Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 40(47): 9137-9147, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33051352

RESUMEN

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 SOD1G93A 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.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Metabolismo de los Lípidos , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Muerte Celular , Ésteres del Colesterol/metabolismo , Sustancia Gris/metabolismo , Humanos , Lisofosfatidilcolinas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Neuronas Motoras/patología , Receptores Acoplados a Proteínas G/genética , Receptores de Fosfolipasa A2/metabolismo , Médula Espinal/metabolismo , Superóxido Dismutasa-1/genética , Triglicéridos/metabolismo
2.
Proc Natl Acad Sci U S A ; 114(10): 2699-2704, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28223512

RESUMEN

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, GbaD409V/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.


Asunto(s)
Carbamatos/farmacología , Inhibidores Enzimáticos/administración & dosificación , Glucosiltransferasas/antagonistas & inhibidores , Enfermedad de Parkinson/tratamiento farmacológico , Quinuclidinas/farmacología , alfa-Sinucleína/genética , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Glucosiltransferasas/genética , Humanos , Ratones , Mutación , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/patología , Agregación Patológica de Proteínas/tratamiento farmacológico , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología , Ubiquitina/metabolismo , Proteínas tau/metabolismo
3.
Hum Mol Genet ; 25(13): 2645-2660, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27126635

RESUMEN

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.


Asunto(s)
Glucosilceramidasa/metabolismo , Glucosilceramidasa/fisiología , Animales , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Dopamina , Enfermedad de Gaucher/genética , Expresión Génica , Glucosilceramidasa/genética , Glucosilceramidasa/uso terapéutico , Humanos , Ratones , Actividad Motora/efectos de los fármacos , Mutación , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , alfa-Sinucleína/líquido cefalorraquídeo , alfa-Sinucleína/metabolismo
4.
Proc Natl Acad Sci U S A ; 112(26): 8100-5, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26056266

RESUMEN

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 ß-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.


Asunto(s)
Esclerosis Amiotrófica Lateral/fisiopatología , Glicoesfingolípidos/fisiología , Esclerosis Amiotrófica Lateral/enzimología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Gangliósido G(M3)/administración & dosificación , Glucosiltransferasas/antagonistas & inhibidores , Humanos , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Transgénicos , Médula Espinal/fisiopatología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
5.
Proc Natl Acad Sci U S A ; 110(9): 3537-42, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23297226

RESUMEN

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.


Asunto(s)
Encéfalo/enzimología , Enfermedad de Gaucher/tratamiento farmacológico , Enfermedad de Gaucher/enzimología , Glucosilceramidasa/metabolismo , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/enzimología , alfa-Sinucleína/metabolismo , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Enfermedad de Gaucher/patología , Enfermedad de Gaucher/fisiopatología , Glucosilceramidasa/administración & dosificación , Glucosilceramidasa/genética , Glucosilceramidasa/uso terapéutico , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Humanos , Memoria , Ratones , Ratones Transgénicos , Trastornos Parkinsonianos/fisiopatología , Estructura Cuaternaria de Proteína , Psicosina/análogos & derivados , Psicosina/metabolismo , alfa-Sinucleína/genética , Proteínas tau/química , Proteínas tau/metabolismo
6.
Proc Natl Acad Sci U S A ; 110(26): 10812-7, 2013 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-23754387

RESUMEN

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.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Acidosis/etiología , Acidosis/genética , Acidosis/metabolismo , Esclerosis Amiotrófica Lateral/etiología , Esclerosis Amiotrófica Lateral/genética , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Glucógeno/metabolismo , Humanos , Ratones , Ratones Transgénicos , Mutación , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
7.
Mol Ther ; 22(8): 1450-1459, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24814151

RESUMEN

Spinal muscular atrophy is a progressive motor neuron disease caused by a deficiency of survival motor neuron. In this study, we evaluated the efficacy of intravenous administration of a recombinant adeno-associated virus (AAV1) vector encoding human insulin-like growth factor-1 (IGF-1) in a severe mouse model of spinal muscular atrophy. Measurable quantities of human IGF-1 transcripts and protein were detected in the liver (up to 3 months postinjection) and in the serum indicating that IGF-1 was secreted from the liver into systemic circulation. Spinal muscular atrophy mice administered AAV1-IGF-1 on postnatal day 1 exhibited a lower extent of motor neuron degeneration, cardiac and muscle atrophy as well as a greater extent of innervation at the neuromuscular junctions compared to untreated controls at day 8 posttreatment. Importantly, treatment with AAV1-IGF-1 prolonged the animals' lifespan, increased their body weights and improved their motor coordination. Quantitative polymerase chain reaction and western blot analyses showed that AAV1-mediated expression of IGF-1 led to an increase in survival motor neuron transcript and protein levels in the spinal cord, brain, muscles, and heart. These data indicate that systemically delivered AAV1-IGF-1 can correct several of the biochemical and behavioral deficits in spinal muscular atrophy mice through increasing tissue levels of survival motor neuron.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/genética , Atrofia Muscular Espinal/fisiopatología , Atrofia Muscular Espinal/terapia , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Humanos , Inyecciones Intravenosas , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Hígado/metabolismo , Ratones , Atrofia Muscular Espinal/sangre , Atrofia Muscular Espinal/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Resultado del Tratamiento
8.
Mol Ther ; 20(9): 1713-23, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22735381

RESUMEN

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.


Asunto(s)
Anticuerpos Antivirales/inmunología , Encéfalo/inmunología , Dependovirus/genética , Terapia Genética/métodos , Enfermedad de Niemann-Pick Tipo A/terapia , Adulto , Animales , Anticuerpos Antivirales/metabolismo , Biomarcadores/metabolismo , Encéfalo/metabolismo , Dependovirus/inmunología , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Inmunización , Ratones , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/inmunología , Enfermedad de Niemann-Pick Tipo A/metabolismo , Transgenes
9.
Mol Ther ; 20(10): 1893-901, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22828503

RESUMEN

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.


Asunto(s)
Terapia Genética , Enfermedad de Niemann-Pick Tipo A/terapia , Esfingomielina Fosfodiesterasa/deficiencia , Animales , Encéfalo/enzimología , Dependovirus/genética , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos/genética , Inyecciones , Macaca fascicularis , Masculino , Ratones , Ratones Noqueados , Enfermedad de Niemann-Pick Tipo A/patología , Primates/metabolismo , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/metabolismo
10.
Sci Rep ; 13(1): 19529, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37945646

RESUMEN

Multiple sclerosis (MS), a chronic neurodegenerative disease driven by damage to the protective myelin sheath, is currently incurable. Today, all clinically available treatments modulate the immune-mediated symptoms of the disease but they fail to stop neurodegeneration in many patients. Remyelination, the regenerative process of myelin repair by oligodendrocytes, which is considered a necessary step to protect demyelinated axons and stop neuronal death, is impaired in MS patients. One of the major obstacles to finding effective remyelinating drugs is the lack of biomimetic drug screening platforms that enable quantification of compounds' potential to stimulate 3D myelination in the physiologically relevant axon-like environment. To address this need, we built a unique myelination drug discovery platform, by expanding our previously developed technology, artificial axons (AAs), which enables 3D-printing of synthetic axon mimics with the geometry and mechanical properties closely resembling those of biological axons. This platform allows for high-throughput phenotypic myelination assay based on quantification of 3D wrapping of myelin membrane around axons in response to compounds. Here, we demonstrate quantification of 3D myelin wrapping by rat oligodendrocytes around the axon mimics in response to a small library of known pro-myelinating compounds. This assay shows pro-myelinating activity for all tested compounds consistent with the published in vitro and in vivo data, demonstrating predictive power of AA platform. We find that stimulation of myelin wrapping by these compounds is dose-dependent, providing a facile means to quantify the compounds' potency and efficacy in promoting myelin wrapping. Further, the ranking of relative efficacy among these compounds differs in this 3D axon-like environment as compared to a traditional oligodendrocyte 2D differentiation assay quantifying area of deposited myelin membrane. Together, we demonstrate that the artificial axons platform and associated phenotypic myelin wrapping assay afford direct evaluation of myelin wrapping by oligodendrocytes in response to soluble compounds in an axon-like environment, providing a predictive tool for the discovery of remyelinating therapies.


Asunto(s)
Esclerosis Múltiple , Enfermedades Neurodegenerativas , Humanos , Ratas , Animales , Biomimética , Axones/fisiología , Vaina de Mielina/fisiología , Oligodendroglía/fisiología , Esclerosis Múltiple/tratamiento farmacológico
11.
iScience ; 26(3): 106156, 2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36852281

RESUMEN

Promoting myelination capacity of endogenous oligodendrocyte precursor cells (OPCs) is a promising therapeutic approach for CNS demyelinating disorders such as Multiple Sclerosis (MS). To aid in the discovery of myelination-promoting compounds, we generated a genome-engineered human pluripotent stem cell (hPSC) line that consists of three reporters: identification-and-purification tag, GFP, and secreted-NanoLuc, driven by the endogenous PDGFRA, PLP1, and MBP genes, respectively. Using this cell line, we established a high-throughput drug screening platform and performed a small-molecule screen, which identified at least two myelination-promoting small-molecule (Ro1138452 and SR2211) that target prostacyclin (IP) receptor and retinoic acid receptor-related orphan receptor γ (RORγ), respectively. Single-cell-transcriptomic analysis of differentiating OPCs treated with these molecules further confirmed that they promote oligodendrocyte differentiation and revealed several pathways that are potentially modulated by them. The molecules and their target pathways provide promising targets for the possible development of remyelination-based therapy for MS and other demyelinating disorders.

12.
Nat Neurosci ; 26(1): 12-26, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36536241

RESUMEN

Iron dysregulation has been implicated in multiple neurodegenerative diseases, including Parkinson's disease (PD). Iron-loaded microglia are frequently found in affected brain regions, but how iron accumulation influences microglia physiology and contributes to neurodegeneration is poorly understood. Here we show that human induced pluripotent stem cell-derived microglia grown in a tri-culture system are highly responsive to iron and susceptible to ferroptosis, an iron-dependent form of cell death. Furthermore, iron overload causes a marked shift in the microglial transcriptional state that overlaps with a transcriptomic signature found in PD postmortem brain microglia. Our data also show that this microglial response contributes to neurodegeneration, as removal of microglia from the tri-culture system substantially delayed iron-induced neurotoxicity. To elucidate the mechanisms regulating iron response in microglia, we performed a genome-wide CRISPR screen and identified novel regulators of ferroptosis, including the vesicle trafficking gene SEC24B. These data suggest a critical role for microglia iron overload and ferroptosis in neurodegeneration.


Asunto(s)
Ferroptosis , Células Madre Pluripotentes Inducidas , Sobrecarga de Hierro , Enfermedad de Parkinson , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Hierro/metabolismo , Sobrecarga de Hierro/metabolismo , Microglía/metabolismo , Enfermedad de Parkinson/genética
13.
Neurobiol Dis ; 45(1): 272-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21884794

RESUMEN

The efficacy of administering a recombinant adeno-associated virus (AAV) vector encoding human IGF-1 (AAV2/1-hIGF-1) into the deep cerebellar nucleus (DCN) of a type III SMA mouse model was evaluated. High levels of IGF-1 transcripts and protein were detected in the spinal cord at 2 months post-injection demonstrating that axonal connections between the cerebellum and spinal cord were able to act as conduits for the viral vector and protein to the spinal cord. Mice treated with AAV2/1-hIGF-1 and analyzed 8 months later showed changes in endogenous Bax and Bcl-xl levels in spinal cord motor neurons that were consistent with IGF-1-mediated anti-apoptotic effects on motor neurons. However, although AAV2/1-hIGF-1 treatment reduced the extent of motor neuron cell death, the majority of rescued motor neurons were non-functional, as they lacked axons that innervated the muscles. Furthermore, treated SMA mice exhibited abnormal muscle fibers, aberrant neuromuscular junction structure, and impaired performance on motor function tests. These data indicate that although CNS-directed expression of IGF-1 could reduce motor neuron cell death, this did not translate to improvements in motor function in an adult mouse model of type III SMA.


Asunto(s)
Muerte Celular/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Actividad Motora/efectos de los fármacos , Neuronas Motoras/efectos de los fármacos , Atrofia Muscular Espinal/terapia , Animales , Muerte Celular/fisiología , Cerebelo/efectos de los fármacos , Cerebelo/patología , Cerebelo/fisiopatología , Terapia Genética , Vectores Genéticos , Factor I del Crecimiento Similar a la Insulina/farmacología , Ratones , Actividad Motora/fisiología , Neuronas Motoras/patología , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/patología , Atrofia Muscular Espinal/patología , Atrofia Muscular Espinal/fisiopatología , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Médula Espinal/fisiopatología
14.
FASEB J ; 25(12): 4369-77, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21876068

RESUMEN

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.


Asunto(s)
Esclerosis Amiotrófica Lateral/etiología , Corticosterona/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Corticosterona/sangre , Corticosterona/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Mutantes , Ratones Transgénicos , Modelos Biológicos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Restricción Física/efectos adversos , Estrés Fisiológico , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
15.
Alzheimers Res Ther ; 14(1): 19, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35105352

RESUMEN

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 Aß42 accumulation in the brains of AD model mice when treatment was started prophylactically. Remarkably, GCSi treatment also reduced soluble Aß42 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 Aß42 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 Aß neuropathology and that glycosphingolipid homeostasis plays a critical role in the consolidation of remote memories.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides , Animales , Modelos Animales de Enfermedad , Gangliósido G(M3) , Glucosiltransferasas , Memoria a Largo Plazo , Ratones , Ratones Transgénicos , Placa Amiloide
16.
Mol Ther ; 18(12): 2075-84, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20859261

RESUMEN

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.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Terapia Genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Sistema Nervioso Central/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Supervivencia sin Enfermedad , Células Madre Embrionarias , Femenino , Inmunohistoquímica , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/genética
17.
Sci Rep ; 11(1): 803, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436868

RESUMEN

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 SOD1G93A 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.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Neuronas Motoras/patología , Enfermedades del Sistema Nervioso/patología , Médula Espinal/patología , Esteroles/química , Superóxido Dismutasa-1/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Muerte Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Heces/química , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Médula Espinal/metabolismo , Esteroles/metabolismo , Superóxido Dismutasa-1/genética
18.
Mol Ther ; 16(4): 649-56, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18362923

RESUMEN

Late infantile neuronal ceroid lipofuscinosis (LINCL) is an autosomal recessive neurodegenerative disease caused by mutations in CLN2, which encodes the lysosomal protease tripeptidyl peptidase 1 (TPP1). LINCL is characterized clinically by progressive motor and cognitive decline, and premature death. Enzyme-replacement therapy (ERT) is currently available for lysosomal storage diseases affecting peripheral tissues, but has not been used in patients with central nervous system (CNS) involvement. Enzyme delivery through the cerebrospinal fluid is a potential alternative route to the CNS, but has not been studied for LINCL. In this study, we identified relevant neuropathological and behavioral hallmarks of disease in a mouse model of LINCL and correlated those findings with tissues from LINCL patients. Subsequently, we tested if intraventricular delivery of TPP1 to the LINCL mouse was efficacious. We found that infusion of recombinant human TPP1 through an intraventricular cannula led to enzyme distribution in several regions of the brain of treated mice. In vitro activity assays confirm increased TPP1 activity throughout the rostral-caudal extent of the brain. Importantly, treated mice showed attenuated neuropathology, and decreased resting tremor relative to vehicle-treated mice. This data demonstrates that intraventricular enzyme delivery to the CNS is feasible and may be of therapeutic value.


Asunto(s)
Endopeptidasas/uso terapéutico , Lipofuscinosis Ceroideas Neuronales/terapia , Adulto , Aminopeptidasas , Animales , Astrocitos/metabolismo , Encéfalo/enzimología , Encéfalo/patología , Ventrículos Cerebrales , Niño , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas , Modelos Animales de Enfermedad , Endopeptidasas/genética , Endopeptidasas/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Lactante , Ratones , Ratones Noqueados , Lipofuscinosis Ceroideas Neuronales/patología , Lipofuscinosis Ceroideas Neuronales/fisiopatología , Neuronas/metabolismo , Fenotipo , Células de Purkinje/metabolismo , Células de Purkinje/patología , Proteínas Recombinantes/uso terapéutico , Serina Proteasas , Tripeptidil Peptidasa 1
19.
Mol Ther ; 16(6): 1056-64, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18388910

RESUMEN

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.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/terapia , Sistema Nervioso Central/citología , Dependovirus/genética , Terapia Genética/métodos , Factor I del Crecimiento Similar a la Insulina/genética , Neuroglía/citología , Neuroglía/metabolismo , Animales , Supervivencia Celular , Sistema Nervioso Central/metabolismo , Cerebelo/metabolismo , Femenino , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Enfermedades Neurodegenerativas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
20.
J Neuropathol Exp Neurol ; 67(8): 803-18, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18648322

RESUMEN

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.


Asunto(s)
Conducta Animal/fisiología , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Fenotipo , Factores de Edad , Animales , Sistema Nervioso Central/ultraestructura , Progresión de la Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión/métodos , Actividad Motora/fisiología , Fuerza Muscular/fisiología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Desempeño Psicomotor/fisiología , Tiempo de Reacción/fisiología , alfa-Glucosidasas/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA