Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 14(12): e0226245, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31887144

RESUMO

Antibody therapies for Alzheimer's Disease (AD) hold promise but have been limited by the inability of these proteins to migrate efficiently across the blood brain barrier (BBB). Central nervous system (CNS) gene transfer by vectors like adeno-associated virus (AAV) overcome this barrier by allowing the bodies' own cells to produce the therapeutic protein, but previous studies using this method to target amyloid-ß have shown success only with truncated single chain antibodies (Abs) lacking an Fc domain. The Fc region mediates effector function and enhances antigen clearance from the brain by neonatal Fc receptor (FcRn)-mediated reverse transcytosis and is therefore desirable to include for such treatments. Here, we show that single chain Abs fused to an Fc domain retaining FcRn binding, but lacking Fc gamma receptor (FcγR) binding, termed a silent scFv-IgG, can be expressed and released into the CNS following gene transfer with AAV. While expression of canonical IgG in the brain led to signs of neurotoxicity, this modified Ab was efficiently secreted from neuronal cells and retained target specificity. Steady state levels in the brain exceeded peak levels obtained by intravenous injection of IgG. AAV-mediated expression of this scFv-IgG reduced cortical and hippocampal plaque load in a transgenic mouse model of progressive ß-amyloid plaque accumulation. These findings suggest that CNS gene delivery of a silent anti-Aß scFv-IgG was well-tolerated, durably expressed and functional in a relevant disease model, demonstrating the potential of this modality for the treatment of Alzheimer's disease.


Assuntos
Doença de Alzheimer/terapia , Sistema Nervoso Central/metabolismo , Vetores Genéticos/administração & dosagem , Fragmentos Fc das Imunoglobulinas/genética , Anticorpos de Cadeia Única/genética , Doença de Alzheimer/genética , Animais , Barreira Hematoencefálica , Linhagem Celular , Dependovirus/genética , Modelos Animais de Doenças , Progressão da Doença , Terapia Genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/metabolismo , Camundongos , Camundongos Transgênicos , Domínios Proteicos , Receptores Fc/metabolismo , Receptores de IgG/metabolismo , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/metabolismo
2.
PLoS One ; 10(6): e0130517, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26107373

RESUMO

Respiratory syncytial virus (RSV) is the principal cause of bronchiolitis in infants and a significant healthcare problem. The RSV Glycoprotein (G) mediates attachment of the virus to the cell membrane, which facilitates interaction of the RSV Fusion (F) protein with nucleolin, thereby triggering fusion of the viral and cellular membranes. However, a host protein ligand for G has not yet been identified. Here we show that CX3CR1 is expressed in the motile cilia of differentiated human airway epithelial (HAE) cells, and that CX3CR1 co-localizes with RSV particles. Upon infection, the distribution of CX3CR1 in these cells is significantly altered. Complete or partial deletion of RSV G results in viruses binding at least 72-fold less efficiently to cells, and reduces virus replication. Moreover, an antibody targeting an epitope near the G protein's CX3CR1-binding motif significantly inhibits binding of the virus to airway cells. Given previously published evidence of the interaction of G with CX3CR1 in human lymphocytes, these findings suggest a role for G in the interaction of RSV with ciliated lung cells. This interpretation is consistent with past studies showing a protective benefit in immunizing against G in animal models of RSV infection, and would support targeting the CX3CR1-G protein interaction for prophylaxis or therapy. CX3CR1 expression in lung epithelial cells may also have implications for other respiratory diseases such as asthma.


Assuntos
Células Epiteliais/metabolismo , Receptores de Quimiocinas/genética , Mucosa Respiratória/metabolismo , Vírus Sincicial Respiratório Humano/genética , Proteínas do Envelope Viral/genética , Proteínas Virais de Fusão/genética , Anticorpos/farmacologia , Sequência de Bases , Sítios de Ligação , Receptor 1 de Quimiocina CX3C , Diferenciação Celular , Criança , Cílios/metabolismo , Cílios/patologia , Cílios/virologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Epitopos/química , Epitopos/imunologia , Expressão Gênica , Humanos , Dados de Sequência Molecular , Cultura Primária de Células , Ligação Proteica , Receptores de Quimiocinas/antagonistas & inibidores , Receptores de Quimiocinas/química , Receptores de Quimiocinas/metabolismo , Mucosa Respiratória/patologia , Mucosa Respiratória/virologia , Vírus Sincicial Respiratório Humano/metabolismo , Deleção de Sequência , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo
3.
Nucleic Acid Ther ; 23(2): 109-17, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23308382

RESUMO

Expansions of CUG trinucleotide sequences in RNA transcripts provide the basis for toxic RNA gain-of-function that leads to detrimental changes in RNA metabolism. A CTG repeat element normally resides in the 3' untranslated region of the dystrophia myotonica-protein kinase (DMPK) gene, but when expanded it is the genetic lesion of myotonic dystrophy type 1 (DM1), a hereditary neuromuscular disease. The pathogenic DMPK transcript containing the CUG expansion is retained in ribonuclear foci as part of a complex with RNA-binding proteins such as muscleblind-like 1 (MBNL1), resulting in aberrant splicing of numerous RNA transcripts and consequent physiological abnormalities including myotonia. Herein, we demonstrate molecular and physiological amelioration of the toxic effects of mutant RNA in the HSA(LR) mouse model of DM1 by systemic administration of peptide-linked morpholino (PPMO) antisense oligonucleotides bearing a CAG repeat sequence. Intravenous administration of PPMO conjugates to HSA(LR) mice led to redistribution of Mbnl1 protein in myonuclei and corrections in abnormal RNA splicing. Additionally, myotonia was completely eliminated in PPMO-treated HSA(LR) mice. These studies provide proof of concept that neutralization of RNA toxicity by systemic delivery of antisense oligonucleotides that target the CUG repeat is an effective therapeutic approach for treating the skeletal muscle aspects of DM1 pathology.


Assuntos
Morfolinos/administração & dosagem , Distrofia Miotônica/genética , Peptídeos/administração & dosagem , Proteínas de Ligação a RNA/genética , Regiões 3' não Traduzidas/genética , Animais , Humanos , Camundongos , Morfolinos/química , Mutação , Distrofia Miotônica/metabolismo , Distrofia Miotônica/patologia , Miotonina Proteína Quinase , Oligonucleotídeos Antissenso/administração & dosagem , Peptídeos/química , Proteínas Serina-Treonina Quinases/genética , RNA/genética , RNA/toxicidade , Splicing de RNA/genética , Expansão das Repetições de Trinucleotídeos/genética , Repetições de Trinucleotídeos/genética
4.
J Inherit Metab Dis ; 34(3): 795-809, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21416197

RESUMO

Lysosomal storage diseases (LSDs) are metabolic disorders caused by enzyme deficiencies that lead to lysosomal accumulation of undegraded substrates. Enzyme replacement therapies (ERT) have been developed as treatments for patients with Gaucher, Niemann-Pick, Fabry, and Pompe diseases. Depending on the disease, the corresponding therapeutic enzyme is designed to be internalized by diseased cells through receptor-mediated endocytosis via macrophage mannose receptors (MMR) or mannose-6-phosphate receptors (M6PR). Enzymes developed to treat Gaucher and Niemann-Pick diseases are meant to target MMR-expressing cells, and in the case of Cerezyme [recombinant human ß-glucocerebrosidase (rhßGC)] for treating Gaucher disease, glycans on the enzyme are modified to increase specificity toward this receptor. Due to heterogeneity in glycosylation on enzymes intended to target the M6PR, however, there may also be some unintended targeting to MMR-expressing cells, which could act as unwanted sinks. Examples include Fabrazyme [recombinant human α-galactosidase A (rhαGal)] for treating Fabry disease and Myozyme [recombinant human acid α-glucosidase (rhGAA)] for treating Pompe disease. It is therefore of great interest to better understand the cell type and tissue distribution of MMR in murine LSD models used to evaluate ERT efficacy and mechanism of action. In this study, we generated affinity-purified polyclonal antibody against murine MMR and used it to carry out a systematic examination of MMR protein localization in murine models of Gaucher, Niemann-Pick, Fabry, and Pompe diseases. Using immunohistochemistry, immunofluorescence, and confocal microscopy, we examined MMR distribution in liver, spleen, lung, kidney, heart, diaphragm, quadriceps, and triceps in these animal models and compared them with MMR distribution in wild-type mice.


Assuntos
Lectinas Tipo C/análise , Lectinas Tipo C/metabolismo , Doenças por Armazenamento dos Lisossomos/metabolismo , Lectinas de Ligação a Manose/análise , Lectinas de Ligação a Manose/metabolismo , Receptores de Superfície Celular/análise , Receptores de Superfície Celular/metabolismo , Animais , Anticorpos , Formação de Anticorpos , Linhagem Celular , Modelos Animais de Doenças , Descoberta de Drogas/métodos , Humanos , Fígado/metabolismo , Fígado/patologia , Doenças por Armazenamento dos Lisossomos/patologia , Macrófagos/metabolismo , Receptor de Manose , Camundongos , Camundongos Knockout , Terapia de Alvo Molecular/métodos , Músculos/metabolismo , Músculos/patologia , Baço/metabolismo , Baço/patologia , Distribuição Tecidual
5.
Microsc Res Tech ; 73(7): 694-703, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19953667

RESUMO

Efficient targeting of therapeutic reagents to tissues and cell types of interest is critical to achieving therapeutic efficacy and avoiding unwanted side effects due to offtarget uptake. To increase assay efficiency and reduce the number of animals used per experiment during preclinical development, we used a combination of direct fluorescence labeling and confocal microscopy to simultaneously examine the biodistribution of two therapeutic proteins, Cerezyme and Ceredase, in the same animals. We show that the fluorescent tags do not interfere with protein uptake and localization. We are able to detect Cerezyme and Ceredase in intact cells and organs and demonstrate colocalization within target cells using confocal microscopy. In addition, the relative amount of protein internalized by different cell types can be quantified using cell type-specific markers and morphometric analysis. This approach provides an easy and straightforward means of assessing the tissue and cell type-specific biodistribution of multiple protein therapeutics in target organs using a minimal number of animals.


Assuntos
Terapia de Reposição de Enzimas/métodos , Fluorescência , Glucosilceramidase/farmacocinética , Microscopia Confocal/métodos , Coloração e Rotulagem/métodos , Estruturas Animais/química , Estruturas Animais/citologia , Animais , Glucosilceramidase/administração & dosagem , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos C57BL
6.
Am J Pathol ; 174(6): 2137-49, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19406991

RESUMO

Transforming growth factor-beta (TGF-beta) is a pleiotropic growth factor; its overexpression has been implicated in many diseases, making it a desirable target for therapeutic neutralization. In initial safety studies, mice were chronically treated (three times per week) with high doses (50 mg/kg) of a murine, pan-neutralizing, anti-TGF-beta antibody. Nine weeks after the initiation of treatment, a subset of mice exhibited weight loss that was concurrent with decreased food intake. Histopathology revealed a unique, nonneoplastic cystic epithelial hyperplasia and tongue inflammation, as well as dental dysplasia and epithelial hyperplasia and inflammation of both the gingiva and esophagus. In an effort to determine the cause of this site-specific pathology, we examined TGF-beta expression in these tissues and saliva under normal conditions. By immunostaining, we found higher expression levels of active TGF-beta1 and TGF-beta3 in normal tongue and esophageal submucosa compared with gut mucosal tissues, as well as detectable TGF-beta1 in normal saliva by Western blot analysis. Interestingly, mast cells within the tongue, esophagus, and skin co-localized predominantly with the TGF-beta1 expressed in these tissues. Our findings demonstrate a novel and restricted pathology in oral and esophageal tissues of mice chronically treated with anti-TGF-beta that is associated with basal TGF-beta expression in saliva and by mast cells within these tissues. These studies illustrate a previously unappreciated biological role of TGF-beta in maintaining homeostasis within both oral and esophageal tissues.


Assuntos
Esôfago/metabolismo , Homeostase/fisiologia , Mastócitos/metabolismo , Boca/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Western Blotting , Esôfago/imunologia , Esôfago/patologia , Feminino , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Boca/imunologia , Boca/patologia , Saliva/química , Saliva/imunologia
7.
Glycobiology ; 17(5): 467-78, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17251309

RESUMO

Recombinant human glucocerebrosidase (imiglucerase, Cerezyme) is used in enzyme replacement therapy for Gaucher disease. Complex oligosaccharides present on Chinese hamster ovary cell-expressed glucocerebrosidase (GCase) are enzymatically remodeled into a mannose core, facilitating mannose receptor-mediated uptake into macrophages. Alternative expression systems could be used to produce GCase containing larger oligomannose structures, offering the possibility of an improvement in targeting to macrophages. A secondary advantage of these expression systems would be to eliminate the need for carbohydrate remodeling. Here, multiple expression systems were used to produce GCase containing primarily terminal oligomannose, from Man2 to Man9. GCase from these multiple expression systems was compared to Cerezyme with respect to affinity for mannose receptor and serum mannose-binding lectin (MBL), macrophage uptake, and intracellular half-life. In vivo studies comparing clearance and targeting of Cerezyme and the Man9 form of GCase were carried out in a Gaucher mouse model (D409V/null). Mannose receptor binding, macrophage uptake, and in vivo targeting were similar for all forms of GCase. Increased MBL binding was observed for all forms of GCase having larger mannose structures than those of Cerezyme, which could influence pharmacokinetic behavior. These studies demonstrate that although alternative cell expression systems are effective for producing oligomannose-terminated glucocerebrosidase, there is no biochemical or pharmacological advantage in producing GCase with an increased number of mannose residues. The display of alternative carbohydrate structures on GCase expressed in these systems also runs the risk of undesirable consequences, such as an increase in MBL binding or a possible increase in immunogenicity due to the presentation of non-mammalian glycans.


Assuntos
Doença de Gaucher/enzimologia , Glucosilceramidase/biossíntese , Manose/metabolismo , Oligossacarídeos/biossíntese , Modificação Traducional de Proteínas/fisiologia , Animais , Células CHO , Cricetinae , Cricetulus , Sistemas de Liberação de Medicamentos , Doença de Gaucher/tratamento farmacológico , Doença de Gaucher/genética , Doença de Gaucher/imunologia , Expressão Gênica , Glucosilceramidase/administração & dosagem , Glucosilceramidase/genética , Glucosilceramidase/imunologia , Glicosilação , Humanos , Lectinas Tipo C/imunologia , Lectinas Tipo C/metabolismo , Manose/genética , Manose/imunologia , Receptor de Manose , Lectina de Ligação a Manose/imunologia , Lectina de Ligação a Manose/metabolismo , Lectinas de Ligação a Manose/imunologia , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Knockout , Oligossacarídeos/genética , Oligossacarídeos/imunologia , Polissacarídeos/imunologia , Polissacarídeos/metabolismo , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Especificidade da Espécie
8.
Mol Ther ; 11(5): 754-62, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15851014

RESUMO

Niemann-Pick A disease (NPA) is a fatal lysosomal storage disorder caused by a deficiency in acid sphingomyelinase (ASM) activity. The lack of functional ASM results in cellular accumulation of sphingomyelin and cholesterol within distended lysosomes throughout the brain. In this study, we investigated the potential of AAV-mediated expression of ASM to correct the brain pathology in an ASM knockout (ASMKO) mouse model of NPA. An AAV serotype 2 vector encoding human ASM (AAV2-hASM) was injected directly into the adult ASMKO hippocampus of one hemisphere. This resulted in expression of human ASM in all major cell layers of the ipsilateral hippocampus for at least 15 weeks postinjection. Transduced cells were also present in the entorhinal cortex, medial septum, and contralateral hippocampus in a pattern consistent with retrograde axonal transport of AAV2. There was a substantial reduction of distended lysosomes and an almost complete reversal of cholesterol accumulation in all areas of the brain that were targeted by AAV2-hASM. These findings show that the ASMKO brain is responsive to ASM replacement and that retrograde transport of AAV2 functions as a platform for widespread gene delivery and reversal of pathology in affected brain.


Assuntos
Encéfalo/patologia , Dependovirus/genética , Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos/genética , Doenças de Niemann-Pick/genética , Doenças de Niemann-Pick/patologia , Animais , Encéfalo/metabolismo , Colesterol/metabolismo , Humanos , Lisossomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças de Niemann-Pick/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Esfingomielina Fosfodiesterase/deficiência , Esfingomielina Fosfodiesterase/genética , Esfingomielina Fosfodiesterase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...