Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Curr Gene Ther ; 22(4): 352-365, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35249485

RESUMO

BACKGROUND: GM1 gangliosidosis (GM1) is an autosomal recessive disorder characterized by the deficiency of beta-galactosidase (ß-gal), a ubiquitous lysosomal enzyme that catalyzes the hydrolysis of GM1 ganglioside. OBJECTIVE: The study aims to explore the application of the AAV9-coGLB1 for effective treatment in a GM1 gangliosidosis mutant mouse model. METHODS: We designed a novel adeno-associated virus 9 (AAV9) vector expressing ß-gal (AAV9- coGLB1) to treat GM1 gangliosidosis. The vector, injected via the caudal vein at 4 weeks of age, drove the widespread and sustained expression of ß-gal for up to 32 weeks in the Glb1G455R/G455R mutant mice (GM1 mice). RESULTS: The increased levels of ß-gal reduced the pathological damage occurring in GM1 mice. Histological analyses showed that myelin deficits and neuron-specific pathology were reduced in the cerebral cortex region of AAV9-coGLB1-treated mice. Immunohistochemical staining showed that the accumulation of GM1 ganglioside was also reduced after gene therapy. The reduction of the storage in these regions was accompanied by a decrease in activated microglia. In addition, AAV9 treatment reversed the blockade of autophagic flux in GM1 mice. CONCLUSION: These results show that AAV9-coGLB1 reduces the pathological signs of GM1 gangliosidosis in a mouse model.


Assuntos
Gangliosidose GM1 , Animais , Sistema Nervoso Central , Dependovirus/genética , Modelos Animais de Doenças , Gangliosídeo G(M1) , Gangliosidose GM1/genética , Gangliosidose GM1/metabolismo , Gangliosidose GM1/terapia , Inflamação/genética , Inflamação/terapia , Lisossomos/genética , Lisossomos/patologia , Camundongos
2.
Hum Gene Ther ; 31(21-22): 1169-1177, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33045869

RESUMO

GM1 gangliosidosis is a rare neurodegenerative lysosomal storage disease caused by loss-of-function mutations in the gene encoding beta-galactosidase (ß-gal). There are no approved treatments for GM1 gangliosidosis. Previous studies in animal models have demonstrated that adeno-associated viral (AAV) vector-mediated gene transfer to the brain can restore ß-gal expression and prevent the onset of neurological signs. We developed an optimized AAV vector expressing human ß-gal and evaluated the efficacy of a single intracerebroventricular injection of this vector into the cerebrospinal fluid (CSF) of a murine disease model. The AAV vector administration into the CSF increased ß-gal activity in the brain, reduced neuronal lysosomal storage lesions, prevented the onset of neurological signs and gait abnormalities, and increased survival. These findings demonstrate the potential therapeutic activity of this vector and support its subsequent development for the treatment of GM1 gangliosidosis.


Assuntos
Encéfalo/metabolismo , Líquido Cefalorraquidiano/metabolismo , Dependovirus/genética , Gangliosidose GM1/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , beta-Galactosidase/fisiologia , Animais , Encéfalo/patologia , Líquido Cefalorraquidiano/citologia , Modelos Animais de Doenças , Gangliosidose GM1/genética , Gangliosidose GM1/metabolismo , Gangliosidose GM1/patologia , Vetores Genéticos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , beta-Galactosidase/administração & dosagem , beta-Galactosidase/genética
3.
Mol Ther ; 25(4): 892-903, 2017 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-28236574

RESUMO

GM1 gangliosidosis is a fatal neurodegenerative disease that affects individuals of all ages. Favorable outcomes using adeno-associated viral (AAV) gene therapy in GM1 mice and cats have prompted consideration of human clinical trials, yet there remains a paucity of objective biomarkers to track disease status. We developed a panel of biomarkers using blood, urine, cerebrospinal fluid (CSF), electrodiagnostics, 7 T MRI, and magnetic resonance spectroscopy in GM1 cats-either untreated or AAV treated for more than 5 years-and compared them to markers in human GM1 patients where possible. Significant alterations were noted in CSF and blood of GM1 humans and cats, with partial or full normalization after gene therapy in cats. Gene therapy improved the rhythmic slowing of electroencephalograms (EEGs) in GM1 cats, a phenomenon present also in GM1 patients, but nonetheless the epileptiform activity persisted. After gene therapy, MR-based analyses revealed remarkable preservation of brain architecture and correction of brain metabolites associated with microgliosis, neuroaxonal loss, and demyelination. Therapeutic benefit of AAV gene therapy in GM1 cats, many of which maintain near-normal function >5 years post-treatment, supports the strong consideration of human clinical trials, for which the biomarkers described herein will be essential for outcome assessment.


Assuntos
Biomarcadores , Gangliosidose GM1/genética , Gangliosidose GM1/metabolismo , Terapia Genética , Animais , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Biomarcadores/urina , Gatos , Dependovirus/classificação , Dependovirus/genética , Modelos Animais de Doenças , Eletroencefalografia , Gangliosidose GM1/mortalidade , Gangliosidose GM1/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Hipocalcemia/metabolismo , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Resultado do Tratamento
4.
Chem Commun (Camb) ; 52(32): 5497-515, 2016 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-27043200

RESUMO

Lysosomal storage disorders (LSDs) are often caused by mutations that destabilize native folding and impair the trafficking of enzymes, leading to premature endoplasmic reticulum (ER)-associated degradation, deficiencies of specific hydrolytic functions and aberrant storage of metabolites in the lysosomes. Enzyme replacement therapy (ERT) and substrate reduction therapy (SRT) are available for a few of these conditions, but most remain orphan. A main difficulty is that virtually all LSDs involve neurological decline and neither proteins nor the current SRT drugs can cross the blood-brain barrier. Twenty years ago a new therapeutic paradigm better suited for neuropathic LSDs was launched, namely pharmacological chaperone (PC) therapy. PCs are small molecules capable of binding to the mutant protein at the ER, inducing proper folding, restoring trafficking and increasing enzyme activity and substrate processing in the lysosome. In many LSDs the mutated protein is a glycosidase and the accumulated substrate is an oligo- or polysaccharide or a glycoconjugate, e.g. a glycosphingolipid. Although it might appear counterintuitive, substrate analogues (glycomimetics) behaving as competitive glycosidase inhibitors are good candidates to perform PC tasks. The advancements in the knowledge of the molecular basis of LSDs, including enzyme structures, binding modes, trafficking pathways and substrate processing mechanisms, have been put forward to optimize PC selectivity and efficacy. Moreover, the chemical versatility of glycomimetics and the variety of structures at hand allow simultaneous optimization of chaperone and pharmacokinetic properties. In this Feature Article we review the advancements made in this field in the last few years and the future outlook through the lessons taught by three archetypical LSDs: Gaucher disease, GM1-gangliosidosis and Fabry disease.


Assuntos
Carboidratos/química , Doença de Fabry/tratamento farmacológico , Gangliosidose GM1/tratamento farmacológico , Doença de Gaucher/tratamento farmacológico , Chaperonas Moleculares/uso terapêutico , Mimetismo Molecular , Doença de Fabry/metabolismo , Gangliosidose GM1/metabolismo , Doença de Gaucher/metabolismo , Humanos
5.
Hum Mol Genet ; 24(15): 4353-64, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25964428

RESUMO

GM1 gangliosidosis (GM1) is an autosomal recessive lysosomal storage disease where GLB1 gene mutations result in a reduction or absence of lysosomal acid ß-galactosidase (ßgal) activity. ßgal deficiency leads to accumulation of GM1-ganglioside in the central nervous system (CNS). GM1 is characterized by progressive neurological decline resulting in generalized paralysis, extreme emaciation and death. In this study, we assessed the therapeutic efficacy of an adeno-associated virus (AAV) 9-mßgal vector infused systemically in adult GM1 mice (ßGal(-/-)) at 1 × 10(11) or 3 × 10(11) vector genomes (vg). Biochemical analysis of AAV9-treated GM1 mice showed high ßGal activity in liver and serum. Moderate ßGal levels throughout CNS resulted in a 36-76% reduction in GM1-ganglioside content in the brain and 75-86% in the spinal cord. Histological analyses of the CNS of animals treated with 3 × 10(11) vg dose revealed increased presence of ßgal and clearance of lysosomal storage throughout cortex, hippocampus, brainstem and spinal cord. Storage reduction in these regions was accompanied by a marked decrease in astrogliosis. AAV9 treatment resulted in improved performance in multiple tests of motor function and behavior. Also the majority of GM1 mice in the 3 × 10(11) vg cohort retained ambulation and rearing despite reaching the humane endpoint due to weight loss. Importantly, the median survival of AAV9 treatment groups (316-576 days) was significantly increased over controls (250-264 days). This study shows that moderate widespread expression of ßgal in the CNS of GM1 gangliosidosis mice is sufficient to achieve significant biochemical impact with phenotypic amelioration and extension in lifespan.


Assuntos
Sistema Nervoso Central/metabolismo , Gangliosidose GM1/genética , Terapia Genética , beta-Galactosidase/genética , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Tronco Encefálico/metabolismo , Tronco Encefálico/patologia , Sistema Nervoso Central/patologia , Dependovirus/genética , Modelos Animais de Doenças , Gangliosídeos/metabolismo , Gangliosidose GM1/metabolismo , Gangliosidose GM1/terapia , Vetores Genéticos , Humanos , Camundongos , Medula Espinal/metabolismo , Medula Espinal/patologia , beta-Galactosidase/biossíntese , beta-Galactosidase/sangue
6.
J Lipid Res ; 56(5): 1006-13, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25795792

RESUMO

Bis(monoacylglycero)phosphate (BMP) is a negatively charged glycerophospholipid with an unusual sn-1;sn-1' structural configuration. BMP is primarily enriched in endosomal/lysosomal membranes. BMP is thought to play a role in glycosphingolipid degradation and cholesterol transport. Elevated BMP levels have been found in many lysosomal storage diseases (LSDs), suggesting an association with lysosomal storage material. The gangliosidoses are a group of neurodegenerative LSDs involving the accumulation of either GM1 or GM2 gangliosides resulting from inherited deficiencies in ß-galactosidase or ß-hexosaminidase, respectively. Little information is available on BMP levels in gangliosidosis brain tissue. Our results showed that the content of BMP in brain was significantly greater in humans and in animals (mice, cats, American black bears) with either GM1 or GM2 ganglioside storage diseases, than in brains of normal subjects. The storage of BMP and ganglioside GM2 in brain were reduced similarly following adeno-associated viral-mediated gene therapy in Sandhoff disease mice. We also found that C22:6, C18:0, and C18:1 were the predominant BMP fatty acid species in gangliosidosis brains. The results show that BMP accumulates as a secondary storage material in the brain of a broad range of mammals with gangliosidoses.


Assuntos
Doenças do Gato/metabolismo , Gangliosidose GM1/veterinária , Lisofosfolipídeos/metabolismo , Monoglicerídeos/metabolismo , Doença de Sandhoff/veterinária , Animais , Encéfalo/metabolismo , Gatos , Feminino , Gangliosidose GM1/metabolismo , Humanos , Metabolismo dos Lipídeos , Masculino , Camundongos da Linhagem 129 , Camundongos Knockout , Doença de Sandhoff/metabolismo , Ursidae
7.
Mol Genet Metab ; 114(2): 274-80, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25557439

RESUMO

BACKGROUND: The gangliosidoses (Tay-Sachs disease, Sandhoff disease, and GM1-gangliosidosis) are progressive neurodegenerative diseases caused by lysosomal enzyme activity deficiencies and consequent accumulation of gangliosides in the central nervous system (CNS). The infantile forms are distinguished from the juvenile forms by age of onset, rate of disease progression, and age of death. There are no approved treatments for the gangliosidoses. In search of potential biomarkers of disease, we quantified 188 analytes in CSF and serum from living human patients with longitudinal (serial) measurements. Notably, several associated with inflammation were elevated in the CSF of infantile gangliosidosis patients, and less so in more slowly progressing forms of juvenile gangliosidosis, but not in MPS disease. Thirteen CSF and two serum biomarker candidates were identified. Five candidate biomarkers were distinguished by persistent elevation in the CSF of patients with the severe infantile phenotype: ENA-78, MCP-1, MIP-1α, MIP-1ß, and TNFR2. Correspondence of abnormal elevation with other variables of disease-i.e., severity of clinical phenotype, differentiation from changes in serum, and lack of abnormality in other neurodegenerative lysosomal diseases-identifies these analytes as biomarkers of neuropathology specific to the gangliosidosis diseases.


Assuntos
Biomarcadores/líquido cefalorraquidiano , Gangliosidoses/diagnóstico , Inflamação/diagnóstico , Adolescente , Biomarcadores/sangue , Sistema Nervoso Central/metabolismo , Quimiocina CCL2/líquido cefalorraquidiano , Quimiocina CCL4/líquido cefalorraquidiano , Quimiocina CXCL5/líquido cefalorraquidiano , Criança , Pré-Escolar , Feminino , Gangliosidoses/metabolismo , Gangliosidose GM1/diagnóstico , Gangliosidose GM1/metabolismo , Humanos , Lactente , Masculino , Receptores Tipo II do Fator de Necrose Tumoral/líquido cefalorraquidiano , Doença de Sandhoff/diagnóstico , Doença de Sandhoff/metabolismo , Doença de Tay-Sachs/diagnóstico , Doença de Tay-Sachs/metabolismo , Fatores de Transcrição/líquido cefalorraquidiano
8.
Sci Signal ; 7(338): pe19, 2014 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-25118326

RESUMO

Lipocalins are a class of proteins that scavenge hydrophobic molecules in diverse contexts, including the immune system, the nervous system, and cancer. A recent study by Watanbe et al. identifies lipocalin 2 produced by the female mouse reproductive tract as a sperm-capacitating agent that alters the membrane properties of sperm in preparation for fertilization. The potential for lipocalins to act as general modulators of plasma membrane bioactivity is discussed.


Assuntos
Proteínas de Fase Aguda/metabolismo , Regulação Alostérica/fisiologia , Membrana Celular/metabolismo , Lipocalinas/metabolismo , Microdomínios da Membrana/metabolismo , Modelos Biológicos , Proteínas Oncogênicas/metabolismo , Capacitação Espermática/fisiologia , Proteínas de Fase Aguda/química , Animais , Feminino , Gangliosidose GM1/metabolismo , Lipocalina-2 , Lipocalinas/química , Masculino , Camundongos , Proteínas Oncogênicas/química
9.
Mol Genet Metab ; 107(1-2): 203-12, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22784478

RESUMO

Deficiencies of lysosomal ß-D-galactosidase can result in GM1 gangliosidosis, a severe neurodegenerative disease characterized by massive neuronal storage of GM1 ganglioside in the brain. Currently there are no available therapies that can even slow the progression of this disease. Enzyme enhancement therapy utilizes small molecules that can often cross the blood brain barrier, but are also often competitive inhibitors of their target enzyme. It is a promising new approach for treating diseases, often caused by missense mutations, associated with dramatically reduced levels of functionally folded enzyme. Despite a number of positive reports based on assays performed with patient cells, skepticism persists that an inhibitor-based treatment can increase mutant enzyme activity in vivo. To date no appropriate animal model, i.e., one that recapitulates a responsive human genotype and clinical phenotype, has been reported that could be used to validate enzyme enhancement therapy. In this report, we identify a novel enzyme enhancement-agent, N-nonyl-deoxygalactonojirimycin, that enhances the mutant ß-galactosidase activity in the lysosomes of a number of patient cell lines containing a variety of missense mutations. We then demonstrate that treatment of cells from a previously described, naturally occurring feline model (that biochemically, clinically and molecularly closely mimics GM1 gangliosidosis in humans) with this molecule, results in a robust enhancement of their mutant lysosomal ß-galactosidase activity. These data indicate that the feline model could be used to validate this therapeutic approach and determine the relationship between the disease stage at which this therapy is initiated and the maximum clinical benefits obtainable.


Assuntos
1-Desoxinojirimicina/análogos & derivados , Terapia de Reposição de Enzimas , Gangliosidose GM1/metabolismo , Proteínas Mutantes/metabolismo , beta-Galactosidase/metabolismo , 1-Desoxinojirimicina/administração & dosagem , 1-Desoxinojirimicina/farmacologia , Animais , Gatos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Gangliosidose GM1/tratamento farmacológico , Gangliosidose GM1/genética , Temperatura Alta , Humanos , Concentração de Íons de Hidrogênio , Proteínas Mutantes/antagonistas & inibidores , Proteínas Mutantes/química , Mutação , Desnaturação Proteica/efeitos dos fármacos , Resultado do Tratamento , beta-Galactosidase/antagonistas & inibidores , beta-Galactosidase/química
10.
J Virol ; 86(13): 7028-42, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22514351

RESUMO

Polyomaviruses are nonenveloped viruses with capsids composed primarily of 72 pentamers of the viral VP1 protein, which forms the outer shell of the capsid and binds to cell surface oligosaccharide receptors. Highly conserved VP1 proteins from closely related polyomaviruses recognize different oligosaccharides. To determine whether amino acid changes restricted to the oligosaccharide binding site are sufficient to determine receptor specificity and how changes in receptor usage affect tropism, we studied the primate polyomavirus simian virus 40 (SV40), which uses the ganglioside GM1 as a receptor that mediates cell binding and entry. Here, we used two sequential genetic screens to isolate and characterize viable SV40 mutants with mutations in the VP1 GM1 binding site. Two of these mutants were completely resistant to GM1 neutralization, were no longer stimulated by incorporation of GM1 into cell membranes, and were unable to bind to GM1 on the cell surface. In addition, these mutant viruses displayed an infection defect in monkey cells with high levels of cell surface GM1. Interestingly, one mutant infected cells with low cell surface GM1 more efficiently than wild-type virus, apparently by utilizing a different ganglioside receptor. Our results indicate that a small number of mutations in the GM1 binding site are sufficient to alter ganglioside usage and change tropism, and they suggest that VP1 divergence is driven primarily by a requirement to accommodate specific receptors. In addition, our results suggest that GM1 binding is required for vacuole formation in permissive monkey CV-1 cells. Further study of these mutants will provide new insight into polyomavirus entry, pathogenesis, and evolution.


Assuntos
Gangliosidose GM1/metabolismo , Receptores Virais/metabolismo , Vírus 40 dos Símios/fisiologia , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo , Tropismo Viral , Ligação Viral , Substituição de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Humanos , Vírus 40 dos Símios/genética
11.
Virology ; 422(1): 114-24, 2012 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-22056390

RESUMO

Repression of human papillomavirus (HPV) E6 and E7 oncogenes in established cervical carcinoma cell lines causes senescence due to reactivation of cellular tumor suppressor pathways. Here, we determined whether ongoing expression of HPV16 or HPV18 oncogenes is required for the proliferation of primary human cervical carcinoma cells in serum-free conditions at low passage number after isolation from patients. We used an SV40 viral vector expressing the bovine papillomavirus E2 protein to repress E6 and E7 in these cells. To enable efficient SV40 infection and E2 gene delivery, we first incubated the primary cervical cancer cells with the ganglioside GM1, a cell-surface receptor for SV40 that is limiting in these cells. Repression of HPV in primary cervical carcinoma cells caused them to undergo senescence, but the E2 protein had little effect on HPV-negative primary cells. These data suggest that E6 and E7 dependence is an inherent property of human cervical cancer cells.


Assuntos
Papillomavirus Humano 16/genética , Papillomavirus Humano 18/genética , Proteínas Oncogênicas Virais/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/virologia , Proliferação de Células , Senescência Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Gangliosidose GM1/metabolismo , Papillomavirus Humano 16/fisiologia , Papillomavirus Humano 18/fisiologia , Humanos , Proteínas Oncogênicas Virais/biossíntese , Proteínas Oncogênicas Virais/genética , Proteínas E7 de Papillomavirus/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Vírus 40 dos Símios/genética , Vírus 40 dos Símios/fisiologia , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/genética
12.
J Neurochem ; 118(3): 399-406, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21574998

RESUMO

G(M1) -gangliosidosis is a fatal neurodegenerative disorder caused by deficiency of lysosomal acid ß-galactosidase (ß-gal). Accumulation of its substrate ganglioside G(M1) (G(M1) ) in lysosomes and other parts of the cell leads to progressive neurodegeneration, but underlying mechanisms remain unclear. Previous studies demonstrated an essential role for interaction of G(M1) with tropomyosin receptor kinase (Trk) receptors in neuronal growth, survival and differentiation. In this study we demonstrate accumulation of G(M1) in the cell-surface rafts and lysosomes of the ß-gal knockout (ß-gal-/-) mouse brain association with accumulation of Trk receptors and enhancement of its downstream signaling. Immunofluorescence and subcellular fractionation analysis revealed accumulation of Trk receptors in the late endosomes/lysosomes of the ß-gal-/- mouse brain and their association with ubiquitin and p62. Administration of a chemical chaperone to ß-gal-/- mouse expressing human mutant R201C protein resulted in a marked reduction of intracellular storage of G(M1) and phosphorylated Trk. These findings indicate that G(M1) accumulation in rafts causes activation of Trk signaling, which may participate in the pathogenesis of G(M1) -gangliosidosis.


Assuntos
Química Encefálica/fisiologia , Gangliosidose GM1/metabolismo , Hexosaminas/farmacologia , Lisossomos/metabolismo , Chaperonas Moleculares/farmacologia , Receptor trkA/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Encéfalo/patologia , Química Encefálica/efeitos dos fármacos , Células Cultivadas , Imunofluorescência , Humanos , Imunoprecipitação , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Crescimento Neural/fisiologia , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , beta-Galactosidase/genética , beta-Galactosidase/farmacologia
13.
Mol Cell ; 36(3): 500-11, 2009 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-19917257

RESUMO

Mitochondria-associated ER membranes, or MAMs, define the sites of endoplasmic reticulum/mitochondria juxtaposition that control Ca(2+) flux between these organelles. We found that in a mouse model of the human lysosomal storage disease GM1-gangliosidosis, GM1-ganglioside accumulates in the glycosphingolipid-enriched microdomain (GEM) fractions of MAMs, where it interacts with the phosphorylated form of IP3 receptor-1, influencing the activity of this channel. Ca(2+) depleted from the ER is then taken up by the mitochondria, leading to Ca(2+) overload in this organelle. The latter induces mitochondrial membrane permeabilization (MMP), opening of the permeability transition pore, and activation of the mitochondrial apoptotic pathway. This study identifies the GEMs as the sites of Ca(2+) diffusion between the ER and the mitochondria. We propose a new mechanism of Ca(2+)-mediated apoptotic signaling whereby GM1 accumulation at the GEMs alters Ca(2+) dynamics and acts as a molecular effector of both ER stress-induced and mitochondria-mediated apoptosis of neuronal cells.


Assuntos
Apoptose , Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Gangliosídeo G(M1)/metabolismo , Mitocôndrias/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Encéfalo/metabolismo , Cálcio/farmacologia , Células Cultivadas , Citocromos c/metabolismo , Modelos Animais de Doenças , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Gangliosídeo G(M1)/farmacologia , Gangliosidose GM1/genética , Gangliosidose GM1/metabolismo , Gangliosidose GM1/patologia , Glicoesfingolipídeos/metabolismo , Humanos , Immunoblotting , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Microdomínios da Membrana/metabolismo , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Mitocôndrias/fisiologia , Mitocôndrias/ultraestrutura , Espécies Reativas de Oxigênio/metabolismo , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
14.
J Neurochem ; 109(5): 1237-49, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19476542

RESUMO

The insertion and removal of NMDA receptors from the synapse are critical events that modulate synaptic plasticity. While a great deal of progress has been made on understanding the mechanisms that modulate trafficking of NMDA receptors, we do not currently understand the molecular events required for the fusion of receptor containing vesicles with the plasma membrane. Here, we show that sphingomyelin phosphodiesterase 3 (also known as neutral sphingomyelinase-2) is critical for tumor necrosis factor (TNF) alpha-induced trafficking of NMDA receptors and synaptic plasticity. TNFalpha initiated a rapid increase in ceramide that was associated with increased surface localization of NMDA receptor NR1 subunits and a specific clustering of NR1 phosphorylated on serines 896 and 897 into lipid rafts. Brief applications of TNFalpha increased the rate and amplitude of NMDA-evoked calcium bursts and enhanced excitatory post-synaptic currents. Pharmacological inhibition or genetic mutation of neutral sphingomyelinase-2 prevented TNFalpha-induced generation of ceramide, phosphorylation of NR1 subunits, clustering of NR1, enhancement of NMDA-evoked calcium flux and excitatory post-synaptic currents.


Assuntos
Plasticidade Neuronal/fisiologia , Neurônios/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Compostos de Anilina/farmacologia , Animais , Compostos de Benzilideno/farmacologia , Cálcio/metabolismo , Células Cultivadas , Ceramidas/metabolismo , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Ácidos Graxos Monoinsaturados/farmacologia , Transferência Ressonante de Energia de Fluorescência/métodos , Gangliosidose GM1/metabolismo , Hipocampo/citologia , Técnicas In Vitro , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Mutação/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Técnicas de Patch-Clamp/métodos , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Serina/metabolismo , Espectrometria de Massas por Ionização por Electrospray/métodos , Esfingomielina Fosfodiesterase/genética
15.
J Neurosci ; 29(13): 4228-38, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19339617

RESUMO

17-beta-Estradiol (E2) is a steroid hormone involved in numerous bodily functions, including several brain functions. In particular, E2 is neuroprotective against excitotoxicity and other forms of brain injuries, a property that requires the extracellular signal-regulated kinase (ERK) pathway and possibly that of other signaling molecules. The mechanism and identity of the receptor(s) involved remain unclear, although it has been suggested that E2 receptor alpha (ERalpha) and G proteins are involved. We, therefore, investigated whether E2-mediated neuroprotection and ERK activation were linked to pertussis toxin (PTX)-sensitive G-protein-coupled effector systems. Biochemical and image analysis of organotypic hippocampal slices and cortical neuronal cultures showed that E2-mediated neuroprotection as well as E2-induced ERK activation were sensitive to PTX. The sensitivity to PTX suggested a possible role of G-protein- and beta-arrestin-mediated mechanisms. Western immunoblots from E2-treated cortical neuronal cultures revealed an increase in phosphorylation of both G-protein-coupled receptor-kinase 2 and beta-arrestin-1, a G-protein-coupled receptor adaptor protein. Transfection of neurons with beta-arrestin-1 small interfering RNA prevented E2-induced ERK activation. Coimmunoprecipitation experiments indicated that E2 increased the recruitment of beta-arrestin-1 and c-Src to ERalpha. These findings suggested that ERalpha is regulated by a mechanism associated with receptor desensitization and downregulation. In support of this idea, we found that E2 treatment of cortical synaptoneurosomes resulted in internalization of ERalpha, whereas treatment of cortical neurons with the ER agonists E-6-BSA-FITC [beta-estradiol-6-(O-carboxymethyl)oxime-bovine serum albumin conjugated with fluorescein isothiocyanate] and E-6-biotin [1,3,5(10)-estratrien-3,17beta-diol-6-one-6-carboxymethloxime-NH-propyl-biotin] resulted in agonist internalization. These results demonstrate that E2-mediated neuroprotection and ERK activation involve ERalpha activation of G-protein- and beta-arrestin-mediated mechanisms.


Assuntos
Arrestinas/metabolismo , Estradiol/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Animais Recém-Nascidos , Biotina/metabolismo , Encéfalo/citologia , Técnicas de Cultura de Células , Morte Celular/efeitos dos fármacos , Células Cultivadas , Toxina da Cólera/metabolismo , Interações Medicamentosas , Ativação Enzimática/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/toxicidade , Feminino , Gangliosidose GM1/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , N-Metilaspartato/toxicidade , Toxina Pertussis/farmacologia , Gravidez , Transporte Proteico/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , beta-Arrestina 1 , beta-Arrestinas
16.
J Neurochem ; 109(5): 1250-60, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19317854

RESUMO

The assembly of amyloid beta-protein to amyloid fibrils is a critical event in Alzheimer's disease. Evidence exists that endocytic pathway abnormalities, including the enlargement of early endosomes, precede the extraneuronal amyloid fibril deposition in the brain. We determined whether endocytic dysfunction potently promotes the assembly of amyloid beta-protein on the surface of cultured cells. Blocking the early endocytic pathway by clathrin suppression, inactivation of small GTPases, removal of membrane cholesterol, and Rab5 knockdown did not result in amyloid fibril formation on the cell surface from exogenously added soluble amyloid beta-protein. In contrast, blocking the late endocytic pathway by Rab7 suppression markedly induced the amyloid fibril formation in addition to the enlargement of early endosomes. Notably, a monoclonal antibody specific to GM1-ganglioside-bound amyloid beta-protein, an endogenous seed for Alzheimer amyloid, completely blocks the amyloid fibril formation. Our results suggest that late but not early endocytic dysfunction contributes to the amyloid fibril formation by facilitating the generation of amyloid seed in the Alzheimer's brain.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Endocitose/fisiologia , Peptídeos beta-Amiloides/farmacologia , Animais , Proteínas de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Cadeias Pesadas de Clatrina/genética , Cadeias Pesadas de Clatrina/metabolismo , Relação Dose-Resposta a Droga , Endocitose/efeitos dos fármacos , Gangliosidose GM1/metabolismo , Imunoprecipitação/métodos , Mutação/genética , Células PC12/citologia , Células PC12/efeitos dos fármacos , Células PC12/metabolismo , RNA Interferente Pequeno/farmacologia , Ratos , Fatores de Tempo , Transfecção/métodos , beta-Ciclodextrinas/farmacologia , Proteína cdc42 de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7 , Proteínas rac1 de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/genética
17.
Subcell Biochem ; 49: 441-67, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18751922

RESUMO

Glycosphingolipids, comprising a ceramide lipid backbone linked to one/more saccharides, are particularly abundant on the outer leaflet of the eukaryotic plasma membrane and play a role in a wide variety of essential cellular processes. Biosynthesis and subsequently degradation of these lipids is tightly regulated via the involvement of numerous enzymes, and failure of an enzyme to participate in the metabolism results in storage of the enzyme's substrate, giving rise to a lysosomal storage disease. The characteristics, severity and onset of the disease are dependent on the enzyme deficient and the residual activity. Most lysosomal storage disorders found thus far are caused by a defect in the catabolic activity of a hydrolase, causing progressive accumulation of its substrate, predominantly in the lysosome. Storage of gangliosides, sialic acid containing glycosphingolipids, mostly found in the central nervous system, is a hallmark of neuronopathic forms of the disease, that include GM1 and GM2 gangliosidoses, Gaucher type II and III and Niemann-Pick C. Models for these diseases have provided valuable insight into the disease pathology and potential treatment methods.Treatment of these rare but severe disorders proves challenging due to restricted access of therapeutics through the blood-brain barrier. However, recent advances in enzyme replacement, bone marrow transplantation, gene transfer, substrate reduction and chaperon-mediated therapy provide great potential in treating these devastating disorders.


Assuntos
Glicoesfingolipídeos/metabolismo , Doenças por Armazenamento dos Lisossomos do Sistema Nervoso/metabolismo , Animais , Transplante de Medula Óssea , Gangliosidoses GM2/metabolismo , Gangliosidose GM1/metabolismo , Doença de Gaucher/metabolismo , Terapia Genética , Humanos , Doenças por Armazenamento dos Lisossomos do Sistema Nervoso/terapia , Modelos Animais , Chaperonas Moleculares/uso terapêutico , Doenças de Niemann-Pick/metabolismo , Sialiltransferases/deficiência
18.
Electrophoresis ; 28(17): 3100-4, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17668449

RESUMO

We report an ultrasensitive method for the analysis of glycosphingolipid catabolism. The substrate G(M1) and the set of seven metabolites into which it can be degraded (G(A1), G(M2), G(A2), G(M3), LacCer, GlcCer, and Cer) were labeled with the highly fluorescent dye tetramethylrhodamine. CE with LIF detection was used to assay these compounds with 150 +/- 80 yoctomole mass (1 ymol = 10(-24) mol = 0.6 copies) detection limits and 5 +/- 3 pM concentration detection limits. An alignment algorithm based on migration of two components was employed to correct for drift in the separation. The within-day and between-day precision in peak height was 20%, in peak width 15%, and in adjusted migration time 0.03%. After normalization to total sample injected, the RSD in peak height reduced to 2-6%, which approaches the limit set by molecular shot noise in the number of molecules taken for analysis. PC12 cells were incubated with the labeled G(M1). Fluorescent microscopy demonstrated uptake by the cells. CE was used to separate a cellular homogenate prepared from these cells. A set of peaks was observed, which were tentatively identified based on comigration with the standards. Roughly 120 pL of homogenate was injected, which contained a total of 150 zmol of labeled substrate and products. Metabolite that preserves the fluorescent label can be detected at the yoctomole level, which should allow characterization of this metabolic pathway in single cells.


Assuntos
Gangliosidose GM1/metabolismo , Animais , Eletroforese Capilar/métodos , Corantes Fluorescentes/química , Células PC12 , Ratos , Rodaminas/química
19.
Traffic ; 7(11): 1482-94, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16984405

RESUMO

Lipid rafts are membrane microdomains rich in cholesterol and glycosphingolipids that have been implicated in the regulation of intracellular protein trafficking. During exocytosis, a class of proteins termed SNAREs mediate secretory granule-plasma membrane fusion. To investigate the role of lipid rafts in secretory granule exocytosis, we examined the raft association of SNARE proteins and SNARE complexes in rat basophilic leukemia (RBL) mast cells. The SNARE protein SNAP-23 co-localized with a lipid raft marker and was present in detergent-insoluble lipid raft microdomains in RBL cells. By contrast, only small amounts (<20%) of the plasma membrane SNARE syntaxin 4 or the granule-associated SNARE vesicle-associated membrane protein (VAMP)-2 were present in these microdomains. Despite this, essentially all syntaxin 4 and most of VAMP-2 in these rafts were present in SNARE complexes containing SNAP-23, while essentially none of these complexes were present in nonraft membranes. Whereas SNAP-23 is membrane anchored by palmitoylation, the association of the transmembrane protein syntaxin 4 with lipid rafts was because of its binding to SNAP-23. After stimulating mast cells exocytosis, the amount of syntaxin 4 and VAMP-2 present in rafts increased twofold, and these proteins were now present in raft-associated phospho-SNAP-23/syntaxin 4/VAMP-2 complexes, revealing differential association of SNARE fusion complexes during the process of regulated exocytosis.


Assuntos
Exocitose/fisiologia , Mastócitos/metabolismo , Microdomínios da Membrana/metabolismo , Proteínas SNARE/metabolismo , Animais , Toxina da Cólera/metabolismo , Dinitrofenóis/imunologia , Dinitrofenóis/farmacologia , Exocitose/efeitos dos fármacos , Gangliosidose GM1/metabolismo , Células HeLa , Humanos , Imunoglobulina E/imunologia , Imunoglobulina E/farmacologia , Mastócitos/efeitos dos fármacos , Camundongos , Fosforilação/efeitos dos fármacos , Ligação Proteica , Proteínas Qa-SNARE/metabolismo , Ratos , Receptores de IgE/agonistas , Receptores de IgE/metabolismo , Proteínas SNARE/genética , Soroalbumina Bovina/imunologia , Soroalbumina Bovina/farmacologia , Proteína 25 Associada a Sinaptossoma/genética , Proteína 25 Associada a Sinaptossoma/metabolismo , Transfecção , Proteína 2 Associada à Membrana da Vesícula/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
20.
Blood ; 106(7): 2259-68, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15941905

RESUMO

Bone marrow cells (BMCs) could correct some pathologic conditions of the central nervous system (CNS) if these cells would effectively repopulate the brain. One such condition is G(M1)-gangliosidosis, a neurodegenerative glycosphingolipidosis due to deficiency of lysosomal beta-galactosidase (beta-gal). In this disease, abnormal build up of G(M1)-ganglioside in the endoplasmic reticulum of brain cells results in calcium imbalance, induction of an unfolded protein response (UPR), and neuronal apoptosis. These processes are accompanied by the activation/proliferation of microglia and the production of inflammatory cytokines. Here we demonstrate that local neuroinflammation promotes the selective activation of chemokines, such as stromal-cell-derived factor 1 (SDF-1), macrophage inflammatory protein 1-alpha (MIP-1alpha), and MIP-1beta, which chemoattract genetically modified BMCs into the CNS. Mice that underwent bone marrow transplantation showed increased beta-gal activity in different brain regions and reduced lysosomal storage. Decreased production of chemokines and effectors of the UPR as well as restoration of neurologic functions accompanied this phenotypic reversion. Our results suggest that beta-gal-expressing bone marrow (BM)-derived cells selectively migrate to the CNS under a gradient of chemokines and become a source of correcting enzyme to deficient neurons. Thus, a disease condition such as G(M1)-gangliosidosis, which is characterized by neurodegeneration and neuroinflammation, may influence the response of the CNS to ex vivo gene therapy.


Assuntos
Células da Medula Óssea/metabolismo , Sistema Nervoso Central/metabolismo , Quimiocinas/metabolismo , Gangliosidose GM1/metabolismo , Animais , Apoptose , Células da Medula Óssea/citologia , Encéfalo/metabolismo , Cálcio/metabolismo , Movimento Celular , Proliferação de Células , Transplante de Células , Quimiocina CCL3 , Quimiocina CCL4 , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Cromatografia em Camada Fina , Regulação para Baixo , Retículo Endoplasmático/metabolismo , Ensaio de Imunoadsorção Enzimática , Gangliosidose GM1/genética , Terapia Genética/métodos , Vetores Genéticos , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imuno-Histoquímica , Inflamação , Lisossomos/enzimologia , Proteínas Inflamatórias de Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Doenças Neurodegenerativas/patologia , Neurônios/patologia , Dobramento de Proteína , RNA/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleases/metabolismo , Fatores de Tempo , Regulação para Cima , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA