Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 9(11)2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38753465

RESUMO

Glycogen storage disease type III (GSDIII) is a rare metabolic disorder due to glycogen debranching enzyme (GDE) deficiency. Reduced GDE activity leads to pathological glycogen accumulation responsible for impaired hepatic metabolism and muscle weakness. To date, there is no curative treatment for GSDIII. We previously reported that 2 distinct dual AAV vectors encoding for GDE were needed to correct liver and muscle in a GSDIII mouse model. Here, we evaluated the efficacy of rapamycin in combination with AAV gene therapy. Simultaneous treatment with rapamycin and a potentially novel dual AAV vector expressing GDE in the liver and muscle resulted in a synergic effect demonstrated at biochemical and functional levels. Transcriptomic analysis confirmed synergy and suggested a putative mechanism based on the correction of lysosomal impairment. In GSDIII mice livers, dual AAV gene therapy combined with rapamycin reduced the effect of the immune response to AAV observed in this disease model. These data provide proof of concept of an approach exploiting the combination of gene therapy and rapamycin to improve efficacy and safety and to support clinical translation.


Assuntos
Dependovirus , Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos , Fígado , Sirolimo , Animais , Sirolimo/farmacologia , Sirolimo/uso terapêutico , Dependovirus/genética , Terapia Genética/métodos , Camundongos , Fígado/metabolismo , Vetores Genéticos/genética , Vetores Genéticos/administração & dosagem , Músculo Esquelético/metabolismo , Fenótipo , Sistema da Enzima Desramificadora do Glicogênio/genética , Sistema da Enzima Desramificadora do Glicogênio/metabolismo , Humanos , Masculino
2.
J Clin Invest ; 134(2)2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38015640

RESUMO

Glycogen storage disease type III (GSDIII) is a rare inborn error of metabolism affecting liver, skeletal muscle, and heart due to mutations of the AGL gene encoding for the glycogen debranching enzyme (GDE). No curative treatment exists for GSDIII. The 4.6 kb GDE cDNA represents the major technical challenge toward the development of a single recombinant adeno-associated virus-derived (rAAV-derived) vector gene therapy strategy. Using information on GDE structure and molecular modeling, we generated multiple truncated GDEs. Among them, an N-terminal-truncated mutant, ΔNter2-GDE, had a similar efficacy in vivo compared with the full-size enzyme. A rAAV vector expressing ΔNter2-GDE allowed significant glycogen reduction in heart and muscle of Agl-/- mice 3 months after i.v. injection, as well as normalization of histology features and restoration of muscle strength. Similarly, glycogen accumulation and histological features were corrected in a recently generated Agl-/- rat model. Finally, transduction with rAAV vectors encoding ΔNter2-GDE corrected glycogen accumulation in an in vitro human skeletal muscle cellular model of GSDIII. In conclusion, our results demonstrated the ability of a single rAAV vector expressing a functional mini-GDE transgene to correct the muscle and heart phenotype in multiple models of GSDIII, supporting its clinical translation to patients with GSDIII.


Assuntos
Sistema da Enzima Desramificadora do Glicogênio , Doença de Depósito de Glicogênio Tipo III , Humanos , Camundongos , Ratos , Animais , Doença de Depósito de Glicogênio Tipo III/genética , Doença de Depósito de Glicogênio Tipo III/terapia , Sistema da Enzima Desramificadora do Glicogênio/genética , Músculo Esquelético/metabolismo , Glicogênio/metabolismo , Transgenes
3.
Nat Commun ; 12(1): 6393, 2021 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-34737297

RESUMO

Pompe disease (PD) is a severe neuromuscular disorder caused by deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA). PD is currently treated with enzyme replacement therapy (ERT) with intravenous infusions of recombinant human GAA (rhGAA). Although the introduction of ERT represents a breakthrough in the management of PD, the approach suffers from several shortcomings. Here, we developed a mouse model of PD to compare the efficacy of hepatic gene transfer with adeno-associated virus (AAV) vectors expressing secretable GAA with long-term ERT. Liver expression of GAA results in enhanced pharmacokinetics and uptake of the enzyme in peripheral tissues compared to ERT. Combination of gene transfer with pharmacological chaperones boosts GAA bioavailability, resulting in improved rescue of the PD phenotype. Scale-up of hepatic gene transfer to non-human primates also successfully results in enzyme secretion in blood and uptake in key target tissues, supporting the ongoing clinical translation of the approach.


