Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Hum Gene Ther ; 33(23-24): 1293-1304, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36094106

RESUMEN

Ex vivo gene therapy procedures targeting hematopoietic stem and progenitor cells (HSPCs) predominantly utilize lentivirus-based vectors for gene transfer. We provide the first pre-clinical evidence of the therapeutic utility of a foamy virus vector (FVV) for the genetic correction of human leukocyte adhesion deficiency type 1 (LAD-1), an inherited primary immunodeficiency resulting from mutation of the ß2 integrin common chain, CD18. CD34+ HSPCs isolated from a severely affected LAD-1 patient were transduced under a current good manufacturing practice-compatible protocol with FVV harboring a therapeutic CD18 transgene. LAD-1-associated cellular chemotactic defects were ameliorated in transgene-positive, myeloid-differentiated LAD-1 cells assayed in response to a strong neutrophil chemoattractant in vitro. Xenotransplantation of vector-transduced LAD-1 HSPCs in immunodeficient (NSG) mice resulted in long-term (∼5 months) human cell engraftment within murine bone marrow. Moreover, engrafted LAD-1 myeloid cells displayed in vivo levels of transgene marking previously reported to ameliorate the LAD-1 phenotype in a large animal model of the disease. Vector insertion site analysis revealed a favorable vector integration profile with no overt evidence of genotoxicity. These results coupled with the unique biological features of wild-type foamy virus support the development of FVVs for ex vivo gene therapy of LAD-1.


Asunto(s)
Síndrome de Deficiencia de Adhesión del Leucocito , Spumavirus , Humanos , Ratones , Animales , Spumavirus/genética , Vectores Genéticos/genética , Síndrome de Deficiencia de Adhesión del Leucocito/genética , Síndrome de Deficiencia de Adhesión del Leucocito/terapia , Células Madre Hematopoyéticas , Antígenos CD18/genética , Antígenos CD34/genética
2.
Sci Rep ; 11(1): 17377, 2021 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-34462463

RESUMEN

Down syndrome (DS) is caused by the trisomy of chromosome 21. Among the many disabilities found in individuals with DS is an increased risk of early-onset Alzheimer's disease (AD). Although higher oxidative stress and an upregulation of amyloid ß (Aß) peptides from an extra copy of the APP gene are attributed to the AD susceptibility, the relationship between the two factors is unclear. To address this issue, we established an in vitro cellular model using neurons differentiated from DS patient-derived induced pluripotent stem cells (iPSCs) and isogenic euploid iPSCs. Neurons differentiated from DS patient-derived iPSCs secreted more Aß compared to those differentiated from the euploid iPSCs. Treatment of the neurons with an antioxidant, N-acetylcysteine, significantly suppressed the Aß secretion. These findings suggest that oxidative stress has an important role in controlling the Aß level in neurons differentiated from DS patient-derived iPSCs and that N-acetylcysteine can be a potential therapeutic option to ameliorate the Aß secretion.


Asunto(s)
Acetilcisteína/farmacología , Péptidos beta-Amiloides/metabolismo , Antioxidantes/farmacología , Síndrome de Down/genética , Regulación hacia Abajo/efectos de los fármacos , Enfermedad de Alzheimer/etiología , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Síndrome de Down/complicaciones , Síndrome de Down/patología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos
3.
Mol Ther ; 29(2): 680-690, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33554867

RESUMEN

Adeno-associated virus (AAV) integrates into host genomes at low frequency, but when integration occurs in oncogenic hotspots it can cause hepatocellular carcinoma (HCC). Given the possibility of recombinant AAV (rAAV) integration leading to HCC, common causes of liver inflammation like non-alcoholic fatty liver disease (NAFLD) may increase the risk of rAAV-induced HCC. A rAAV targeting the oncogenic mouse Rian locus was used, and as expected led to HCC in all mice infected as neonates, likely due to growth-related hepatocyte proliferation in young mice. Mice infected with rAAV as adults did not develop HCC unless they were fed a diet leading to NAFLD, with increased inflammation and hepatocyte proliferation. Female mice were less susceptible to rAAV-induced HCC, and male mice with NAFLD treated with estrogen exhibited less inflammation and immune exhaustion associated with oncogenesis compared to those without estrogen. Adult NAFLD mice infected with a non-targeted control rAAV also developed HCC, though only half as frequently as those exposed to the Rian targeted rAAV. This study shows that adult mice exposed to rAAV gene therapy in the context of chronic liver disease developed HCC at high frequency, and thus warrants further study in humans given the high prevalence of NAFLD in the population.


Asunto(s)
Carcinoma Hepatocelular/etiología , Dependovirus/genética , Terapia Genética/efectos adversos , Vectores Genéticos/genética , Hepatopatías/complicaciones , Hepatopatías/etiología , Neoplasias Hepáticas/etiología , Animales , Carcinoma Hepatocelular/diagnóstico , Modelos Animales de Enfermedad , Terapia Genética/métodos , Incidencia , Hepatopatías/patología , Neoplasias Hepáticas/diagnóstico , Ratones
4.
Nat Rev Immunol ; 19(12): 723-733, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31417198

RESUMEN

The prospect of transplanting cells and tissues without the risk of immune rejection or the need for powerful immunosuppressive drugs is the 'holy grail' of transplantation medicine. Now, with the advent of pluripotent stem cells, CRISPR-Cas9 and other gene-editing technologies, the race to create 'off-the-shelf' donor cells that are invisible to the immune system ('universal cells') has started. One important approach for creating such cells involves the manipulation of genes required for immune recognition, in particular HLA class I and II proteins. Other approaches leverage knowledge of immune-cloaking strategies used by certain bacteria, viruses, parasites, the fetus and cancer cells to induce tolerance to allogeneic cell-based therapies by modifying cells to express immune-suppressive molecules such as PD-L1 and CTLA4-Ig. Various academic groups as well as biotechnology and pharmaceutical companies are on the verge of bringing these therapies into the clinic.


Asunto(s)
Ingeniería Genética , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase I/genética , Evasión Inmune , Animales , Femenino , Humanos , Neoplasias/inmunología , Placenta/inmunología , Embarazo , Trasplante de Células Madre
5.
JCI Insight ; 52019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31039138

RESUMEN

Monosomy 7 or deletion of 7q (del(7q)) are common clonal cytogenetic abnormalities associated with high grade myelodysplastic syndrome (MDS) arising in inherited and acquired bone marrow failure. Current non-transplant approaches to treat marrow failure may be complicated by stimulation of clonal outgrowth. To study the biological consequences of del(7q) within the context of a failing marrow, we generated induced pluripotent stem cells (iPSCs) derived from patients with Shwachman Diamond Syndrome (SDS), a bone marrow failure disorder with MDS predisposition, and genomically engineered a 7q deletion. The TGFß pathway was the top differentially regulated pathway in transcriptomic analysis of SDS versus SDSdel(7q) iPSCs. SMAD2 phosphorylation was increased in SDS relative to wild type cells consistent with hyperactivation of the TGFbeta pathway in SDS. Phospho-SMAD2 levels were reduced following 7q deletion in SDS cells and increased upon restoration of 7q diploidy. Inhibition of the TGFbeta pathway rescued hematopoiesis in SDS-iPSCs and in bone marrow hematopoietic cells from SDS patients while it had no impact on the SDSdel(7q) cells. These results identified a potential targetable vulnerability to improve hematopoiesis in an MDS-predisposition syndrome, and highlight the importance of the germline context of somatic alterations to inform precision medicine approaches to therapy.


Asunto(s)
Médula Ósea/patología , Síndromes Mielodisplásicos/prevención & control , Medicina de Precisión/métodos , Síndrome de Shwachman-Diamond/terapia , Médula Ósea/efectos de los fármacos , Ingeniería Celular , Deleción Cromosómica , Cromosomas Humanos Par 7/genética , Células HEK293 , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/patología , Cariotipificación , Síndromes Mielodisplásicos/genética , Fosforilación/genética , RNA-Seq , Síndrome de Shwachman-Diamond/diagnóstico , Síndrome de Shwachman-Diamond/genética , Síndrome de Shwachman-Diamond/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
6.
Mol Ther ; 26(5): 1255-1265, 2018 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-29606506

RESUMEN

X-linked severe combined immunodeficiency (X-SCID) has been successfully treated by hematopoietic stem cell (HSC) transduction with retroviral vectors expressing the interleukin-2 receptor subunit gamma gene (IL2RG), but several patients developed malignancies due to vector integration near cellular oncogenes. This adverse side effect could in principle be avoided by accurate IL2RG gene editing with a vector that does not contain a functional promoter or IL2RG gene. Here, we show that adeno-associated virus (AAV) gene editing vectors can insert a partial Il2rg cDNA at the endogenous Il2rg locus in X-SCID murine bone marrow cells and that these ex vivo-edited cells repopulate transplant recipients and produce CD4+ and CD8+ T cells. Circulating, edited lymphocytes increased over time and appeared in secondary transplant recipients, demonstrating successful editing in long-term repopulating cells. Random vector integration events were nearly undetectable, and malignant transformation of the transplanted cells was not observed. Similar editing frequencies were observed in human hematopoietic cells. Our results demonstrate that therapeutically relevant HSC gene editing can be achieved by AAV vectors in the absence of site-specific nucleases and suggest that this may be a safe and effective therapy for hematopoietic diseases where in vivo selection can increase edited cell numbers.


Asunto(s)
Dependovirus/genética , Edición Génica , Vectores Genéticos/genética , Subunidad gamma Común de Receptores de Interleucina/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Alelos , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Orden Génico , Terapia Genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunoterapia Adoptiva , Ratones , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia
7.
Cell ; 171(5): 1057-1071.e11, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29033131

RESUMEN

Type I interferon restrains interleukin-1ß (IL-1ß)-driven inflammation in macrophages by upregulating cholesterol-25-hydroxylase (Ch25h) and repressing SREBP transcription factors. However, the molecular links between lipid metabolism and IL-1ß production remain obscure. Here, we demonstrate that production of 25-hydroxycholesterol (25-HC) by macrophages is required to prevent inflammasome activation by the DNA sensor protein absent in melanoma 2 (AIM2). We find that in response to bacterial infection or lipopolysaccharide (LPS) stimulation, macrophages upregulate Ch25h to maintain repression of SREBP2 activation and cholesterol synthesis. Increasing macrophage cholesterol content is sufficient to trigger IL-1ß release in a crystal-independent but AIM2-dependent manner. Ch25h deficiency results in cholesterol-dependent reduced mitochondrial respiratory capacity and release of mitochondrial DNA into the cytosol. AIM2 deficiency rescues the increased inflammasome activity observed in Ch25h-/-. Therefore, activated macrophages utilize 25-HC in an anti-inflammatory circuit that maintains mitochondrial integrity and prevents spurious AIM2 inflammasome activation.


Asunto(s)
Colesterol/metabolismo , Inflamasomas/metabolismo , Macrófagos/metabolismo , Animales , Colesterol/biosíntesis , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Hidroxicolesteroles/metabolismo , Inflamasomas/inmunología , Inflamación/inmunología , Inflamación/patología , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Listeria monocytogenes/fisiología , Listeriosis/inmunología , Macrófagos/citología , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Mitocondrias/metabolismo , Oxiesteroles/metabolismo
8.
Nat Genet ; 49(8): 1267-1273, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28628105

RESUMEN

Vectors based on adeno-associated virus type 2 (AAV2) are powerful tools for gene transfer and genome editing applications. The level of interest in this system has recently surged in response to reports of therapeutic efficacy in human clinical trials, most notably for those in patients with hemophilia B (ref. 3). Understandably, a recent report drawing an association between AAV2 integration events and human hepatocellular carcinoma (HCC) has generated controversy about the causal or incidental nature of this association and the implications for AAV vector safety. Here we describe and functionally characterize a previously unknown liver-specific enhancer-promoter element in the wild-type AAV2 genome that is found between the stop codon of the cap gene, which encodes proteins that form the capsid, and the right-hand inverted terminal repeat. This 124-nt sequence is within the 163-nt common insertion region of the AAV genome, which has been implicated in the dysregulation of known HCC driver genes and thus offers added insight into the possible link between AAV integration events and the multifactorial pathogenesis of HCC.


Asunto(s)
Regiones no Traducidas 3' , Dependovirus/genética , Elementos de Facilitación Genéticos , Genoma Viral , Hígado/virología , Regiones Promotoras Genéticas , Animales , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Femenino , Vectores Genéticos/genética , Humanos , Neoplasias Hepáticas/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Transgenes
9.
Nat Biotechnol ; 35(8): 765-772, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28504668

RESUMEN

Polymorphisms in the human leukocyte antigen (HLA) class I genes can cause the rejection of pluripotent stem cell (PSC)-derived products in allogeneic recipients. Disruption of the Beta-2 Microglobulin (B2M) gene eliminates surface expression of all class I molecules, but leaves the cells vulnerable to lysis by natural killer (NK) cells. Here we show that this 'missing-self' response can be prevented by forced expression of minimally polymorphic HLA-E molecules. We use adeno-associated virus (AAV)-mediated gene editing to knock in HLA-E genes at the B2M locus in human PSCs in a manner that confers inducible, regulated, surface expression of HLA-E single-chain dimers (fused to B2M) or trimers (fused to B2M and a peptide antigen), without surface expression of HLA-A, B or C. These HLA-engineered PSCs and their differentiated derivatives are not recognized as allogeneic by CD8+ T cells, do not bind anti-HLA antibodies and are resistant to NK-mediated lysis. Our approach provides a potential source of universal donor cells for applications where the differentiated derivatives lack HLA class II expression.


Asunto(s)
Antígenos HLA/inmunología , Células Asesinas Naturales/inmunología , Células Madre Pluripotentes/inmunología , Trasplantes/inmunología , Animales , Femenino , Rechazo de Injerto/inmunología , Antígenos HLA/química , Antígenos HLA/genética , Humanos , Ratones , Células Madre Pluripotentes/química , Células Madre Pluripotentes/citología , Trasplantes/química , Trasplantes/citología
10.
Mol Ther ; 24(3): 582-91, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26444081

RESUMEN

Many applications of pluripotent stem cells (PSCs) require efficient editing of silent chromosomal genes. Here, we show that a major limitation in isolating edited clones is silencing of the selectable marker cassette after homologous recombination and that this can be overcome by using a ubiquitous chromatin opening element (UCOE) promoter-driven transgene. We use this strategy to edit the silent IL2RG locus in human PSCs with a recombinant adeno-associated virus (rAAV)-targeting vector in the absence of potentially genotoxic, site-specific nucleases and show that IL2RG is required for natural killer and T-cell differentiation of human PSCs. Insertion of an active UCOE promoter into a silent locus altered the histone modification and cytosine methylation pattern of surrounding chromatin, but these changes resolved when the UCOE promoter was removed. This same approach could be used to correct IL2RG mutations in X-linked severe combined immunodeficiency patient-derived induced PSCs (iPSCs), to prevent graft versus host disease in regenerative medicine applications, or to edit other silent genes.


Asunto(s)
Edición Génica , Silenciador del Gen , Subunidad gamma Común de Receptores de Interleucina/genética , Células Madre Pluripotentes/metabolismo , Diferenciación Celular , Supervivencia Celular/genética , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Epigénesis Genética , Técnicas de Inactivación de Genes , Marcación de Gen , Sitios Genéticos , Humanos , Células Asesinas Naturales/citología , Células Madre Pluripotentes/citología , Regiones Promotoras Genéticas , Subgrupos de Linfocitos T/citología , Transgenes , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética
11.
Immunity ; 43(6): 1125-36, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26682985

RESUMEN

The interleukin-1 receptor I (IL-1RI) is critical for host resistance to Mycobacterium tuberculosis (Mtb), yet the mechanisms of IL-1RI-mediated pathogen control remain unclear. Here, we show that without IL-1RI, Mtb-infected newly recruited Ly6G(hi) myeloid cells failed to upregulate tumor necrosis factor receptor I (TNF-RI) and to produce reactive oxygen species, resulting in compromised pathogen control. Furthermore, simultaneous ablation of IL-1RI and TNF-RI signaling on either stroma or hematopoietic cells led to early lethality, indicating non-redundant and synergistic roles of IL-1 and TNF in mediating macrophage-stroma cross-talk that was critical for optimal control of Mtb infection. Finally, we show that even in the presence of functional Mtb-specific adaptive immunity, the lack of IL-1α and not IL-1ß led to an exuberant intracellular pathogen replication and progressive non-resolving inflammation. Our study reveals functional interdependence between IL-1 and TNF in enabling Mtb control mechanisms that are critical for host survival.


Asunto(s)
Interleucina-1alfa/inmunología , Tuberculosis/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Separación Celular , Modelos Animales de Enfermedad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium tuberculosis , Receptores Tipo I de Interleucina-1/inmunología
12.
Nat Genet ; 47(10): 1104-5, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26417859

RESUMEN

Adeno-associated virus (AAV) vectors have been widely adopted for use in gene therapy. A new study raises concerns regarding this approach, reporting that chromosomal insertions of AAV serotype 2 seem to activate proto-oncogenes in human hepatocellular carcinoma.


Asunto(s)
Carcinoma Hepatocelular/genética , Dependovirus/genética , Neoplasias Hepáticas/genética , Mutagénesis Insercional , Humanos
13.
J Lipid Res ; 56(3): 722-736, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25598080

RESUMEN

The spectrum of nonalcoholic fatty liver disease (NAFLD) includes steatosis, nonalcoholic steatohepatitis (NASH), and cirrhosis. Recognition and timely diagnosis of these different stages, particularly NASH, is important for both potential reversibility and limitation of complications. Liver biopsy remains the clinical standard for definitive diagnosis. Diagnostic tools minimizing the need for invasive procedures or that add information to histologic data are important in novel management strategies for the growing epidemic of NAFLD. We describe an "omics" approach to detecting a reproducible signature of lipid metabolites, aqueous intracellular metabolites, SNPs, and mRNA transcripts in a double-blinded study of patients with different stages of NAFLD that involves profiling liver biopsies, plasma, and urine samples. Using linear discriminant analysis, a panel of 20 plasma metabolites that includes glycerophospholipids, sphingolipids, sterols, and various aqueous small molecular weight components involved in cellular metabolic pathways, can be used to differentiate between NASH and steatosis. This identification of differential biomolecular signatures has the potential to improve clinical diagnosis and facilitate therapeutic intervention of NAFLD.


Asunto(s)
Lípidos/sangre , Lípidos/orina , Enfermedad del Hígado Graso no Alcohólico , Polimorfismo de Nucleótido Simple , Adulto , Biomarcadores/metabolismo , Biomarcadores/orina , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/epidemiología , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/orina
14.
Nat Struct Mol Biol ; 21(11): 969-75, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25282150

RESUMEN

To determine which genomic features promote homologous recombination, we created a genome-wide map of gene targeting sites. We used an adeno-associated virus vector to target identical loci introduced as transcriptionally active retroviral vectors. A comparison of ~2,000 targeted and untargeted sites showed that targeting occurred throughout the human genome and was not influenced by the presence of nearby CpG islands, sequence repeats or DNase I-hypersensitive sites. Targeted sites were preferentially located within transcription units, especially when the target loci were transcribed in the opposite orientation to their surrounding chromosomal genes. We determined the impact of DNA replication by mapping replication forks, which revealed a preference for recombination at target loci transcribed toward an incoming fork. Our results constitute the first genome-wide screen of gene targeting in mammalian cells and demonstrate a strong recombinogenic effect of colliding polymerases.


Asunto(s)
Replicación del ADN , Desoxirribonucleasa I/genética , Dependovirus/genética , Genoma Humano , Recombinación Homóloga , Transcripción Genética , Línea Celular Tumoral , Mapeo Cromosómico , Islas de CpG , Desoxirribonucleasa I/metabolismo , Sitios Genéticos , Vectores Genéticos , Células HEK293 , Humanos
15.
Science ; 345(6197): 679-84, 2014 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-25104388

RESUMEN

Type I interferon (IFN) protects against viruses, yet it also has a poorly understood suppressive influence on inflammation. Here, we report that activated mouse macrophages lacking the IFN-stimulated gene cholesterol 25-hydroxylase (Ch25h) and that are unable to produce the oxysterol 25-hydroxycholesterol (25-HC) overproduce inflammatory interleukin-1 (IL-1) family cytokines. 25-HC acts by antagonizing sterol response element-binding protein (SREBP) processing to reduce Il1b transcription and to broadly repress IL-1-activating inflammasomes. In accord with these dual actions of 25-HC, Ch25h-deficient mice exhibit increased sensitivity to septic shock, exacerbated experimental autoimmune encephalomyelitis, and a stronger ability to repress bacterial growth. These findings identify an oxysterol, 25-HC, as a critical mediator in the negative-feedback pathway of IFN signaling on IL-1 family cytokine production and inflammasome activity.


Asunto(s)
Hidroxicolesteroles/metabolismo , Inflamación/genética , Interferón Tipo I/inmunología , Esteroide Hidroxilasas/inmunología , Animales , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Retroalimentación Fisiológica , Inflamasomas/genética , Inflamasomas/inmunología , Inflamación/inmunología , Inflamación/microbiología , Interleucina-1/inmunología , Activación de Macrófagos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Elementos de Respuesta/genética , Choque Séptico/genética , Choque Séptico/inmunología , Esteroide Hidroxilasas/genética
16.
Nucleic Acids Res ; 42(5): 3119-24, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24371280

RESUMEN

DNA mismatches that occur between vector homology arms and chromosomal target sequences reduce gene targeting frequencies in several species; however, this has not been reported in human cells. Here we demonstrate that even a single mismatched base pair can significantly decrease human gene targeting frequencies. In addition, we show that homology arm polymorphisms can be used to direct allele-specific targeting or to improve unfavorable vector designs that introduce deletions.


Asunto(s)
Marcación de Gen , Polimorfismo Genético , Disparidad de Par Base , Línea Celular Tumoral , Cromosomas/química , Sitios Genéticos , Vectores Genéticos/química , Humanos , Polimorfismo de Nucleótido Simple
17.
J Biol Chem ; 288(50): 35812-23, 2013 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-24189069

RESUMEN

25-Hydroxycholesterol (25OHC) is an enzymatically derived oxidation product of cholesterol that modulates lipid metabolism and immunity. 25OHC is synthesized in response to interferons and exerts broad antiviral activity by as yet poorly characterized mechanisms. To gain further insights into the basis for antiviral activity, we evaluated time-dependent responses of the macrophage lipidome and transcriptome to 25OHC treatment. In addition to altering specific aspects of cholesterol and sphingolipid metabolism, we found that 25OHC activates integrated stress response (ISR) genes and reprograms protein translation. Effects of 25OHC on ISR gene expression were independent of liver X receptors and sterol-response element-binding proteins and instead primarily resulted from activation of the GCN2/eIF2α/ATF4 branch of the ISR pathway. These studies reveal that 25OHC activates the integrated stress response, which may contribute to its antiviral activity.


Asunto(s)
Hidroxicolesteroles/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Animales , Células de la Médula Ósea/citología , Ésteres del Colesterol/metabolismo , Perfilación de la Expresión Génica , Hidroxicolesteroles/metabolismo , Receptores X del Hígado , Macrófagos/citología , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Muromegalovirus/fisiología , Receptores Nucleares Huérfanos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Esfingolípidos/metabolismo , Proteínas de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores
18.
J Lipid Res ; 54(9): 2525-42, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23776196

RESUMEN

Studies of macrophage biology have been significantly advanced by the availability of cell lines such as RAW264.7 cells. However, it is unclear how these cell lines differ from primary macrophages such as thioglycolate-elicited peritoneal macrophages (TGEMs). We used the inflammatory stimulus Kdo2-lipid A (KLA) to stimulate RAW264.7 and TGEM cells. Temporal changes of lipid and gene expression levels were concomitantly measured and a systems-level analysis was performed on the fold-change data. Here we present a comprehensive comparison between the two cell types. Upon KLA treatment, both RAW264.7 and TGEM cells show a strong inflammatory response. TGEM (primary) cells show a more rapid and intense inflammatory response relative to RAW264.7 cells. DNA levels (fold-change relative to control) are reduced in RAW264.7 cells, correlating with greater downregulation of cell cycle genes. The transcriptional response suggests that the cholesterol de novo synthesis increases considerably in RAW264.7 cells, but 25-hydroxycholesterol increases considerably in TGEM cells. Overall, while RAW264.7 cells behave similarly to TGEM cells in some ways and can be used as a good model for inflammation- and immune function-related kinetic studies, they behave differently than TGEM cells in other aspects of lipid metabolism and phenotypes used as models for various disorders such as atherosclerosis.


Asunto(s)
Metabolismo de los Lípidos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Tioglicolatos/farmacología , Animales , Línea Celular , Citocinas/metabolismo , Perfilación de la Expresión Génica , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Ratones , Transcripción Genética/efectos de los fármacos
19.
Mol Ther ; 21(6): 1232-41, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23629003

RESUMEN

The clinical use of human pluripotent stem cells and their derivatives is limited by the rejection of transplanted cells due to differences in their human leukocyte antigen (HLA) genes. This has led to the proposed use of histocompatible, patient-specific stem cells; however, the preparation of many different stem cell lines for clinical use is a daunting task. Here, we develop two distinct genetic engineering approaches that address this problem. First, we use a combination of gene targeting and mitotic recombination to derive HLA-homozygous embryonic stem cell (ESC) subclones from an HLA-heterozygous parental line. A small bank of HLA-homozygous stem cells with common haplotypes would match a significant proportion of the population. Second, we derive HLA class I-negative cells by targeted disruption of both alleles of the Beta-2 Microglobulin (B2M) gene in ESCs. Mixed leukocyte reactions and peptide-specific HLA-restricted CD8(+) T cell responses were reduced in class I-negative cells that had undergone differentiation in embryoid bodies. These B2M(-/-) ESCs could act as universal donor cells in applications where the transplanted cells do not express HLA class II genes. Both approaches used adeno-associated virus (AAV) vectors for efficient gene targeting in the absence of potentially genotoxic nucleases, and produced pluripotent, transgene-free cell lines.


Asunto(s)
Antígenos HLA/genética , Células Madre Pluripotentes/citología , Alelos , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Línea Celular , Células Cultivadas , Dependovirus/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Marcación de Gen , Ingeniería Genética , Vectores Genéticos , Antígenos HLA/metabolismo , Haplotipos , Histocompatibilidad/genética , Homocigoto , Humanos , Células Madre Pluripotentes/metabolismo , Recombinación Genética , Microglobulina beta-2/genética , Microglobulina beta-2/metabolismo
20.
Mol Ther ; 21(5): 964-72, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23531552

RESUMEN

The development of leukemia following gammaretroviral vector-mediated gene therapy for X-linked severe combined immunodeficiency disease and chronic granulomatous disease (CGD) has emphasized the need for long-term follow-up in animals treated with hematopoietic stem cell gene therapy. In this study, we report the long-term follow-up (4-7 years) of four dogs with canine leukocyte adhesion deficiency (CLAD) treated with foamy viral (FV) vector-mediated gene therapy. All four CLAD dogs previously received nonmyeloablative conditioning with 200 cGy total body irradiation followed by infusion of autologous, CD34(+) hematopoietic stem cells transduced by a FV vector expressing canine CD18 from an internal Murine Stem Cell Virus (MSCV) promoter. CD18(+) leukocyte levels were >2% following infusion of vector-transduced cells leading to ongoing reversal of the CLAD phenotype for >4 years. There was no clinical development of lymphoid or myeloid leukemia in any of the four dogs and integration site analysis did not reveal insertional oncogenesis. These results showing disease correction/amelioration of disease in CLAD without significant adverse events provide support for the use of a FV vector to treat children with leukocyte adhesion deficiency type 1 (LAD-1) in a human gene therapy clinical trial.


Asunto(s)
Terapia Genética , Vectores Genéticos/genética , Síndrome de Deficiencia de Adhesión del Leucocito/genética , Síndrome de Deficiencia de Adhesión del Leucocito/terapia , Spumavirus/genética , Animales , Antígenos CD34/metabolismo , Médula Ósea , Antígenos CD18/metabolismo , Modelos Animales de Enfermedad , Perros , Femenino , Estudios de Seguimiento , Técnicas de Transferencia de Gen , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos , Recuento de Leucocitos , Leucocitos/metabolismo , Masculino , Subgrupos de Linfocitos T/metabolismo , Transducción Genética , Integración Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA