Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Nat Chem Biol ; 11(7): 511-7, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26030728

RESUMEN

Spinal muscular atrophy (SMA), which results from the loss of expression of the survival of motor neuron-1 (SMN1) gene, represents the most common genetic cause of pediatric mortality. A duplicate copy (SMN2) is inefficiently spliced, producing a truncated and unstable protein. We describe herein a potent, orally active, small-molecule enhancer of SMN2 splicing that elevates full-length SMN protein and extends survival in a severe SMA mouse model. We demonstrate that the molecular mechanism of action is via stabilization of the transient double-strand RNA structure formed by the SMN2 pre-mRNA and U1 small nuclear ribonucleic protein (snRNP) complex. The binding affinity of U1 snRNP to the 5' splice site is increased in a sequence-selective manner, discrete from constitutive recognition. This new mechanism demonstrates the feasibility of small molecule-mediated, sequence-selective splice modulation and the potential for leveraging this strategy in other splicing diseases.


Asunto(s)
Empalme Alternativo , Atrofia Muscular Espinal/tratamiento farmacológico , ARN Bicatenario/agonistas , Ribonucleoproteína Nuclear Pequeña U1/agonistas , Bibliotecas de Moléculas Pequeñas/farmacología , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo , Animales , Sitios de Unión , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Modelos Moleculares , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/mortalidad , Atrofia Muscular Espinal/patología , Unión Proteica/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Proteolisis , Precursores del ARN/agonistas , Precursores del ARN/química , Precursores del ARN/metabolismo , ARN Bicatenario/química , ARN Bicatenario/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/química , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/metabolismo , Análisis de Supervivencia , Proteína 2 para la Supervivencia de la Neurona Motora/química , Proteína 2 para la Supervivencia de la Neurona Motora/genética
2.
Nat Chem Biol ; 14(12): 1068-1069, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30323218

Asunto(s)
Empalme del ARN , Exones
3.
FASEB J ; 27(5): 1820-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23325320

RESUMEN

Most neurodegenerative diseases are linked to aberrant accumulation of aggregation-prone proteins. Among them, Huntington's disease (HD) is caused by an expanded polyglutamine repeat stretch in the N terminus of the mutant huntingtin protein (mHTT), which gets cleaved and aggregates in the brain. Recently established human induced pluripotent stem cell-derived HD neurons exhibit some disease-relevant phenotypes and provide tools for HD research. However, they have limitations such as genetic heterogeneity and an absence of mHTT aggregates and lack a robust neurodegeneration phenotype. In addition, the relationship between the phenotype and mHTT levels has not been elucidated. Herein, we present a human embryonic stem cell (hESC)-derived HD neuronal model expressing HTTexon1 fragments, which addresses the deficiencies enumerated above. The wild-type and HD lines are derived from an isogenic background and exhibit insoluble mHTT aggregates and neurodegeneration. We also demonstrate a quantitative relationship between neurodegeneration and soluble monomeric (but not oligomeric or aggregated) mHTT levels. Reduction of ~10% of mHTT is sufficient to prevent toxicity, whereas ~90% reduction of wild-type HTT is safe and well-tolerated in these cells. A known HD toxicity modifier (Rhes) showed expected rescue of neurodegeneration. Therefore, the hESC-derived neuronal models complement existing induced pluripotent stem cell-derived neuronal models and provide valuable tools for HD research.


Asunto(s)
Células Madre Embrionarias/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Proteínas del Tejido Nervioso/genética , Línea Celular , Proteínas de Unión al GTP/genética , Técnicas de Silenciamiento del Gen , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Modelos Biológicos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/metabolismo , Neuronas/patología , Transfección
6.
Nat Commun ; 10(1): 137, 2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30635584

RESUMEN

Dysregulation of RNA splicing by spliceosome mutations or in cancer genes is increasingly recognized as a hallmark of cancer. Small molecule splicing modulators have been introduced into clinical trials to treat solid tumors or leukemia bearing recurrent spliceosome mutations. Nevertheless, further investigation of the molecular mechanisms that may enlighten therapeutic strategies for splicing modulators is highly desired. Here, using unbiased functional approaches, we report that the sensitivity to splicing modulation of the anti-apoptotic BCL2 family genes is a key mechanism underlying preferential cytotoxicity induced by the SF3b-targeting splicing modulator E7107. While BCL2A1, BCL2L2 and MCL1 are prone to splicing perturbation, BCL2L1 exhibits resistance to E7107-induced splicing modulation. Consequently, E7107 selectively induces apoptosis in BCL2A1-dependent melanoma cells and MCL1-dependent NSCLC cells. Furthermore, combination of BCLxL (BCL2L1-encoded) inhibitors and E7107 remarkably enhances cytotoxicity in cancer cells. These findings inform mechanism-based approaches to the future clinical development of splicing modulators in cancer treatment.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Antígenos de Histocompatibilidad Menor/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Empalme del ARN/efectos de los fármacos , Proteína bcl-X/genética , Células A549 , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Doxiciclina/farmacología , Sinergismo Farmacológico , Compuestos Epoxi/farmacología , Femenino , Humanos , Neoplasias Pulmonares/genética , Macrólidos/farmacología , Melanoma/genética , Ratones , Ratones Desnudos , Interferencia de ARN , Empalme del ARN/genética , ARN Interferente Pequeño/genética , Empalmosomas/efectos de los fármacos , Empalmosomas/genética , Secuenciación del Exoma , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cell Rep ; 23(1): 282-296.e4, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29617667

RESUMEN

Hotspot mutations in splicing factor genes have been recently reported at high frequency in hematological malignancies, suggesting the importance of RNA splicing in cancer. We analyzed whole-exome sequencing data across 33 tumor types in The Cancer Genome Atlas (TCGA), and we identified 119 splicing factor genes with significant non-silent mutation patterns, including mutation over-representation, recurrent loss of function (tumor suppressor-like), or hotspot mutation profile (oncogene-like). Furthermore, RNA sequencing analysis revealed altered splicing events associated with selected splicing factor mutations. In addition, we were able to identify common gene pathway profiles associated with the presence of these mutations. Our analysis suggests that somatic alteration of genes involved in the RNA-splicing process is common in cancer and may represent an underappreciated hallmark of tumorigenesis.


Asunto(s)
Tasa de Mutación , Neoplasias/genética , Factores de Empalme de ARN/genética , Línea Celular Tumoral , Genes Supresores de Tumor , Humanos , Mutación con Pérdida de Función , Neoplasias/clasificación , Oncogenes , Empalme del ARN/genética
8.
Cell Syst ; 7(1): 28-40.e4, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-29936182

RESUMEN

Discriminating transcriptional changes that drive disease pathogenesis from nonpathogenic and compensatory responses is a daunting challenge. This is particularly true for neurodegenerative diseases, which affect the expression of thousands of genes in different brain regions at different disease stages. Here we integrate functional testing and network approaches to analyze previously reported transcriptional alterations in the brains of Huntington disease (HD) patients. We selected 312 genes whose expression is dysregulated both in HD patients and in HD mice and then replicated and/or antagonized each alteration in a Drosophila HD model. High-throughput behavioral testing in this model and controls revealed that transcriptional changes in synaptic biology and calcium signaling are compensatory, whereas alterations involving the actin cytoskeleton and inflammation drive disease. Knockdown of disease-driving genes in HD patient-derived cells lowered mutant Huntingtin levels and activated macroautophagy, suggesting a mechanism for mitigating pathogenesis. Our multilayered approach can thus untangle the wealth of information generated by transcriptomics and identify early therapeutic intervention points.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Enfermedad de Huntington/genética , Animales , Encéfalo/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Fibroblastos/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Enfermedad de Huntington/fisiopatología , Células Madre Pluripotentes Inducidas , Masculino , Transcriptoma/genética
9.
Cancer Cell ; 34(2): 225-241.e8, 2018 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-30107174

RESUMEN

Mutations affecting RNA splicing factors are the most common genetic alterations in myelodysplastic syndrome (MDS) patients and occur in a mutually exclusive manner. The basis for the mutual exclusivity of these mutations and how they contribute to MDS is not well understood. Here we report that although different spliceosome gene mutations impart distinct effects on splicing, they are negatively selected for when co-expressed due to aberrant splicing and downregulation of regulators of hematopoietic stem cell survival and quiescence. In addition to this synthetic lethal interaction, mutations in the splicing factors SF3B1 and SRSF2 share convergent effects on aberrant splicing of mRNAs that promote nuclear factor κB signaling. These data identify shared consequences of splicing-factor mutations and the basis for their mutual exclusivity.


Asunto(s)
Mutación , Neoplasias/genética , Empalmosomas , Animales , Caspasa 8/genética , Femenino , Hematopoyesis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/fisiología , Fosfoproteínas/genética , Factores de Empalme de ARN/genética , Factores de Empalme Serina-Arginina/genética
10.
Nat Commun ; 8: 15522, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28541300

RESUMEN

Pladienolide, herboxidiene and spliceostatin have been identified as splicing modulators that target SF3B1 in the SF3b subcomplex. Here we report that PHF5A, another component of this subcomplex, is also targeted by these compounds. Mutations in PHF5A-Y36, SF3B1-K1071, SF3B1-R1074 and SF3B1-V1078 confer resistance to these modulators, suggesting a common interaction site. RNA-seq analysis reveals that PHF5A-Y36C has minimal effect on basal splicing but inhibits the global action of splicing modulators. Moreover, PHF5A-Y36C alters splicing modulator-induced intron-retention/exon-skipping profile, which correlates with the differential GC content between adjacent introns and exons. We determine the crystal structure of human PHF5A demonstrating that Y36 is located on a highly conserved surface. Analysis of the cryo-EM spliceosome Bact complex shows that the resistance mutations cluster in a pocket surrounding the branch point adenosine, suggesting a competitive mode of action. Collectively, we propose that PHF5A-SF3B1 forms a central node for binding to these splicing modulators.


Asunto(s)
Adenosina/química , Empalme Alternativo , Proteínas Portadoras/química , Fosfoproteínas/química , Factores de Empalme de ARN/química , Proliferación Celular , Supervivencia Celular , Microscopía por Crioelectrón , Cristalografía por Rayos X , Compuestos Epoxi/química , Exones , Alcoholes Grasos/química , Células HCT116 , Humanos , Intrones , Macrólidos/química , Espectrometría de Masas , Mutagénesis Sitio-Dirigida , Mutación , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/química , Fosfoproteínas/metabolismo , Unión Proteica , Conformación Proteica , Piranos/química , Interferencia de ARN , Factores de Empalme de ARN/metabolismo , Proteínas de Unión al ARN , Proteínas Recombinantes/química , Análisis de Secuencia de ARN , Compuestos de Espiro/química , Empalmosomas/metabolismo , Transactivadores
11.
Nat Med ; 22(6): 672-8, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27135740

RESUMEN

Mutations in genes encoding splicing factors (which we refer to as spliceosomal genes) are commonly found in patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). These mutations recurrently affect specific amino acid residues, leading to perturbed normal splice site and exon recognition. Spliceosomal gene mutations are always heterozygous and rarely occur together with one another, suggesting that cells may tolerate only a partial deviation from normal splicing activity. To test this hypothesis, we engineered mice to express a mutated allele of serine/arginine-rich splicing factor 2 (Srsf2(P95H))-which commonly occurs in individuals with MDS and AML-in an inducible, hemizygous manner in hematopoietic cells. These mice rapidly succumbed to fatal bone marrow failure, demonstrating that Srsf2-mutated cells depend on the wild-type Srsf2 allele for survival. In the context of leukemia, treatment with the spliceosome inhibitor E7107 (refs. 7,8) resulted in substantial reductions in leukemic burden, specifically in isogenic mouse leukemias and patient-derived xenograft AMLs carrying spliceosomal mutations. Whereas E7107 treatment of mice resulted in widespread intron retention and cassette exon skipping in leukemic cells regardless of Srsf2 genotype, the magnitude of splicing inhibition following E7107 treatment was greater in Srsf2-mutated than in Srsf2-wild-type leukemia, consistent with the differential effect of E7107 on survival. Collectively, these data provide genetic and pharmacologic evidence that leukemias with spliceosomal gene mutations are preferentially susceptible to additional splicing perturbations in vivo as compared to leukemias without such mutations. Modulation of spliceosome function may thus provide a new therapeutic avenue in genetically defined subsets of individuals with MDS or AML.


Asunto(s)
Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/genética , Factores de Empalme Serina-Arginina/genética , Empalmosomas/genética , Anemia Aplásica/genética , Animales , Enfermedades de la Médula Ósea/genética , Trastornos de Fallo de la Médula Ósea , Trasplante de Médula Ósea , Catálisis , Línea Celular Tumoral , Compuestos Epoxi/farmacología , Citometría de Flujo , Técnicas de Sustitución del Gen , Hemicigoto , Hemoglobinuria Paroxística/genética , Humanos , Macrólidos/farmacología , Ratones , Ratones Noqueados , Mutación , Trasplante de Neoplasias , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Elife ; 42015 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-25738228

RESUMEN

Huntington's disease (HD) represents an important model for neurodegenerative disorders and proteinopathies. It is mainly caused by cytotoxicity of the mutant huntingtin protein (Htt) with an expanded polyQ stretch. While Htt is ubiquitously expressed, HD is characterized by selective neurodegeneration of the striatum. Here we report a striatal-enriched orphan G protein-coupled receptor(GPCR) Gpr52 as a stabilizer of Htt in vitro and in vivo. Gpr52 modulates Htt via cAMP-dependent but PKA independent mechanisms. Gpr52 is located within an intron of Rabgap1l, which exhibits epistatic effects on Gpr52-mediated modulation of Htt levels by inhibiting its substrate Rab39B, which co-localizes with Htt and translocates Htt to the endoplasmic reticulum. Finally, reducing Gpr52 suppresses HD phenotypes in both patient iPS-derived neurons and in vivo Drosophila HD models. Thus, our discovery reveals modulation of Htt levels by a striatal-enriched GPCR via its GPCR function, providing insights into the selective neurodegeneration and potential treatment strategies.


Asunto(s)
Cuerpo Estriado/metabolismo , Enfermedad de Huntington/metabolismo , Intrones , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Animales , Animales Modificados Genéticamente , Western Blotting , Células Cultivadas , Cuerpo Estriado/citología , Modelos Animales de Enfermedad , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Femenino , Humanos , Enfermedad de Huntington/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Actividad Motora/genética , Actividad Motora/fisiología , Mutación , Desempeño Psicomotor/fisiología , Interferencia de ARN , Receptores Acoplados a Proteínas G/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
13.
Trends Pharmacol Sci ; 35(2): 53-62, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24388390

RESUMEN

Classical targeted drug discovery is based on targeting druggable targets, typically kinases and receptors of which the function can be agonized or antagonized. This strategy meets difficulties in cases such as Huntington's disease (HD) and similar neurodegenerative disorders, where the pathological function of the protein causing the disease is not clear. HD is caused by mutant HTT protein (mHTT) containing an expanded polyglutamine (polyQ) stretch, but the function of mHTT and how mHTT causes HD are unknown, thus preventing efforts to screen for mHTT 'inhibitors'. However, HD is appealing for drug discovery because the genetic mutation is clear, as compared with other major neurodegenerative disorders. Although mHTT is not a conventional 'druggable' target, one approach that appears promising is lowering its level, which might be applicable to other neurodegenerative disorders and proteinopathies linked to aberrant accumulation of proteins. Here we review mHTT lowering strategies that might provide promising avenues for drugging such diseases.


Asunto(s)
Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/metabolismo , Animales , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Terapia Molecular Dirigida , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo
14.
Nat Neurosci ; 16(5): 562-70, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23525043

RESUMEN

Huntington's disease is caused by expanded CAG repeats in HTT, conferring toxic gain of function on mutant HTT (mHTT) protein. Reducing mHTT amounts is postulated as a strategy for therapeutic intervention. We conducted genome-wide RNA interference screens for genes modifying mHTT abundance and identified 13 hits. We tested 10 in vivo in a Drosophila melanogaster Huntington's disease model, and 6 exhibited activity consistent with the in vitro screening results. Among these, negative regulator of ubiquitin-like protein 1 (NUB1) overexpression lowered mHTT in neuronal models and rescued mHTT-induced death. NUB1 reduces mHTT amounts by enhancing polyubiquitination and proteasomal degradation of mHTT protein. The process requires CUL3 and the ubiquitin-like protein NEDD8 necessary for CUL3 activation. As a potential approach to modulating NUB1 for treatment, interferon-ß lowered mHTT and rescued neuronal toxicity through induction of NUB1. Thus, we have identified genes modifying endogenous mHTT using high-throughput screening and demonstrate NUB1 as an exemplar entry point for therapeutic intervention of Huntington's disease.


Asunto(s)
Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Células Cultivadas , Proteínas Cullin/metabolismo , Modelos Animales de Enfermedad , Drosophila/efectos de los fármacos , Drosophila/metabolismo , Embrión de Mamíferos , Femenino , Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Proteína Huntingtina , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína NEDD8 , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/toxicidad , Neuronas/efectos de los fármacos , Embarazo , Factores de Transcripción/genética , Ubiquitinas/metabolismo
16.
Mol Neurodegener ; 5: 58, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21156064

RESUMEN

BACKGROUND: The mutation in Huntington's disease is a polyglutamine expansion near the N-terminus of huntingtin. Huntingtin expressed in immortalized neurons is cleaved near the N-terminus to form N-terminal polypeptides known as cleavage products A and B (cpA and cpB). CpA and cpB with polyglutamine expansion form inclusions in the nucleus and cytoplasm, respectively. The formation of cpA and cpB in primary neurons has not been established and the proteases involved in the formation of these fragments are unknown. RESULTS: Delivery of htt cDNA into the mouse striatum using adeno-associated virus or into primary cortical neurons using lentivirus generated cpA and cpB, indicating that neurons in brain and in vitro can form these fragments. A screen of small molecule protease inhibitors introduced to clonal striatal X57 cells and HeLa cells identified compounds that reduced levels of cpA and are inhibitors of the aspartyl proteases cathepsin D and cathepsin E. The most effective compound, P1-N031, is a transition state mimetic for aspartyl proteases. By western blot analysis, cathepsin D was easily detected in clonal striatal X57 cells, mouse brain and primary neurons, whereas cathepsin E was only detectible in clonal striatal X57 cells. In primary neurons, levels of cleavage product A were not changed by the same compounds that were effective in clonal striatal cells or by mRNA silencing to partially reduce levels of cathepsin D. Instead, treating primary neurons with compounds that are known to inhibit gamma secretase activity either indirectly (Imatinib mesylate, Gleevec) or selectively (LY-411,575 or DAPT) reduced levels of cpA. LY-411,575 or DAPT also increased survival of primary neurons expressing endogenous full-length mutant huntingtin. CONCLUSION: We show that cpA and cpB are produced from a larger huntingtin fragment in vivo in mouse brain and in primary neuron cultures. The aspartyl protease involved in forming cpA has cathepsin-D like properties in immortalized neurons and gamma secretase-like properties in primary neurons, suggesting that cell type may be a critical factor that specifies the aspartyl protease responsible for cpA. Since gamma secretase inhibitors were also protective in primary neurons, further study of the role of gamma-secretase activity in HD neurons is justified.

17.
J Neurochem ; 97(6): 1585-99, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16805770

RESUMEN

While transplanted neural stem cells (NSCs) have been shown to hold promise for cell replacement in models of a number of neurological disorders, these examples have typically been under conditions where the host cells become dysfunctional due to a cell autonomous etiology, i.e. a 'sick' cell within a relatively supportive environment. It has long been held that cell replacement in a toxic milieu would not likely be possible; donor cells would succumb in much the same way as endogenous cells had. Many metabolic diseases are characterized by this situation, suggesting that they would be poor targets for cell replacement therapies. On the other hand, models of such diseases could prove ideal for testing the capacity for cell replacement under such challenging conditions. In the twitcher (twi ) mouse -- as in patients with Krabbe or globoid cell leukodystrophy (GLD), for which it serves as an authentic model -- loss of galactocerebrosidase (GalC) activity results in the accumulation of psychosine, a toxic glycolipid. Twi mice, like children with GLD, exhibit inexorable neurological deterioration presumably as a result of dysfunctional and ultimately degenerated oligodendrocytes with loss of myelin. It is believed that GLD pathophysiology is related to a psychosine-filled environment that kills not only host oligodendrocytes but theoretically any new cells placed into that milieu. Through the implantation of NSCs into the brains of both neonatal and juvenile/young adult twi mice, we have determined that widespread oligodendrocyte replacement and remyelination is feasible. NSCs appear to be intrinsically resistant to psychosine -- more so in their undifferentiated state than when directed ex vivo to become oligodendrocytes. This resistance can be enhanced by engineering the NSCs to over-express GalC. Some twi mice grafted with such engineered NSCs had thicker white tracts and lived 2-3 times longer than expected. While their brains had detectable levels of GalC, it was probably more significant that their psychosine levels were lower than in twi mice that died at a younger age. This concept of resistance based on differentiation state extended to human NSCs which could similarly survive within the twi brain. Taken together, these results suggest a number of points regarding cellular therapies against degenerative diseases with a prominent cell non-autonomous component: Cell replacement is possible if cells resistant to the toxic environment are employed. Furthermore, an important aspect of successful treatment will likely be not only cell replacement but also cross-correction of host cells to provide them with enzyme activity and hence resistance. While oligodendrocyte replacement alone was not a sufficient treatment for GLD (even when extensive), the replacement of both cells and molecules -- e.g. with NSCs that could both become oligodendrocytes and 'pumps' for GalC -- emerges as a promising basis for a multidisciplinary strategy. Most neurological disease are complex in this way and will likely require multifaceted approaches, perhaps with NSCs serving as the 'glue'.


Asunto(s)
Galactosilceramidasa/biosíntesis , Leucodistrofia de Células Globoides/cirugía , Neuronas/fisiología , Trasplante de Células Madre/métodos , Células Madre/fisiología , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Galactosilceramidasa/deficiencia , Terapia Genética/métodos , Humanos , Inmunohistoquímica , Leucodistrofia de Células Globoides/patología , Ratones , Ratones Mutantes , Microscopía Electrónica de Transmisión/métodos , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/patología , Vaina de Mielina/ultraestructura , Neuronas/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Oligodendroglía/fisiología , Psicosina/toxicidad , Células Madre/efectos de los fármacos , Transducción Genética/métodos
18.
J Biol Chem ; 279(18): 18614-22, 2004 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-14985362

RESUMEN

Loss-of-function mutations in parkin are the predominant cause of familial Parkinson's disease. We previously reported that parkin-/- mice exhibit nigrostriatal deficits in the absence of nigral degeneration. Parkin has been shown to function as an E3 ubiquitin ligase. Loss of parkin function, therefore, has been hypothesized to cause nigral degeneration via an aberrant accumulation of its substrates. Here we employed a proteomic approach to determine whether loss of parkin function results in alterations in abundance and/or modification of proteins in the ventral midbrain of parkin-/- mice. Two-dimensional gel electrophoresis followed by mass spectrometry revealed decreased abundance of a number of proteins involved in mitochondrial function or oxidative stress. Consistent with reductions in several subunits of complexes I and IV, functional assays showed reductions in respiratory capacity of striatal mitochondria isolated from parkin-/- mice. Electron microscopic analysis revealed no gross morphological abnormalities in striatal mitochondria of parkin-/- mice. In addition, parkin-/- mice showed a delayed rate of weight gain, suggesting broader metabolic abnormalities. Accompanying these deficits in mitochondrial function, parkin-/- mice also exhibited decreased levels of proteins involved in protection from oxidative stress. Consistent with these findings, parkin-/- mice showed decreased serum antioxidant capacity and increased protein and lipid peroxidation. The combination of proteomic, genetic, and physiological analyses reveal an essential role for parkin in the regulation of mitochondrial function and provide the first direct evidence of mitochondrial dysfunction and oxidative damage in the absence of nigral degeneration in a genetic mouse model of Parkinson's disease.


Asunto(s)
Mitocondrias/fisiología , Enfermedades Mitocondriales/genética , Estrés Oxidativo , Ubiquitina-Proteína Ligasas/fisiología , Animales , Antioxidantes , Ganglios Basales/ultraestructura , Respiración de la Célula , Ratones , Ratones Noqueados , Mitocondrias/química , Mitocondrias/metabolismo , Proteínas Mitocondriales/análisis , Fosforilación Oxidativa , Enfermedad de Parkinson/etiología , Procesamiento Proteico-Postraduccional , Especies Reactivas de Oxígeno , Ubiquitina-Proteína Ligasas/genética , Aumento de Peso
19.
J Biol Chem ; 278(44): 43628-35, 2003 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-12930822

RESUMEN

Loss-of-function mutations in parkin are the major cause of early-onset familial Parkinson's disease. To investigate the pathogenic mechanism by which loss of parkin function causes Parkinson's disease, we generated a mouse model bearing a germline disruption in parkin. Parkin-/- mice are viable and exhibit grossly normal brain morphology. Quantitative in vivo microdialysis revealed an increase in extracellular dopamine concentration in the striatum of parkin-/- mice. Intracellular recordings of medium-sized striatal spiny neurons showed that greater currents are required to induce synaptic responses, suggesting a reduction in synaptic excitability in the absence of parkin. Furthermore, parkin-/- mice exhibit deficits in behavioral paradigms sensitive to dysfunction of the nigrostriatal pathway. The number of dopaminergic neurons in the substantia nigra of parkin-/- mice, however, is normal up to the age of 24 months, in contrast to the substantial loss of nigral neurons characteristic of Parkinson's disease. Steady-state levels of CDCrel-1, synphilin-1, and alpha-synuclein, which were identified previously as substrates of the E3 ubiquitin ligase activity of parkin, are unaltered in parkin-/- brains. Together these findings provide the first evidence for a novel role of parkin in dopamine regulation and nigrostriatal function, and a non-essential role of parkin in the survival of nigral neurons in mice.


Asunto(s)
Encéfalo/patología , Dopamina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/fisiología , Alelos , Animales , Conducta Animal , Western Blotting , Encéfalo/metabolismo , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Electrofisiología , Mutación de Línea Germinal , Ratones , Ratones Transgénicos , Modelos Genéticos , Neuronas/metabolismo , Enfermedad de Parkinson/genética , Receptores Dopaminérgicos/metabolismo , Sustancia Negra/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA