Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 3804, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714648

RESUMEN

Messenger RNA (mRNA) therapeutics delivered via lipid nanoparticles hold the potential to treat metabolic diseases caused by protein deficiency, including propionic acidemia (PA), methylmalonic acidemia (MMA), and phenylketonuria (PKU). Herein we report results from multiple independent preclinical studies of mRNA-3927 (an investigational treatment for PA), mRNA-3705 (an investigational treatment for MMA), and mRNA-3210 (an investigational treatment for PKU) in murine models of each disease. All 3 mRNA therapeutics exhibited pharmacokinetic/pharmacodynamic (PK/PD) responses in their respective murine model by driving mRNA, protein, and/or protein activity responses, as well as by decreasing levels of the relevant biomarker(s) when compared to control-treated animals. These preclinical data were then used to develop translational PK/PD models, which were scaled allometrically to humans to predict starting doses for first-in-human clinical studies for each disease. The predicted first-in-human doses for mRNA-3927, mRNA-3705, and mRNA-3210 were determined to be 0.3, 0.1, and 0.4 mg/kg, respectively.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos , Modelos Animales de Enfermedad , Fenilcetonurias , Acidemia Propiónica , ARN Mensajero , Acidemia Propiónica/genética , Acidemia Propiónica/terapia , Acidemia Propiónica/tratamiento farmacológico , Animales , Fenilcetonurias/genética , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/terapia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Errores Innatos del Metabolismo de los Aminoácidos/tratamiento farmacológico , Ratones , Humanos , Masculino , Femenino , Nanopartículas/química , Ratones Endogámicos C57BL , Liposomas
2.
Mol Ther Methods Clin Dev ; 28: 129-145, 2023 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-36654798

RESUMEN

Mutations in GUCY2D are associated with severe early-onset retinal dystrophy, Leber congenital amaurosis type 1 (LCA1), a leading cause of blindness in children. Despite a high degree of visual disturbance stemming from photoreceptor dysfunction, patients with LCA1 largely retain normal photoreceptor structure, suggesting that they are good candidates for gene replacement therapy. The purpose of this study was to conduct the preclinical and IND-enabling experiments required to support clinical application of AAV5-hGRK1-GUCY2D in patients harboring biallelic recessive mutations in GUCY2D. Preclinical studies were conducted in mice to evaluate the effect of vector manufacturing platforms and transgene species on the therapeutic response. Dose-ranging studies were conducted in cynomolgus monkeys to establish the minimum dose required for efficient photoreceptor transduction. Good laboratory practice (GLP) studies evaluated systemic biodistribution in rats and toxicology in non-human primates (NHPs). These results expanded our knowledge of dose response for an AAV5-vectored transgene under control of the human rhodopsin kinase (hGRK1) promoter in NHPs with respect to photoreceptor transduction and safety and, in combination with the rat biodistribution and mouse efficacy studies, informed the design of a first-in-human clinical study in patients with LCA1.

3.
CPT Pharmacometrics Syst Pharmacol ; 7(7): 442-452, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29920993

RESUMEN

Acid sphingomyelinase deficiency (ASMD) is a rare lysosomal storage disorder with heterogeneous clinical manifestations, including hepatosplenomegaly and infiltrative pulmonary disease, and is associated with significant morbidity and mortality. Olipudase alfa (recombinant human acid sphingomyelinase) is an enzyme replacement therapy under development for the non-neurological manifestations of ASMD. We present a quantitative systems pharmacology (QSP) model supporting the clinical development of olipudase alfa. The model is multiscale and mechanistic, linking the enzymatic deficiency driving the disease to molecular-level, cellular-level, and organ-level effects. Model development was informed by natural history, and preclinical and clinical studies. By considering patient-specific pharmacokinetic (PK) profiles and indicators of disease severity, the model describes pharmacodynamic (PD) and clinical end points for individual patients. The ASMD QSP model provides a platform for quantitatively assessing systemic pharmacological effects in adult and pediatric patients, and explaining variability within and across these patient populations, thereby supporting the extrapolation of treatment response from adults to pediatrics.


Asunto(s)
Terapia de Reemplazo Enzimático/métodos , Modelos Biológicos , Enfermedades de Niemann-Pick/terapia , Proteínas Recombinantes/uso terapéutico , Esfingomielina Fosfodiesterasa/genética , Esfingomielina Fosfodiesterasa/uso terapéutico , Animales , Calibración , Humanos , Ratones , Ratones Noqueados , Proteínas Recombinantes/farmacocinética , Esfingomielina Fosfodiesterasa/farmacocinética
4.
Mol Genet Metab Rep ; 8: 48-50, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27489778

RESUMEN

Correlations between angiotensin-converting enzyme (ACE) genotype (I/I, I/D, D/D), disease severity at baseline and response to enzyme replacement therapy (ERT) were assessed in the Pompe disease Late-Onset Treatment Study (LOTS). No correlations were observed between ACE genotype and disease severity at baseline. However, D/D patients appeared to have a reduced response to alglucosidase alfa treatment than I/I or I/D patients, suggesting that ACE polymorphisms may influence the response to alglucosidase alfa treatment and warrants further investigation.

5.
Mol Ther ; 21(7): 1306-15, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23689599

RESUMEN

Salutary responses to adeno-associated viral (AAV) gene therapy have been reported in the mouse model of Sandhoff disease (SD), a neurodegenerative lysosomal storage disease caused by deficiency of ß-N-acetylhexosaminidase (Hex). While untreated mice reach the humane endpoint by 4.1 months of age, mice treated by a single intracranial injection of vectors expressing human hexosaminidase may live a normal life span of 2 years. When treated with the same therapeutic vectors used in mice, two cats with SD lived to 7.0 and 8.2 months of age, compared with an untreated life span of 4.5 ± 0.5 months (n = 11). Because a pronounced humoral immune response to both the AAV1 vectors and human hexosaminidase was documented, feline cDNAs for the hexosaminidase α- and ß-subunits were cloned into AAVrh8 vectors. Cats treated with vectors expressing feline hexosaminidase produced enzymatic activity >75-fold normal at the brain injection site with little evidence of an immune infiltrate. Affected cats treated with feline-specific vectors by bilateral injection of the thalamus lived to 10.4 ± 3.7 months of age (n = 3), or 2.3 times as long as untreated cats. These studies support the therapeutic potential of AAV vectors for SD and underscore the importance of species-specific cDNAs for translational research.


Asunto(s)
Enfermedades de los Gatos/enzimología , Enfermedades de los Gatos/terapia , Enfermedad de Sandhoff/enzimología , Enfermedad de Sandhoff/terapia , beta-N-Acetilhexosaminidasas/metabolismo , Animales , Enfermedades de los Gatos/genética , Gatos , Dependovirus/genética , Modelos Animales de Enfermedad , Terapia Genética/métodos , Vectores Genéticos/genética , Enfermedad de Sandhoff/genética , beta-N-Acetilhexosaminidasas/genética
6.
PLoS One ; 5(10): e13468, 2010 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-20976108

RESUMEN

BACKGROUND: GM1-gangliosidosis is a glycosphingolipid (GSL) lysosomal storage disease caused by a genetic deficiency of acid ß-galactosidase (ßgal), which results in the accumulation of GM1-ganglioside and its asialo-form (GA1) primarily in the CNS. Age of onset ranges from infancy to adulthood, and excessive ganglioside accumulation produces progressive neurodegeneration and psychomotor retardation in humans. Currently, there are no effective therapies for the treatment of GM1-gangliosidosis. METHODOLOGY/PRINCIPAL FINDINGS: In this study we examined the effect of thalamic infusion of AAV2/1-ßgal vector in adult GM1 mice on enzyme distribution, activity, and GSL content in the CNS, motor behavior, and survival. Six to eight week-old GM1 mice received bilateral injections of AAV vector in the thalamus, or thalamus and deep cerebellar nuclei (DCN) with pre-determined endpoints at 1 and 4 months post-injection, and the humane endpoint, or 52 weeks of age. Enzyme activity was elevated throughout the CNS of AAV-treated GM1 mice and GSL storage nearly normalized in most structures analyzed, except in the spinal cord which showed ∼50% reduction compared to age-matched untreated GM1 mice spinal cord. Survival was significantly longer in AAV-treated GM1 mice (52 wks) than in untreated mice. However the motor performance of AAV-treated GM1 mice declined over time at a rate similar to that observed in untreated GM1 mice. CONCLUSIONS/SIGNIFICANCE: Our studies show that the AAV-modified thalamus can be used as a 'built-in' central node network for widespread distribution of lysosomal enzymes in the mouse cerebrum. In addition, this study indicates that thalamic delivery of AAV vectors should be combined with additional targets to supply the cerebellum and spinal cord with therapeutic levels of enzyme necessary to achieve complete correction of the neurological phenotype in GM1 mice.


Asunto(s)
Dependovirus/genética , Gangliosidosis GM1/terapia , Vectores Genéticos , Transfección , Animales , Cromatografía en Capa Delgada , Potenciales Evocados Visuales , Gangliosidosis GM1/genética , Ratones , Reacción en Cadena de la Polimerasa , Prueba de Desempeño de Rotación con Aceleración Constante , Análisis de Supervivencia
7.
J Neurochem ; 113(6): 1525-35, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20374428

RESUMEN

Sandhoff disease is an autosomal recessive, neurodegenerative disease involving the storage of brain ganglioside GM2 and asialo-GM2. Previous studies showed that caloric restriction, which augments longevity, and N-butyldeoxynojirimycin (NB-DNJ, Miglustat), an imino sugar that hinders the glucosyltransferase catalyzing the first step in glycosphingolipid biosynthesis, both increase longevity and improve motor behavior in the beta-hexosaminidase (Hexb) knockout (-/-) murine model of Sandhoff disease. In this study, we used a restricted ketogenic diet (KD-R) and NB-DNJ to combat ganglioside accumulation. Adult Hexb-/- mice were placed into one of the following groups: (i) a standard diet (SD), (ii) a SD with NB-DNJ (SD + NB-DNJ), (iii) a KD-R, and (iv) a KD-R with NB-DNJ (KD-R + NB-DNJ). Forebrain GM2 content (mug sialic acid/100 mg dry wt) in the four groups was 375 +/- 15, 312 +/- 8, 340 +/- 28, and 279 +/- 26, respectively, indicating an additive interaction between NB-DNJ and the KD-R. Most interestingly, brain NB-DNJ content was 3.5-fold greater in the KD-R + NB-DNJ mice than in the SD + NB-DNJ mice. These data suggest that the KD-R and NB-DNJ may be a potential combinatorial therapy for Sandhoff disease by enhancing NB-DNJ delivery to the brain and may allow lower dosing to achieve the same degree of efficacy as high dose monotherapy.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Encéfalo/efectos de los fármacos , Dieta Cetogénica/métodos , Gangliósido G(M2)/metabolismo , Enfermedad de Sandhoff , 1-Desoxinojirimicina/uso terapéutico , Ácido 3-Hidroxibutírico/sangre , Análisis de Varianza , Animales , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Encéfalo/citología , Encéfalo/metabolismo , Cromatografía Líquida de Alta Presión/métodos , Cromatografía en Capa Delgada/métodos , Ingestión de Alimentos/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Ratones Noqueados , Vaina de Mielina/metabolismo , Células de Purkinje/metabolismo , Células de Purkinje/patología , Enfermedad de Sandhoff/dietoterapia , Enfermedad de Sandhoff/tratamiento farmacológico , Enfermedad de Sandhoff/patología , beta-N-Acetilhexosaminidasas/deficiencia , beta-N-Acetilhexosaminidasas/genética
8.
Lipids ; 44(3): 197-205, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19034545

RESUMEN

Sandhoff disease (SD) is a glycosphingolipid (GSL) storage disease that arises from an autosomal recessive mutation in the gene for the beta-subunit of beta-Hexosaminidase A (Hexb gene), which catabolizes ganglioside GM2 within lysosomes. Accumulation of GM2 and asialo-GM2 (GA2) occurs primarily in the CNS, leading to neurodegeneration and brain dysfunction. We analyzed the total lipids in the brains of SD mice, cats, and humans. GM2 and GA2 were mostly undetectable in the normal mouse, cat, and human brain. The lipid abnormalities in the SD cat brain were generally intermediate to those observed in the SD mouse and the SD human brains. GM2 comprised 38, 67, and 87% of the total brain ganglioside distribution in the SD mice, cats, and humans, respectively. The ratio of GA2-GM2 was 0.93, 0.13, and 0.27 in the SD mice, cats, and humans, respectively, suggesting that the relative storage of GA2 is greater in the SD mouse than in the SD cat or human. Finally, the myelin-enriched lipids, cerebrosides and sulfatides, were significantly lower in the SD brains than in the control brains. This study is the first comparative analysis of brain lipids in mice, cats, and humans with SD and will be important for designing therapies for Sandhoff disease patients.


Asunto(s)
Encéfalo/metabolismo , Enfermedad de Sandhoff/metabolismo , Animales , Gatos , Cromatografía en Capa Delgada , Gangliósidos/metabolismo , Hexosaminidasa B/genética , Humanos , Ratones , Enfermedad de Sandhoff/genética
9.
J Lipid Res ; 49(5): 929-38, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18287616

RESUMEN

Gangliosides are sialic acid-containing glycosphingolipids that have long been associated with tumor malignancy and metastasis. Mounting evidence suggests that gangliosides also modulate tumor angiogenesis. Tumor cells shed gangliosides into the microenvironment, which produces both autocrine and paracrine effects on tumor cells and tumor-associated host cells. In this study, we show that the simple monosialoganglioside GM3 counteracts the proangiogenic effects of vascular endothelial growth factor (VEGF) and of the complex disialoganglioside GD1a. GM3 suppressed the action of VEGF and GD1a on the proliferation of human umbilical vein endothelial cells (HUVECs) and inhibited the migration of HUVECs toward VEGF as a chemoattractant. Enrichment of added GM3 in the HUVEC membrane also reduced the phosphorylation of vascular endothelial growth factor receptor 2 (VEGFR-2) and downstream Akt. Moreover, GM3 reduced the proangiogenic effects of GD1a and growth factors in the in vivo Matrigel plug assay. Inhibition of GM3 biosynthesis with the glucosyl transferase inhibitor, N-butyldeoxynojirimycin (NB-DNJ), increased HUVEC proliferation and the phosphorylation of VEGFR-2 and Akt. The effects of NB-DNJ on HUVECs were reversed with the addition of GM3. We conclude that GM3 has antiangiogenic action and may possess therapeutic potential for reducing tumor angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Gangliósido G(M1)/análogos & derivados , Gangliósido G(M3)/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Encéfalo , Bovinos , División Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Gangliósido G(M1)/antagonistas & inhibidores , Gangliósido G(M1)/farmacología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Venas Umbilicales/citología , Venas Umbilicales/efectos de los fármacos , Venas Umbilicales/fisiología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
10.
Neurochem Int ; 52(6): 1125-33, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18207611

RESUMEN

Sandhoff disease involves the CNS accumulation of ganglioside GM2 and asialo-GM2 (GA2) due to inherited defects in the beta-subunit gene of beta-hexosaminidase A and B (Hexb gene). Accumulation of these glycosphingolipids (GSLs) produces progressive neurodegeneration, ultimately leading to death. Substrate reduction therapy (SRT) aims to decrease the rate of glycosphingolipid (GSL) biosynthesis to compensate for the impaired rate of catabolism. The imino sugar, N-butyldeoxygalactonojirimycin (NB-DGJ) inhibits the first committed step in GSL biosynthesis. NB-DGJ treatment, administered from postnatal day 2 (p-2) to p-5 (600 mg/kg/day)), significantly reduced total brain ganglioside and GM2 content in the Sandhoff disease (Hexb(-/-)) mice, but did not reduce the content of GA2. We also found that NB-DGJ treatment caused a slight, but significant elevation in brain sialidase activity. The drug had no adverse effects on viability, body weight, brain weight, or brain water content in the mice. No significant alterations in neutral lipids or acidic phospholipids were observed in the NB-DGJ-treated Hexb(-/-) mice. Our results show that NB-DGJ is effective in reducing total brain ganglioside and GM2 content at early neonatal ages.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Encéfalo/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Gangliósido G(M2)/metabolismo , Enfermedad de Sandhoff/tratamiento farmacológico , Enfermedad de Sandhoff/metabolismo , 1-Desoxinojirimicina/farmacología , 1-Desoxinojirimicina/uso terapéutico , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Encéfalo/fisiopatología , Química Encefálica/efectos de los fármacos , Química Encefálica/fisiología , Modelos Animales de Enfermedad , Regulación hacia Abajo/fisiología , Gangliósido G(M2)/genética , Gangliósidos/metabolismo , Glicoesfingolípidos/metabolismo , Ratones , Ratones Noqueados , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/fisiopatología , Degeneración Nerviosa/prevención & control , Neuraminidasa/efectos de los fármacos , Neuraminidasa/metabolismo , Enfermedad de Sandhoff/fisiopatología , Resultado del Tratamiento , Cadena alfa de beta-Hexosaminidasa/genética
11.
J Negat Results Biomed ; 6: 8, 2007 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-17623103

RESUMEN

BACKGROUND: Sandhoff disease is an inherited lysosomal storage disease caused by a mutation in the gene for the beta-subunit (Hexb gene) of beta-hexosaminidase A (alphabeta) and B (beta beta). The beta-subunit together with the GM2 activator protein catabolize ganglioside GM2. This enzyme deficiency results in GM2 accumulation primarily in the central nervous system. To investigate how abnormal GM2 catabolism affects the peripheral nervous system in a mouse model of Sandhoff disease (Hexb-/-), we examined the electrophysiology of dissected sciatic nerves, structure of central and peripheral myelin, and lipid composition of the peripheral nervous system. RESULTS: We detected no significant difference in signal impulse conduction velocity or any consistent change in the frequency-dependent conduction slowing and failure between freshly dissected sciatic nerves from the Hexb+/- and Hexb-/- mice. The low-angle x-ray diffraction patterns from freshly dissected sciatic and optic nerves of Hexb+/- and Hexb-/- mice showed normal myelin periods; however, Hexb-/- mice displayed a approximately 10% decrease in the relative amount of compact optic nerve myelin, which is consistent with the previously established reduction in myelin-enriched lipids (cerebrosides and sulfatides) in brains of Hexb-/- mice. Finally, analysis of lipid composition revealed that GM2 content was present in the sciatic nerve of the Hexb-/- mice (undetectable in Hexb+/-). CONCLUSION: Our findings demonstrate the absence of significant functional, structural, or compositional abnormalities in the peripheral nervous system of the murine model for Sandhoff disease, but do show the potential value of integrating multiple techniques to evaluate myelin structure and function in nervous system disorders.


Asunto(s)
Modelos Animales de Enfermedad , Lípidos/análisis , Vaina de Mielina/química , Conducción Nerviosa , Sistema Nervioso Periférico/fisiopatología , Enfermedad de Sandhoff/fisiopatología , Animales , Ratones , Ratones Noqueados , Ratones Transgénicos , Estructura Molecular , Difracción de Rayos X
12.
Nat Med ; 13(4): 439-47, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17351625

RESUMEN

Intracranial transplantation of neural stem cells (NSCs) delayed disease onset, preserved motor function, reduced pathology and prolonged survival in a mouse model of Sandhoff disease, a lethal gangliosidosis. Although donor-derived neurons were electrophysiologically active within chimeric regions, the small degree of neuronal replacement alone could not account for the improvement. NSCs also increased brain beta-hexosaminidase levels, reduced ganglioside storage and diminished activated microgliosis. Additionally, when oral glycosphingolipid biosynthesis inhibitors (beta-hexosaminidase substrate inhibitors) were combined with NSC transplantation, substantial synergy resulted. Efficacy extended to human NSCs, both to those isolated directly from the central nervous system (CNS) and to those derived secondarily from embryonic stem cells. Appreciating that NSCs exhibit a broad repertoire of potentially therapeutic actions, of which neuronal replacement is but one, may help in formulating rational multimodal strategies for the treatment of neurodegenerative diseases.


Asunto(s)
Encéfalo/citología , Células Madre Embrionarias/citología , Neuronas/citología , Enfermedad de Sandhoff/terapia , Trasplante de Células Madre , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacología , Animales , Humanos , Inmunohistoquímica , Ratones , Ratones Noqueados , Microglía/metabolismo , Técnicas de Placa-Clamp , Enfermedad de Sandhoff/tratamiento farmacológico , beta-N-Acetilhexosaminidasas/antagonistas & inhibidores , beta-N-Acetilhexosaminidasas/genética , beta-N-Acetilhexosaminidasas/metabolismo
13.
J Cell Biol ; 172(3): 469-78, 2006 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-16449196

RESUMEN

The central nervous system (CNS) of terrestrial vertebrates underwent a prominent molecular change when a tetraspan membrane protein, myelin proteolipid protein (PLP), replaced the type I integral membrane protein, P0, as the major protein of myelin. To investigate possible reasons for this molecular switch, we genetically engineered mice to express P0 instead of PLP in CNS myelin. In the absence of PLP, the ancestral P0 provided a periodicity to mouse compact CNS myelin that was identical to mouse PNS myelin, where P0 is the major structural protein today. The PLP-P0 shift resulted in reduced myelin internode length, degeneration of myelinated axons, severe neurological disability, and a 50% reduction in lifespan. Mice with equal amounts of P0 and PLP in CNS myelin had a normal lifespan and no axonal degeneration. These data support the hypothesis that the P0-PLP shift during vertebrate evolution provided a vital neuroprotective function to myelin-forming CNS glia.


Asunto(s)
Sistema Nervioso Central/metabolismo , Evolución Molecular , Proteína P0 de la Mielina/genética , Proteína Proteolipídica de la Mielina/genética , Vaina de Mielina/metabolismo , 2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Axones/metabolismo , Axones/patología , Moléculas de Adhesión Celular Neuronal/metabolismo , Sistema Nervioso Central/patología , Sistema Nervioso Central/fisiopatología , Prueba de Esfuerzo , Expresión Génica/genética , Longevidad/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica , Actividad Motora/genética , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , Proteína P0 de la Mielina/metabolismo , Proteína Proteolipídica de la Mielina/metabolismo , Vaina de Mielina/patología , Vaina de Mielina/ultraestructura , Glicoproteína Asociada a Mielina , Oligodendroglía/metabolismo , Nervio Óptico/química , Hidrolasas Diéster Fosfóricas/metabolismo , Regiones Promotoras Genéticas/genética , Receptores de Superficie Celular/metabolismo , Canales de Sodio/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología , Análisis de Supervivencia , Difracción de Rayos X
14.
J Neuropathol Exp Neurol ; 64(11): 976-90, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16254492

RESUMEN

Peripheral neuropathies often result in abnormalities in the structure of internodal myelin, including changes in period and membrane packing, as observed by electron microscopy (EM). Mutations in the gene that encodes the major adhesive structural protein of internodal myelin in the peripheral nervous system of humans and mice--P0 glycoprotein--correlate with these defects. The mechanisms by which P0 mutations interfere with myelin packing and stability are not well understood and cannot be provided by EM studies that give static and qualitative information on fixed material. To gain insights into the pathogenesis of mutant P0, we used x-ray diffraction, which can detect more subtle and dynamic changes in native myelin, to investigate myelin structure in sciatic nerves from murine models of hereditary neuropathies. We used mice with disruption of one or both copies of the P0 gene (models of Charcot-Marie-Tooth-like neuropathy [CMT1B] or Dejerine-Sottas-like neuropathy) and mice with a CMT1B resulting from a transgene encoding P0 with an amino terminal myc-tag. To directly test the structural role of P0, we also examined a mouse that expresses P0 instead of proteolipid protein in central nervous system myelin. To link our findings on unfixed nerves with EM results, we analyzed x-ray patterns from unembedded, aldehyde-fixed nerves and from plastic-embedded nerves. From the x-ray patterns recorded from whole nerves, we assessed the amount of myelin and its quality (i.e. relative thickness and regularity). Among sciatic nerves having different levels of P0, we found that unfixed nerves and, to a lesser extent, fixed but unembedded nerves gave diffraction patterns of sufficient quality to distinguish periods, sometimes differing by a few Angstroms. Certain packing abnormalities were preserved qualitatively by aldehyde fixation, and the relative amount and structural integrity of myelin among nerves could be distinguished. Measurements from the same nerve over time showed that the amount of P0 affected myelin's stability against swelling, thus directly supporting the hypothesis that packing defects underlie instability in "live" or intact myelin. Our findings demonstrate that diffraction can provide a quantitative basis for understanding, at a molecular level, the membrane packing defects that occur in internodal myelin in demyelinating peripheral neuropathies.