Assuntos
Doença de Depósito de Glicogênio Tipo II/enzimologia , alfa-Glucosidases/metabolismo , Animais , Autofagia , Terapia de Reposição de Enzimas , Feminino , Doença de Depósito de Glicogênio Tipo II/terapia , Fígado/enzimologia , Masculino , Camundongos , alfa-Glucosidases/genética
4.
Mol Ther Methods Clin Dev ; 14: 285-299, 2019 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-31497619

RESUMO

Unexpectedly, the synthetic antioxidant MnTBAP was found to cause a rapid and reversible downregulation of CD4 on T cells in vitro and in vivo. This effect resulted from the internalization of membrane CD4 T cell molecules into clathrin-coated pits and involved disruption of the CD4/p56Lck complex. The CD4 deprivation induced by MnTBAP had functional consequences on CD4-dependent infectious processes or immunological responses as shown in various models, including gene therapy. In cultured human T cells, MnTBAP-induced downregulation of CD4 functionally suppressed gp120- mediated lentiviral transduction in a model relevant for HIV infection. The injection of MnTBAP in mice reduced membrane CD4 on lymphocytes in vivo within 5 days of treatment, preventing OVA peptide T cell immunization while allowing subsequent immunization once treatment was stopped. In a mouse gene therapy model, MnTBAP treatment at the time of adenovirus-associated virus (AAV) vector administration, successfully controlled the induction of anti-transgene and anti-capsid immune responses mediated by CD4+ T cells, enabling the redosing mice with the same vector. These functional data provide new avenues to develop alternative therapeutic immunomodulatory strategies based on temporary regulation of CD4. These could be particularly useful for AAV gene therapy in which novel strategies for redosing are needed.

5.
Acta Biomater ; 64: 259-268, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29017974

RESUMO

Gene transfer using lentiviral vectors has therapeutic applications spanning from monogenic and infectious diseases to cancer. Such gene therapy has to be improved by enhancing the levels of viral infection of target cells and/or reducing the amount of lentivirus for greater safety and reduced costs. Vectofusin-1, a recently developed cationic amphipathic peptide with a pronounced capacity to enhance such viral transduction, strongly promotes the entry of several retroviral pseudotypes into target cells when added to the culture medium. To clarify the molecular basis of its action the peptide was investigated on a molecular and a supramolecular level by a variety of biophysical approaches. We show that in culture medium vectofusin-1 rapidly forms complexes in the 10 nm range that further assemble into annular and extended nanofibrils. These associate with viral particles allowing them to be easily pelleted for optimal virus-cell interaction. Thioflavin T fluorescence, circular dichroism and infrared spectroscopies indicate that these fibrils have a unique α-helical structure whereas most other viral transduction enhancers form ß-amyloid fibrils. A vectofusin-1 derivative (LAH2-A4) is inefficient in biological assays and does not form nanofibrils, suggesting that supramolecular assembly is essential for transduction enhancement. Our observations define vectofusin-1 as a member of a new class of α-helical enhancers of lentiviral infection. Its fibril formation is reversible which bears considerable advantages in handling the peptide in conditions well-adapted to Good Manufacturing Practices and scalable gene therapy protocols.


Assuntos
Lentivirus , Nanofibras/química , Peptídeos , Transdução Genética/métodos , Vírion/química , Linhagem Celular , Humanos , Peptídeos/química , Peptídeos/farmacologia
6.
J Biol Chem ; 292(45): 18672-18681, 2017 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-28928217

RESUMO

Autophagy-related proteins such as Beclin-1 are involved in an array of complex processes, including antiviral responses, and may also modulate the efficiency of gene therapy viral vectors. The Tat-Beclin-1 (TB1) peptide has been reported as an autophagy-inducing factor inhibiting the replication of pathogens such as HIV, type 1 (HIV-1). However, autophagy-related proteins are also essential for the early steps of HIV-1 infection. Therefore, we examined the effects of the Beclin-1 evolutionarily conserved domain in TB1 on viral transduction and autophagy in single-round HIV infection or with nonreplicative HIV-1-derived lentiviral vectors. TB1 enhanced transduction with various pseudotypes but without inducing the autophagy process. TB1 augmented the transduction of human CD34+ hematopoietic stem/progenitor cells while maintaining their capacity to engraft in vivo into humanized mice. TB1 was as effective as other transduction additives and functioned by enhancing the adhesion and fusion of viral particles with target cells but not their aggregation. We also found that the N-terminal L1 loop was critical for TB1 transduction-enhancing activity. Interestingly, the Tat-Beclin-2 (TB2) peptide, derived from the human Beclin-2 protein, was even more potent than TB1 in promoting viral transduction and infection. Taken together, our findings suggest that the TB1 and TB2 peptides enhance the viral entry step. Tat-Beclin peptides therefore represent a new family of viral transduction enhancers for potential use in gene therapy.


Assuntos
Autofagia , Proteína Beclina-1/metabolismo , HIV-1/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lentivirus/fisiologia , Internalização do Vírus , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Sequência de Aminoácidos , Animais , Proteína Beclina-1/química , Proteína Beclina-1/genética , Linhagem Celular Transformada , Linhagem Celular Tumoral , Células Cultivadas , Sequência Conservada , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/virologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos Transgênicos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Organismos Livres de Patógenos Específicos , Regulação para Cima , Produtos do Gene tat do Vírus da Imunodeficiência Humana/química , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
7.
PLoS Biol ; 15(7): e2001867, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28749943

RESUMO

Individual cells take lineage commitment decisions in a way that is not necessarily uniform. We address this issue by characterising transcriptional changes in cord blood-derived CD34+ cells at the single-cell level and integrating data with cell division history and morphological changes determined by time-lapse microscopy. We show that major transcriptional changes leading to a multilineage-primed gene expression state occur very rapidly during the first cell cycle. One of the 2 stable lineage-primed patterns emerges gradually in each cell with variable timing. Some cells reach a stable morphology and molecular phenotype by the end of the first cell cycle and transmit it clonally. Others fluctuate between the 2 phenotypes over several cell cycles. Our analysis highlights the dynamic nature and variable timing of cell fate commitment in hematopoietic cells, links the gene expression pattern to cell morphology, and identifies a new category of cells with fluctuating phenotypic characteristics, demonstrating the complexity of the fate decision process (which is different from a simple binary switch between 2 options, as it is usually envisioned).


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Multipotentes/metabolismo , Transcrição Gênica , Antígeno AC133/genética , Antígeno AC133/metabolismo , Antígenos CD34/genética , Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Forma Celular , Rastreamento de Células , Células Cultivadas , Sangue Fetal/citologia , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Humanos , Processamento de Imagem Assistida por Computador , Microscopia Confocal , Células-Tronco Multipotentes/citologia , Análise de Componente Principal , Análise de Célula Única , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo , Imagem com Lapso de Tempo
8.
Cytometry A ; 91(3): 254-260, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28248454

RESUMO

Cell differentiation is a longitudinal and dynamic process. Studying and quantifying such a process require tools combining precise time resolution and statistical power. Imaging flow cytometry (IFC) provides statistically significant number of microscopy images of individual cells in a sample at a given time point. Time-lapse microscopy (TLM) is the method of choice for studying the dynamics of cell processes at a high temporal, but low statistical resolution. In this work, we show that the dynamic changes of cord-blood derived CD34+ cells in response to cytokine stimulation can be successfully studied, in a label-free way, by the combination of the IFCs statistical power and the TLM's high time resolution. Cell morphology phenotypes were quantified through roundness and surface area, measured both in IFC and with a homemade segmentation algorithm in TLM. Two distinct morphologies-polarized and round-were observed in cord-blood derived CD34+. We show that some cells have the ability to fluctuate between these morphologies, suggesting that the apparent stable composition of round and polarized cells may actually represent a dynamic equilibrium. This example demonstrates that the different resolutions and modalities of IFC and TLM are complementary and allow the study of complex dynamic biological processes. © 2017 International Society for Advancement of Cytometry.


Assuntos
Antígenos CD34/isolamento & purificação , Citometria de Fluxo/métodos , Microscopia/métodos , Imagem com Lapso de Tempo/métodos , Antígenos CD34/metabolismo , Contagem de Células/métodos , Diferenciação Celular/genética , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Humanos , Processamento de Imagem Assistida por Computador/métodos
9.
J Vis Exp ; (113)2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27501019

RESUMO

Although bioluminescence imaging (BLI) shows promise for monitoring tumor burden in animal models of cancer, these analyses remain mostly qualitative. Here we describe a method for bioluminescence imaging to obtain a semi-quantitative analysis of tumor burden and treatment response. This method is based on the calculation of a luminoscore, a value that allows comparisons of two animals from the same or different experiments. Current BLI instruments enable the calculation of this luminoscore, which relies mainly on the acquisition conditions (back and front acquisitions) and the drawing of the region of interest (manual markup around the mouse). Using two previously described mouse lymphoma models based on cell engraftment, we show that the luminoscore method can serve as a noninvasive way to verify successful tumor cell inoculation, monitor tumor burden, and evaluate the effects of in situ cancer treatment (CpG-DNA). Finally, we show that this method suits different experimental designs. We suggest that this method be used for early estimates of treatment response in preclinical small-animal studies.


Assuntos
Linfoma , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Medições Luminescentes , Camundongos
10.
Exp Cell Res ; 341(2): 187-95, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26844629

RESUMO

Myostatin, also known as growth differentiation factor 8, is a member of the transforming growth factor-beta superfamily that has been shown to play a key role in the regulation of the skeletal muscle mass. Indeed, while myostatin deletion or loss of function induces muscle hypertrophy, its overexpression or systemic administration causes muscle atrophy. Since myostatin blockade is effective in increasing skeletal muscle mass, myostatin inhibitors have been actively sought after. Decorin, a member of the small leucine-rich proteoglycan family is a metalloprotein that was previously shown to bind and inactivate myostatin in a zinc-dependent manner. Furthermore, the myostatin-binding site has been shown to be located in the decorin N-terminal domain. In the present study, we investigated the anti-myostatin activity of short and soluble fragments of decorin. Our results indicate that the murine decorin peptides DCN48-71 and 42-65 are sufficient for inactivating myostatin in vitro. Moreover, we show that the interaction of mDCN48-71 to myostatin is strictly zinc-dependent. Binding of myostatin to activin type II receptor results in the phosphorylation of Smad2/3. Addition of the decorin peptide 48-71 decreased in a dose-dependent manner the myostatin-induced phosphorylation of Smad2 demonstrating thereby that the peptide inhibits the activation of the Smad signaling pathway. Finally, we found that mDCN48-71 displays a specificity towards myostatin, since it does not inhibit other members of the transforming growth factor-beta family.


Assuntos
Decorina/metabolismo , Músculo Esquelético/metabolismo , Miostatina/genética , Transdução de Sinais , Proteínas Smad/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Humanos , Peptídeos/metabolismo , Proteoglicanas/metabolismo , Fator de Crescimento Transformador beta/metabolismo
11.
J Neurooncol ; 121(1): 9-18, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25300908

RESUMO

Primary central nervous system lymphomas (PCNSL) include ocular and cerebral lymphomas and are rare aggressive malignancies with poor prognoses. Compared with other lymphomas, they are a challenge for clinicians and scientists, for diagnosis and therapeutic progress and their prognosis remains unsatisfactory, because of the lack of molecular and biological knowledge. Indeed, several limitations of human sample present a major obstacle to the identification of the particular microenvironment of the sanctuary sites where these tumor cells grow. In addition, the generally poor overall condition and performance status of patients with PCNSL limit their participation in prospective trials. Therefore, animal models of PCNSL are essential for tumor microenvironment characterization and for antitumor response studying. In this review, we have compiled the B-and T-cell PCNSL mouse models that are used to improve our understanding of the lymphoma microenvironment, tropism and migration and to investigate novel therapeutic strategies.


