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










Base de datos
Intervalo de año de publicación
1.
Environ Mol Mutagen ; 62(5): 306-318, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34050964

RESUMEN

The organotypic human air-liquid-interface (ALI) airway tissue model has been used as an in vitro cell culture system for evaluating the toxicity of inhaled substances. ALI airway cultures are highly differentiated, which has made it challenging to evaluate genetic toxicology endpoints. In the current study, we assayed DNA damage with the high-throughput CometChip assay and quantified mutagenesis with Duplex Sequencing, an error-corrected next-generation sequencing method capable of detecting a single mutation per 107 base pairs. Fully differentiated human ALI airway cultures were treated from the basolateral side with 6.25 to 100 µg/mL ethyl methanesulfonate (EMS) over a period of 28 days. CometChip assays were conducted after 3 and 28 days of treatment, and Duplex Sequencing after 28 days of treatment. Treating the airway cultures with EMS resulted in time- and concentration-dependent increases in DNA damage and a concentration-dependent increase in mutant frequency. The mutations observed in the EMS-treated cultures were predominantly C → T transitions and exhibited a unique trinucleotide signature relative to the negative control. Measurement of physiological endpoints indicated that the EMS treatments had no effect on anti-p63-positive basal cell frequency, but produced concentration-responsive increases in cytotoxicity and perturbations in cell morphology, along with concentration-responsive decreases in culture viability, goblet cell and anti-Ki67-positive proliferating cell frequency, cilia beating frequency, and mucin secretion. The results indicate that a unified 28-day study can be used to measure several important safety endpoints in physiologically relevant human in vitro ALI airway cultures, including DNA damage, mutagenicity, and tissue-specific general toxicity.


Asunto(s)
Daño del ADN , Células Epiteliales/patología , Metanosulfonato de Etilo/efectos adversos , Mutagénesis , Pruebas de Mutagenicidad/métodos , Mutación , Sistema Respiratorio/patología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Mutágenos/efectos adversos , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/metabolismo
2.
Stem Cell Reports ; 13(1): 91-104, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31204301

RESUMEN

Myeloid-differentiated hematopoietic stem cells (HSCs) have contributed to a number of novel treatment approaches for lysosomal storage diseases of the central nervous system (CNS), and may also be applied to patients infected with HIV. We quantified hematopoietic stem and progenitor cell (HSPC) trafficking to 20 tissues including lymph nodes, spleen, liver, gastrointestinal tract, CNS, and reproductive tissues. We observed efficient marking of multiple macrophage subsets, including CNS-associated myeloid cells, suggesting that HSPC-derived macrophages are a viable approach to target gene-modified cells to tissues. Gene-marked cells in the CNS were unique from gene-marked cells at any other physiological sites including peripheral blood. This novel finding suggests that these cells were derived from HSPCs, migrated to the brain, were compartmentalized, established myeloid progeny, and could be targeted for lifelong delivery of therapeutic molecules. Our findings have highly relevant implications for the development of novel therapies for genetic and infectious diseases of the CNS.


Asunto(s)
Sistema Nervioso Central/citología , Trasplante de Células Madre Hematopoyéticas , Células Mieloides/citología , Animales , Diferenciación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Terapia Genética/métodos , Células Madre Hematopoyéticas , Estudios Longitudinales , Enfermedades por Almacenamiento Lisosomal/patología , Enfermedades por Almacenamiento Lisosomal/terapia , Macaca nemestrina , Macrófagos/citología
3.
JCI Insight ; 3(13)2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29997284

RESUMEN

The genomic integration of HIV into cells results in long-term persistence of virally infected cell populations. This integration event acts as a heritable mark that can be tracked to monitor infected cells that persist over time. Previous reports have documented clonal expansion in people and have linked them to proto-oncogenes; however, their significance or contribution to the latent reservoir has remained unclear. Here, we demonstrate that a directed pattern of clonal expansion occurs in vivo, specifically in gene pathways important for viral replication and persistence. These biological processes include cellular division, transcriptional regulation, RNA processing, and posttranslational modification pathways. This indicates preferential expansion when integration events occur within genes or biological pathways beneficial for HIV replication and persistence. Additionally, these expansions occur quickly during unsuppressed viral replication in vivo, reinforcing the importance of early intervention for individuals to limit reservoir seeding of clonally expanded HIV-infected cells.


Asunto(s)
Genes Virales/genética , Infecciones por VIH/genética , VIH-1/genética , Integración Viral/genética , Replicación Viral/genética , Vacunas contra el SIDA , Animales , Linfocitos T CD4-Positivos , División Celular , Cromosomas Humanos/genética , Regulación Viral de la Expresión Génica , Genoma Viral , Infecciones por VIH/inmunología , VIH-1/patogenicidad , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/fisiología , Humanos , Masculino , Ratones Endogámicos NOD , Integración Viral/fisiología
4.
Blood Adv ; 2(9): 987-999, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29720491

RESUMEN

Hematopoietic stem-cell gene therapy is a promising treatment of X-linked severe combined immunodeficiency disease (SCID-X1), but currently, it requires recipient conditioning, extensive cell manipulation, and sophisticated facilities. With these limitations in mind, we explored a simpler therapeutic approach to SCID-X1 treatment by direct IV administration of foamy virus (FV) vectors in the canine model. FV vectors were used because they have a favorable integration site profile and are resistant to serum inactivation. Here, we show improved efficacy of our in vivo gene therapy platform by mobilization with granulocyte colony-stimulating factor (G-CSF) and AMD3100 before injection of an optimized FV vector incorporating the human phosphoglycerate kinase enhancerless promoter. G-CSF/AMD3100 mobilization before FV vector delivery accelerated kinetics of CD3+ lymphocyte recovery, promoted thymopoiesis, and increased immune clonal diversity. Gene-corrected T lymphocytes exhibited a normal CD4:CD8 ratio and a broad T-cell receptor repertoire and showed restored γC-dependent signaling function. Treated animals showed normal primary and secondary antibody responses to bacteriophage immunization and evidence for immunoglobulin class switching. These results demonstrate safety and efficacy of an accessible, portable, and translatable platform with no conditioning regimen for the treatment of SCID-X1 and other genetic diseases.


Asunto(s)
Enfermedades de los Perros , Terapia Genética , Vectores Genéticos/farmacología , Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética , Compuestos Heterocíclicos/farmacología , Spumavirus , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X , Animales , Bencilaminas , Relación CD4-CD8 , Ciclamas , Modelos Animales de Enfermedad , Enfermedades de los Perros/sangre , Enfermedades de los Perros/genética , Enfermedades de los Perros/terapia , Perros , Humanos , Fosfoglicerato Quinasa/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/sangre , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/veterinaria
5.
Mol Ther Methods Clin Dev ; 8: 75-86, 2018 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-29276718

RESUMEN

Reactivation of fetal hemoglobin (HbF) is a promising approach for the treatment of ß-hemoglobinopathies and the targeting of genes involved in HbF regulation is under intensive investigation. Here, we established a nonhuman primate (NHP) transplantation model to evaluate hematopoietic stem cell (HSC)-based gene editing strategies aimed at reactivating HbF. We first characterized the transient HbF induction to autologous HSC transplantation in pigtailed macaques, which was comparable in duration and amplitude to that of human patients. After validating function of the HbF repressor BCL11A in NHPs, we transplanted a pigtailed macaque with CD34+ cells electroporated with TALE nuclease mRNA targeting the BCL11A coding sequence. In vivo gene editing levels were low, but some BCL11A deletions were detected as late as 200 days post-transplantation. HbF production, as determined by F-cell staining and γ-globin expression, was slightly increased in this animal as compared to transplant controls. We also provided proof-of-concept results for the selection of edited NHP CD34+ cells in culture following integration of the P140K/MGMT cassette at the BCL11A locus. In summary, the NHP model described here will allow the testing of novel therapeutic approaches for hemoglobinopathies and should facilitate clinical translation.

6.
Sci Transl Med ; 9(414)2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-29093179

RESUMEN

Hematopoietic reconstitution after bone marrow transplantation is thought to be driven by committed multipotent progenitor cells followed by long-term engrafting hematopoietic stem cells (HSCs). We observed a population of early-engrafting cells displaying HSC-like behavior, which persisted long-term in vivo in an autologous myeloablative transplant model in nonhuman primates. To identify this population, we characterized the phenotype and function of defined nonhuman primate hematopoietic stem and progenitor cell (HSPC) subsets and compared these to human HSPCs. We demonstrated that the CD34+CD45RA-CD90+ cell phenotype is highly enriched for HSCs. This population fully supported rapid short-term recovery and robust multilineage hematopoiesis in the nonhuman primate transplant model and quantitatively predicted transplant success and time to neutrophil and platelet recovery. Application of this cell population has potential in the setting of HSC transplantation and gene therapy/editing approaches.


Asunto(s)
Linaje de la Célula , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Animales , Antígenos CD/metabolismo , Plaquetas/citología , Células Clonales , Humanos , Macaca nemestrina , Neutrófilos/citología , Fenotipo , Transcriptoma/genética , Trasplante Autólogo
7.
Mol Ther Methods Clin Dev ; 6: 17-30, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28649577

RESUMEN

Adoptive cellular immunotherapy is a promising and powerful method for the treatment of a broad range of malignant and infectious diseases. Although the concept of cellular immunotherapy was originally proposed in the 1990s, it has not seen successful clinical application until recent years. Despite significant progress in creating engineered receptors against both malignant and viral epitopes, no efficient preclinical animal models exist for rapidly testing and directly comparing these engineered receptors. The use of matured human T cells in mice usually leads to graft-versus-host disease (GvHD), which severely limits the effectiveness of such studies. Alternatively, adult apheresis CD34+ cells engraft in neonatal non-obese diabetic (NOD)-severe combined immunodeficiency (SCID)-common γ chain-/- (NSG) mice and lead to the development of CD3+ T cells in peripheral circulation. We demonstrate that these in vivo murine-matured autologous CD3+ T cells from humans (MATCH) can be collected from the mice, engineered with lentiviral vectors, reinfused into the mice, and detected in multiple lymphoid compartments at stable levels over 50 days after injection. Unlike autologous CD3+ cells collected from human donors, these MATCH mice did not exhibit GvHD after T cell administration. This novel mouse model offers the opportunity to screen different immunotherapy-based treatments in a preclinical setting.

8.
Stem Cells Transl Med ; 6(3): 864-876, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28297579

RESUMEN

Successful expansion of bone marrow (BM) hematopoietic stem and progenitor cells (HSPCs) would benefit many HSPC transplantation and gene therapy/editing applications. However, current expansion technologies have been limited by a loss of multipotency and self-renewal properties ex vivo. We hypothesized that an ex vivo vascular niche would provide prohematopoietic signals to expand HSPCs while maintaining multipotency and self-renewal. To test this hypothesis, BM autologous CD34+ cells were expanded in endothelial cell (EC) coculture and transplanted in nonhuman primates. CD34+ C38- HSPCs cocultured with ECs expanded up to 17-fold, with a significant increase in hematopoietic colony-forming activity compared with cells cultured with cytokines alone (colony-forming unit-granulocyte-erythroid-macrophage-monocyte; p < .005). BM CD34+ cells that were transduced with green fluorescent protein lentivirus vector and expanded on ECs engrafted long term with multilineage polyclonal reconstitution. Gene marking was observed in granulocytes, lymphocytes, platelets, and erythrocytes. Whole transcriptome analysis indicated that EC coculture altered the expression profile of 75 genes in the BM CD34+ cells without impeding the long-term engraftment potential. These findings show that an ex vivo vascular niche is an effective platform for expansion of adult BM HSPCs. Stem Cells Translational Medicine 2017;6:864-876.


Asunto(s)
Células de la Médula Ósea/citología , Células Endoteliales/citología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Animales , Antígenos CD34/metabolismo , Linaje de la Célula , Proliferación Celular , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Humanos , Primates , Factores de Tiempo
9.
Blood ; 127(20): 2416-26, 2016 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-26980728

RESUMEN

Genome editing in hematopoietic stem and progenitor cells (HSPCs) is a promising novel technology for the treatment of many human diseases. Here, we evaluated whether the disruption of the C-C chemokine receptor 5 (CCR5) locus in pigtailed macaque HSPCs by zinc finger nucleases (ZFNs) was feasible. We show that macaque-specific CCR5 ZFNs efficiently induce CCR5 disruption at levels of up to 64% ex vivo, 40% in vivo early posttransplant, and 3% to 5% in long-term repopulating cells over 6 months following HSPC transplant. These genome-edited HSPCs support multilineage engraftment and generate progeny capable of trafficking to secondary tissues including the gut. Using deep sequencing technology, we show that these ZFNs are highly specific for the CCR5 locus in primary cells. Further, we have adapted our clonal tracking methodology to follow individual CCR5 mutant cells over time in vivo, reinforcing that CCR5 gene-edited HSPCs are capable of long-term engraftment. Together, these data demonstrate that genome-edited HSPCs engraft, and contribute to multilineage repopulation after autologous transplantation in a clinically relevant large animal model, an important step toward the development of stem cell-based genome-editing therapies for HIV and potentially other diseases as well.


Asunto(s)
Trasplante de Médula Ósea , Linaje de la Célula , Edición Génica , Trasplante de Células Madre Hematopoyéticas , Macaca nemestrina/genética , Receptores CCR5/genética , Secuencia de Aminoácidos , Animales , Línea Celular , Electroporación , Estudios de Factibilidad , Técnicas de Silenciamiento del Gen , Supervivencia de Injerto , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Datos de Secuencia Molecular , Mutación , Reacción en Cadena de la Polimerasa/métodos , ARN Mensajero/genética , Receptores CCR5/deficiencia , Análisis de Secuencia de ADN , Acondicionamiento Pretrasplante , Trasplante Autólogo , Irradiación Corporal Total , Dedos de Zinc
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...