Asunto(s)
Trastornos Heredodegenerativos del Sistema Nervioso/metabolismo , Proteínas de la Mielina/química , Proteínas de la Mielina/metabolismo , Enfermedades del Sistema Nervioso Periférico/metabolismo , Aldehídos/farmacología , Animales , Modelos Animales de Enfermedad , Microanálisis por Sonda Electrónica/métodos , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Ratones , Ratones Transgénicos , Modelos Moleculares , Proteína P0 de la Mielina/genética , Proteína P0 de la Mielina/metabolismo , Proteínas de la Mielina/efectos de los fármacos , Proteínas de la Mielina/ultraestructura , Nervio Óptico/efectos de los fármacos , Nervio Óptico/metabolismo , Nervio Óptico/patología , Nervio Óptico/ultraestructura , Enfermedades del Sistema Nervioso Periférico/genética , Nervio Ciático/efectos de los fármacos , Nervio Ciático/metabolismo , Nervio Ciático/patología , Nervio Ciático/ultraestructura , Factores de Tiempo
15.
J Biol Chem ; 277(37): 34349-58, 2002 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-12068006

RESUMEN

GM3 ganglioside inhibits tetraspanin CD9-facilitated cell motility in various cell lines (Ono, M., Handa, K., Sonnino, S., Withers, D. A., Nagai, H., and Hakomori, S. (2001) Biochemistry 40, 6414-6421). We now report the following: (i) CD9 has the novel feature of being soluble in chloroform/methanol, and classifiable as "proteolipid"; (ii) CD9 and alpha(3) integrin were concentrated together in the low-density glycolipid-enriched microdomain (GEM) of ldlD/CD9 cells, and the alpha(3) expression ratio (value for cells grown under +Gal condition divided by the value for cells grown under -Gal condition) in GEM of ldlD/CD9 cells was higher than that in control ldlD/moc cells, suggesting that CD9 recruits alpha(3) in GEM under +Gal condition, whereby GM3 is present. (iii) Chemical levels of alpha(3) and CD9 in the total extract or membrane fractions from cells grown under +Gal versus -Gal condition were nearly identical, whereas alpha(3) expressed at the cell surface, probed by antibody binding in flow cytometry, was higher under -Gal than +Gal condition. These results suggest that GM3 synthesized under +Gal condition promotes interaction of alpha(3) with CD9, which restricts alpha(3) binding to its antibody. A concept of the alpha(3)/CD9 interaction promoted by GM3 was further supported by (i) co-immunoprecipitation of CD9 and alpha(3) under +Gal but not -Gal condition, (ii) enhanced co-immunoprecipitation of CD9 and alpha(3) when GM3 was added exogenously to cells under -Gal condition, and (iii) the co-localization images of CD9 with alpha(3) and of GM3 with CD9 in fluorescence laser scanning confocal microscopy. Based on the promotion of alpha(3)/CD9 interaction by GM3 and the status of laminin-5 as a true ligand for alpha(3), the laminin-5/alpha(3)-dependent motility of ldlD/CD9 cells was found to be greatly enhanced under -Gal condition, but strongly inhibited under +Gal condition. Such a motility difference under +Gal versus -Gal condition was not observed for ldlD/moc cells. The inhibitory effect observed in ldlD/CD9 cells under +Gal condition was reversed upon addition of anti-alpha(3) antibody and is therefore based on interaction between alpha(3), CD9, and GM3 in GEM.


Asunto(s)
Antígenos CD/química , Moléculas de Adhesión Celular/fisiología , Movimiento Celular/fisiología , Gangliósido G(M3)/fisiología , Integrina alfa3/química , Glicoproteínas de Membrana/química , Animales , Antígenos CD/análisis , Células CHO , Cricetinae , Glicosilación , Integrina alfa3/análisis , Glicoproteínas de Membrana/análisis , Pruebas de Precipitina , Tetraspanina 29 , Kalinina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...