Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
N Engl J Med ; 386(2): 138-147, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34898140

RESUMEN

Gene therapy with LentiGlobin for sickle cell disease (bb1111, lovotibeglogene autotemcel) consists of autologous transplantation of a patient's hematopoietic stem cells transduced with the BB305 lentiviral vector that encodes the ßA-T87Q-globin gene. Acute myeloid leukemia developed in a woman approximately 5.5 years after she had received LentiGlobin for sickle cell disease as part of the initial cohort (Group A) of the HGB-206 study. An analysis of peripheral-blood samples revealed that blast cells contained a BB305 lentiviral vector insertion site. The results of an investigation of causality indicated that the leukemia was unlikely to be related to vector insertion, given the location of the insertion site, the very low transgene expression in blast cells, and the lack of an effect on expression of surrounding genes. Several somatic mutations predisposing to acute myeloid leukemia were present after diagnosis, which suggests that patients with sickle cell disease are at increased risk for hematologic malignant conditions after transplantation, most likely because of a combination of risks associated with underlying sickle cell disease, transplantation procedure, and inadequate disease control after treatment. (Funded by Bluebird Bio.).


Asunto(s)
Anemia de Células Falciformes/terapia , Expresión Génica , Terapia Genética/efectos adversos , Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda/etiología , Globinas beta/genética , Adulto , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/genética , Carcinogénesis , Femenino , Vectores Genéticos , Humanos , Lentivirus , Factores de Riesgo , Análisis de Secuencia de ARN , Transgenes , Trasplante Autólogo
2.
Proc Natl Acad Sci U S A ; 111(18): 6804-9, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24753614

RESUMEN

Parkinson disease (PD) is characterized by the selective loss of dopaminergic neurons of the substantia nigra pars compacta (SNpc). Although growing evidence indicates that endoplasmic reticulum (ER) stress is a hallmark of PD, its exact contribution to the disease process is not well understood. Here we report that developmental ablation of X-Box binding protein 1 (XBP1) in the nervous system, a key regulator of the unfolded protein response (UPR), protects dopaminergic neurons against a PD-inducing neurotoxin. This survival effect was associated with a preconditioning condition that resulted from induction of an adaptive ER stress response in dopaminergic neurons of the SNpc, but not in other brain regions. In contrast, silencing XBP1 in adult animals triggered chronic ER stress and dopaminergic neuron degeneration. Supporting this finding, gene therapy to deliver an active form of XBP1 provided neuroprotection and reduced striatal denervation in animals injected with 6-hydroxydopamine. Our results reveal a physiological role of the UPR in the maintenance of protein homeostasis in dopaminergic neurons that may help explain the differential neuronal vulnerability observed in PD.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Factores de Transcripción/metabolismo , Animales , Supervivencia Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Neuronas Dopaminérgicas/efectos de los fármacos , Estrés del Retículo Endoplásmico , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa , Neurotoxinas/toxicidad , Oxidopamina/toxicidad , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Factores de Transcripción del Factor Regulador X , Sustancia Negra/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Respuesta de Proteína Desplegada , Proteína 1 de Unión a la X-Box
3.
Biochem Biophys Res Commun ; 420(3): 558-63, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22445760

RESUMEN

Huntington's disease (HD) is caused by mutations that expand a polyglutamine region in the amino-terminal domain of Huntingtin (Htt), leading to the accumulation of intracellular inclusions and progressive neurodegeneration. Recent reports indicate the engagement of endoplasmic reticulum (ER) stress responses in human HD post mortem samples and animal models of the disease. Adaptation to ER stress is mediated by the activation of the unfolded protein response (UPR), an integrated signal transduction pathway that attenuates protein folding stress by controlling the expression of distinct transcription factors including X-Box binding protein 1 (XBP1). Here we targeted the expression of XBP1 on a novel viral-based model of HD. We delivered an active form of XBP1 locally into the striatum of adult mice using adeno-associated vectors (AAVs) and co-expressed this factor with a large fragment of mutant Htt as a fusion protein with RFP (Htt588(Q95)-mRFP) to directly visualize the accumulation of Htt inclusions in the brain. Using this approach, we observed a significant reduction in the accumulation of Htt588(Q95)-mRFP intracellular inclusion when XBP1 was co-expressed in the striatum. These results contrast with recent findings indicating a protective effect of XBP1 deficiency in neurodegeneration using knockout mice, and suggest a potential use of gene therapy strategies to manipulate the UPR in the context of HD.


Asunto(s)
Cuerpo Estriado/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Enfermedad de Huntington/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada/genética , Animales , Dependovirus , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/genética , Terapia Genética/métodos , Vectores Genéticos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/terapia , Masculino , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Factores de Transcripción del Factor Regulador X , Transducción Genética , Proteína 1 de Unión a la X-Box
4.
Hum Gene Ther ; 33(23-24): 1279-1292, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36226412

RESUMEN

Mucopolysaccharidosis type II (MPS II, Hunter syndrome) is an X-linked recessive lysosomal disease caused by deficiency of iduronate-2-sulfatase (IDS). The absence of IDS results in the accumulation of the glycosaminoglycans (GAGs) heparan sulfate and dermatan sulfate. Currently, the only approved treatment option for MPS II is enzyme replacement therapy (ERT), Elaprase. However, ERT is demanding for the patient and does not ameliorate neurological manifestations of the disease. Using an IDS-deficient mouse model that phenocopies the human disease, we evaluated hematopoietic stem and progenitor cells (HSPCs) transduced with a lentiviral vector (LVV) carrying a codon-optimized human IDS coding sequence regulated by a ubiquitous MNDU3 promoter (MNDU3-IDS). Mice treated with MNDU3-IDS LVV-transduced cells showed supraphysiological levels of IDS enzyme activity in plasma, peripheral blood mononuclear cells, and in most analyzed tissues. These enzyme levels were sufficient to normalize GAG storage in analyzed tissues. Importantly, IDS levels in the brains of MNDU3-IDS-engrafted animals were restored to 10-20% than that of wild-type mice, sufficient to normalize GAG content and prevent emergence of cognitive deficit as evaluated by neurobehavioral testing. These results demonstrate the potential effectiveness of ex vivo MNDU3-IDS LVV-transduced HSPCs for treatment of MPS II.


Asunto(s)
Iduronato Sulfatasa , Mucopolisacaridosis II , Animales , Ratones , Humanos , Mucopolisacaridosis II/genética , Mucopolisacaridosis II/terapia , Leucocitos Mononucleares , Iduronato Sulfatasa/genética , Terapia de Reemplazo Enzimático , Modelos Animales de Enfermedad , Células Madre Hematopoyéticas
5.
Blood Adv ; 5(13): 2701-2706, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34196676

RESUMEN

Betibeglogene autotemcel (beti-cel) gene therapy (GT) for patients with transfusion-dependent ß-thalassemia uses autologous CD34+ cells transduced with BB305 lentiviral vector (LVV), which encodes a modified ß-globin gene. BB305 LVV also contains select HIV sequences for viral packaging, reverse transcription, and integration. This case report describes a patient successfully treated with beti-cel in a phase 1/2 study (HGB-204; #NCT01745120) and subsequently diagnosed with wild-type (WT) HIV infection. From 3.5 to 21 months postinfusion, the patient stopped chronic red blood cell transfusions; total hemoglobin (Hb) and GT-derived HbAT87Q levels were 6.6 to 9.5 and 2.8 to 3.8 g/dL, respectively. At 21 months postinfusion, the patient resumed transfusions for anemia that coincided with an HIV-1 infection diagnosis. Quantitative polymerase chain reaction assays detected no replication-competent lentivirus. Next-generation sequencing confirmed WT HIV sequences. Six months after starting antiretroviral therapy, total Hb and HbAT87Q levels recovered to 8.6 and 3.6 g/dL, respectively, and 3.5 years postinfusion, 13.4 months had elapsed since the patient's last transfusion. To our knowledge, this is the first report of WT HIV infection in an LVV-based GT recipient and demonstrates persistent long-term hematopoiesis after treatment with beti-cel and the ability to differentiate between WT HIV and BB305-derived sequences.


Asunto(s)
Infecciones por VIH , Talasemia beta , Terapia Genética , Vectores Genéticos/genética , Infecciones por VIH/complicaciones , Infecciones por VIH/terapia , Humanos , Lentivirus/genética , Talasemia beta/genética , Talasemia beta/terapia
6.
Sci Rep ; 6: 21709, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26906090

RESUMEN

Although protein-folding stress at the endoplasmic reticulum (ER) is emerging as a driver of neuronal dysfunction in models of spinal cord injury and neurodegeneration, the contribution of this pathway to peripheral nerve damage remains poorly explored. Here we targeted the unfolded protein response (UPR), an adaptive reaction against ER stress, in mouse models of sciatic nerve injury and found that ablation of the transcription factor XBP1, but not ATF4, significantly delay locomotor recovery. XBP1 deficiency led to decreased macrophage recruitment, a reduction in myelin removal and axonal regeneration. Conversely, overexpression of XBP1s in the nervous system in transgenic mice enhanced locomotor recovery after sciatic nerve crush, associated to an improvement in key pro-regenerative events. To assess the therapeutic potential of UPR manipulation to axonal regeneration, we locally delivered XBP1s or an shRNA targeting this transcription factor to sensory neurons of the dorsal root ganglia using a gene therapy approach and found an enhancement or reduction of axonal regeneration in vivo, respectively. Our results demonstrate a functional role of specific components of the ER proteostasis network in the cellular changes associated to regeneration and functional recovery after peripheral nerve injury.


Asunto(s)
Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/fisiopatología , Nervio Ciático/fisiopatología , Respuesta de Proteína Desplegada , Factor de Transcripción Activador 4/metabolismo , Animales , Axones/fisiología , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Estrés del Retículo Endoplásmico , Expresión Génica , Locomoción , Macrófagos/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Recuperación de la Función , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Proteína 1 de Unión a la X-Box/genética , Proteína 1 de Unión a la X-Box/metabolismo
8.
Brain Res Dev Brain Res ; 153(1): 19-27, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15464214

RESUMEN

Canavan disease (CD) is an autosomal recessive disorder that leads to spongy degeneration in the white matter of the brain. Aspartoacylase (ASPA) synthesizing cells, oligodendrocytes, are lost in CD. Transplantation of neural progenitor cells (NPCs) offers an interesting therapeutic approach for treating neurodegenerative diseases by replacing the lost cells. Therefore, the NPCs transplantation to the brain of the CD mouse was studied. Injection of mouse NPCs to the striatum and cerebellum of juvenile CD mouse showed numerous BrdU positive cells at 1 month after injection. The same result was also observed in the adult CD mouse brain after 5 weeks of post-transplantation period. The implanted cells differentiated into oligodendrocytes and fibrous astrocytes, as observed using glial cell marker. This is the first report to describe the survival, distribution and differentiation of NPCs within the brain of CD mouse and a first step toward the potential clinical use of cell therapy to treat CD.


Asunto(s)
Encéfalo/citología , Enfermedad de Canavan/terapia , Diferenciación Celular/fisiología , Oligodendroglía/citología , Trasplante de Células Madre , Amidohidrolasas/genética , Animales , Supervivencia Celular , Trasplante de Células , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Células Madre/citología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA