Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Hepatol Commun ; 7(2): e0009, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-37074875

RESUMO

BACKGROUND AND AIMS: Adeno-associated virus (AAV) vectors are widely used to deliver therapeutic transgenes to distinct tissues, including the liver. Vectors based on naturally occurring AAV serotypes as well as vectors using engineered capsids have shown variations in tissue tropism and level of transduction between different mouse models. Moreover, results obtained in rodents frequently lack translatability into large animal studies. In light of the increasing interest in AAV vectors for human gene therapy, an increasing number of studies are being performed in nonhuman primates. To keep animal numbers to a minimum and thus optimize the process of AAV capsid selection, we developed a multiplex barcoding approach to simultaneously evaluate the in vivo vector performance for a set of serotypes and capsid-engineered AAV vectors across multiple organs. APPROACH AND RESULTS: Vector biodistribution and transgene expression were assessed by quantitative PCR, quantitative reverse transcription PCR, vector DNA amplicon Illumina sequencing and vRNAseq in male and female rhesus macaques simultaneously dosed with a mixture of barcoded naturally occurring or engineered AAV vectors encoding the same transgene. As expected, our findings show animal-to-animal variation in both the biodistribution and tissue transduction pattern, which was partly influenced by each animal's distinctive serological status. CONCLUSIONS: This method offers a robust approach to AAV vector optimization that can be used to identify and validate AAV vectors for gene delivery to potentially any anatomical site or cell type.


Assuntos
Capsídeo , Dependovirus , Animais , Camundongos , Feminino , Masculino , Humanos , Capsídeo/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Distribuição Tecidual , Macaca mulatta/genética , Macaca mulatta/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Terapia Genética/métodos
2.
Mol Ther ; 31(4): 1059-1073, 2023 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-36760126

RESUMO

We aim to develop an in vivo hematopoietic stem cell (HSC) gene therapy approach for persistent control/protection of HIV-1 infection based on the stable expression of a secreted decoy protein for HIV receptors CD4 and CCR5 (eCD4-Ig) from blood cells. HSCs in mice and a rhesus macaque were mobilized from the bone marrow and transduced by an intravenous injection of HSC-tropic, integrating HDAd5/35++ vectors expressing rhesus eCD4-Ig. In vivo HSC transduction/selection resulted in stable serum eCD4-Ig levels of ∼100 µg/mL (mice) and >20 µg/mL (rhesus) with half maximal inhibitory concentrations (IC50s) of 1 µg/mL measured by an HIV neutralization assay. After simian-human-immunodeficiency virus D (SHIV.D) challenge of rhesus macaques injected with HDAd-eCD4-Ig or a control HDAd5/35++ vector, peak plasma viral load levels were ∼50-fold lower in the eCD4-Ig-expressing animal. Furthermore, the viral load was lower in tissues with the highest eCD4-Ig expression, specifically the spleen and lymph nodes. SHIV.D challenge triggered a selective expansion of transduced CD4+CCR5+ cells, thereby increasing serum eCD4-Ig levels. The latter, however, broke immune tolerance and triggered anti-eCD4-Ig antibody responses, which could have contributed to the inability to eliminate SHIV.D. Our data will guide us in the improvement of the in vivo approach. Clearly, our conclusions need to be validated in larger animal cohorts.


Assuntos
Infecções por HIV , HIV-1 , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Humanos , Animais , Camundongos , Macaca mulatta , Vírus da Imunodeficiência Símia/genética , Células-Tronco Hematopoéticas , Síndrome de Imunodeficiência Adquirida dos Símios/terapia
3.
Nat Commun ; 9(1): 4438, 2018 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-30361514

RESUMO

Allogeneic transplantation (allo-HCT) has led to the cure of HIV in one individual, raising the question of whether transplantation can eradicate the HIV reservoir. To test this, we here present a model of allo-HCT in SHIV-infected, cART-suppressed nonhuman primates. We infect rhesus macaques with SHIV-1157ipd3N4, suppress them with cART, then transplant them using MHC-haploidentical allogeneic donors during continuous cART. Transplant results in ~100% myeloid donor chimerism, and up to 100% T-cell chimerism. Between 9 and 47 days post-transplant, terminal analysis shows that while cell-associated SHIV DNA levels are reduced in the blood and in lymphoid organs post-transplant, the SHIV reservoir persists in multiple organs, including the brain. Sorting of donor-vs.-recipient cells reveals that this reservoir resides in recipient cells. Moreover, tetramer analysis indicates a lack of virus-specific donor immunity post-transplant during continuous cART. These results suggest that early post-transplant, allo-HCT is insufficient for recipient reservoir eradication despite high-level donor chimerism and GVHD.


Assuntos
Reservatórios de Doenças/virologia , Transplante de Células-Tronco Hematopoéticas , Complexo Principal de Histocompatibilidade , Vírus da Imunodeficiência Símia/fisiologia , Transplante Haploidêntico , Animais , Terapia Antirretroviral de Alta Atividade , Linfócitos T CD8-Positivos/imunologia , DNA Viral/metabolismo , Macaca mulatta , RNA Viral/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Transplante Homólogo
4.
Nature ; 510(7504): 273-7, 2014 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-24776797

RESUMO

Pluripotent stem cells provide a potential solution to current epidemic rates of heart failure by providing human cardiomyocytes to support heart regeneration. Studies of human embryonic-stem-cell-derived cardiomyocytes (hESC-CMs) in small-animal models have shown favourable effects of this treatment. However, it remains unknown whether clinical-scale hESC-CM transplantation is feasible, safe or can provide sufficient myocardial regeneration. Here we show that hESC-CMs can be produced at a clinical scale (more than one billion cells per batch) and cryopreserved with good viability. Using a non-human primate model of myocardial ischaemia followed by reperfusion, we show that cryopreservation and intra-myocardial delivery of one billion hESC-CMs generates extensive remuscularization of the infarcted heart. The hESC-CMs showed progressive but incomplete maturation over a 3-month period. Grafts were perfused by host vasculature, and electromechanical junctions between graft and host myocytes were present within 2 weeks of engraftment. Importantly, grafts showed regular calcium transients that were synchronized to the host electrocardiogram, indicating electromechanical coupling. In contrast to small-animal models, non-fatal ventricular arrhythmias were observed in hESC-CM-engrafted primates. Thus, hESC-CMs can remuscularize substantial amounts of the infarcted monkey heart. Comparable remuscularization of a human heart should be possible, but potential arrhythmic complications need to be overcome.


Assuntos
Células-Tronco Embrionárias/citologia , Coração , Infarto do Miocárdio/patologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Regeneração , Animais , Arritmias Cardíacas/fisiopatologia , Cálcio/metabolismo , Sobrevivência Celular , Vasos Coronários/fisiologia , Criopreservação , Modelos Animais de Doenças , Eletrocardiografia , Humanos , Macaca nemestrina , Masculino , Camundongos , Medicina Regenerativa/métodos
5.
Exp Hematol ; 42(7): 497-504, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24704161

RESUMO

Umbilical cord blood transplant continues to increase in prevalence as a treatment option for various hematopoietic and immune disorders. Because of the limited number of cells available in a single cord blood unit, investigators have explored methods of increasing cell dose before transplant, including overexpression of the homeobox B4 (HOXB4) transcription factor. We have previously reported the development of leukemia in several nonhuman primate (NHP) subjects transplanted with HOXB4-expanded bone marrow cells at approximately 2 years posttransplant. Here, we provide long-term data for a NHP receiving a HOXB4-expanded cord blood graft. Longitudinal follow-up included gene marking analysis, complete blood counts, morphologic/pathologic assessment, phenotypic analysis of subsets, and retroviral integration site analysis. In each of these independent assays, we saw no indication of clonal dominance, and all signs pointed toward normal, healthy hematopoiesis. Furthermore, in-depth clonal analysis of an animal that developed leukemia after transplantation of HOXB4-modified bone marrow cells showed that dominant clones could be detected as early as 6 months posttransplant using the genomic analysis technique detailed here. Parallel analysis of the cord blood transplant macaque showed no such sites. These findings demonstrate the ability to study the use of gene-modified and expanded cord blood cells in a NHP model.


Assuntos
Proteínas de Homeodomínio/genética , Modelos Animais , Transplante de Células-Tronco , Fatores de Transcrição/genética , Animais , Marcadores Genéticos , Humanos , Macaca nemestrina , Primatas
6.
Comp Med ; 64(1): 63-7, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24512963

RESUMO

A 2.25-y-old male pigtailed macaque (Macaca nemestrina) was experimentally irradiated and received a bone marrow transplant. After transplantation and engraftment, the macaque had unexpected recurring pancytopenia and dependent edema of the prepuce, scrotum, and legs. The diagnostic work-up included a blood smear, which revealed a trypomastigote consistent with Trypanosoma cruzi, the causative agent of Chagas disease (CD). We initially hypothesized that the macaque had acquired the infection when it lived in Georgia. However, because the animal had received multiple blood transfusions, all blood donors were screened for CD. One male pigtailed macaque blood donor, which was previously housed in Louisiana, was positive for T. cruzi antibodies via serology. Due to the low prevalence of infection in Georgia, the blood transfusion was hypothesized to be the source of T. cruzi infection. The transfusion was confirmed as the mechanism of transmission when screening of archived serum revealed seroconversion after blood transfusion from the seropositive blood donor. The macaque made a full clinical recovery, and further follow-up including thoracic radiography, echocardiography, and gross necropsy did not show any abnormalities associated with CD. Other animals that received blood transfusions from the positive blood donor were tested, and one additional pigtailed macaque on the same research protocol was positive for T. cruzi. Although CD has been reported to occur in many nonhuman primate species, especially pigtailed macaques, the transmission of CD via blood transfusion in nonhuman primates has not been reported previously.


Assuntos
Transfusão de Sangue/veterinária , Doença de Chagas/veterinária , Hospedeiro Imunocomprometido , Macaca nemestrina/parasitologia , Doenças dos Macacos/parasitologia , Trypanosoma cruzi/isolamento & purificação , Animais , Anticorpos Antiprotozoários/sangue , Biomarcadores/sangue , Doença de Chagas/sangue , Doença de Chagas/imunologia , Doença de Chagas/transmissão , Fracionamento da Dose de Radiação , Terapia Genética , Macaca nemestrina/sangue , Macaca nemestrina/imunologia , Masculino , Modelos Animais , Doenças dos Macacos/sangue , Doenças dos Macacos/imunologia , Transplante de Células-Tronco , Reação Transfusional , Trypanosoma cruzi/imunologia
7.
Exp Hematol ; 42(2): 114-25.e4, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24513167

RESUMO

Insertional mutagenesis resulting from the integration of retroviral vectors has led to the discovery of many oncogenes associated with leukemia. We investigated the role of HOXC6, identified by proximal provirus integration in a large animal hematopoietic stem cell gene therapy study, for a potential involvement in hematopoietic stem cell activity and hematopoietic cell fate decision. HOXC6 was overexpressed in the murine bone marrow transplantation model and tested in a competitive repopulation assay in comparison to the known hematopoietic stem cell expansion factor, HOXB4. We have identified HOXC6 as a factor that enhances competitive repopulation capacity in vivo and colony formation in vitro. Ectopic HOXC6 expression also induced strong myeloid differentiation and expansion of granulocyte-macrophage progenitors/common myeloid progenitors (GMPs/CMPs) in vivo, resulting in myeloid malignancies with low penetrance (3 of 17 mice), likely in collaboration with Meis1 because of a provirus integration mapped to the 3' region in the malignant clone. We characterized the molecular basis of HOXC6-induced myeloid differentiation and malignant cell transformation with complementary DNA microarray analysis. Overexpression of HOXC6 induced a gene expression signature similar to several acute myeloid leukemia subtypes when compared with normal GMPs/CMPs. These results demonstrate that HOXC6 acts as a regulator in hematopoiesis and is involved in malignant transformation.


Assuntos
Medula Óssea/patologia , Diferenciação Celular/genética , Transformação Celular Neoplásica , Proteínas de Homeodomínio/genética , Animais , Transplante de Células-Tronco Hematopoéticas , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos
8.
Mol Ther ; 21(6): 1270-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23587923

RESUMO

Umbilical cord blood (CB) transplantation is a promising therapeutic approach but continues to be associated with delayed engraftment and infections. Here, we explored in our macaque CB transplant model expansion and engraftment kinetics of cells expanded with the combination of HOXB4 and Delta-1. CB cells were divided into two equal fractions; one fraction was transduced with HOXB4 yellow fluorescent protein (YFP) and expanded on control OP9 cells, and the other was transduced with HOXB4 green fluorescent protein (GFP) and expanded on Delta-expressing OP9 cells (OP9-DL1). Both fractions were transplanted into myeloablated subjects. Neutrophil and platelet recovery occurred within 7 and 19 days respectively, which was significantly earlier than in our previous study using cells expanded with HOXB4 alone, which resulted in neutrophil recovery within 12 days (P = 0.05) and platelet recovery within 37 days (P = 0.02). Furthermore, two of three animals in the current study remained fully transfusion-independent after transplantation. By day 30, reconstitution of lymphocytes was significantly greater with the HOXB4/OP9-DL1 expanded cells in all animals (P = 0.05). In conclusion, our data show that the combination of OP9-DL1 and HOXB4 can result in increased numbers of repopulating cells, thus leading to rapid engraftment and transfusion independence in macaques transplanted with autologous, expanded CB cells.


Assuntos
Proteínas de Homeodomínio/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Macaca nemestrina/genética , Proteínas de Membrana/genética , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Animais , Antígenos CD34/metabolismo , Plaquetas/metabolismo , Transfusão de Sangue/métodos , Diferenciação Celular , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Feminino , Sangue Fetal/citologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Transplante de Células-Tronco Hematopoéticas/métodos , Proteínas de Homeodomínio/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macaca nemestrina/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Fatores de Transcrição/metabolismo
9.
Exp Hematol ; 40(3): 187-96, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22155723

RESUMO

The use of umbilical cord blood for allogeneic transplantation has increased dramatically over the past years. However, the limited number of cells available in a single cord blood unit remains a serious obstacle. Here, we wished to establish a nonhuman primate cord blood transplantation model that would allow us to test various hematopoietic stem cell expansion and gene therapy strategies. We implemented HOXB4-mediated expansion based on our previous experience with HOXB4 in autologous cells. Cord blood units were divided into two equal parts; half of the cells were transduced with a yellow fluorescent protein control vector and cryopreserved, and half were transduced with a HOXB4GFP vector, expanded, and cryopreserved. Both fractions of cells were transplanted into Macaca nemestrina subjects. We found that neutrophil recovery occurred within 19 days in all animals, and both neutrophil and platelet recovery were substantially accelerated compared to human single unit cord blood transplants. In addition, HOXB4-transduced and expanded cells resulted in superior engraftment of all hematopoietic lineages in all animals over nonexpanded controls. In conclusion, we have successfully established a nonhuman primate cord blood transplantation model and demonstrated that HOXB4 stimulates expansion and engraftment of repopulating cells. The availability of such a model has significant implications for developing and testing strategies to improve clinical cord blood transplantation, as it will allow comparison of different stem cell expansion methodologies within a single animal. Furthermore, it can be used in long-term follow-up studies to determine how specific expansion techniques affect engraftment of various hematopoietic lineages.


Assuntos
Linhagem da Célula , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Animais , Proteínas de Bactérias/genética , Ensaio de Unidades Formadoras de Colônias , Feminino , Genes Reporter , Vetores Genéticos/genética , Sobrevivência de Enxerto , Proteínas de Fluorescência Verde/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Proteínas Luminescentes/genética , Macaca nemestrina , Masculino , Modelos Animais , Mielopoese , Complicações Pós-Operatórias , Gravidez , Proteínas Recombinantes de Fusão/fisiologia , Retroviridae/genética , Trombopoese , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Transdução Genética , Condicionamento Pré-Transplante , Integração Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA