not only delays disease progression but mediates a sustained reversal of disease phenotype that persists longer than the huntingtin knockdown. Reduction of wild-type huntingtin, along with mutant huntingtin, produces the same sustained disease reversal. Similar ASO infusion into nonhuman primates is shown to effectively lower huntingtin in many brain regions targeted by HD pathology. Rather than requiring continuous treatment, our findings establish a therapeutic strategy for sustained HD disease reversal produced by transient ASO-mediated diminution of huntingtin synthesis., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
34. Marked differences in neurochemistry and aggregates despite similar behavioural and neuropathological features of Huntington disease in the full-length BACHD and YAC128 mice.
- Author
-
Pouladi MA, Stanek LM, Xie Y, Franciosi S, Southwell AL, Deng Y, Butland S, Zhang W, Cheng SH, Shihabuddin LS, and Hayden MR
- Subjects
- Animals, Disease Models, Animal, Female, Huntingtin Protein, Huntington Disease metabolism, Huntington Disease pathology, Male, Mice, Mice, Transgenic, Nerve Tissue Proteins metabolism, Neurons pathology, Transgenes, Huntington Disease genetics, Nerve Tissue Proteins genetics, Neurons metabolism
- Abstract
The development of animal models of Huntington disease (HD) has enabled studies that help define the molecular aberrations underlying the disease. The BACHD and YAC128 transgenic mouse models of HD harbor a full-length mutant huntingtin (mHTT) and recapitulate many of the behavioural and neuropathological features of the human condition. Here, we demonstrate that while BACHD and YAC128 animals exhibit similar deficits in motor learning and coordination, depressive-like symptoms, striatal volume loss and forebrain weight loss, they show obvious differences in key features characteristic of HD. While YAC128 mice exhibit significant and widespread accumulation of mHTT striatal aggregates, these mHTT aggregates are absent in BACHD mice. Furthermore, the levels of several striatally enriched mRNA for genes, such as DARPP-32, enkephalin, dopamine receptors D1 and D2 and cannabinoid receptor 1, are significantly decreased in YAC128 but not BACHD mice. These findings may reflect sequence differences in the human mHTT transgenes harboured by the BACHD and YAC128 mice, including both single nucleotide polymorphisms as well as differences in the nature of CAA interruptions of the CAG tract. Our findings highlight a similar profile of HD-like behavioural and neuropathological deficits and illuminate differences that inform the use of distinct endpoints in trials of therapeutic agents in the YAC128 and BACHD mice.
- Published
- 2012
- Full Text
- View/download PDF
35. Mutant GBA1 expression and synucleinopathy risk: first insights from cellular and mouse models.
- Author
-
Sardi SP, Singh P, Cheng SH, Shihabuddin LS, and Schlossmacher MG
- Subjects
- Animals, Brain metabolism, Brain pathology, Disease Models, Animal, Gaucher Disease metabolism, Glucosylceramidase metabolism, Humans, Mice, Gaucher Disease genetics, Gene Expression Regulation genetics, Glucosylceramidase genetics, Mutation genetics, alpha-Synuclein metabolism
- Abstract
Heterozygous mutations in the glucocerebrosidase gene (GBA1) are associated with increased risk for α-synuclein aggregation disorders ('synucleinopathies'), which include Parkinson's disease (PD) and dementia with Lewy bodies (DLB). Homozygous GBA1 mutations lead to reduced GBA1 lysosomal activity underlying three variants of Gaucher disease (GD). Despite the wealth of clinical and genetic evidence supporting the association between mutant genotypes and synucleinopathy risk, the precise mechanisms by which GBA1 mutations lead to PD and DLB remain unclear. Here, we summarize recent findings that highlight the complexity of this pathogenetic link. In neural cells, both gain and loss of function mechanisms, as conferred by mutant GBA1 expression and activity loss, respectively, seem to promote aberrant α-synuclein processing. In addition, we draw attention to recent insights gleaned from GD animal models regarding axonal pathology, brain inflammation and memory dysfunction. From a translational perspective, we discuss the concepts of neural enzyme replacement therapy and pharmacological agents as potential treatment strategies for GBA1-associated synucleinopathies. Finally, we touch on the issue whether aberrant α-synuclein species may coregulate GBA1 activity in the vertebrate brain, thereby providing a reverse link, i.e., between an important synucleinopathy risk factor and the enzyme's lysosomal function. In summary, several leads connecting GBA1 mutations with α-synuclein misprocessing have emerged as potential targets for the treatment of GBA1-related synucleinopathies, and possibly, for non-GBA1-associated neurodegenerative diseases., (Copyright © 2012 S. Karger AG, Basel.)
- Published
- 2012
- Full Text
- View/download PDF
36. Disease progression in a mouse model of amyotrophic lateral sclerosis: the influence of chronic stress and corticosterone.
- Author
-
Fidler JA, Treleaven CM, Frakes A, Tamsett TJ, McCrate M, Cheng SH, Shihabuddin LS, Kaspar BK, and Dodge JC
- Subjects
- Amyotrophic Lateral Sclerosis genetics, Amyotrophic Lateral Sclerosis metabolism, Animals, Corticosterone blood, Corticosterone pharmacology, Disease Models, Animal, Disease Progression, Female, Humans, Male, Mice, Mice, Mutant Strains, Mice, Transgenic, Models, Biological, Mutant Proteins genetics, Mutant Proteins metabolism, Restraint, Physical adverse effects, Stress, Physiological, Superoxide Dismutase genetics, Superoxide Dismutase metabolism, Amyotrophic Lateral Sclerosis etiology, Corticosterone metabolism
- Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by motor neuron cell loss, muscular atrophy, and a shortened life span. Survival is highly variable, as some patients die within months, while others live for many years. Exposure to stress or the development of a nonoptimal stress response to disease might account for some of this variability. We show in the SOD1(G93A) mouse model of ALS that recurrent exposure to restraint stress led to an earlier onset of astrogliosis and microglial activation within the spinal cord, accelerated muscular weakness, and a significant decrease in median survival (105 vs. 122 d) when compared to nonstressed animals. Moreover, during normal disease course, ALS mice display a cacostatic stress response by developing an aberrant serum corticosterone circadian rhythm. Interestingly, we also found that higher corticosterone levels were significantly correlated with both an earlier onset of paralysis (males: r(2)=0.746; females: r(2)=0.707) and shorter survival times (males: r(2)=0.680; females: r(2)=0.552) in ALS mice. These results suggest that stress is capable of accelerating disease progression and that strategies that modulate glucocorticoid metabolism might be a viable treatment approach for ALS.
- Published
- 2011
- Full Text
- View/download PDF
37. Neural stem cell transplantation as a therapeutic approach for treating lysosomal storage diseases.
- Author
-
Shihabuddin LS and Cheng SH
- Subjects
- Animals, Humans, Lysosomal Storage Diseases surgery, Neural Stem Cells physiology, Stem Cell Transplantation methods
- Abstract
Treating the central nervous system manifestations of subjects with neuropathic lysosomal storage diseases remains a major technical challenge. This is because of the low efficiency by which lysosomal enzymes in systemic circulation are able to traverse the blood brain barrier into the central nervous system. Intracranial transplantation of neural stems cells genetically modified to overexpress the respective deficient enzymes represents a potential approach to addressing this group of diseases. The unique properties of neural stem cells and progenitor cells, such as their ability to migrate to distal sites, differentiate into various cell types and integrate within the host brain without disrupting normal function, making them particularly attractive therapeutic agents. In addition, neural stem cells are amenable to ex vivo propagation and modification by gene transfer vectors. In this regard, transplanted cells can serve not only as a source of lysosomal enzymes but also as a means to potentially repair the injured brain by replenishing the organ with healthy cells and effecting the release of neuroprotective factors. This review discusses some of the well-characterized neural stem cell types and their possible use in treating neuropathic lysosomal storage diseases such as the Niemann Pick A disease.
- Published
- 2011
- Full Text
- View/download PDF
38. CNS expression of glucocerebrosidase corrects alpha-synuclein pathology and memory in a mouse model of Gaucher-related synucleinopathy.
- Author
-
Sardi SP, Clarke J, Kinnecom C, Tamsett TJ, Li L, Stanek LM, Passini MA, Grabowski GA, Schlossmacher MG, Sidman RL, Cheng SH, and Shihabuddin LS
- Subjects
- Analysis of Variance, Animals, Blotting, Western, Dependovirus, Endopeptidase K metabolism, Gaucher Disease metabolism, Gene Transfer Techniques, Genetic Vectors genetics, Glucosylceramidase genetics, Hippocampus cytology, Immunohistochemistry, Mice, Gaucher Disease pathology, Glucosylceramidase metabolism, Hippocampus enzymology, alpha-Synuclein metabolism
- Abstract
Emerging genetic and clinical evidence suggests a link between Gaucher disease and the synucleinopathies Parkinson disease and dementia with Lewy bodies. Here, we provide evidence that a mouse model of Gaucher disease (Gba1(D409V/D409V)) exhibits characteristics of synucleinopathies, including progressive accumulation of proteinase K-resistant α-synuclein/ubiquitin aggregates in hippocampal neurons and a coincident memory deficit. Analysis of homozygous (Gba1(D409V/D409V)) and heterozygous (Gba1(D409V/+) and Gba1(+/-)) Gaucher mice indicated that these pathologies are a result of the combination of a loss of glucocerebrosidase activity and a toxic gain-of-function resulting from expression of the mutant enzyme. Importantly, adeno-associated virus-mediated expression of exogenous glucocerebrosidase injected into the hippocampus of Gba1(D409V/D409V) mice ameliorated both the histopathological and memory aberrations. The data support the contention that mutations in GBA1 can cause Parkinson disease-like α-synuclein pathology, and that rescuing brain glucocerebrosidase activity might represent a therapeutic strategy for GBA1-associated synucleinopathies.
- Published
- 2011
- Full Text
- View/download PDF
39. Acid β-glucosidase mutants linked to Gaucher disease, Parkinson disease, and Lewy body dementia alter α-synuclein processing.
- Author
-
Cullen V, Sardi SP, Ng J, Xu YH, Sun Y, Tomlinson JJ, Kolodziej P, Kahn I, Saftig P, Woulfe J, Rochet JC, Glicksman MA, Cheng SH, Grabowski GA, Shihabuddin LS, and Schlossmacher MG
- Subjects
- Animals, Cathepsin D deficiency, Cathepsin D genetics, Cell Line, Disease Models, Animal, Dose-Response Relationship, Drug, Enzyme-Linked Immunosorbent Assay methods, Gene Expression Regulation drug effects, Gene Expression Regulation genetics, Green Fluorescent Proteins genetics, Humans, Immunosuppressive Agents pharmacology, Mice, Mice, Knockout, Mutagenesis, Site-Directed methods, Rats, Sirolimus pharmacology, Transfection, alpha-Synuclein genetics, Gaucher Disease genetics, Glucosylceramidase genetics, Lewy Body Disease genetics, Mutation genetics, Parkinson Disease genetics, alpha-Synuclein metabolism
- Abstract
Objective: Heterozygous mutations in the GBA1 gene elevate the risk of Parkinson disease and dementia with Lewy bodies; both disorders are characterized by misprocessing of α-synuclein (SNCA). A loss in lysosomal acid-β-glucosidase enzyme (GCase) activity due to biallelic GBA1 mutations underlies Gaucher disease. We explored mechanisms for the gene's association with increased synucleinopathy risk., Methods: We analyzed the effects of wild-type (WT) and several GBA mutants on SNCA in cellular and in vivo models using biochemical and immunohistochemical protocols., Results: We observed that overexpression of all GBA mutants examined (N370S, L444P, D409H, D409V, E235A, and E340A) significantly raised human SNCA levels to 121 to 248% of vector control (p < 0.029) in neural MES23.5 and PC12 cells, but without altering GCase activity. Overexpression of WT GBA in neural and HEK293-SNCA cells increased GCase activity, as expected (ie, to 167% in MES-SNCA, 128% in PC12-SNCA, and 233% in HEK293-SNCA; p < 0.002), but had mixed effects on SNCA. Nevertheless, in HEK293-SNCA cells high GCase activity was associated with SNCA reduction by ≤32% (p = 0.009). Inhibition of cellular GCase activity (to 8-20% of WT; p < 0.0017) did not detectably alter SNCA levels. Mutant GBA-induced SNCA accumulation could be pharmacologically reversed in D409V-expressing PC12-SNCA cells by rapamycin, an autophagy-inducer (≤40%; 10μM; p < 0.02). Isofagomine, a GBA chaperone, showed a related trend. In mice expressing two D409Vgba knockin alleles without signs of Gaucher disease (residual GCase activity, ≥20%), we recorded an age-dependent rise of endogenous Snca in hippocampal membranes (125% vs WT at 52 weeks; p = 0.019). In young Gaucher disease mice (V394Lgba+/+//prosaposin[ps]-null//ps-transgene), which demonstrate neurological dysfunction after age 10 weeks (GCase activity, ≤10%), we recorded no significant change in endogenous Snca levels at 12 weeks of age. However, enhanced neuronal ubiquitin signals and axonal spheroid formation were already present. The latter changes were similar to those seen in three week-old cathepsin D-deficient mice., Interpretation: Our results demonstrate that GBA mutants promote SNCA accumulation in a dose- and time-dependent manner, thereby identifying a biochemical link between GBA1 mutation carrier status and increased synucleinopathy risk. In cell culture models, this gain of toxic function effect can be mitigated by rapamycin. Loss in GCase activity did not immediately raise SNCA concentrations, but first led to neuronal ubiquitinopathy and axonal spheroids, a phenotype shared with other lysosomal storage disorders., (Copyright © 2011 American Neurological Association.)
- Published
- 2011
- Full Text
- View/download PDF
40. Antisense oligonucleotides delivered to the mouse CNS ameliorate symptoms of severe spinal muscular atrophy.
- Author
-
Passini MA, Bu J, Richards AM, Kinnecom C, Sardi SP, Stanek LM, Hua Y, Rigo F, Matson J, Hung G, Kaye EM, Shihabuddin LS, Krainer AR, Bennett CF, and Cheng SH
- Subjects
- Animals, Disease Models, Animal, Drug Delivery Systems, Humans, Macaca fascicularis, Mice, Motor Neurons physiology, Muscular Atrophy, Spinal physiopathology, Neuromuscular Junction ultrastructure, Oligonucleotides, Antisense administration & dosage, Oligonucleotides, Antisense genetics, Oligonucleotides, Antisense pharmacokinetics, RNA Splicing, Spinal Cord physiopathology, Survival of Motor Neuron 1 Protein genetics, Survival of Motor Neuron 2 Protein genetics, Muscular Atrophy, Spinal genetics, Muscular Atrophy, Spinal pathology, Muscular Atrophy, Spinal therapy, Oligonucleotides, Antisense therapeutic use, Spinal Cord pathology
- Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disorder caused by mutations in the SMN1 gene that result in a deficiency of SMN protein. One approach to treat SMA is to use antisense oligonucleotides (ASOs) to redirect the splicing of a paralogous gene, SMN2, to boost production of functional SMN. Injection of a 2'-O-2-methoxyethyl-modified ASO (ASO-10-27) into the cerebral lateral ventricles of mice with a severe form of SMA resulted in splice-mediated increases in SMN protein and in the number of motor neurons in the spinal cord, which led to improvements in muscle physiology, motor function and survival. Intrathecal infusion of ASO-10-27 into cynomolgus monkeys delivered putative therapeutic levels of the oligonucleotide to all regions of the spinal cord. These data demonstrate that central nervous system-directed ASO therapy is efficacious and that intrathecal infusion may represent a practical route for delivering this therapeutic in the clinic.
- Published
- 2011
- Full Text
- View/download PDF
41. Relationship between neuropathology and disease progression in the SOD1(G93A) ALS mouse.
- Author
-
Yang WW, Sidman RL, Taksir TV, Treleaven CM, Fidler JA, Cheng SH, Dodge JC, and Shihabuddin LS
- Subjects
- Alanine genetics, Amino Acid Substitution genetics, Amyotrophic Lateral Sclerosis genetics, Animals, Disease Models, Animal, Female, Glycine genetics, Humans, Male, Mice, Mice, Transgenic, Superoxide Dismutase genetics, Amyotrophic Lateral Sclerosis enzymology, Amyotrophic Lateral Sclerosis pathology, Disease Progression, Superoxide Dismutase biosynthesis
- Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of upper and lower motor neurons. However, recent reports suggest an active role of non-neuronal cells in the pathogenesis of the disease. Here, we examined quantitatively the temporal development of neuropathologic features in the brain and spinal cord of a mouse model of ALS (SOD1(G93A)). Four phases of the disease were studied in both male and female SOD1(G93A) mice: presymptomatic (PRE-SYM), symptomatic (SYM), endstage (ES) and moribund (MB). Compared to their control littermates, SOD1(G93A) mice showed an increase in astrogliosis in the motor cortex, spinal cord and motor trigeminal nucleus in the SYM phase that worsened progressively in ES and MB animals. Associated with this increase in astrogliosis was a concomitant increase in motor neuron cell death in the spinal cord and motor trigeminal nucleus in both ES and MB mice, as well as in the ventrolateral thalamus in MB animals. In contrast, microglial activation was significantly increased in all the same regions but only when the mice were in the MB phase. These results suggest that astrogliosis preceded or occurred concurrently with neuronal degeneration whereas prominent microgliosis was evident later (MB stage), after significant motor neuron degeneration had occurred. Hence, our findings support a role for astrocytes in modulating the progression of non-cell autonomous degeneration of motor neurons, with microglia playing a role in clearing degenerating neurons., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
42. Comparative analysis of acid sphingomyelinase distribution in the CNS of rats and mice following intracerebroventricular delivery.
- Author
-
Treleaven CM, Tamsett T, Fidler JA, Taksir TV, Cheng SH, Shihabuddin LS, and Dodge JC
- Subjects
- Animals, Dose-Response Relationship, Drug, Infusions, Intraventricular, Mice, Organ Specificity, Rats, Central Nervous System enzymology, Sphingomyelin Phosphodiesterase metabolism
- Abstract
Niemann-Pick A (NPA) disease is a lysosomal storage disorder (LSD) caused by a deficiency in acid sphingomyelinase (ASM) activity. Previously, we reported that biochemical and functional abnormalities observed in ASM knockout (ASMKO) mice could be partially alleviated by intracerebroventricular (ICV) infusion of hASM. We now show that this route of delivery also results in widespread enzyme distribution throughout the rat brain and spinal cord. However, enzyme diffusion into CNS parenchyma did not occur in a linear dose-dependent fashion. Moreover, although the levels of hASM detected in the rat CNS were determined to be within the range shown to be therapeutic in ASMKO mice, the absolute amounts represented less than 1% of the total dose administered. Finally, our results also showed that similar levels of enzyme distribution are achieved across rodent species when the dose is normalized to CNS weight as opposed to whole body weight. Collectively, these data suggest that the efficacy observed following ICV delivery of hASM in ASMKO mice could be scaled to CNS of the rat.
- Published
- 2011
- Full Text
- View/download PDF
43. AAV4-mediated expression of IGF-1 and VEGF within cellular components of the ventricular system improves survival outcome in familial ALS mice.
- Author
-
Dodge JC, Treleaven CM, Fidler JA, Hester M, Haidet A, Handy C, Rao M, Eagle A, Matthews JC, Taksir TV, Cheng SH, Shihabuddin LS, and Kaspar BK
- Subjects
- Amyotrophic Lateral Sclerosis genetics, Amyotrophic Lateral Sclerosis metabolism, Animals, Central Nervous System metabolism, Dependovirus genetics, Disease Models, Animal, Disease-Free Survival, Embryonic Stem Cells, Female, Immunohistochemistry, Insulin-Like Growth Factor I genetics, Male, Mice, Mice, Transgenic, Reverse Transcriptase Polymerase Chain Reaction, Vascular Endothelial Growth Factor A genetics, Amyotrophic Lateral Sclerosis therapy, Genetic Therapy, Insulin-Like Growth Factor I metabolism, Vascular Endothelial Growth Factor A metabolism
- Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by motor neuron cell death in the cortex, brainstem, and spinal cord. Extensive efforts have been made to develop trophic factor-based therapies to enhance motor neuron survival; however, achievement of adequate therapeutic delivery to all regions of the corticospinal tract has remained a significant challenge. Here, we show that adeno-associated virus serotype 4 (AAV4)-mediated expression of insulin-like growth factor-1 (IGF-1) or vascular endothelial growth factor (VEGF)-165 in the cellular components of the ventricular system including the ependymal cell layer, choroid plexus [the primary cerebrospinal fluid (CSF)-producing cells of the central nervous system (CNS)] and spinal cord central canal leads to trophic factor delivery throughout the CNS, delayed motor decline and a significant extension of survival in SOD1(G93A) transgenic mice. Interestingly, when IGF-1- and VEGF-165-expressing AAV4 vectors were given in combination, no additional benefit in efficacy was observed suggesting that these trophic factors are acting on similar signaling pathways to modestly slow disease progression. Consistent with these findings, experiments conducted in a recently described in vitro cell culture model of ALS led to a similar result, with both IGF-1 and VEGF-165 providing significant motor neuron protection but in a nonadditive fashion. These findings support the continued investigation of trophic factor-based therapies that target the CNS as a potential treatment of ALS.
- Published
- 2010
- Full Text
- View/download PDF
44. Magnetic resonance imaging-guided delivery of adeno-associated virus type 2 to the primate brain for the treatment of lysosomal storage disorders.
- Author
-
Salegio EA, Kells AP, Richardson RM, Hadaczek P, Forsayeth J, Bringas J, Sardi SP, Passini MA, Shihabuddin LS, Cheng SH, Fiandaca MS, and Bankiewicz KS
- Subjects
- Animals, Brain pathology, Humans, Intraoperative Care, Neurons metabolism, Primates, Sphingomyelin Phosphodiesterase genetics, Sphingomyelin Phosphodiesterase therapeutic use, Transduction, Genetic, Transgenes genetics, Brain metabolism, Dependovirus genetics, Gene Transfer Techniques, Genetic Therapy, Lysosomal Storage Diseases genetics, Lysosomal Storage Diseases therapy, Magnetic Resonance Imaging
- Abstract
Gene replacement therapy for the neurological deficits caused by lysosomal storage disorders, such as in Niemann-Pick disease type A, will require widespread expression of efficacious levels of acid sphingomyelinase (ASM) in the infant human brain. At present there is no treatment available for this devastating pediatric condition. This is partly because of inherent constraints associated with the efficient delivery of therapeutic agents into the CNS of higher order models. In this study we used an adeno-associated virus type 2 (AAV2) vector encoding human acid sphingomyelinase tagged with a viral hemagglutinin epitope (AAV2-hASM-HA) to transduce highly interconnected CNS regions such as the brainstem and thalamus. On the basis of our data showing global cortical expression of a secreted reporter after thalamic delivery in nonhuman primates (NHPs), we set out to investigate whether such widespread expression could be enhanced after brainstem infusion. To maximize delivery of the therapeutic transgene throughout the CNS, we combined a single brainstem infusion with bilateral thalamic infusions in naive NHPs. We found that enzymatic augmentation in brainstem, thalamic, cortical, as well subcortical areas provided convincing evidence that much of the large NHP brain can be transduced with as few as three injection sites.
- Published
- 2010
- Full Text
- View/download PDF
45. Stem cell transplantation for neurometabolic and neurodegenerative diseases.
- Author
-
Shihabuddin LS and Aubert I
- Subjects
- Animals, Humans, Metabolic Diseases surgery, Lysosomal Storage Diseases, Nervous System surgery, Neurodegenerative Diseases surgery, Stem Cell Transplantation methods
- Abstract
Over the last decade, the potential for therapeutic use of stem cell transplantation for cell replacement or as cellular vectors for gene delivery for neurometabolic and neurodegenerative diseases has received a great deal of interest. There has been substantial progress in our understanding of stem cell biology. Potential applications of cell-mediated therapy include direct cell replacement or protection and repair of the host nervous system. Given the complexities of the cellular organization of the nervous system, especially in diseased states, it seems that using stem cells as cellular vectors to prevent or ameliorate neurological disorders rather than cell replacement and the regrowth of damaged circuitry is more likely to succeed in the near term. Recent success in the treatment of lysosomal storage diseases with genetically modified stem cells support this notion. In Alzheimer's and Parkinson's diseases, stem cell therapy is at its early stages and data generated in animal models and clinical trials using other cell types suggest that a combination of gene and stem cell therapy may be an optimal therapeutic paradigm., (Copyright 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
46. CNS-targeted gene therapy improves survival and motor function in a mouse model of spinal muscular atrophy.
- Author
-
Passini MA, Bu J, Roskelley EM, Richards AM, Sardi SP, O'Riordan CR, Klinger KW, Shihabuddin LS, and Cheng SH
- Subjects
- Animals, Disease Models, Animal, Humans, Mice, Muscle Strength, Muscle, Skeletal pathology, Muscular Atrophy, Spinal mortality, Muscular Atrophy, Spinal physiopathology, Neurites metabolism, Neuromuscular Junction pathology, Genetic Therapy, Motor Neurons physiology, Muscular Atrophy, Spinal therapy, Survival of Motor Neuron 1 Protein genetics
- Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disease caused by a deficiency of survival motor neuron (SMN) due to mutations in the SMN1 gene. In this study, an adeno-associated virus (AAV) vector expressing human SMN (AAV8-hSMN) was injected at birth into the CNS of mice modeling SMA. Western blot analysis showed that these injections resulted in widespread expression of SMN throughout the spinal cord, and this translated into robust improvement in skeletal muscle physiology, including increased myofiber size and improved neuromuscular junction architecture. Treated mice also displayed substantial improvements on behavioral tests of muscle strength, coordination, and locomotion, indicating that the neuromuscular junction was functional. Treatment with AAV8-hSMN increased the median life span of mice with SMA-like disease to 50 days compared with 15 days for untreated controls. Moreover, injecting mice with SMA-like disease with a human SMN-expressing self-complementary AAV vector - a vector that leads to earlier onset of gene expression compared with standard AAV vectors - led to improved efficacy of gene therapy, including a substantial extension in median survival to 157 days. These data indicate that CNS-directed, AAV-mediated SMN augmentation is highly efficacious in addressing both neuronal and muscular pathologies in a severe mouse model of SMA.
- Published
- 2010
- Full Text
- View/download PDF
47. Intracerebroventricular infusion of acid sphingomyelinase corrects CNS manifestations in a mouse model of Niemann-Pick A disease.
- Author
-
Dodge JC, Clarke J, Treleaven CM, Taksir TV, Griffiths DA, Yang W, Fidler JA, Passini MA, Karey KP, Schuchman EH, Cheng SH, and Shihabuddin LS
- Subjects
- Animals, Brain metabolism, Cholesterol metabolism, Disease Models, Animal, Humans, Injections, Intraventricular methods, Lysosomes drug effects, Lysosomes metabolism, Mice, Mice, Knockout, Niemann-Pick Disease, Type A genetics, Niemann-Pick Disease, Type A pathology, Sphingomyelin Phosphodiesterase deficiency, Sphingomyelin Phosphodiesterase metabolism, Sphingomyelins metabolism, Time Factors, Niemann-Pick Disease, Type A drug therapy, Sphingomyelin Phosphodiesterase administration & dosage
- Abstract
Niemann-Pick A (NPA) disease is a lysosomal storage disorder (LSD) caused by a deficiency in acid sphingomyelinase (ASM) activity. Previously, we showed that the storage pathology in the ASM knockout (ASMKO) mouse brain could be corrected by intracerebral injections of cell, gene and protein based therapies. However, except for instances where distal areas were targeted with viral vectors, correction of lysosomal storage pathology was typically limited to a region within a few millimeters from the injection site. As NPA is a global neurometabolic disease, the development of delivery strategies that maximize the distribution of the enzyme throughout the CNS is likely necessary to arrest or delay progression of the disease. To address this challenge, we evaluated the effectiveness of intracerebroventricular (ICV) delivery of recombinant human ASM into ASMKO mice. Our findings showed that ICV delivery of the enzyme led to widespread distribution of the hydrolase throughout the CNS. Moreover, a significant reduction in lysosomal accumulation of sphingomyelin was observed throughout the brain and also within the spinal cord and viscera. Importantly, we demonstrated that repeated ICV infusions of ASM were effective at improving the disease phenotype in the ASMKO mouse as indicated by a partial alleviation of the motor abnormalities. These findings support the continued exploration of ICV delivery of recombinant lysosomal enzymes as a therapeutic modality for LSDs such as NPA that manifests substrate accumulation within the CNS.
- Published
- 2009
- Full Text
- View/download PDF
48. Polysialic acid regulates the clustering, migration, and neuronal differentiation of progenitor cells in the adult hippocampus.
- Author
-
Burgess A, Wainwright SR, Shihabuddin LS, Rutishauser U, Seki T, and Aubert I
- Subjects
- Analysis of Variance, Animals, Bromodeoxyuridine metabolism, Cell Count, Cell Movement drug effects, Doublecortin Domain Proteins, Epidermal Growth Factor pharmacology, Fibroblast Growth Factors pharmacology, Glycoside Hydrolases pharmacology, Ki-67 Antigen metabolism, Membrane Transport Proteins metabolism, Microscopy, Confocal, Microtubule-Associated Proteins metabolism, Monocarboxylic Acid Transporters, Neurons drug effects, Neuropeptides metabolism, Phosphopyruvate Hydratase metabolism, Rats, Rats, Wistar, Adult Stem Cells drug effects, Cell Differentiation drug effects, Cell Movement physiology, Hippocampus cytology, Neurons physiology, Sialic Acids metabolism
- Abstract
Newborn cells of the adult dentate gyrus in the hippocampus are characterized by their abundant expression of polysialic acid (PSA), a carbohydrate attached to the neural cell adhesion molecule (NCAM). PSA+ newborn cells of the dentate gyrus form clusters with proliferating neural progenitor cells, migrate away from these clusters, and terminally differentiate. To identify the roles of PSA in the development of adult progenitors of the dentate gyrus, we injected endoneuraminidase N (endoN) into the hippocampus of adult rats to specifically cleave PSA from NCAM. Two days later, we administered the mitotic marker, 5-bromo-2'-deoxyuridine (BrdU). Three days after BrdU injection, BrdU+ cells were found inside and outside the clusters of newborn cells. In endoN-treated animals, the total number of BrdU+ cells was not changed but significantly more BrdU+ cells were present within clusters, suggesting that PSA normally facilitates the migration of progenitors away from the clusters. Seven days post-BrdU injection, endoN-treated animals had significantly more BrdU+ cells which were also positive for the mature neuronal nuclear marker NeuN compared with controls, indicating that the loss of PSA from progenitor cells increases neuronal differentiation. This report is the first demonstration that PSA is involved in controlling the spatio-temporal neuronal maturation of adult hippocampal progenitors in the normal brain. In vitro, the removal of PSA from adult-derived neural progenitors significantly enhanced neuronal differentiation, strengthening our in vivo findings and indicating that PSA removal on isolated progenitor cells, apart from a complex in vivo environment, induces neuronal maturation., ((c) 2008 Wiley Periodicals, Inc.)
- Published
- 2008
- Full Text
- View/download PDF
49. Temporal neuropathologic and behavioral phenotype of 6neo/6neo Pompe disease mice.
- Author
-
Sidman RL, Taksir T, Fidler J, Zhao M, Dodge JC, Passini MA, Raben N, Thurberg BL, Cheng SH, and Shihabuddin LS
- Subjects
- Age Factors, Animals, Central Nervous System ultrastructure, Disease Progression, Glial Fibrillary Acidic Protein metabolism, Glycogen metabolism, Glycogen Storage Disease Type II genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Microscopy, Electron, Transmission methods, Motor Activity physiology, Muscle Strength physiology, Muscle, Skeletal pathology, Muscle, Skeletal physiopathology, Psychomotor Performance physiology, Reaction Time physiology, alpha-Glucosidases deficiency, Behavior, Animal physiology, Central Nervous System pathology, Disease Models, Animal, Glycogen Storage Disease Type II pathology, Glycogen Storage Disease Type II physiopathology, Phenotype
- Abstract
Pompe disease (glycogen storage disease II) is caused by mutations in the acid alpha-glucosidase gene. The most common form is rapidly progressive with glycogen storage, particularly in muscle, which leads to profound weakness, cardiac failure, and death by the age of 2 years. Although usually considered a muscle disease, glycogen storage also occurs in the CNS. We evaluated the progression of neuropathologic and behavioral abnormalities in a Pompe disease mouse model (6neo/6neo) that displays many features of the human disease. Homozygous mutant mice store excess glycogen within large neurons of hindbrain, spinal cord, and sensory ganglia by the age of 1 month; accumulations then spread progressively within many CNS cell types. "Silver degeneration" and Fluoro-Jade C stains revealed severe degeneration in axon terminals of primary sensory neurons at 3 to 9 months. These abnormalities were accompanied by progressive behavioral impairment on rotorod, wire hanging, and foot fault tests. The extensive neuropathologic alterations in this model suggest that therapy of skeletal and cardiac muscle disorders by systemic enzyme replacement therapy may not be sufficient to reverse functional deficits due to CNS glycogen storage, particularly early-onset, rapidly progressive disease. A better understanding of the basis for clinical manifestations is needed to correlate CNS pathology with Pompe disease manifestations.
- Published
- 2008
- Full Text
- View/download PDF
50. Delivery of AAV-IGF-1 to the CNS extends survival in ALS mice through modification of aberrant glial cell activity.
- Author
-
Dodge JC, Haidet AM, Yang W, Passini MA, Hester M, Clarke J, Roskelley EM, Treleaven CM, Rizo L, Martin H, Kim SH, Kaspar R, Taksir TV, Griffiths DA, Cheng SH, Shihabuddin LS, and Kaspar BK
- Subjects
- Animals, Cell Survival, Central Nervous System metabolism, Cerebellum metabolism, Female, Insulin-Like Growth Factor I metabolism, Male, Mice, Neurodegenerative Diseases metabolism, Tumor Necrosis Factor-alpha metabolism, Amyotrophic Lateral Sclerosis genetics, Amyotrophic Lateral Sclerosis therapy, Central Nervous System cytology, Dependovirus genetics, Genetic Therapy methods, Insulin-Like Growth Factor I genetics, Neuroglia cytology, Neuroglia metabolism
- Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease of the motor system. Recent work in rodent models of ALS has shown that insulin-like growth factor-1 (IGF-1) slows disease progression when delivered at disease onset. However, IGF-1's mechanism of action along the neuromuscular axis remains unclear. In this study, symptomatic ALS mice received IGF-1 through stereotaxic injection of an IGF-1-expressing viral vector to the deep cerebellar nuclei (DCN), a region of the cerebellum with extensive brain stem and spinal cord connections. We found that delivery of IGF-1 to the central nervous system (CNS) reduced ALS neuropathology, improved muscle strength, and significantly extended life span in ALS mice. To explore the mechanism of action of IGF-1, we used a newly developed in vitro model of ALS. We demonstrate that IGF-1 is potently neuroprotective and attenuates glial cell-mediated release of tumor necrosis factor-alpha (TNF-alpha) and nitric oxide (NO). Our results show that delivering IGF-1 to the CNS is sufficient to delay disease progression in a mouse model of familial ALS and demonstrate for the first time that IGF-1 attenuates the pathological activity of non-neuronal cells that contribute to disease progression. Our findings highlight an innovative approach for delivering IGF-1 to the CNS.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.