Assuntos
Neoplasias do Sistema Nervoso Central/fisiopatologia , Neoplasias do Sistema Nervoso Central/terapia , Linfoma/fisiopatologia , Linfoma/terapia , Animais , Neoplasias do Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Humanos , Linfoma/patologia , Camundongos
12.
Invest Ophthalmol Vis Sci ; 54(5): 3657-65, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23611989

RESUMO

PURPOSE: Primary cerebral lymphoma (PCL) and primary intraocular lymphoma (PIOL) belong to the systemic diffuse large B-cell lymphoma family and are characterized by the presence of CD20(+) lymphoma B cells in the brain or the eye. These highly aggressive malignancies have a poor prognosis and no specific therapy. The presence of effector immune cells in the damaged brain and vitreous suggests that treatment with anti-human CD20 (hCD20) monoclonal antibodies might be effective. We developed murine models of PCL and PIOL to assess the intracerebral and intraocular antitumor effect of ublituximab, a promising glycoengineered anti-hCD20 mAb with a high affinity for FcγRIIIa (CD16) receptors. METHODS: The murine lymphoma B-cell line A20.IIA-GFP-hCD20 (H-2(d)) was injected into the right cerebral striatum or the vitreous of immunocompetent adult BALB/c mice (H-2(d)). Four to 7 days later, ublituximab was injected intracerebrally or intravitreously into the tumor site. Rituximab was the reference compound. Survival was monitored for injected mice; histopathological and flow cytometric analyses were performed to study tumor growth and T-cell infiltration. RESULTS: Single doses of ublituximab, injected intracerebrally or intravitreously, had a marked antitumor effect, more pronounced than that obtained with the same dose of rituximab in these conditions. The reduction in tumor cells was correlated with an increased proportion of CD8(+) T cells. This efficacy was observed only against lymphoma B cells expressing hCD20. CONCLUSIONS: These in vivo results confirm the potential of the glycoengineered anti-hCD20 mAb ublituximab as an innovative therapeutic approach to treat primary central nervous system lymphoma and other B-cell lymphomas.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos CD20/imunologia , Neoplasias do Sistema Nervoso Central/tratamento farmacológico , Neoplasias Oculares/tratamento farmacológico , Linfoma de Células B/tratamento farmacológico , Animais , Anticorpos Monoclonais Murinos/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Neoplasias do Sistema Nervoso Central/imunologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Neoplasias Oculares/imunologia , Feminino , Humanos , Linfoma de Células B/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Engenharia de Proteínas , Rituximab , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Exp Clin Cancer Res ; 32: 18, 2013 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-23561041

RESUMO

BACKGROUND: Toll-like receptor (TLR) agonists have important properties that can be exploited for immunotherapy against tumors. Locally injected immunostimulatory oligodeoxynucleotides containing CpG motifs (CpG-ODNs), which are TLR9 agonists, have shown promise in cancer models. Several studies have demonstrated that these motifs have immunologic effects similar to those of bacterial DNA and can stimulate monocytes, macrophages, dendritic, and B cells, which then produce several proinflammatory cytokines. However, these CpG-ODNs appear to produce opposite effects on tumor B cells. METHODS: In this study, we investigated the direct effects of a murine class B CpG (1826) ODNs on lymphoma B cells in vitro and in vivo, using mouse models of non-Hodgkin B lymphomas developing in immunoprivileged sites, specifically the brain and the eye, and in subcutaneous sites. RESULTS: In vitro, CpG-ODNs produced antiproliferative and proapoptotic effects on lymphoma B cells. In vivo, it had an antitumor effect when injected into tumors in murine models of subcutaneous lymphoma (SCL) and primary cerebral lymphoma (PCL). However, its intravitreal administration into a primary intraocular lymphoma (PIOL) mouse model did not produce an antitumor effect. In vitro experiments using supernatant from mouse PIOL samples demonstrated that the PIOL molecular microenvironment inhibits the antiproliferative effect of CpG-ODNs on lymphoma B-cells. CONCLUSIONS: Responsiveness to CpG stimulation differs in subcutaneous, cerebral, and ocular tumors, according to the tumoral and molecular microenvironment, and this should be considered for further therapeutic approaches.


Assuntos
DNA de Neoplasias/genética , Linfoma de Células B/tratamento farmacológico , Linfoma de Células B/genética , Oligodesoxirribonucleotídeos/farmacologia , Microambiente Tumoral/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/genética , Ilhas de CpG , Modelos Animais de Doenças , Neoplasias Oculares/tratamento farmacológico , Neoplasias Oculares/genética , Neoplasias Oculares/patologia , Feminino , Humanos , Linfoma de Células B/patologia , Camundongos , Camundongos Endogâmicos BALB C , Receptor Toll-Like 9/agonistas , Microambiente Tumoral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA