Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Immunother ; 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38775140

RESUMEN

Cancer remains a leading cause of death worldwide, but immunotherapies hold promises to cure it by awaking the patient's immune system to provide long-term protection. Cell therapies, involving the infusion of immune cells, either directly or genetically modified, are being developed to recognize and destroy cancer cells. Here, we explored the potential of a new synthetic circuit to reprogram B cells to cure cancers. This circuit consists in a sensor (a membrane-anchored IgG1), a transducer (a fragment of the NR4A1 promoter) and an effector molecule. Upon recognition of its target, this sensor triggers signaling pathways leading to the activation of the transducer and to effector expression (here, a reporter molecule). We showed that this circuit could discriminate tumors expressing the target antigen from those that did not, in a dose dependent manner in vitro. Going further, we replaced the original membrane-anchored sensor by an immunoglobulin expression cassette that can not only be membrane-anchored but also be secreted depending on B-cell maturation status. This allowed concomitant activation of the circuit and secretion of transgenic antibodies directed against the targeted antigen. Of note, these antibodies could correctly bind their target and were recognized by FcR expressed at the surface of immune cells, which should synergically amplify the action of the effector. The potential of reprogrammed B cells remains to be assessed in vivo by implementing a therapeutic effector. In the future, B-cell reprogramming platforms should allow personalized cancer treatment by adapting both the sensor and the therapeutic effectors to patients.

2.
Mol Ther Nucleic Acids ; 33: 1-14, 2023 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-37359346

RESUMEN

The expansion of genetic engineering has brought a new dimension for synthetic immunology. Immune cells are perfect candidates because of their ability to patrol the body, interact with many cell types, proliferate upon activation, and differentiate in memory cells. This study aimed at implementing a new synthetic circuit in B cells, allowing the expression of therapeutic molecules in a temporally and spatially restricted manner that is induced by the presence of specific antigens. This should enhance endogenous B cell functions in terms of recognition and effector properties. We developed a synthetic circuit encoding a sensor (a membrane-anchored B cell receptor targeting a model antigen), a transducer (a minimal promoter induced by the activated sensor), and effector molecules. We isolated a 734-bp-long fragment of the NR4A1 promoter, specifically activated by the sensor signaling cascade in a fully reversible manner. We demonstrate full antigen-specific circuit activation as its recognition by the sensor induced the activation of the NR4A1 promoter and the expression of the effector. Overall, such novel synthetic circuits offer huge possibilities for the treatment of many pathologies, as they are completely programmable; thus, the signal-specific sensors and effector molecules can be adapted to each disease.

3.
Reprod Fertil Dev ; 35(4): 307-320, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36593258

RESUMEN

CONTEXT: Mammalian target of rapamycin complex 1 (mTORC1) is an essential sensor that regulates fundamental biological processes like cell growth, proliferation and energy metabolism. The treatment of disease by sirolimus, a mTORC1 inhibitor, causes adverse effects, such as female fertility disorders. AIMS: The objective of the study was to decipher the reproductive consequences of a downregulation of mTORC1 in the hypothalamus. METHODS: The reduced expression of mTORC1 was induced after intracerebroventricular injection of lentivirus expressing a short hairpin RNA (shRNA) against regulatory associated protein of TOR (raptor) in adult female mice (ShRaptor mice). KEY RESULTS: The ShRaptor mice were fertile and exhibited a 15% increase in the litter size compared with control mice. The histological analysis showed an increase in antral, preovulatory follicles and ovarian cysts. In the hypothalamus, the GnRH mRNA and FSH levels in ShRaptor mice were significantly elevated. CONCLUSIONS: These results support the hypothesis that mTORC1 in the central nervous system participates in the regulation of female fertility and ovarian function by influencing the GnRH neuronal activity. IMPLICATIONS: These results suggest that a lower mTORC1 activity directly the central nervous system leads to a deregulation in the oestrous cycle and an induction of ovarian cyst development.


Asunto(s)
Quistes Ováricos , Rapaces , Femenino , Animales , Ratones , Humanos , Serina-Treonina Quinasas TOR/metabolismo , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Factores de Transcripción/metabolismo , ARN Interferente Pequeño , Hipotálamo/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Rapaces/genética , Rapaces/metabolismo , Mamíferos/genética
4.
Cancer Immunol Immunother ; 71(7): 1771-1775, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34748076

RESUMEN

Here, we report a novel experimental setup to perform adoptive transfer of gene-edited B cells using humanized immune system mice by infusing autologous HIS mouse-derived human B cells "educated" in a murine context and thus rendered tolerant to the host. The present approach presents two advantages over the conventional humanized PBMC mouse models: (i) it circumvents the risk of xenogeneic graft-versus-host reaction and (ii) it mimics more closely human immune responses, thus favoring clinical translation. We show that the frequencies and numbers of transduced B cells in recipient's spleens one week post-transfer are within the range of the size of the pre-immune B cell population specific for a given protein antigen in the mouse. They are also compatible with the B cell numbers required to elicit a sizeable immune response upon immunization. Altogether, our findings pave the way for future studies aiming at assessing therapeutic interventions involving B cell reprogramming for instance by an antibody transgene in a "humanized" hematopoietic setting.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Leucocitos Mononucleares , Traslado Adoptivo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones SCID
5.
Blood Adv ; 3(3): 461-475, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30755435

RESUMEN

T cells represent a valuable tool for treating cancers and infectious and inherited diseases; however, they are mainly short-lived in vivo. T-cell therapies would strongly benefit from gene transfer into long-lived persisting naive T cells or T-cell progenitors. Here we demonstrate that baboon envelope glycoprotein pseudotyped lentiviral vectors (BaEV-LVs) far outperformed other LV pseudotypes for transduction of naive adult and fetal interleukin-7-stimulated T cells. Remarkably, BaEV-LVs efficiently transduced thymocytes and T-cell progenitors generated by culture of CD34+ cells on Delta-like ligand 4 (Dll4). Upon NOD/SCIDγC-/- engraftment, high transduction levels (80%-90%) were maintained in all T-cell subpopulations. Moreover, T-cell lineage reconstitution was accelerated in NOD/SCIDγC-/- recipients after T-cell progenitor injection compared with hematopoietic stem cell transplantation. Furthermore, γC-encoding BaEV-LVs very efficiently transduced Dll4-generated T-cell precursors from a patient with X-linked severe combined immunodeficiency (SCID-X1), which fully rescued T-cell development in vitro. These results indicate that BaEV-LVs are valuable tools for the genetic modification of naive T cells, which are important targets for gene therapy. Moreover, they allowed for the generation of gene-corrected T-cell progenitors that rescued SCID-X1 T-cell development in vitro. Ultimately, the coinjection of LV-corrected T-cell progenitors and hematopoietic stem cells might accelerate T-cell reconstitution in immunodeficient patients.


Asunto(s)
Lentivirus/genética , Células Madre/metabolismo , Animales , Ratones , Ratones Endogámicos NOD , Ratones SCID , Papio
6.
Blood Adv ; 1(23): 2088-2104, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-29296856

RESUMEN

Hematopoietic stem cell (HSC)-based gene therapy trials are now moving toward the use of lentiviral vectors (LVs) with success. However, one challenge in the field remains: efficient transduction of HSCs without compromising their stem cell potential. Here we showed that measles virus glycoprotein-displaying LVs (hemagglutinin and fusion protein LVs [H/F-LVs]) were capable of transducing 100% of early-acting cytokine-stimulated human CD34+ (hCD34+) progenitor cells upon a single application. Strikingly, these H/F-LVs also allowed transduction of up to 70% of nonstimulated quiescent hCD34+ cells, whereas conventional vesicular stomatitis virus G (VSV-G)-LVs reached 5% at the most with H/F-LV entry occurring exclusively through the CD46 complement receptor. Importantly, reconstitution of NOD/SCIDγc-/- (NSG) mice with H/F-LV transduced prestimulated or resting hCD34+ cells confirmed these high transduction levels in all myeloid and lymphoid lineages. Remarkably, for resting CD34+ cells, secondary recipients exhibited increasing transduction levels of up to 100%, emphasizing that H/F-LVs efficiently gene-marked HSCs in the resting state. Because H/F-LVs promoted ex vivo gene modification of minimally manipulated CD34+ progenitors that maintained stemness, we assessed their applicability in Fanconi anemia, a bone marrow (BM) failure with chromosomal fragility. Notably, only H/F-LVs efficiently gene-corrected minimally stimulated hCD34+ cells in unfractionated BM from these patients. These H/F-LVs improved HSC gene delivery in the absence of cytokine stimulation while maintaining their stem cell potential. Thus, H/F-LVs will facilitate future clinical applications requiring HSC gene modification, including BM failure syndromes, for which treatment has been very challenging up to now.

7.
mBio ; 6(2): e02427, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25759505

RESUMEN

UNLABELLED: Nipah virus and Hendra virus are emerging, highly pathogenic, zoonotic paramyxoviruses that belong to the genus Henipavirus. They infect humans as well as numerous mammalian species. Both viruses use ephrin-B2 and -B3 as cell entry receptors, and following initial entry into an organism, they are capable of rapid spread throughout the host. We have previously reported that Nipah virus can use another attachment receptor, different from its entry receptors, to bind to nonpermissive circulating leukocytes, thereby promoting viral dissemination within the host. Here, this attachment molecule was identified as heparan sulfate for both Nipah virus and Hendra virus. Cells devoid of heparan sulfate were not able to mediate henipavirus trans-infection and showed reduced permissivity to infection. Virus pseudotyped with Nipah virus glycoproteins bound heparan sulfate and heparin but no other glycosaminoglycans in a surface plasmon resonance assay. Furthermore, heparin was able to inhibit the interaction of the viruses with the heparan sulfate and to block cell-mediated trans-infection of henipaviruses. Moreover, heparin was shown to bind to ephrin-B3 and to restrain infection of permissive cells in vitro. Consequently, treatment with heparin devoid of anticoagulant activity improved the survival of Nipah virus-infected hamsters. Altogether, these results reveal heparan sulfate as a new attachment receptor for henipaviruses and as a potential therapeutic target for the development of novel approaches against these highly lethal infections. IMPORTANCE: The Henipavirus genus includes two closely related, highly pathogenic paramyxoviruses, Nipah virus and Hendra virus, which cause elevated morbidity and mortality in animals and humans. Pathogenesis of both Nipah virus and Hendra virus infection is poorly understood, and efficient antiviral treatment is still missing. Here, we identified heparan sulfate as a novel attachment receptor used by both viruses to bind host cells. We demonstrate that heparin was able to inhibit the interaction of the viruses with heparan sulfate and to block cell-mediated trans-infection of henipaviruses. Moreover, heparin also bound to the viral entry receptor and thereby restricted infection of permissive cells in vitro. Consequently, heparin treatment improved survival of Nipah virus-infected hamsters. These results uncover an important role of heparan sulfate in henipavirus infection and open novel perspectives for the development of heparan sulfate-targeting therapeutic approaches for these emerging infections.


Asunto(s)
Virus Hendra/fisiología , Infecciones por Henipavirus/patología , Infecciones por Henipavirus/virología , Heparitina Sulfato/metabolismo , Virus Nipah/fisiología , Acoplamiento Viral , Animales , Células Cultivadas , Cricetinae , Humanos , Leucocitos/virología , Resonancia por Plasmón de Superficie , Análisis de Supervivencia
8.
Viral Immunol ; 27(10): 512-20, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25343523

RESUMEN

As a prelude to immunization studies in nonhuman primates, we compared in mice the immunogenicity of a simian immunodeficiency virus (SIV)-based integrase (IN)-defective lentiviral vector (IDLV) encoding the model antigen-enhanced green fluorescence protein (eGFP) in the presence or absence of the murine granulocyte-macrophage colony-stimulating factor (mGM-CSF) expressed from an internal ribosomal entry site (IRES) sequence. BALB/c mice were immunized once intramuscularly with IDLV expressing eGFP alone or eGFP and mGM-CSF and immune responses were evaluated up to 90 days from the single intramuscular immunization. Results indicated that the mGM-CSF was unable to improve the magnitude and quality of the immune response against the eGFP transgene in the context of the SIV-based IDLV, as evaluated by enzyme-linked immunosorbent spot (ELISPOT) assays for interferon-γ (IFN-γ) and by intracellular cytokine staining for IFN-γ, interleukin-2 (IL-2), and tumor necrosis factor-alpha (TNF-α). These findings suggest that for vaccination purposes, the presence of mGM-CSF expressed after the IRES in a SIV-based IDLV system does not favor the improvement of the immunological response against the transgene of interest. Further studies should investigate whether the selection of a different cytokine gene might improve the immune response against the transgene.


Asunto(s)
Portadores de Fármacos , Expresión Génica , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Virus de la Inmunodeficiencia de los Simios/genética , Linfocitos T/inmunología , Vacunas Virales/inmunología , Animales , Ensayo de Immunospot Ligado a Enzimas , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/inmunología , Inyecciones Intramusculares , Integrasas/deficiencia , Interferón gamma/análisis , Interleucina-2/análisis , Ratones Endogámicos BALB C , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Virus de la Inmunodeficiencia de los Simios/enzimología , Coloración y Etiquetado , Factor de Necrosis Tumoral alfa/análisis , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
9.
Blood ; 124(8): 1221-31, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-24951430

RESUMEN

Hematopoietic stem cell (HSC)-based gene therapy holds promise for the cure of many diseases. The field is now moving toward the use of lentiviral vectors (LVs) as evidenced by 4 successful clinical trials. These trials used vesicular-stomatitis-virus-G protein (VSV-G)-LVs at high doses combined with strong cytokine-cocktail stimulation to obtain therapeutically relevant transduction levels; however, they might compromise the HSC character. Summarizing all these disadvantages, alternatives to VSV-G-LVs are urgently needed. We generated here high-titer LVs pseudotyped with a baboon retroviral envelope glycoprotein (BaEV-LVs), resistant to human complement. Under mild cytokine prestimulation to preserve the HSC characteristics, a single BaEV-LV application at a low dose, resulted in up to 90% of hCD34(+) cell transduction. Even more striking was that these new BaEV-LVs allowed, at low doses, efficient transduction of up to 30% of quiescent hCD34(+) cells, whereas high-dose VSV-G-LVs were insufficient. Importantly, reconstitution of NOD/Lt-SCID/γc(-/-) (NSG) mice with BaEV-LV-transduced hCD34(+) cells maintained these high transduction levels in all myeloid and lymphoid lineages, including early progenitors. This transduction pattern was confirmed or even increased in secondary NSG recipient mice. This suggests that BaEV-LVs efficiently transduce true HSCs and could improve HSC-based gene therapy, for which high-level HSC correction is needed for life-long cure.


Asunto(s)
Betaretrovirus/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Células Madre Hematopoyéticas , Lentivirus/genética , Transducción Genética , Proteínas del Envoltorio Viral/genética , Animales , Antígenos CD34 , Línea Celular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Macaca , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID
10.
Development ; 141(10): 2096-107, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24803656

RESUMEN

The four related mammalian MEX-3 RNA-binding proteins are evolutionarily conserved molecules for which the in vivo functions have not yet been fully characterized. Here, we report that male mice deficient for the gene encoding Mex3b are subfertile. Seminiferous tubules of Mex3b-deficient mice are obstructed as a consequence of the disrupted phagocytic capacity of somatic Sertoli cells. In addition, both the formation and the integrity of the blood-testis barrier are compromised owing to mislocalization of N-cadherin and connexin 43 at the surface of Sertoli cells. We further establish that Mex3b acts to regulate the cortical level of activated Rap1, a small G protein controlling phagocytosis and cell-cell interaction, through the activation and transport of Rap1GAP. The active form of Rap1 (Rap1-GTP) is abnormally increased at the membrane cortex and chemically restoring Rap1-GTP to physiological levels rescues the phagocytic and adhesion abilities of Sertoli cells. Overall, these findings implicate Mex3b in the spatial organization of the Rap1 pathway that orchestrates Sertoli cell functions.


Asunto(s)
Proteínas de Unión al ARN/fisiología , Células de Sertoli/fisiología , Proteínas de Unión al GTP rap1/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos , Femenino , Humanos , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas de Unión al ARN/genética , Epitelio Seminífero/metabolismo , Células de Sertoli/metabolismo , Transducción de Señal , Distribución Tisular/genética , Proteínas de Unión al GTP rap1/genética
12.
Biotechniques ; 55(3): 137-40, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24003946

RESUMEN

Efficient production of transgenic animals using low-titer lentiviral constructs remains challenging. Here we demonstrate that microinjection of simian immundeficiency virus-derived lentiviral constructs can produce transgenic mice and rats with high efficiency even when using low-titer virus preparations.


Asunto(s)
Animales Modificados Genéticamente/genética , Técnicas de Transferencia de Gen , Virus de la Inmunodeficiencia de los Simios/genética , Animales , Embrión de Mamíferos , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células Jurkat , Ratones , Micromanipulación , Ratas , Ratas Wistar
13.
Genesis ; 51(3): 193-200, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23281269

RESUMEN

Immune tolerance to self-antigens is a complex process that utilizes multiple mechanisms working in concert to maintain homeostasis and prevent autoimmunity. Considerable progress in deciphering the mechanisms controlling the activation or deletion of T cells has been made by using T cell receptor (TCR) transgenic mice. One such model is the F5 model in which CD8 T cells express a TCR specific for an epitope derived from the influenza NP68 protein. Our aim was to create transgenic mouse models expressing constitutively the NP68 epitope fused to enhanced green fluorescent protein (EGFP) in order to assess unambiguously the relative levels of NP68 epitope expressed by single cells. We used a lentiviral-based approach to generate two independent transgenic mouse strains expressing the fusion protein EGFP-NP68 under the control of CAG (CMV immediate early enhancer and the chicken ß-actin promoter) or spleen focus-forming virus (SFFV) promoters. Analysis of the pattern of EGFP expression in the hematopoietic compartment showed that CAG and SFFV promoters are differentially regulated during T cell development. However, both promoters drove high EGFP-NP68 expression in dendritic cells (pDCs, CD8α(+) cDCs, and CD8α(-) cDCs) from spleen or generated in vitro following differentiation from bone-marrow progenitors. NP68 epitope was properly processed and successfully presented by dendritic cells (DCs) by direct presentation and cross-presentation to F5 CD8 T cells. The models presented here are valuable tools to investigate the priming of F5 CD8 T cells by different subsets of DCs.


Asunto(s)
Epítopos de Linfocito T/genética , Proteínas Fluorescentes Verdes/genética , Transgenes , Proteínas Virales/genética , Animales , Células Dendríticas/metabolismo , Ingeniería Genética/métodos , Vectores Genéticos , Antígenos de Histocompatibilidad Clase I/metabolismo , Lentivirus/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas , Linfocitos T/metabolismo
14.
PLoS One ; 7(11): e49065, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23145071

RESUMEN

Regulation of the extracellular matrix (ECM) plays an important functional role either in physiological or pathological conditions. The plasminogen activation (PA) system, comprising the uPA and tPA proteases and their inhibitor PAI-1, is one of the main suppliers of extracellular proteolytic activity contributing to tissue remodeling. Although its function in development is well documented, its precise role in mouse embryonic stem cell (ESC) differentiation in vitro is unknown. We found that the PA system components are expressed at very low levels in undifferentiated ESCs and that upon differentiation uPA activity is detected mainly transiently, whereas tPA activity and PAI-1 protein are maximum in well differentiated cells. Adipocyte formation by ESCs is inhibited by amiloride treatment, a specific uPA inhibitor. Likewise, ESCs expressing ectopic PAI-1 under the control of an inducible expression system display reduced adipogenic capacities after induction of the gene. Furthermore, the adipogenic differentiation capacities of PAI-1(-/-) induced pluripotent stem cells (iPSCs) are augmented as compared to wt iPSCs. Our results demonstrate that the control of ESC adipogenesis by the PA system correspond to different successive steps from undifferentiated to well differentiated ESCs. Similarly, skeletal myogenesis is decreased by uPA inhibition or PAI-1 overexpression during the terminal step of differentiation. However, interfering with uPA during days 0 to 3 of the differentiation process augments ESC myotube formation. Neither neurogenesis, cardiomyogenesis, endothelial cell nor smooth muscle formation are affected by amiloride or PAI-1 induction. Our results show that the PA system is capable to specifically modulate adipogenesis and skeletal myogenesis of ESCs by successive different molecular mechanisms.


Asunto(s)
Adipogénesis/fisiología , Células Madre Embrionarias/fisiología , Desarrollo de Músculos/fisiología , Activadores Plasminogénicos/genética , Activadores Plasminogénicos/metabolismo , Plasminógeno/genética , Plasminógeno/metabolismo , Adipocitos/metabolismo , Animales , Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Serpina E2/genética , Serpina E2/metabolismo
15.
Hum Gene Ther ; 23(7): 754-68, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22463386

RESUMEN

Although clinical benefits have been reported in several human hematopoietic gene therapy trials, a remaining important goal is the transition to nonmyeloablative pretransplantation conditioning to decrease toxicity. Previous attempts at reduced intensity conditioning in nonhuman primates have resulted in only temporary vector marking of autologous blood cells or their persistence at low levels, well below the thresholds for clinical efficacy. In addition, we reasoned that lentiviral vector particles displaying cytokines at their surface have the potential to preserve stem cell fitness better than current ex vivo transduction protocols, which involve exposure to cytokine overstimulation. Here we show that the classically nonmyeloablative agent fludarabine (30 mg/m(2)/day for 3 days) together with low-level total body irradiation (2 Gy) and the use of a stem cell factor-displaying simian immunodeficiency virus-based vector, resulted in sustained, single-copy vector marking of autologous blood cells in two macaques over 3 years posttransplantation at levels averaging 1% of all lineages. This percentage is within the range of anticipated efficacy levels for hemophilia and related diseases and forms a basis for further improvement.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Virus de la Inmunodeficiencia de los Simios/genética , Factor de Células Madre/genética , Acondicionamiento Pretrasplante , Animales , Antígenos CD34/metabolismo , Quimerismo , Terapia Genética , Vectores Genéticos , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Hemofilia A/terapia , Humanos , Recuento de Linfocitos , Macaca fascicularis , Masculino , Agonistas Mieloablativos/administración & dosificación , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Transducción Genética , Trasplante Autólogo , Vidarabina/administración & dosificación , Vidarabina/análogos & derivados , Irradiación Corporal Total
16.
Blood ; 119(5): 1139-50, 2012 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-22117040

RESUMEN

In vivo lentiviral vector (LV)-mediated gene delivery would represent a great step forward in the field of gene therapy. Therefore, we have engineered a novel LV displaying SCF and a mutant cat endogenous retroviral glycoprotein, RDTR. These RDTR/SCF-LVs outperformed RDTR-LVs for transduction of human CD34(+) cells (hCD34(+)). For in vivo gene therapy, these novel RDTR/SCF-displaying LVs can distinguish between the target hCD34(+) cells of interest and nontarget cells. Indeed, they selectively targeted transduction to 30%-40% of the hCD34(+) cells in cord blood mononuclear cells and in the unfractionated BM of healthy and Fanconi anemia donors, resulting in the correction of CD34(+) cells in the patients. Moreover, RDTR/SCF-LVs targeted transduction to CD34(+) cells with 95-fold selectivity compared with T cells in total cord blood. Remarkably, in vivo injection of the RDTR/SCF-LVs into the BM cavity of humanized mice resulted in the highly selective transduction of candidate hCD34(+)Lin(-) HSCs. In conclusion, this new LV will facilitate HSC-based gene therapy by directly targeting these primitive cells in BM aspirates or total cord blood. Most importantly, in the future, RDTR/SCF-LVs might completely obviate ex vivo handling and simplify gene therapy for many hematopoietic defects because of their applicability to direct in vivo inoculation.


Asunto(s)
Médula Ósea/metabolismo , Terapia Genética/métodos , Vectores Genéticos/fisiología , Células Madre Hematopoyéticas/metabolismo , Hemoglobinuria Paroxística/terapia , Lentivirus/genética , Anemia Aplásica , Animales , Animales Recién Nacidos , Médula Ósea/patología , Enfermedades de la Médula Ósea , Trastornos de Fallo de la Médula Ósea , Células Cultivadas , Proteínas de Unión al ADN/genética , Marcación de Gen/métodos , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Hemoglobinuria Paroxística/genética , Hemoglobinuria Paroxística/patología , Humanos , Cadenas gamma de Inmunoglobulina/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Transducción Genética
17.
J Virol ; 85(12): 5975-85, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21450813

RESUMEN

Gene transfer into quiescent T and B cells is of importance for gene therapy and immunotherapy approaches to correct hematopoietic disorders. Previously, we generated lentiviral vectors (LVs) pseudotyped with the Edmonston measles virus (MV) hemagglutinin and fusion glycoproteins (Hgps and Fgps) (H/F-LVs), which, for the first time, allowed efficient transduction of quiescent human B and T cells. These target cells express both MV entry receptors used by the vaccinal Edmonston strain, CD46 and signaling lymphocyte activation molecule (SLAM). Interestingly, LVs pseudotyped with an MV Hgp, blind for the CD46 binding site, were completely inefficient for resting-lymphocyte transduction. Similarly, SLAM-blind H mutants that recognize only CD46 as the entry receptor did not allow stable LV transduction of resting T cells. The CD46-tropic LVs accomplished vector-cell binding, fusion, entry, and reverse transcription at levels similar to those achieved by the H/F-LVs, but efficient proviral integration did not occur. Our results indicate that both CD46 and SLAM binding sites need to be present in cis in the Hgp to allow successful stable transduction of quiescent lymphocytes. Moreover, the entry mechanism utilized appears to be crucial: efficient transduction was observed only when CD46 and SLAM were correctly engaged and an entry mechanism that strongly resembles macropinocytosis was triggered. Taken together, our results suggest that although vector entry can occur through the CD46 receptor, SLAM binding and subsequent signaling are also required for efficient LV transduction of quiescent lymphocytes to occur.


Asunto(s)
Antígenos CD/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Glicoproteínas/genética , Lentivirus/genética , Activación de Linfocitos , Virus del Sarampión/genética , Proteína Cofactora de Membrana/metabolismo , Receptores de Superficie Celular/metabolismo , Adulto , Antígenos CD/genética , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/virología , Humanos , Lentivirus/metabolismo , Virus del Sarampión/química , Proteína Cofactora de Membrana/genética , Pinocitosis , Receptores de Superficie Celular/genética , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología , Transducción Genética , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
19.
Stem Cells Dev ; 20(7): 1233-46, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20954847

RESUMEN

Embryonic stem (ES) cells differentiate in vitro into all cell lineages. We previously found that the p38 mitogen activated kinase (p38MAPK) pathway controls the commitment of ES cells toward either cardiomyogenesis (p38 on) or neurogenesis (p38 off ). In this study, we show that p38α knock-out ES cells do not differentiate into cardiac, endothelial, smooth muscle, and skeletal muscle lineages. Reexpression of p38MAPK in these cells partially rescues their mesodermal differentiation defects and corrects the high level of spontaneous neurogenesis of knock-out cells. Wild-type ES cells were treated with a p38MAPK-specific inhibitor during the differentiation process. These experiments allowed us to identify 2 early independent successive p38MAPK functions in the formation of mesodermal lineages. Further, the first one correlates with the regulation of the expression of Brachyury, an essential mesodermal-specific transcription factor, by p38MAPK. In conclusion, by genetic and biochemical approaches, we demonstrate that p38MAPK activity is essential for the commitment of ES cell into cardiac, endothelial, smooth muscle, and skeletal muscle mesodermal lineages.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Mesodermo/citología , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Animales , Western Blotting , Células Cultivadas , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Proteínas Fetales/genética , Proteínas Fetales/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Imidazoles/farmacología , Mesodermo/metabolismo , Ratones , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Desarrollo de Músculos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
20.
Blood ; 114(15): 3173-80, 2009 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-19667401

RESUMEN

Up to now, no lentiviral vector (LV) tool existed to govern efficient and stable gene delivery into quiescent B lymphocytes, which hampers its application in gene therapy and immunotherapy areas. Here, we report that LVs incorporating measles virus (MV) glycoproteins, H and F, on their surface allowed transduction of 50% of quiescent B cells, which are not permissive to VSVG-LV transduction. This high transduction level correlated with B-cell SLAM expression and was not at cost of cell-cycle entry or B-cell activation. Moreover, the naive and memory phenotypes of transduced resting B cells were maintained. Importantly, H/F-LVs represent the first tool permitting stable transduction of leukemic cancer cells, B-cell chronic lymphocytic leukemia cells, blocked in G(0)/G(1) early phase of the cell cycle. Thus, H/F-LV transduction overcomes the limitations of current LVs by making B cell-based gene therapy and immunotherapy applications feasible. These new LVs will facilitate antibody production and the study of gene functions in these healthy and cancer immune cells.


Asunto(s)
Linfocitos B/metabolismo , Vectores Genéticos , Glicoproteínas/metabolismo , VIH-1 , Virus del Sarampión , Transducción Genética/métodos , Proteínas Virales de Fusión/metabolismo , Antígenos CD/biosíntesis , Linfocitos B/patología , Línea Celular , Femenino , Fase G1 , Regulación de la Expresión Génica , Terapia Genética/métodos , Glicoproteínas/genética , Humanos , Leucemia Linfocítica Crónica de Células B , Activación de Linfocitos , Masculino , Receptores de Superficie Celular/biosíntesis , Fase de Descanso del Ciclo Celular , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Proteínas Virales de Fusión/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...