Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Blood ; 121(11): 2144-53, 2013 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-23305740

RESUMEN

Donor hematopoietic stem cells (HSCs) can correct T-cell deficiencies in patients with severe combined immunodeficiency by replacing resident thymus cells. However, as those progenitors that naturally migrate to the thymus are not capable of supporting long-term thymopoiesis, a successful transplant is thought to require the ongoing migration of donor progenitors. We previously showed that the forced intrathymic administration of histocompatible HSCs can sustain long-term thymopoiesis in ZAP-70-immunodeficient mice. However, it is not known whether T-cell reconstitution across histocompatibility barriers is modulated by intrathymic vs intravenous administration of HSCs. In the absence of conditioning, long-term thymopoiesis by semiallogeneic progenitors was detected in mice transplanted via the intrathymic, but not the intravenous, route. In intrathymic-transplanted mice, ongoing thymopoiesis was associated with a 10-fold higher level of early thymic progenitors (ETPs). The enhanced reconstitution capacity of these intrathymic-derived ETPs was corroborated by their significantly augmented myeloid lineage potential compared with endogenous ETPs. Notably, though, myeloablative conditioning resulted in a reduced expansion of intrathymic-administered donor ETPs. Thus, in the absence of conditioning, the forced thymic entry of HSCs results in a sustained T-cell development across histocompatibility barriers, highlighting the capacity of the thymus to support cells with long-term renewal potential.


Asunto(s)
Diferenciación Celular/inmunología , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas/métodos , Histocompatibilidad/fisiología , Células Progenitoras Linfoides/fisiología , Linfocitos T/fisiología , Timo , Animales , Células Cultivadas , Supervivencia de Injerto/inmunología , Supervivencia de Injerto/fisiología , Hematopoyesis/inmunología , Hematopoyesis/fisiología , Histocompatibilidad/inmunología , Prueba de Histocompatibilidad , Infusiones Intravenosas , Células Progenitoras Linfoides/citología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Timo/citología , Acondicionamiento Pretrasplante/métodos , Proteína Tirosina Quinasa ZAP-70/deficiencia , Proteína Tirosina Quinasa ZAP-70/genética , Proteína Tirosina Quinasa ZAP-70/inmunología
2.
PLoS Pathog ; 6(6): e1000948, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20548955

RESUMEN

Antiviral monoclonal antibodies (mAbs) represent promising therapeutics. However, most mAbs-based immunotherapies conducted so far have only considered the blunting of viral propagation and not other possible therapeutic effects independent of virus neutralization, namely the modulation of the endogenous immune response. As induction of long-term antiviral immunity still remains a paramount challenge for treating chronic infections, we have asked here whether neutralizing mAbs can, in addition to blunting viral propagation, exert immunomodulatory effects with protective outcomes. Supporting this idea, we report here that mice infected with the FrCas(E) murine retrovirus on day 8 after birth die of leukemia within 4-5 months and mount a non-protective immune response, whereas those rapidly subjected to short immunotherapy with a neutralizing mAb survive healthy and mount a long-lasting protective antiviral immunity with strong humoral and cellular immune responses. Interestingly, the administered mAb mediates lysis of infected cells through an antibody-dependent cell cytotoxicity (ADCC) mechanism. In addition, it forms immune complexes (ICs) with infected cells that enhance antiviral CTL responses through Fc gammaR-mediated binding to dendritic cells (DCs). Importantly, the endogenous antiviral antibodies generated in mAb-treated mice also display the same properties, allowing containment of viral propagation and enhancement of memory cellular responses after disappearance of the administered mAb. Thus, our data demonstrate that neutralizing antiviral mAbs can act as immunomodulatory agents capable of stimulating a protective immunity lasting long after the end of the treatment. They also show an important role of infected-cells/antibody complexes in the induction and the maintenance of protective immunity through enhancement of both primary and memory antiviral T-cell responses. They also indicate that targeting infected cells, and not just viruses, by antibodies can be crucial for elicitation of efficient, long-lasting antiviral T-cell responses. This must be considered when designing antiviral mAb-based immunotherapies.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antivirales/inmunología , Inmunización Pasiva , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/terapia , Retroviridae/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Complejo Antígeno-Anticuerpo , Proliferación Celular , Citometría de Flujo , Ratones
3.
Blood ; 115(10): 1913-20, 2010 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-20040762

RESUMEN

The sustained differentiation of T cells in the thymus cannot be maintained by resident intrathymic (IT) precursors and requires that progenitors be replenished from the bone marrow (BM). In patients with severe combined immunodeficiency (SCID) treated by hematopoietic stem cell transplantation, late T-cell differentiation defects are thought to be due to an insufficient entry of donor BM progenitors into the thymus. Indeed, we find that the intravenous injection of BM progenitors into nonconditioned zeta-chain-associated protein kinase 70 (ZAP-70)-deficient mice with SCID supports short- but not long-term thymopoiesis. Remarkably, we now show that the IT administration of these progenitors produces a significant level of donor-derived thymopoiesis for more than 6 months after transplantation. In contrast to physiologic thymopoiesis, long-term donor thymopoiesis was not due to the continued recruitment of progenitors from the BM. Rather, IT transplantation resulted in the unique generation of a large population of early c-Kit(high) donor precursors within the thymus. These ZAP-70-deficient mice that received an IT transplant had a significantly increased prothymocyte niche compared with their untreated counterparts; this phenotype was associated with the generation of a medulla. Thus, IT administration of BM progenitors results in the filling of an expanded precursor niche and may represent a strategy for enhancing T-cell differentiation in patients with SCID.


Asunto(s)
Trasplante de Médula Ósea/métodos , Trasplante de Médula Ósea/fisiología , Células Progenitoras Linfoides/trasplante , Linfopoyesis/fisiología , Timo/citología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Linaje de la Célula/genética , Células Cultivadas , Infusiones Intravenosas , Recuento de Linfocitos , Células Progenitoras Linfoides/fisiología , Linfopoyesis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nicho de Células Madre/citología , Linfocitos T/citología , Linfocitos T/fisiología , Timo/fisiología , Factores de Tiempo , Proteína Tirosina Quinasa ZAP-70/genética
4.
J Immunol ; 182(6): 3398-405, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19265117

RESUMEN

Tumor cell-based vaccines are currently used in clinical trails, but they are in general poorly immunogenic because they are composed of cell extracts or apoptotic cells. Live tumor cells should be much better Ags provided that they are properly processed by the host immune system. We show herein that stable expression of a small hairpin RNA for ERK5 (shERK5) decreases ERK5 levels in human and mouse leukemic cells and leads to their elimination by NK cells in vivo. The shERK5 cells show down-regulation of MHC class I expression at the plasma membrane. Accordingly, ectopic activation of the ERK5 pathway induces MHC class I gene expression. Coinjection of shERK5-expressing cells into the peritoneum diminishes survival of engrafted wild-type tumor cells. Moreover, s.c. injection of shERK5-expressing cells strongly diminishes tumor development by wild-type cells. Our results show that shERK5 expression in leukemia cells effectively attenuates their tumor activity and allows their use as a tumor cell-based vaccine.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Técnicas de Silenciamiento del Gen , Antígenos de Histocompatibilidad Clase I/metabolismo , Células Asesinas Naturales/inmunología , Leucemia L1210/prevención & control , Activación de Linfocitos/inmunología , Proteína Quinasa 7 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 7 Activada por Mitógenos/genética , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Células Cultivadas , Citotoxicidad Inmunológica/genética , Antígenos de Histocompatibilidad Clase I/biosíntesis , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Células Jurkat , Células Asesinas Naturales/metabolismo , Leucemia L1210/enzimología , Leucemia L1210/genética , Leucemia L1210/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteína Quinasa 7 Activada por Mitógenos/biosíntesis , ARN Interferente Pequeño/fisiología , Transducción de Señal/genética , Transducción de Señal/inmunología
5.
Blood ; 112(12): 4729-38, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18796630

RESUMEN

Glucose is a major source of energy for living organisms, and its transport in vertebrates is a universally conserved property. Of all cell lineages, human erythrocytes express the highest level of the Glut1 glucose transporter with more than 200,000 molecules per cell. However, we recently reported that erythrocyte Glut1 expression is a specific trait of vitamin C-deficient mammalian species, comprising only higher primates, guinea pigs, and fruit bats. Here, we show that in all other tested mammalian species, Glut1 was transiently expressed in erythrocytes during the neonatal period. Glut1 was up-regulated during the erythroblast stage of erythroid differentiation and was present on the vast majority of murine red blood cells (RBCs) at birth. Notably though, Glut1 was not induced in adult mice undergoing anemia-induced erythropoiesis, and under these conditions, the up-regulation of a distinct transporter, Glut4, was responsible for an increased glucose transport. Sp3 and Sp1 transcriptions factors have been proposed to regulate Glut1 transcription, and we find that the concomitant repression of Glut1 and induction of Glut4 was associated with a significantly augmented Sp3/Sp1 ratio. Glucose transporter expression patterns in mice and human erythrocytes are therefore distinct. In mice, there is a postnatal switch from Glut1 to Glut4, with Glut4 further up-regulated under anemic conditions.


Asunto(s)
Eritropoyesis/genética , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 4/genética , Anemia/genética , Anemia/patología , Animales , Animales Recién Nacidos , Bovinos , Perros , Eritrocitos/metabolismo , Eritrocitos/patología , Exosomas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas/fisiología , Ratas
6.
Mol Ther ; 17(3): 472-9, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19088703

RESUMEN

The thymus is the primary site of T-cell development and plays a key role in the induction of self-tolerance. We previously showed that the intrathymic (i.t.) injection of a transgene-expressing lentiviral vector (LV) in mice can result in the correction of a T cell-specific genetic defect. Nevertheless, the efficiency of thymocyte transduction did not exceed 0.1-0.3% and we were unable to detect any thymus transduction in macaques. As such, we initiated studies to assess the capacity of recombinant adeno-associated virus (rAAV) vectors to transduce murine and primate thymic cells. In vivo administration of AAV serotype 2-derived single-stranded AAV (ssAAV) and self-complementary AAV (scAAV) vectors pseudotyped with capsid proteins of serotypes 1, 2, 4, 5, and 8 demonstrated that murine thymus transduction was significantly enhanced by scAAV2/8. Transgene expression was detected in 5% of thymocytes and, notably, transduced cells represented 1% of peripheral T lymphocytes. Moreover, i.t. administration of scAAV2/8 particles in macaques, by endoscopic-mediated guidance, resulted in significant gene transfer. Thus, in healthy animals, where thymic gene transfer does not provide a selective advantage, scAAV2/8 is a unique tool promoting the in situ transduction of thymocytes with the subsequent export of gene-modified lymphocytes to the periphery.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Macaca fascicularis/metabolismo , Timo/metabolismo , Transgenes/genética , Animales , Diferenciación Celular , Línea Celular , Movimiento Celular , Dependovirus/clasificación , Vectores Genéticos/farmacología , Genoma Viral/genética , Humanos , Cinética , Ratones , Fenotipo , Timo/citología
7.
J Virol ; 82(3): 1339-49, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18032505

RESUMEN

Neutralizing monoclonal antibodies (MAbs) are increasingly being considered for blunting human viral infections. However, whether they can also exert indirect effects on endogenous antiviral immune responses has been essentially overlooked. We have recently shown that a short (several-day) period of immunotherapy with the neutralizing 667 MAb of mouse neonates shortly after infection with the lethal FrCas(E) retrovirus not only has an immediate effect on the viral load but also permits an endogenous antiviral immunity to emerge. Even though passive immunotherapy was administered during the particular period of immunocompetence acquisition, the endogenous response eventually arising was protective and persisted long (>1 year) after the MAb has disappeared. As very high levels of anti-FrCas(E) antibodies, predominantly of the immunoglobulin G2a (IgG2a) isotype and showing strong neutralization activity, were found in the sera of MAb-treated mice, it was necessary to address whether this humoral immunity was sufficient on its own to confer full protection against FrCas(E) or whether a cytotoxic T-lymphocyte (CTL) response was also necessary. Using a variety of in vivo assays in young and adult animals previously infected by FrCas(E) and treated by 667, we show here that transient 667 immunotherapy is associated with the emergence of a CTL response against virus-infected cells. This cytotoxic activity is indispensable for long-term antiviral protective immunity, as high neutralizing antibody titers, even enhanced in in vivo CD8(+) cell depletion experiments, cannot prevent the FrCas(E)-induced death of infected/treated mice. Our work may have important therapeutic consequences, as it indicates that a short period of MAb-based immunotherapy conducted at a stage where the immune system is still developing can be associated with the mounting of a functional Th1-type immune response characterized by both CTL and IgG2a-type humoral contributions, the cooperation of which is known to be essential for the containment of chronic infections by a variety of viruses.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Inmunización Pasiva , Infecciones por Retroviridae/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Animales Recién Nacidos , Anticuerpos Antivirales/inmunología , Depleción Linfocítica , Ratones , Pruebas de Neutralización
8.
Methods Mol Biol ; 506: 171-90, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19110627

RESUMEN

The thymus provides a specialized environment allowing the differentiation of T lymphocytes from bone marrow-derived progenitor cells. We and others have demonstrated that gene transfer into distinct thymocyte populations can be obtained, both in vivo and ex vivo, using lentiviral vectors. Here, we describe techniques for intrathymic lentiviral transduction in mice, using a surgical approach wherein the thoracic cavity is exposed as well as a significantly less invasive strategy wherein virions are directly injected through the skin. Moreover, thymocyte differentiation from murine and human progenitors is now feasible in vitro, under conditions wherein the Notch and IL-7 signaling pathways are activated. We describe methods allowing transduction of murine and human progenitors and their subsequent differentiation into more mature thymocytes. Conditions for lentiviral gene transfer into more differentiated human thymocyte subsets are also presented. Optimization of technologies for HIV-based gene transfer into murine and human thymocyte progenitors will advance strategies aimed at modulating T-cell differentiation and function in-vivo; approaches potentially targeting patients with genetic and acquired immunodeficiencies as well as immune-sensitive tumors. Furthermore, this technology will foster the progression of basic research aimed at elucidating molecular aspects of T-cell differentiation in mice and humans.


Asunto(s)
Técnicas de Transferencia de Gen , Timo/metabolismo , Animales , Antígenos CD34/inmunología , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Ratones , Timo/citología , Timo/inmunología , Transducción Genética
9.
Mol Ther ; 16(8): 1372-81, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18578013

RESUMEN

Cystinosis belongs to a growing class of lysosomal storage disorders (LSDs) caused by defective transmembrane proteins. The causative CTNS gene encodes the lysosomal cystine transporter, cystinosin. Currently the aminothiol cysteamine is the only drug available for reducing cystine storage but this treatment has non-negligible side effects and administration constraints. In this study, for the first time, we report viral vector-mediated CTNS gene transfer and evaluate the feasibility of this strategy as a complementary treatment. Initially, we transduced human CTNS(-/-) fibroblast cell lines and primary murine Ctns(-/-) hepatocyte cultures in vitro and demonstrated that gene transfer can reduce cystine storage. Because of age-related increase in cystine levels, we transduced hepatocytes from young (/=5 months of age) mice. Our in vitro data suggested that the efficiency of correction was age-dependent. We tested these observations in vivo: short-term (1 week) and long-term (4 weeks) CTNS-transduction significantly reduced hepatic cystine levels in young, but not older, Ctns(-/-) mice. Our data provide the proof-of-concept that gene transfer is feasible for correcting defective lysosomal transport, but suggest that, in the case of cystinosis, it could be preventive but not curative in some tissues.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros/fisiología , Cistina/metabolismo , Cistinosis/terapia , Terapia Genética/métodos , Adenovirus Caninos/genética , Factores de Edad , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Animales , Western Blotting , Línea Celular , Células Cultivadas , Cistinosis/genética , Cistinosis/metabolismo , Perros , Estudios de Factibilidad , Fibroblastos/citología , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Hepatocitos/citología , Hepatocitos/metabolismo , Hepatocitos/ultraestructura , Humanos , Macrófagos del Hígado/citología , Macrófagos del Hígado/metabolismo , Lisosomas/metabolismo , Masculino , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión
10.
J Leukoc Biol ; 84(2): 380-8, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18483203

RESUMEN

The TNF family member, a proliferation-inducing ligand (APRIL), has been suggested to act as a costimulatory molecule in T cell responses. However, studies addressing this role in vivo are largely lacking. Here, we evaluated the effects of APRIL on physiological T cell responses in vivo. Although receptors for APRIL are expressed on a subset of T cells, neither TCR transgenic (Tg) T cell responses nor endogenous TCR responses were affected by Tg APRIL expression in vivo. Moreover, APRIL did not significantly enhance the induction of T cell lymphomas upon Moloney murine leukemia virus (MLV) infection. This clearly contrasts current belief and indicates that APRIL does not serve a major role in T cell immunity or lymphomagenesis. However, we did observe a strong increase in erythroleukemia formation after MLV inoculation of APRIL Tg mice. Strikingly, this erythroleukemia-facilitating property of APRIL was confirmed using the erythroleukemogenic Friend-MLV. Erythroleukemia in APRIL Tg mice was characterized by low hematocrits and grossly enlarged spleens with an increased percentage of erythroid precursors. Altogether, these results unveil new proerythroleukemogenic properties of APRIL.


Asunto(s)
Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/virología , Linfoma de Células T/fisiopatología , Linfocitos T/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Autoinmunidad , Citometría de Flujo , Hematócrito , Heterocigoto , Homocigoto , Linfoma de Células T/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Virus de la Leucemia Murina de Moloney/inmunología , Virus de la Leucemia Murina de Moloney/patogenicidad , Ovalbúmina/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Células Madre/fisiología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
11.
Mol Immunol ; 45(12): 3463-9, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18462800

RESUMEN

The cancer immunosurveillance hypothesis has found strong experimental support in recent years. It is believed that cytotoxic lymphocytes are important effectors in this process. PKCtheta plays an essential role in proliferation, activation and survival of these cells, but also proliferation and survival of leukemic T cells. In light of this, we tested the role of PKCtheta in T cell leukemia progression by inducing this disease in wild-type (wt) and PKCtheta-deficient mice with moloney-murine leukemia virus (M-MuLV). Leukemic PKCtheta(-/-) and wild-type (wt) mice showed the same profile of leukemic cell types, similar spleen and thymus sizes and comparable hematocrits. In contrast, disease incidence was higher and disease onset more rapid in PKCtheta(-/-) mice. Transfer of leukemic T cells from wt donors into PKCtheta-deficient and wt recipients induced leukemia in 100% and 40% of the mice, respectively. Interestingly, leukemic cells from PKCtheta(-/-) donors induced the disease in only 50% of the PKCtheta-deficient and 10% of the wt recipients. Intravenous injection of low numbers of EL4 cells induced tumors earlier in PKCtheta(-/-) mice. Taken together, our results show that PKCtheta is essential for the immune response to leukemia in mice and raise questions about the chronic treatment of humans with PKCtheta inhibitors.


Asunto(s)
Isoenzimas/deficiencia , Leucemia/enzimología , Leucemia/inmunología , Proteína Quinasa C/deficiencia , Animales , Animales Recién Nacidos , Isoenzimas/metabolismo , Leucemia/patología , Leucemia/virología , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Fenotipo , Proteína Quinasa C/metabolismo , Proteína Quinasa C-theta , Análisis de Supervivencia
12.
J Clin Invest ; 115(8): 2287-95, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16075064

RESUMEN

SCID patients have been successfully treated by administration of ex vivo gene-corrected stem cells. However, despite its proven efficacy, such treatment carries specific risks and difficulties. We hypothesized that some of these drawbacks may be overcome by in situ gene correction of T lymphoid progenitors in the thymus. Indeed, in vivo intrathymic transfer of a gene that provides a selective advantage for transduced prothymocytes should result in the generation of functional T lymphocyte progeny, allowing long-term immune reconstitution. We assessed the feasibility of this approach in a murine model of ZAP-70-deficient SCID. A T cell-specific ZAP-70-expressing lentiviral vector was injected into thymi of adult ZAP-70-/- mice without prior conditioning. This resulted in the long-term differentiation of mature TCR-alphabeta+ thymocytes, indicating that the vector had integrated into progenitor cells. Moreover, peripheral ZAP-70-expressing T cells demonstrated a partially diversified receptor repertoire and were responsive to alloantigens in vitro and in vivo. Improved treatment efficacy was achieved in infant ZAP-70-/- mice, in which the thymus is proportionately larger and a higher percentage of prothymocytes are in cycle. Thus, intrathymic injection of a lentiviral vector could represent a simplified and potentially safer alternative to ex vivo gene-modified hematopoietic stem cell transplantation for gene therapy of T cell immunodeficiencies.


Asunto(s)
Terapia Genética , Trasplante de Células Madre Hematopoyéticas , Lentivirus , Proteínas Tirosina Quinasas/genética , Inmunodeficiencia Combinada Grave/terapia , Timo , Transducción Genética , Animales , Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/inmunología , Humanos , Linfopoyesis/genética , Linfopoyesis/inmunología , Ratones , Ratones Noqueados , Proteínas Tirosina Quinasas/inmunología , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Linfocitos T/inmunología , Timo/inmunología , Transducción Genética/métodos , Proteína Tirosina Quinasa ZAP-70
13.
Neuropsychopharmacology ; 33(7): 1584-602, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17805310

RESUMEN

Chromatin remodeling by posttranslational modification of histones plays an important role in brain plasticity, including memory, response to stress and depression. The importance of H3/4 histones acetylation by CREB-binding protein (CBP) or related histone acetyltransferase, including p300, was specifically demonstrated using knockout (KO) mouse models. The physiological role of a related protein that also acts as a transcriptional coactivator with intrinsic histone acetylase activity, the p300/CBP-associated factor (PCAF), is poorly documented. We analyzed the behavioral phenotype of homozygous male and female PCAF KO mice and report a marked impact of PCAF deletion on memory processes and stress response. PCAF KO animals showed short-term memory deficits at 2 months of age, measured using spontaneous alternation, object recognition, or acquisition of a daily changing platform position in the water maze. Acquisition of a fixed platform location was delayed, but preserved, and no passive avoidance deficit was noted. No gender-related difference was observed. These deficits were associated with hippocampal alterations in pyramidal cell layer organization, basal levels of Fos immunoreactivity, and MAP kinase activation. PCAF KO mice also showed an exaggerated response to acute stress, forced swimming, and conditioned fear, associated with increased plasma corticosterone levels. Moreover, learning and memory impairments worsened at 6 and 12 months of age, when animals failed to acquire the fixed platform location in the water maze and showed passive avoidance deficits. These observations demonstrate that PCAF histone acetylase is involved lifelong in the chromatin remodeling necessary for memory formation and response to stress.


Asunto(s)
Memoria/fisiología , Estrés Psicológico/genética , Estrés Psicológico/fisiopatología , Factores de Transcripción p300-CBP/deficiencia , Factores de Edad , Análisis de Varianza , Animales , Reacción de Prevención/fisiología , Conducta Animal , Condicionamiento Psicológico/fisiología , Corticosterona/sangre , Modelos Animales de Enfermedad , Emociones/fisiología , Conducta Exploratoria/fisiología , Miedo , Femenino , Hipocampo/patología , Masculino , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/genética , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Noqueados , Reconocimiento Visual de Modelos/fisiología , Factores Sexuales , Estrés Psicológico/patología
14.
Hum Gene Ther ; 13(10): 1179-88, 2002 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12133271

RESUMEN

The adeno-associated viruses (AAV) offer new perspectives for cytokine gene transfer in rheumatoid arthritis (RA) because they are nonpathogenic and allow long-term transgene expression in vivo. Moreover, the use of a tetracycline-inducible promoter allows regulation of therapeutic gene expression. This study assessed the potential long-term gene regulation of a recombinant AAV vector expressing viral interleukin-10 (vIL-10) in human rheumatoid synovium and the therapeutic efficiency in a mouse model of RA. We constructed a recombinant AAV vector in which the transcription of vIL-10 cDNA is controlled by the TetON system. Transduction of human primary RA synovial cells with AAV-tetON-vIL10 conferred in vitro controlled vIL-10 expression. After intramuscular injection, both incidence and severity of collagen-induced arthritis were significantly reduced at macroscopic, radiological, and histological levels in the group of DBA1 mice treated with AAV-TetON-vIL10 vector plus doxycycline after immunization and boosting compared to control groups. When coinjecting two separate AAV vectors, one encoding the inducible vIL-10 and the other the transcriptional activator, a 10 times excess of the transactivator vector dose allowed efficient control of vIL-10 secretion by doxycycline administration or withdrawal, over an 8-week period. Our results supported, for the first time, the utility of AAV-tetON-vIL10 as a therapeutic tool for gene therapy in RA.


Asunto(s)
Artritis Experimental/terapia , Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética , Interleucina-10/genética , Tetraciclina/farmacología , Animales , Expresión Génica , Regulación de la Expresión Génica , Vectores Genéticos , Genoma Viral , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos DBA , Especificidad de Órganos
15.
Hum Gene Ther ; 13(12): 1483-93, 2002 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-12215269

RESUMEN

The therapeutic potential of monoclonal antibodies (MAbs) for treating a variety of severe or life-threatening diseases is high. Although intravenous infusion appears to be the simplest and most obvious mode of administration, it is not applicable in many long-term treatments. It might, however, be advantageously replaced by gene/cell therapies, rendering treatments cost-effective and eliminating the short- and long-term side effects associated with injection of massive doses of antibodies. Grafting of ex vivo genetically modified cells of various types has already been used for in vivo production and systemic delivery of MAbs in mice. However, although sustained for long periods of time, serum levels of ectopic MAbs were low. We show here that in vivo administration to mice of a first-generation adenoviral vector expressing a model MAb also permits achievement of the same goal, but with 100 to 200 times better efficiency that in any other case of gene transfer described thus far. We also investigated for possible anti-idiotypic response against the ectopic MAb. None was detected in the animals expressing the lowest levels of ectopic MAb production; a response was detected among the highest producers. In the latter case, however, the response was low and could not exert any significant neutralizing activity. In conclusion, our work indicates that high levels of circulating ectopic MAb can be obtained on direct in vivo gene transfer without inducing an anti-idiotypic response sufficiently robust to exert a neutralizing effect. This observation is encouraging in the perspective of clinical applications of this technology.


Asunto(s)
Adenoviridae , Anticuerpos Monoclonales/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Animales , Anticuerpos Monoclonales/biosíntesis , Vectores Genéticos/administración & dosificación , Hepatocitos/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/metabolismo
16.
J Invest Dermatol ; 118(2): 288-94, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11841546

RESUMEN

The therapeutic potential of monoclonal antibodies for treating a variety of severe or life-threatening diseases is high. Although intravenous infusion appears the simplest and most obvious mode of administration, it is not applicable to many long-term treatments. It might be advantageously replaced by gene/cell therapies, however, rendering treatments cost-effective and eliminating the short- and long-term side-effects associated with injection of massive doses of antibodies. We have tested whether skin can potentially be used as an organ for production and systemic delivery of ectopic antibodies. Normal human primary keratinocytes were shown to be capable of synthesis and secretion of a model monoclonal antibody directed against human thyroglobulin upon retroviral gene transduction in vitro. Neo- epidermis reconstructed in vitro, either in cell culture inserts or on dermal substrates, from such modified keratinocytes also produced the monoclonal antibody. Interestingly, the latter could cross the epidermis basal layer and be released in culture fluids. Finally, grafting of epidermis reconstituted in vitro on dermal substrates to SCID mice permitted sustained monoclonal antibody delivery into the bloodstream to be achieved. Our data thus show that genetically engineered keratinocytes can potentially be used for genetic antibody-based immunotherapies. They also indicate that proteins as big as 150 kDa, after release by engineered keratinocytes into skin intercellular spaces, can migrate to the general circulation, which is potentially important for a number of other gene-based therapies.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Piel/inmunología , Animales , Células Cultivadas , Células Epidérmicas , Epidermis/inmunología , Técnicas de Transferencia de Gen , Vectores Genéticos , Técnicas Histológicas , Humanos , Queratinocitos/inmunología , Queratinocitos/trasplante , Ratones , Ratones SCID/fisiología , Retroviridae/genética , Tiroglobulina/inmunología , Distribución Tisular
17.
Oncoimmunology ; 1(5): 600-608, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22934252

RESUMEN

Vav1 is expressed exclusively in hematopoietic cells and is required for T cell development and activation. Vav1-deficient mice show thymic hypocellularity due to a partial block during thymocyte development at the DN3 stage and between the double positive (DP) and single positive (SP) transition. Vav1 has been shown to play a significant role in several non-hematopoietic tumors but its role in leukemogenesis is unknown. To address this question, we investigated the role of Vav1 in retrovirus-induced T cell leukemogenesis. Infection of Vav1-deficient mice with the Moloney strain of murine leukemia virus (M-MuLV) significantly affected tumor phenotype without modulating tumor incidence or latency. M-MuLV-infected Vav1-deficient mice showed reduced splenomegaly, higher hematocrit levels and hypertrophic thymi. Notably, Vav1-deficient mice with M-MuLV leukemias presented with markedly lower TCRß/CD3 levels, indicating that transformation occurred at an earlier stage of T cell development than in WT mice. Thus, impaired T cell development modulates the outcome of retrovirus-induced T cell leukemias, demonstrating a link between T cell development and T cell leukemogenesis.

18.
J Exp Med ; 208(7): 1403-17, 2011 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-21708927

RESUMEN

The multifunctional E4F1 protein was originally discovered as a target of the E1A viral oncoprotein. Growing evidence indicates that E4F1 is involved in key signaling pathways commonly deregulated during cell transformation. In this study, we investigate the influence of E4F1 on tumorigenesis. Wild-type mice injected with fetal liver cells from mice lacking CDKN2A, the gene encoding Ink4a/Arf, developed histiocytic sarcomas (HSs), a tumor originating from the monocytic/macrophagic lineage. Cre-mediated deletion of E4F1 resulted in the death of HS cells and tumor regression in vivo and extended the lifespan of recipient animals. In murine and human HS cell lines, E4F1 inactivation resulted in mitochondrial defects and increased production of reactive oxygen species (ROS) that triggered massive cell death. Notably, these defects of E4F1 depletion were observed in HS cells but not healthy primary macrophages. Short hairpin RNA-mediated depletion of E4F1 induced mitochondrial defects and ROS-mediated death in several human myeloid leukemia cell lines. E4F1 protein is overexpressed in a large subset of human acute myeloid leukemia samples. Together, these data reveal a role for E4F1 in the survival of myeloid leukemic cells and support the notion that targeting E4F1 activities might have therapeutic interest.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Proteínas Represoras/deficiencia , Factores de Transcripción/deficiencia , Animales , Autofagia/fisiología , Secuencia de Bases , Muerte Celular/fisiología , Línea Celular Tumoral , Transformación Celular Neoplásica , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Sarcoma Histiocítico/genética , Sarcoma Histiocítico/metabolismo , Sarcoma Histiocítico/patología , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Ratones Transgénicos , Estrés Oxidativo , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas
19.
Neurosci Lett ; 475(3): 179-83, 2010 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-20371377

RESUMEN

P300/CBP associated factor (PCAF) acts as an acetyltransferase that acetylates specific lysine residues in histones, thereby remodelling chromatin structure. The possible involvement of PCAF in learning and memory processes or mood disorders was recently assessed by characterizing the behavioural phenotype of PCAF KO mice bred on a CD1 background and revealed short-term memory deficits that evolved with age towards long-term memory alteration and an exaggerated response to stress [10]. PCAF KO mice have been backcrossed on a C57BL/6j strain for 15 generations and we report here the first data regarding their behavioural phenotype. PCAF KO mice bred on a C57 background showed short-term memory deficits in terms of decreased spontaneous alternation and absence of acquisition of a daily changing platform position in the water-maze. Acquisition of a fixed platform location or passive avoidance response was preserved. PCAF KO mice showed no difference with WT C57BL/6j controls in their performances in the forced swimming and light/dark exploration box, suggesting no particular phenotype on anxiety and stress responses. We therefore evidenced marked phenotypic differences in PCAF KO mice depending on the genetic background strain confirming that PCAF histone acetyltransferase is involved lifelong in the chromatin remodelling necessary for memory formation but differentially involved in anxiety and response to stress.


Asunto(s)
Ansiedad/psicología , Histona Acetiltransferasas/genética , Memoria a Corto Plazo , Estrés Psicológico/psicología , Factores de Transcripción p300-CBP/genética , Animales , Ansiedad/genética , Reacción de Prevención , Femenino , Histona Acetiltransferasas/fisiología , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tiempo de Reacción , Conducta Espacial , Estrés Psicológico/genética , Factores de Transcripción p300-CBP/fisiología
20.
PLoS One ; 5(9): e12659, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20856822

RESUMEN

BACKGROUND: Lymphopenia results in the proliferation and differentiation of naïve T cells into memory-like cells in the apparent absence of antigenic stimulation. This response, at least in part due to a greater availability of cytokines, is thought to promote anti-self responses. Although potentially autoreactive memory-like CD8(+) T cells generated in a lymphopenic environment are subject to the mechanisms of peripheral tolerance, they can induce autoimmunity in the presence of antigen-specific memory-like CD4(+) T helper cells. METHODOLOGY/PRINCIPAL FINDINGS: Here, we studied the mechanisms underlying CD4 help under lymphopenic conditions in transgenic mice expressing a model antigen in the beta cells of the pancreas. Surprisingly, we found that the self-reactivity mediated by the cooperation of memory-like CD8(+) and CD4(+) T cells was not abrogated by CD40L blockade. In contrast, treatment with anti-IL-2 antibodies inhibited the onset of autoimmunity. IL-2 neutralization prevented the CD4-mediated differentiation of memory-like CD8(+) T cells into pathogenic effectors in response to self-antigen cross-presentation. Furthermore, in the absence of helper cells, induction of IL-2 signaling by an IL-2 immune complex was sufficient to promote memory-like CD8(+) T cell self-reactivity. CONCLUSIONS/SIGNIFICANCE: IL-2 mediates the cooperation of memory-like CD4(+) and CD8(+) T cells in the breakdown of cross-tolerance, resulting in effector cytotoxic T lymphocyte differentiation and the induction of autoimmune disease.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Tolerancia Inmunológica , Memoria Inmunológica , Interleucina-2/inmunología , Linfopenia/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Autoinmunidad , Linfocitos T CD8-positivos/citología , Diferenciación Celular , Reactividad Cruzada , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Linfocitos T Colaboradores-Inductores/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA