Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 15(685): eabo3823, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36857431

RESUMO

Hexanucleotide repeat expansions in C9ORF72 are the most common genetic cause of familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Studies have shown that the hexanucleotide expansions cause the noncanonical translation of C9ORF72 transcripts into neurotoxic dipeptide repeat proteins (DPRs) that contribute to neurodegeneration. We show that a cell-penetrant peptide blocked the nuclear export of C9ORF72-repeat transcripts in HEK293T cells by competing with the interaction between SR-rich splicing factor 1 (SRSF1) and nuclear export factor 1 (NXF1). The cell-penetrant peptide also blocked the translation of toxic DPRs in neurons differentiated from induced neural progenitor cells (iNPCs), which were derived from individuals carrying C9ORF72-linked ALS mutations. This peptide also increased survival of iNPC-differentiated C9ORF72-ALS motor neurons cocultured with astrocytes. Oral administration of the cell-penetrant peptide reduced DPR translation and rescued locomotor deficits in a Drosophila model of mutant C9ORF72-mediated ALS/FTD. Intrathecal injection of this peptide into the brains of ALS/FTD mice carrying a C9ORF72 mutation resulted in reduced expression of DPRs in mouse brains. These findings demonstrate that disrupting the production of DPRs in cellular and animal models of ALS/FTD might be a strategy to ameliorate neurodegeneration in these diseases.


Assuntos
Esclerose Lateral Amiotrófica , Demência Frontotemporal , Humanos , Animais , Camundongos , Dipeptídeos , Proteína C9orf72 , Transporte Ativo do Núcleo Celular , Células HEK293 , Peptídeos , Neurônios Motores , RNA , Fatores de Processamento de Serina-Arginina
2.
Nat Rev Neurol ; 19(1): 39-52, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36481799

RESUMO

Amyotrophic lateral sclerosis (ALS) is a devastating and incurable neurodegenerative disease characterized by the progressive loss of upper and lower motor neurons. ALS causes death, usually within 2-5 years of diagnosis. Riluzole, the only drug currently approved in Europe for the treatment of this condition, offers only a modest benefit, increasing survival by 3 months on average. Recent advances in our understanding of causative or disease-modifying genetic variants and in the development of genetic therapy strategies present exciting new therapeutic opportunities for ALS. In addition, the approval of adeno-associated virus-mediated delivery of functional copies of the SMN1 gene to treat spinal muscular atrophy represents an important therapeutic milestone and demonstrates the potential of gene replacement therapies for motor neuron disorders. In this Review, we describe the current landscape of genetic therapies in ALS, highlighting achievements and critical challenges. In particular, we discuss opportunities for gene replacement therapy in subgroups of people with ALS, and we describe loss-of-function mutations that are known to contribute to the pathophysiology of ALS and could represent novel targets for gene replacement therapies.


Assuntos
Esclerose Lateral Amiotrófica , Doença dos Neurônios Motores , Doenças Neurodegenerativas , Humanos , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/terapia , Doenças Neurodegenerativas/terapia , Neurônios Motores , Terapia Genética
3.
Mol Cell ; 80(5): 876-891.e6, 2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33217318

RESUMO

Stress granules (SGs) are cytoplasmic assemblies of proteins and non-translating mRNAs. Whereas much has been learned about SG formation, a major gap remains in understanding the compositional changes SGs undergo during normal disassembly and under disease conditions. Here, we address this gap by proteomic dissection of the SG temporal disassembly sequence using multi-bait APEX proximity proteomics. We discover 109 novel SG proteins and characterize distinct SG substructures. We reveal dozens of disassembly-engaged proteins (DEPs), some of which play functional roles in SG disassembly, including small ubiquitin-like modifier (SUMO) conjugating enzymes. We further demonstrate that SUMOylation regulates SG disassembly and SG formation. Parallel proteomics with amyotrophic lateral sclerosis (ALS)-associated C9ORF72 dipeptides uncovered attenuated DEP recruitment during SG disassembly and impaired SUMOylation. Accordingly, SUMO activity ameliorated C9ORF72-ALS-related neurodegeneration in Drosophila. By dissecting the SG spatiotemporal proteomic landscape, we provide an in-depth resource for future work on SG function and reveal basic and disease-relevant mechanisms of SG disassembly.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Proteína C9orf72/metabolismo , Grânulos Citoplasmáticos/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Proteína C9orf72/genética , Linhagem Celular Tumoral , Grânulos Citoplasmáticos/genética , Grânulos Citoplasmáticos/patologia , Dipeptídeos/genética , Dipeptídeos/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster , Humanos , Camundongos , Proteômica , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética
4.
Brain ; 142(3): 586-605, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30698736

RESUMO

As clinical evidence supports a negative impact of dysfunctional energy metabolism on the disease progression in amyotrophic lateral sclerosis, it is vital to understand how the energy metabolic pathways are altered and whether they can be restored to slow disease progression. Possible approaches include increasing or rerouting catabolism of alternative fuel sources to supplement the glycolytic and mitochondrial pathways such as glycogen, ketone bodies and nucleosides. To analyse the basis of the catabolic defect in amyotrophic lateral sclerosis we used a novel phenotypic metabolic array. We profiled fibroblasts and induced neuronal progenitor-derived human induced astrocytes from C9orf72 amyotrophic lateral sclerosis patients compared to normal controls, measuring the rates of production of reduced nicotinamide adenine dinucleotides from 91 potential energy substrates. This approach shows for the first time that C9orf72 human induced astrocytes and fibroblasts have an adenosine to inosine deamination defect caused by reduction of adenosine deaminase, which is also observed in induced astrocytes from sporadic patients. Patient-derived induced astrocyte lines were more susceptible to adenosine-induced toxicity, which could be mimicked by inhibiting adenosine deaminase in control lines. Furthermore, adenosine deaminase inhibition in control induced astrocytes led to increased motor neuron toxicity in co-cultures, similar to the levels observed with patient derived induced astrocytes. Bypassing metabolically the adenosine deaminase defect by inosine supplementation was beneficial bioenergetically in vitro, increasing glycolytic energy output and leading to an increase in motor neuron survival in co-cultures with induced astrocytes. Inosine supplementation, in combination with modulation of the level of adenosine deaminase may represent a beneficial therapeutic approach to evaluate in patients with amyotrophic lateral sclerosis.


Assuntos
Adenosina Desaminase/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Neurônios Motores/metabolismo , Adenosina Desaminase/fisiologia , Adulto , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Astrócitos/metabolismo , Proteína C9orf72/metabolismo , Morte Celular , Sobrevivência Celular , Células Cultivadas , Técnicas de Cocultura , Progressão da Doença , Metabolismo Energético/fisiologia , Feminino , Fibroblastos/metabolismo , Humanos , Inosina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Ratos , Ratos Sprague-Dawley , Células-Tronco/metabolismo
5.
Mol Ther Nucleic Acids ; 12: 75-88, 2018 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-30195799

RESUMO

Of familial amyotrophic lateral sclerosis (fALS) cases, 20% are caused by mutations in the gene encoding human cytosolic Cu/Zn superoxide dismutase (hSOD1). Efficient translation of the therapeutic potential of RNAi for the treatment of SOD1-ALS patients requires the development of vectors that are free of significant off-target effects and with reliable biomarkers to discern sufficient target engagement and correct dosing. Using adeno-associated virus serotype 9 to deliver RNAi against hSOD1 in the SOD1G93A mouse model, we found that intrathecal injection of the therapeutic vector via the cisterna magna delayed onset of disease, decreased motor neuron death at end stage by up to 88%, and prolonged the median survival of SOD1G93A mice by up to 42%. To our knowledge, this is the first report to demonstrate no significant off-target effects linked to hSOD1 silencing, providing further confidence in the specificity of this approach. We also report the measurement of cerebrospinal fluid (CSF) hSOD1 protein levels as a biomarker of effective dosing and efficacy of hSOD1 knockdown. Together, these data provide further confidence in the safety of the clinical therapeutic vector. The CSF biomarker will be a useful measure of biological activity for translation into human clinical trials.

6.
Sci Rep ; 8(1): 8097, 2018 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-29802307

RESUMO

TAR DNA-binding protein 43 (TDP-43) is a key player in neurodegenerative diseases including frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). Accumulation of TDP-43 is associated with neuronal death in the brain. How increased and disease-causing mutant forms of TDP-43 induce cell death remains unclear. Here we addressed the role of TDP-43 during neural development and show that reduced TDP-43 causes defects in neural stem/progenitor cell proliferation but not cell death. However, overexpression of wild type and TDP-43A315T proteins induce p53-dependent apoptosis of neural stem/progenitors and human induced pluripotent cell (iPS)-derived immature cortical neurons. We show that TDP-43 induces expression of the proapoptotic BH3-only genes Bbc3 and Bax, and that p53 inhibition rescues TDP-43 induced cell death of embryonic mouse, and human cortical neurons, including those derived from TDP-43G298S ALS patient iPS cells. Hence, an increase in wild type and mutant TDP-43 induces p53-dependent cell death in neural progenitors developing neurons and this can be rescued. These findings may have important implications for accumulated or mutant TDP-43 induced neurodegenerative diseases.


Assuntos
Apoptose , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Neurais/citologia , Neurônios/citologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Ciclo Celular , Inibidor de Quinase Dependente de Ciclina p21/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Mutação , Neurogênese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
7.
Dis Model Mech ; 10(7): 859-868, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28550099

RESUMO

Intronic GGGGCC repeat expansions in C9orf72 are the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Two major pathologies stemming from the hexanucleotide RNA expansions (HREs) have been identified in postmortem tissue: intracellular RNA foci and repeat-associated non-ATG dependent (RAN) dipeptides, although it is unclear how these and other hallmarks of disease contribute to the pathophysiology of neuronal injury. Here, we describe two novel lines of mice that overexpress either 10 pure or 102 interrupted GGGGCC repeats mediated by adeno-associated virus (AAV) and recapitulate the relevant human pathology and disease-related behavioural phenotypes. Similar levels of intracellular RNA foci developed in both lines of mice, but only mice expressing 102 repeats generated C9orf72 RAN pathology, neuromuscular junction (NMJ) abnormalities, dispersal of the hippocampal CA1, enhanced apoptosis, and deficits in gait and cognition. Neither line of mice, however, showed extensive TAR DNA-binding protein 43 (TDP-43) pathology or neurodegeneration. Our data suggest that RNA foci pathology is not a good predictor of C9orf72 RAN dipeptide formation, and that RAN dipeptides and NMJ dysfunction are drivers of C9orf72 disease pathogenesis. These AAV-mediated models of C9orf72-associated ALS/FTD will be useful tools for studying disease pathophysiology and developing new therapeutic approaches.


Assuntos
Comportamento Animal , Encéfalo/patologia , Proteína C9orf72/genética , Expansão das Repetições de DNA/genética , Dependovirus/metabolismo , Técnicas de Transferência de Genes , Animais , Biomarcadores/metabolismo , Encéfalo/fisiopatologia , Região CA1 Hipocampal/patologia , Morte Celular , Núcleo Celular/metabolismo , Cognição , Marcha , Células HEK293 , Humanos , Camundongos , Junção Neuromuscular/metabolismo , Junção Neuromuscular/patologia , Junção Neuromuscular/fisiopatologia , Neurônios/metabolismo , Neurônios/patologia , RNA/metabolismo , Proteína Sequestossoma-1/metabolismo , Regulação para Cima
8.
Hum Mol Genet ; 26(6): 1133-1145, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28158451

RESUMO

Amyotrophic lateral sclerosis (ALS) is a devastating and incurable neurodegenerative disease, characterised by progressive failure of the neuromuscular system. A (G4C2)n repeat expansion in C9ORF72 is the most common genetic cause of ALS and frontotemporal dementia (FTD). To date, the balance of evidence indicates that the (G4C2)n repeat causes toxicity and neurodegeneration via a gain-of-toxic function mechanism; either through direct RNA toxicity or through the production of toxic aggregating dipeptide repeat proteins. Here, we have generated a stable and isogenic motor neuronal NSC34 cell model with inducible expression of a (G4C2)102 repeat, to investigate the gain-of-toxic function mechanisms. The expression of the (G4C2)102 repeat produces RNA foci and also undergoes RAN translation. In addition, the expression of the (G4C2)102 repeat shows cellular toxicity. Through comparison of transcriptomic data from the cellular model with laser-captured spinal motor neurons from C9ORF72-ALS cases, we also demonstrate that the PI3K/Akt cell survival signalling pathway is dysregulated in both systems. Furthermore, partial knockdown of Pten rescues the toxicity observed in the NSC34 (G4C2)102 cellular gain-of-toxic function model of C9ORF72-ALS. Our data indicate that PTEN may provide a potential therapeutic target to ameliorate toxic effects of the (G4C2)n repeat.


Assuntos
Esclerose Lateral Amiotrófica/genética , Expansão das Repetições de DNA/genética , Demência Frontotemporal/genética , PTEN Fosfo-Hidrolase/genética , Proteínas/genética , Esclerose Lateral Amiotrófica/patologia , Proteína C9orf72 , Linhagem Celular , Sobrevivência Celular , Demência Frontotemporal/patologia , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , RNA/genética
9.
Brain ; 137(Pt 7): 2040-51, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24866055

RESUMO

GGGGCC repeat expansions of C9orf72 represent the most common genetic variant of amyotrophic lateral sclerosis and frontotemporal degeneration, but the mechanism of pathogenesis is unclear. Recent reports have suggested that the transcribed repeat might form toxic RNA foci that sequester various RNA processing proteins. Consensus as to the identity of the binding partners is missing and whole neuronal proteome investigation is needed. Using RNA fluorescence in situ hybridization we first identified nuclear and cytoplasmic RNA foci in peripheral and central nervous system biosamples from patients with amyotrophic lateral sclerosis with a repeat expansion of C9orf72 (C9orf72+), but not from those patients without a repeat expansion of C9orf72 (C9orf72-) or control subjects. Moreover, in the cases examined, the distribution of foci-positive neurons correlated with the clinical phenotype (t-test P < 0.05). As expected, RNA foci are ablated by RNase treatment. Interestingly, we identified foci in fibroblasts from an asymptomatic C9orf72+ carrier. We next performed pulldown assays, with GGGGCC5, in conjunction with mass spectrometry analysis, to identify candidate binding partners of the GGGGCC repeat expansion. Proteins containing RNA recognition motifs and involved in splicing, messenger RNA nuclear export and/or translation were significantly enriched. Immunohistochemistry in central nervous system tissue from C9orf72+ patients with amyotrophic lateral sclerosis demonstrated co-localization of RNA foci with SRSF2, hnRNP H1/F, ALYREF and hnRNP A1 in cerebellar granule cells and with SRSF2, hnRNP H1/F and ALYREF in motor neurons, the primary target of pathology in amyotrophic lateral sclerosis. Direct binding of proteins to GGGGCC repeat RNA was confirmed in vitro by ultraviolet-crosslinking assays. Co-localization was only detected in a small proportion of RNA foci, suggesting dynamic sequestration rather than irreversible binding. Additional immunohistochemistry demonstrated that neurons with and without RNA foci were equally likely to show nuclear depletion of TDP-43 (χ(2) P = 0.75) or poly-GA dipeptide repeat protein inclusions (χ(2) P = 0.46). Our findings suggest two non-exclusive pathogenic mechanisms: (i) functional depletion of RNA-processing proteins resulting in disruption of messenger RNA splicing; and (ii) licensing of expanded C9orf72 pre-messenger RNA for nuclear export by inappropriate association with messenger RNA export adaptor protein(s) leading to cytoplasmic repeat associated non-ATG translation and formation of potentially toxic dipeptide repeat protein.


Assuntos
Esclerose Lateral Amiotrófica/genética , Expansão das Repetições de DNA/genética , Proteínas/genética , Proteínas de Ligação a RNA/metabolismo , Trifosfato de Adenosina/farmacocinética , Esclerose Lateral Amiotrófica/patologia , Biotinilação , Encéfalo/patologia , Proteína C9orf72 , Feminino , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Humanos , Masculino , Espectrometria de Massas , Neurônios/patologia , Proteínas Nucleares/metabolismo , Isótopos de Fósforo/farmacocinética , Ligação Proteica/efeitos dos fármacos , Proteínas de Ligação a RNA/genética , Ribonucleoproteínas/metabolismo , Fatores de Processamento de Serina-Arginina , Fatores de Transcrição/metabolismo
10.
Neurobiol Aging ; 35(6): 1499-509, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24439480

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder involving the progressive degeneration of motor neurons in the brain and spinal cord. Mitochondrial dysfunction plays a key role in ALS disease progression and has been observed in several ALS cellular and animal models. Here, we show that fibroblasts isolated from ALS cases with a Cu/Zn superoxide dismutase (SOD1) I113T mutation recapitulate these mitochondrial defects. Using a novel technique, which measures mitochondrial respiration and glycolytic flux simultaneously in living cells, we have shown that SOD1 mutation causes a reduction in mitochondrial respiration and an increase in glycolytic flux. This causes a reduction in adenosine triphosphate produced by oxidative phosphorylation and an increase in adenosine triphosphate produced by glycolysis. Switching the energy source from glucose to galactose caused uncoupling of mitochondria with increased proton leak in SOD1(I113T) fibroblasts. Assessment of the contribution of fatty acid oxidation to total respiration, suggested that fatty acid oxidation is reduced in SOD1 ALS fibroblasts, an effect which can be mimicked by starving the control cells of glucose. These results highlight the importance of understanding the interplay between the major metabolic pathways, which has the potential to lead to strategies to correct the metabolic dysregulation observed in ALS cases.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Metabolismo Energético/genética , Fibroblastos/metabolismo , Glicólise/genética , Mutação , Fosforilação Oxidativa , Superóxido Dismutase/genética , Superóxido Dismutase/fisiologia , Trifosfato de Adenosina/metabolismo , Adulto , Células Cultivadas , Fibroblastos/enzimologia , Fibroblastos/ultraestrutura , Humanos , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Pele/citologia , Superóxido Dismutase-1
11.
PLoS One ; 9(12): e116289, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25551757

RESUMO

Multinucleated giant cells, formed by the fusion of monocytes/macrophages, are features of chronic granulomatous inflammation associated with infections or the persistent presence of foreign material. The tetraspanins CD9 and CD81 regulate multinucleated giant cell formation: soluble recombinant proteins corresponding to the large extracellular domain (EC2) of human but not mouse CD9 can inhibit multinucleated giant cell formation, whereas human CD81 EC2 can antagonise this effect. Tetraspanin EC2 are all likely to have a conserved three helix sub-domain and a much less well-conserved or hypervariable sub-domain formed by short helices and interconnecting loops stabilised by two or more disulfide bridges. Using CD9/CD81 EC2 chimeras and point mutants we have mapped the specific regions of the CD9 EC2 involved in multinucleated giant cell formation. These were primarily located in two helices, one in each sub-domain. The cysteine residues involved in the formation of the disulfide bridges in CD9 EC2 were all essential for inhibitory activity but a conserved glycine residue in the tetraspanin-defining 'CCG' motif was not. A tyrosine residue in one of the active regions that is not conserved between human and mouse CD9 EC2, predicted to be solvent-exposed, was found to be only peripherally involved in this activity. We have defined two spatially-distinct sites on the CD9 EC2 that are required for inhibitory activity. Agents that target these sites could have therapeutic applications in diseases in which multinucleated giant cells play a pathogenic role.


Assuntos
Células Gigantes/metabolismo , Tetraspanina 29/fisiologia , Sequência de Aminoácidos , Animais , Diferenciação Celular , Sequência Conservada , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação Puntual , Estrutura Terciária de Proteína , Alinhamento de Sequência , Tetraspanina 29/química , Tetraspanina 29/genética
12.
Mol Ther ; 21(8): 1486-96, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23732987

RESUMO

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder with no effective treatment to date. Despite its multi-factorial aetiology, oxidative stress is hypothesized to be one of the key pathogenic mechanisms. It is thus proposed that manipulation of the expression of antioxidant genes that are downregulated in the presence of mutant SOD1 may serve as a therapeutic strategy for motor neuronal protection. Lentiviral vectors expressing either PRDX3 or NRF2 genes were tested in the motor neuronal-like NSC34 cell line, and in the ALS tissue culture model, NSC34 cells expressing the human SOD1(G93A) mutation. The NSC34 SOD1(G93A) cells overexpressing either PRDX3 or NRF2 showed a significant decrease in endogenous oxidation stress levels by 40 and 50% respectively compared with controls, whereas cell survival was increased by 30% in both cases. The neuroprotective potential of those two genes was further investigated in vivo in the SOD1(G93A) ALS mouse model, by administering intramuscular injections of adenoassociated virus serotype 6 (AAV6) expressing either of the target genes at a presymptomatic stage. Despite the absence of a significant effect in survival, disease onset or progression, which can be explained by the inefficient viral delivery, the promising in vitro data suggest that a more widespread CNS delivery is needed.


Assuntos
Esclerose Lateral Amiotrófica/genética , Vetores Genéticos/genética , Estresse Oxidativo/genética , Transgenes , Esclerose Lateral Amiotrófica/terapia , Animais , Astrócitos/metabolismo , Linhagem Celular , Dependovirus/genética , Modelos Animais de Doenças , Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética , Humanos , Lentivirus/genética , Camundongos , Camundongos Transgênicos , Neurônios Motores/metabolismo , Fator 2 Relacionado a NF-E2/genética , Peroxirredoxina III/genética , Transdução de Sinais , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Transdução Genética
13.
Hum Mol Genet ; 19(16): 3159-68, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20525971

RESUMO

Phosphatase and tensin homolog (PTEN), a negative regulator of the mammalian target of rapamycin (mTOR) pathway, is widely involved in the regulation of protein synthesis. Here we show that the PTEN protein is enriched in cell bodies and axon terminals of purified motor neurons. We explored the role of the PTEN pathway by manipulating PTEN expression in healthy and diseased motor neurons. PTEN depletion led to an increase in growth cone size, promotion of axonal elongation and increased survival of these cells. These changes were associated with alterations of downstream signaling pathways for local protein synthesis as revealed by an increase in pAKT and p70S6. Most notably, this treatment also restores beta-actin protein levels in axonal growth cones of SMN-deficient motor neurons. Furthermore, we report here that a single injection of adeno-associated virus serotype 6 (AAV6) expressing siPTEN into hind limb muscles at postnatal day 1 in SMNDelta7 mice leads to a significant PTEN depletion and robust improvement in motor neuron survival. Taken together, these data indicate that PTEN-mediated regulation of protein synthesis in motor neurons could represent a target for therapy in spinal muscular atrophy.


Assuntos
Axônios/fisiologia , Neurônios Motores/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Actinas/genética , Análise de Variância , Animais , Axônios/metabolismo , Western Blotting , Sobrevivência Celular , Células Cultivadas , Cones de Crescimento/metabolismo , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Knockout , Neurônios Motores/citologia , PTEN Fosfo-Hidrolase/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Serina-Treonina Quinases TOR
14.
Immunology ; 127(2): 237-48, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19489128

RESUMO

Members of the tetraspanin superfamily of proteins are implicated in a variety of complex cell processes including cell fusion. However, the contribution of individual tetraspanins to these processes has proved difficult to define. Here we report the use of recombinant extracellular regions of tetraspanins to investigate the role of specific members of this family in the fusion of monocytes to form multinucleated giant cells (MGC). In contrast to their positive requirement in sperm-egg fusion, previous studies using antibodies and knockout mice have indicated a negative regulatory role for tetraspanins CD9 and CD81 in this process. In an in vitro model of fusion using human monocytes, we have confirmed observations that antibodies to CD9 and CD81 enhance MGC formation; however, in contrast to previous investigations, we found that all members of a panel of antibodies to CD63 inhibited fusion. Moreover, recombinant proteins corresponding to the large extracellular domains (EC2s) of CD63 and CD9 inhibited MGC formation, whereas the EC2s of CD81 and CD151 had no effect. The potent inhibition of fusion and binding of labelled CD63 EC2 to monocytes under fusogenic conditions suggest a direct interaction with a membrane component required for fusion. Our findings indicate that the tetraspanins CD9, CD63 and CD81 are all involved in MGC formation, but play distinct roles.


Assuntos
Antígenos CD/fisiologia , Células Gigantes/imunologia , Proteínas de Membrana/fisiologia , Adesão Celular/imunologia , Agregação Celular/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Concanavalina A/imunologia , Glutationa Transferase/imunologia , Humanos , Glicoproteínas de Membrana/fisiologia , Monócitos/imunologia , Glicoproteínas da Membrana de Plaquetas/fisiologia , Tetraspanina 28 , Tetraspanina 29 , Tetraspanina 30
15.
Mol Immunol ; 44(10): 2507-17, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17258808

RESUMO

The chemotaxis inhibitory protein of Staphylococcus aureus (CHIPS) is reported to bind to the receptors for C5a and formylated peptides and has been proposed as a promising lead for the development of new anti-inflammatory compounds. Here we have examined the receptor specificity and mode of action of recombinant CHIPS(28-149) and also the immune response to CHIPS(28-149) in patients with S. aureus infections and in uninfected controls. Recombinant CHIPS(28-149) bound with high affinity to the human C5a receptor (C5aR), but had low affinity for the second C5a receptor, C5L2, and the formyl peptide receptor, FPR. Although ligand binding to C5aR was potently inhibited, CHIPS(28-149) had much weaker effects on ligand binding to C5L2 and FPR. Similarly, CHIPS(28-149) potently inhibited the ligand-induced activation of C5aR but was less potent at inhibition via FPR. NMR studies showed that CHIPS(28-149) bound directly to the N-terminus of C5aR but not C5L2, and CHIPS(28-149) residues involved in the interaction were identified by chemical shift analysis. All human sera examined contained high titres of IgG and IgA reactivity against CHIPS(28-149), and no correlation was observed between infection status at the time of serum collection and antibody titre. Individual serum samples promoted or inhibited the binding of CHIPS(28-149) to C5aR, or had no effect. IgG depletion of serum samples abrogated the effects on CHIPS binding, demonstrating that these were antibody mediated. Sera from infected individuals were more likely to inhibit CHIPS(28-149) binding than sera from healthy controls. However, high antibody titres correlated well with both inhibition and enhancement of CHIPS(28-149) binding to C5aR; this suggests that the inhibitory effect relates to epitope specificity rather than greater antibody binding. We conclude that CHIPS is likely to be too immunogenic to be used as an anti-inflammatory treatment but that some antibodies against CHIPS may be useful in the treatment of S. aureus infections.


Assuntos
Proteínas de Bactérias/imunologia , Imunidade , Proteínas de Membrana/imunologia , Receptores de Complemento/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus , Animais , Anticorpos Antibacterianos/sangue , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Calorimetria , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Ressonância Magnética Nuclear Biomolecular , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Conformação Proteica , Mapeamento de Interação de Proteínas , Receptor da Anafilatoxina C5a , Receptores de Complemento/química , Receptores de Complemento/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
16.
J Biol Chem ; 282(6): 3664-71, 2007 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-17158873

RESUMO

C5L2 is a new cellular receptor found to interact with the human anaphylatoxins complement factor C5a and its C-terminal cleavage product C5a des Arg. The classical human C5a receptor (C5aR) preferentially binds C5a, with a 10-100-fold lower affinity for C5a des Arg. In contrast, C5L2 binds both ligands with nearly equal affinity. C5aR presents acidic and tyrosine residues in its N terminus that interact with the core of C5a while a hydrophobic pocket formed by the transmembrane helices interacts with residues in the C terminus of C5a. Here, we have investigated the molecular basis for the increased affinity of C5L2 for C5a des Arg. Rat and mouse C5L2 preferentially bound C5a des Arg, whereas rodent C5aR showed much higher affinity for intact C5a. Effective peptidic and non-peptidic ligands for the transmembrane hydrophobic pocket of C5aR were poor inhibitors of ligand binding to C5L2. An antibody raised against the N terminus of human C5L2 did not affect the binding of C5a to C5L2 but did inhibit C5a des Arg binding. A chimeric C5L2, containing the N terminus of C5aR, had little effect on the affinity for C5a des Arg. Mutation of acidic and tyrosine residues in the N terminus of human C5L2 revealed that 3 residues were critical for C5a des Arg binding but had little involvement in C5a binding. C5L2 thus appears to bind C5a and C5a des Arg by different mechanisms, and, unlike C5aR, C5L2 uses critical residues in its N-terminal domain for binding only to C5a des Arg.


Assuntos
Fragmentos de Peptídeos/fisiologia , Receptor da Anafilatoxina C5a/fisiologia , Substituição de Aminoácidos/genética , Animais , Sítios de Ligação/genética , Células CHO , Linhagem Celular Tumoral , Complemento C5a des-Arginina/metabolismo , Cricetinae , Cricetulus , Humanos , Ligantes , Camundongos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Ligação Proteica/genética , Ratos , Receptor da Anafilatoxina C5a/genética , Receptor da Anafilatoxina C5a/metabolismo
17.
J Virol ; 80(13): 6487-96, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16775336

RESUMO

Human immunodeficiency virus type 1 (HIV-1) infection of human macrophages can be inhibited by antibodies which bind to the tetraspanin protein CD63, but not by antibodies that bind to other members of the tetraspanin family. This inhibitory response was limited to CCR5 (R5)-tropic virus and was only observed using macrophages, but not T cells. Here, we show that recombinant soluble forms of the large extracellular domain (EC2) of human tetraspanins CD9, CD63, CD81, and CD151 produced as fusion proteins with glutathione S-transferase (GST) can all potently and completely inhibit R5 HIV-1 infection of macrophages with 50% inhibitory concentration values of 0.11 to 1.2 nM. Infection of peripheral blood mononuclear cells could also be partly inhibited, although higher concentrations of EC2 proteins were required. Inhibition was largely coreceptor independent, as macrophage infections by virions pseudotyped with CXCR4 (X4)-tropic HIV-1 or vesicular stomatitis virus (VSV)-G glycoproteins were also inhibited, but was time dependent, since addition prior to or during, but not after, virus inoculation resulted in potent inhibition. Incubation with tetraspanins did not decrease CD4 or HIV-1 coreceptor expression but did block virion uptake. Colocalization of fluorescently labeled tetraspanin EC2 proteins and HIV-1 virions within, and with CD4 and CXCR4 at the cell surfaces of, macrophages could be detected, and internalized tetraspanin EC2 proteins were directed to vesicular compartments that contained internalized dextran and transferrin. Collectively, the data suggest that the mechanism of inhibition of HIV-1 infection by tetraspanins is at the step of virus entry, perhaps via interference with binding and/or the formation of CD4-coreceptor complexes within microdomains that are required for membrane fusion events.


Assuntos
Antígenos CD/farmacologia , Infecções por HIV/metabolismo , HIV-1/metabolismo , Macrófagos/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Anticorpos/farmacologia , Antígenos CD/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Endocitose/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Infecções por HIV/tratamento farmacológico , Humanos , Macrófagos/virologia , Microdomínios da Membrana/metabolismo , Estrutura Terciária de Proteína , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/virologia , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/farmacologia , Inativação de Vírus/efeitos dos fármacos
18.
Brain ; 129(Pt 7): 1693-709, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16702190

RESUMO

There is now compelling evidence of mitochondrial dysfunction in motor neuron disease (MND), but the molecular basis of these abnormalities is unknown. It is also unclear whether the observed mitochondrial dysfunction plays a central role in disease pathogenesis, and if so, whether its amelioration might present therapeutic opportunities. We adopted a candidate generation approach using proteomics to screen for changes in mitochondrial protein expression in a well-validated cell-culture model of superoxide dismutase 1 (SOD1) related familial MND (fMND). Changed proteins were identified by matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectroscopy. Protein candidates included apoptotic regulators, anti-oxidants and components of the electron transport chain. Confirmatory Western blotting was performed, and validated protein expression changes were further investigated. Peroxiredoxin 3 (Prx3), a mitochondrial thioredoxin-dependent hydroperoxidase, is downregulated in the presence of mutant SOD1 in both our cell-culture model and in the spinal cord mitochondria of mutant SOD1 transgenic mice. We confirm the expression of Prx3 within the mitochondria of spinal motor neurons in mouse and humans by immunohistochemistry. Using quantitative real-time PCR (Q-PCR), we show that Prx3 is also downregulated in spinal motor neurons from patients with both sporadic (sMND) and SOD1-related fMND. In a disease characterized by oxidative stress, this represents a potentially important deficit in mitochondrial anti-oxidant defence. Recent evidence suggests that oxidative stress from aberrant copper chemistry may not play a major part in the pathogenesis of SOD1-related fMND. From the results of this study we propose disruption of mitochondrial anti-oxidant defence as an alternative mechanism whereby mutant SOD1 may generate oxidative stress within motor neurons. We further demonstrate that ebselen, an anti-oxidant drug already safely used in human studies and that acts as a Prx mimic, is able to ameliorate the toxicity of mutant SOD1 in our cell-culture model. We conclude by showing that ebselen is capable of inducing transcription of the anti-oxidant response element (ARE) and postulate that ebselen may act both by the transcriptional upregulation of anti-oxidant proteins, and directly as an anti-oxidant in its own right.


Assuntos
Antioxidantes/farmacologia , Azóis/farmacologia , Doenças Mitocondriais/metabolismo , Doença dos Neurônios Motores/metabolismo , Compostos Organosselênicos/farmacologia , Superóxido Dismutase/genética , Animais , Antioxidantes/metabolismo , Western Blotting/métodos , Morte Celular/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo , Eletroforese em Gel de Poliacrilamida/métodos , Humanos , Isoindóis , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Doenças Mitocondriais/genética , Doença dos Neurônios Motores/genética , Proteínas de Neoplasias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Peroxidases/metabolismo , Peroxirredoxina III , Peroxirredoxinas , Reação em Cadeia da Polimerase/métodos , Proteômica , Elementos de Resposta/efeitos dos fármacos , Medula Espinal/metabolismo , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
19.
Blood ; 105(7): 2852-61, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15591117

RESUMO

Tetraspanins associate with several transmembrane proteins forming microdomains involved in intercellular adhesion and migration. Here, we show that endothelial tetraspanins relocalize to the contact site with transmigrating leukocytes and associate laterally with both intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1). Alteration of endothelial tetraspanin microdomains by CD9-large extracellular loop (LEL)-glutathione S-transferase (GST) peptides or CD9/CD151 siRNA oligonucleotides interfered with ICAM-1 and VCAM-1 function, preventing lymphocyte transendothelial migration and increasing lymphocyte detachment under shear flow. Heterotypic intercellular adhesion mediated by VCAM-1 or ICAM-1 was augmented when expressed exogenously in the appropriate tetraspanin environment. Therefore, tetraspanin microdomains have a crucial role in the proper adhesive function of ICAM-1 and VCAM-1 during leukocyte adhesion and transendothelial migration.


Assuntos
Antígenos CD/metabolismo , Adesão Celular/imunologia , Endotélio Vascular/citologia , Leucócitos/citologia , Glicoproteínas de Membrana/metabolismo , Microdomínios da Membrana/imunologia , Antígenos CD/química , Antígenos CD/genética , Movimento Celular/imunologia , Células Cultivadas , Endotélio Vascular/imunologia , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Leucócitos/fisiologia , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Estrutura Terciária de Proteína , RNA Interferente Pequeno , Tetraspanina 24 , Tetraspanina 29 , Veias Umbilicais/citologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
20.
J Virol ; 78(3): 1448-55, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14722300

RESUMO

CD81 has been described as a putative receptor for hepatitis C virus (HCV); however, its role in HCV cell entry has not been characterized due to the lack of an efficient cell culture system. We have examined the role of CD81 in HCV glycoprotein-dependent entry by using a recently developed retroviral pseudotyping system. Human immunodeficiency virus (HIV) pseudotypes bearing HCV E1E2 glycoproteins show a restricted tropism for human liver cell lines. Although all of the permissive cell lines express CD81, CD81 expression alone is not sufficient to allow viral entry. CD81 is required for HIV-HCV pseudotype infection since (i) a monoclonal antibody specific for CD81 inhibited infection of susceptible target cells and (ii) silencing of CD81 expression in Huh-7.5 hepatoma cells by small interfering RNAs inhibited HIV-HCV pseudotype infection. Furthermore, expression of CD81 in human liver cells that were previously resistant to infection, HepG2 and HH29, conferred permissivity of HCV pseudotype infection. The characterization of chimeric CD9/CD81 molecules confirmed that the large extracellular loop of CD81 is a determinant for viral entry. These data suggest a functional role for CD81 as a coreceptor for HCV glycoprotein-dependent viral cell entry.


Assuntos
Antígenos CD/metabolismo , Hepacivirus/patogenicidade , Proteínas de Membrana/metabolismo , Receptores Virais/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Estruturais Virais/metabolismo , Animais , Linhagem Celular , Células Epiteliais/virologia , Proteína gp160 do Envelope de HIV/genética , Proteína gp160 do Envelope de HIV/metabolismo , HIV-1/genética , HIV-1/fisiologia , Hepacivirus/genética , Hepacivirus/fisiologia , Hepatócitos/virologia , Humanos , Linfócitos/virologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Tetraspanina 28 , Proteínas do Envelope Viral/genética , Proteínas Estruturais Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA