Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Cell Rep ; 43(3): 113879, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38416647

RESUMEN

Maintenance of CD4 T cells during chronic infections is vital for limiting pathogen burden and disease recrudescence. Although inhibitory receptor expression by CD4 T cells is commonly associated with immune suppression and exhaustion, such cell-intrinsic mechanisms that control activation are also associated with cell survival. Using a mouse model of visceral leishmaniasis (VL), we discovered a subset of lymphocyte activation gene 3 (LAG-3)-expressing CD4 T cells that co-express CXCR5. Although LAG3+CXCR5+ CD4 T cells are present in naive mice, they expand during VL. These cells express gene signatures associated with self-renewal capacity, suggesting progenitor-like properties. When transferred into Rag1-/- mice, these LAG3+CXCR5+ CD4 T cells differentiated into multiple effector types upon Leishmania donovani infection. The transcriptional repressor B cell lymphoma-6 was partially required for their maintenance. Altogether, we propose that the LAG3+CXCR5+ CD4 T cell subset could play a role in maintaining CD4 T cell responses during persistent infections.


Asunto(s)
Linfocitos T CD4-Positivos , Leishmaniasis Visceral , Humanos , Subgrupos de Linfocitos T , Factores de Transcripción , Receptores CXCR5
2.
Virology ; 567: 77-86, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35032866

RESUMEN

Type-I interferon (IFN-I) signals exert a critical role in disease progression during viral infections. However, the immunomodulatory mechanisms by which IFN-I dictates disease outcomes remain to be fully defined. Here we report that IFN-I signals mediate thymic atrophy in viral infections, with more severe and prolonged loss of thymic output and unique kinetics and subtypes of IFN-α/ß expression in chronic infection compared to acute infection. Loss of thymic output was linked to inhibition of early stages of thymopoiesis (DN1-DN2 transition, and DN3 proliferation) and pronounced apoptosis during the late DP stage. Notably, infection-associated thymic defects were largely abrogated upon ablation of IFNαßR and partially mitigated in the absence of CD8 T cells, thus implicating direct as well as indirect effects of IFN-I on thymocytes. These findings provide mechanistic underpinnings for immunotherapeutic strategies targeting IFN-1 signals to manipulate disease outcomes during chronic infections and cancers.


Asunto(s)
Atrofia/virología , Interferón-alfa/inmunología , Interferón beta/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Timocitos/virología , Timo/virología , Animales , Atrofia/genética , Atrofia/inmunología , Atrofia/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Enfermedad Crónica , Femenino , Regulación de la Expresión Génica , Humanos , Memoria Inmunológica , Interferón-alfa/genética , Interferón beta/genética , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ganglios Linfáticos/virología , Depleción Linfocítica , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal/inmunología , Análisis de la Célula Individual , Timocitos/inmunología , Timocitos/patología , Timo/inmunología , Timo/patología
3.
Immunohorizons ; 4(4): 217-230, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32332052

RESUMEN

CD8+ T cell-mediated immunity is critical for host defense against viruses and requires mitochondria-mediated type I IFN (IFN-I) signaling for optimal protection. Cyclophilin D (CypD) is a mitochondrial matrix protein that modulates the mitochondrial permeability transition pore, but its role in IFN-I signaling and CD8+ T cell responses to viral infection has not been previously explored. In this study, we demonstrate that CypD plays a critical extrinsic role in the survival of Ag-specific CD8+ T cell following acute viral infection with lymphocytic choriomeningitis virus in mice. CypD deficiency resulted in reduced IFN-I and increased CD8+ T cell death, resulting in a reduced antiviral CD8+ T cell response. In addition, CypD deficiency was associated with an increase in pathogen burden at an early time-point following infection. Furthermore, our data demonstrate that transfer of wild-type macrophages (expressing CypD) to CypD-deficient mice can partially restore CD8+ T cell responses. These results establish that CypD plays an extrinsic role in regulating optimal effector CD8+ T cell responses to viral infection. Furthermore, this suggests that, under certain circumstances, inhibition of CypD function may have a detrimental impact on the host's ability to respond to viral infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Supervivencia Celular/genética , Interacciones Microbiota-Huesped/genética , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Peptidil-Prolil Isomerasa F/metabolismo , Traslado Adoptivo/métodos , Animales , Antígenos CD8/metabolismo , Peptidil-Prolil Isomerasa F/genética , Femenino , Interacciones Microbiota-Huesped/inmunología , Coriomeningitis Linfocítica/terapia , Coriomeningitis Linfocítica/virología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/inmunología , Mitocondrias/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/genética
4.
Front Immunol ; 8: 1885, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29354118

RESUMEN

Although vaccination has been an effective way of preventing infections ever since the eighteenth century, the generation of therapeutic vaccines and immunotherapies is still a work in progress. A number of challenges impede the development of these therapeutic approaches such as safety issues related to the administration of whole pathogens whether attenuated or inactivated. One safe alternative to classical vaccination methods gaining recognition is the use of nanoparticles, whether synthetic or naturally derived. We have recently demonstrated that the papaya mosaic virus (PapMV)-like nanoparticle can be used as a prophylactic vaccine against various viral and bacterial infections through the induction of protective humoral and cellular immune responses. Moreover, PapMV is also very efficient when used as an immune adjuvant in an immunotherapeutic setting at slowing down the growth of aggressive mouse melanoma tumors in a type I interferon (IFN-I)-dependent manner. In the present study, we were interested in exploiting the capacity of PapMV of inducing robust IFN-I production as treatment for the chronic viral infection model lymphocytic choriomeningitis virus (LCMV) clone 13 (Cl13). Treatment of LCMV Cl13-infected mice with two systemic administrations of PapMV was ineffective, as shown by the lack of changes in viral titers and immune response to LCMV following treatment. Moreover, IFN-α production following PapMV administration was almost completely abolished in LCMV-infected mice. To better isolate the mechanisms at play, we determined the influence of a pretreatment with PapMV on secondary PapMV administration, therefore eliminating potential variables emanating from the infection. Pretreatment with PapMV led to the same outcome as an LCMV infection in that IFN-α production following secondary PapMV immunization was abrogated for up to 50 days while immune activation was also dramatically impaired. We showed that two distinct and overlapping mechanisms were responsible for this outcome. While short-term inhibition was partially the result of interleukin-1 receptor-associated kinase 1 degradation, a crucial component of the toll-like receptor 7 signaling pathway, long-term inhibition was mainly due to interference by PapMV-specific antibodies. Thus, we identified a possible pitfall in the use of virus-like particles for the systemic treatment of chronic viral infections and discuss mitigating alternatives to circumvent these potential problems.

5.
Nano Lett ; 16(3): 1826-32, 2016 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-26891174

RESUMEN

The recent development of novel immunotherapies is revolutionizing cancer treatment. These include, for example, immune checkpoint blockade, immunomodulation, or therapeutic vaccination. Although effective on their own, combining multiple approaches will most likely be required in order to achieve the maximal therapeutic benefit. In this regard, the papaya mosaic virus nanoparticle (PapMV) has shown tremendous potential as (i) an immunostimulatory molecule, (ii) an adjuvant, and (iii) a vaccine platform through its intrinsic capacity to activate the innate immune response in an IFN-α-dependent manner. Here, we demonstrate that intratumor administration of PapMV significantly slows down melanoma progression and prolongs survival. This correlates with enhanced chemokine and pro-inflammatory-cytokine production in the tumor and increased immune-cell infiltration. Proportions of total and tumor-specific CD8(+) T cells dramatically increase following PapMV treatment whereas those of myeloid-derived suppressor cells (MDSC) concomitantly decrease. Moreover, systemic PapMV administration prevents metastatic tumor-implantation in the lungs. Importantly, PapMV also synergistically improves the therapeutic benefit of dendritic cell (DC)-based vaccination and PD-1 blockade by potentiating antitumor immune responses. This study illustrates the immunostimulatory potential of a plant virus-derived nanoparticle for cancer therapy either alone or in conjunction with other promising immunotherapies in clinical development.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Melanoma/prevención & control , Virus del Mosaico/inmunología , Nanopartículas , Animales , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Carica/virología , Línea Celular Tumoral , Citocinas/inmunología , Femenino , Inmunoterapia , Melanoma/inmunología , Melanoma/patología , Ratones Endogámicos C57BL , Virus del Mosaico/química , Nanopartículas/química
6.
Vaccines (Basel) ; 3(3): 620-37, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26350598

RESUMEN

Vaccines are considered one of the greatest medical achievements in the battle against infectious diseases. However, the intractability of various diseases such as hepatitis C, HIV/AIDS, malaria, tuberculosis, and cancer poses persistent hurdles given that traditional vaccine-development methods have proven to be ineffective; as such, these challenges have driven the emergence of novel vaccine design approaches. In this regard, much effort has been put into the development of new safe adjuvants and vaccine platforms. Of particular interest, the utilization of plant virus-like nanoparticles and recombinant plant viruses has gained increasing significance as an effective tool in the development of novel vaccines against infectious diseases and cancer. The present review summarizes recent advances in the use of plant viruses as nanoparticle-based vaccines and adjuvants and their mechanism of action. Harnessing plant-virus immunogenic properties will enable the design of novel, safe, and efficacious prophylactic and therapeutic vaccines against disease.

7.
Cell Mol Gastroenterol Hepatol ; 1(3): 325-341.e1, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-28210682

RESUMEN

BACKGROUND & AIMS: The constant exposure of the liver to food and bacterial antigens through the mesenteric circulation requires it to maintain tolerance while preserving the ability to mount an effective immune response against pathogens. We investigated the contribution of the liver's tolerogenic nature on the establishment of chronic viral infections. METHODS: TTR-NP mice, which express the nucleoprotein (NP) of lymphocytic choriomeningitis virus (LCMV) specifically in hepatocytes under control of a modified transthyretin (TTR) promoter, were infected with the Armstrong (Arm) or WE acute strains of LCMV. RESULTS: The infection persisted for at least 147 days in TTR-NP mice. Expression of NP by the liver induced a strong peripheral tolerance against NP that was mediated by interleukin-10-secreting CD4+ regulatory T cells, leading to high PD-1 (programmed death-1) expression and reduced effector function of virus-specific T cells. Despite an active immune response against LCMV, peripheral tolerance against a single viral protein was sufficient to induce T-cell exhaustion and chronic LCMV Armstrong (Arm) or WE infection by limiting the antiviral T-cell response in an otherwise immunocompetent host. Regulatory T-cell depletion of chronically infected TTR-NP mice led to functional restoration of LCMV-specific CD4+ and CD8+ T cell responses and viral clearance. CONCLUSIONS: Expression of a viral antigen by hepatocytes can induce a state of peripheral tolerance mediated by regulatory T cells that can lead to the establishment of a chronic viral infection. Strategies targeting regulatory T cells in patients chronically infected with hepatotropic viruses could represent a promising approach to restore functional antiviral immunity and clear infection.

9.
Oncoimmunology ; 3(1): e27897, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24800174

RESUMEN

The role of the complement system in oncogenesis and tumor progression remains poorly understood. We have recently demonstrated that the induction of a tumor-specific CD8+ T-cell response is improved upon transient inhibition of the complement system, which is coupled to an increased availability of natural killer cells. The complement system may therefore turn out to constitute a promising target for the development of novel anticancer therapeutics.

10.
Cancer Immunol Res ; 2(3): 200-6, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24778316

RESUMEN

Although the role of the complement system in cancer development has been studied, its involvement in the development of an antitumoral immune response remains poorly understood. Using cobra venom factor (CVF) to inhibit the complement cascade via C3 molecule exhaustion in immunocompetent mice bearing B16gp33 melanoma tumors, we show that transient inhibition of the complement system allowed for the development of a more robust gp33-specific antitumoral CD8(+) T-cell response. This immune response proved to be natural killer (NK) dependent, suggesting an interaction of complement proteins with this cellular subset leading to T lymphocyte activation and enhanced cytotoxic T-cell activity against tumor cells. This study demonstrates for the first time the implication of the complement system in the development of NK-mediated cytotoxic T-cell-dependent antitumoral immune responses. The complement pathway could therefore be a potent therapeutic target to improve NK-dependent antitumoral immune responses in patients with cancer.


Asunto(s)
Complemento C3/inmunología , Inactivadores del Complemento/farmacología , Venenos Elapídicos/farmacología , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Bazo/inmunología
11.
Mol Ther ; 22(6): 1198-1210, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24590047

RESUMEN

Cancer therapy using oncolytic viruses has gained interest in the last decade. Vesicular stomatitis virus is an attractive candidate for this alternative treatment approach. The importance of the immune response against tumor antigens in virotherapy efficacy is now well recognized, however, its relative contribution versus the intrinsic oncolytic capacity of viruses has been difficult to evaluate. To start addressing this question, we compared glycoprotein and matrix mutants of vesicular stomatitis virus (VSV), showing different oncolytic potentials for B16/B16gp33 melanoma tumor cells in vitro, with the wild-type virus in their ability to induce tumor-specific CD8(+) T cell responses and control tumor progression in vivo. Despite the fact that wild-type and G mutants induced a stronger gp33-specific immune response compared to the MM51R mutant, all VSV strains showed a similar capacity to slow down tumor progression. The effectiveness of the matrix mutant treatment proved to be CD8(+) dependent and directed against tumor antigens other than gp33 since adoptive transfer of isolated CD8(+) T lymphocytes from treated B16gp33-bearing mice resulted in significant protection of naive mice against challenge with the parental tumor. Remarkably, the VSV matrix mutant induced the upregulation of major histocompatibility class-I antigen at the tumor cell surface thus favoring recognition by CD8(+) T cells. These results demonstrate that VSV mutants induce an antitumor immune response using several mechanisms. A better understanding of these mechanisms will prove useful for the rational design of viruses with improved therapeutic efficacy.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Glicoproteínas de Membrana/genética , Virus Oncolíticos/genética , Vesiculovirus/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas de la Matriz Viral/genética , Animales , Línea Celular , Chlorocebus aethiops , Genes MHC Clase I , Células Hep G2 , Humanos , Inmunoterapia Adoptiva , Melanoma Experimental/virología , Antígenos Específicos del Melanoma/metabolismo , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Células Vero , Vesiculovirus/metabolismo , Proteínas del Envoltorio Viral/inmunología , Proteínas de la Matriz Viral/inmunología
12.
J Immunol ; 192(3): 1071-8, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24376264

RESUMEN

Developing new adjuvants and vaccination strategies is of paramount importance to successfully fight against many life-threatening infectious diseases and cancer. Very few adjuvants are currently authorized for human use, and these mainly stimulate a humoral response. However, specific Abs are not sufficient to confer protection against persisting infections or cancer. Therefore, development of adjuvants and immunomodulators able to enhance cell-mediated immune responses represents a major medical need. We recently showed that papaya mosaic virus nanoparticles (PapMV), self-assembled from the coat protein of a plant virus and a noncoding ssRNA molecule, are highly immunogenic in mice. PapMV can be used either as a vaccine delivery platform, through fusion of various epitopes to the coat protein or as adjuvant to enhance humoral immune responses against coadministered Ags or vaccines. However, the mechanisms that confer these immunomodulatory properties to PapMV and its ability to enhance T cell vaccines remain unknown. Using immunization studies in mice, we demonstrate in this paper that PapMV represents a novel TLR7 agonist with strong immunostimulatory properties. More importantly, pretreatment with PapMV significantly improves effector and memory CD8(+) T cell responses generated through dendritic cell vaccination increasing protection against a Listeria monocytogenes challenge.


Asunto(s)
Adyuvantes Inmunológicos , Linfocitos T CD8-positivos/inmunología , Listeria monocytogenes/inmunología , Listeriosis/prevención & control , Glicoproteínas de Membrana/agonistas , Subgrupos de Linfocitos T/inmunología , Receptor Toll-Like 7/agonistas , Tymovirus/inmunología , Vacunación , Inmunidad Adaptativa , Animales , Células Dendríticas/inmunología , Evaluación Preclínica de Medicamentos , Femenino , Inmunoglobulina G/biosíntesis , Memoria Inmunológica , Interferón Tipo I/inmunología , Listeriosis/inmunología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/inmunología , Nanopartículas , Ovalbúmina/inmunología , ARN Viral/inmunología , Receptor de Interferón alfa y beta/deficiencia , Receptor Toll-Like 7/deficiencia , Receptor Toll-Like 7/inmunología , Tymovirus/genética
13.
J Immunol ; 191(11): 5420-9, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24140642

RESUMEN

TNF-like ligand 1A (TL1A), also known as TNFSF15, is a member of the TNF superfamily. Its known receptor is death receptor 3 (DR3). In humans, TL1A also binds to a secreted TNF family member called decoy receptor 3, which interferes with the interaction between TL1A and DR3. TL1A/DR3 signal has been implicated in several autoimmune diseases in animal models as well as in clinical conditions. We generated TL1A gene knockout (KO) mice to assess its role in collagen-induced arthritis (CIA), a mouse model of human rheumatoid arthritis. The KO mice were fertile and had no visible anomalies. Their lymphoid organ size and cellularity, T and B cell subpopulations, Th cell and regulatory T cell development in vivo and in vitro, and antiviral immune responses were comparable to those of wild-type mice. However, the KO mice presented ameliorated CIA in terms of clinical scores, disease incidence, and pathological scores. The KO mice had reduced titers of pathogenic anti-collagen Abs in the sera. No apparent defect was found in the function of follicular Th cells. We revealed that plasma cells but not B cells expressed high levels of DR3 and were direct targets of TL1A. In the presence of TL1A, they survived better and produced more pathogenic Ab. This study presented novel knowledge about the role of TL1A in humoral immune responses and its mechanism of action in CIA pathogenesis.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Células Plasmáticas/inmunología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Formación de Anticuerpos/genética , Artritis Experimental/genética , Artritis Reumatoide/genética , Supervivencia Celular/genética , Células Cultivadas , Colágeno/inmunología , Humanos , Inmunidad Humoral , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 25 de Receptores de Factores de Necrosis Tumoral/genética , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Regulación hacia Arriba
14.
Med Sci (Paris) ; 29(2): 175-82, 2013 Feb.
Artículo en Francés | MEDLINE | ID: mdl-23452604

RESUMEN

Cancer is a complex disease that affects more and more people around the world. Unfortunately, existing treatments are only partially efficient and often induce major side effects. Thus, the use of viruses to selectively kill cancer cells is a new promising therapeutic approach. Recently, VSV has been used in oncolytic virotherapy because of its capacity to preferentially infect most human tumor cells. However, despite the availability of good oncolytic VSV mutants, the large variability of tumor cell types and the multiple ways in which they can evade viral infection suggests that therapeutic combinations of various viruses will be necessary to efficiently treat most cancers. A better understanding of the infection mechanisms and immune system recruitment by oncolytic viruses will be of great value for the development of safe and efficient strategies for cancer treatment.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica/métodos , Vesiculovirus/fisiología , Humanos , Sistema Inmunológico/fisiología , Modelos Biológicos , Neoplasias/inmunología , Viroterapia Oncolítica/tendencias , Virus Oncolíticos/genética , Estomatitis Vesicular/virología , Vesiculovirus/genética , Vesiculovirus/inmunología
15.
J Autoimmun ; 42: 19-28, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23137675

RESUMEN

CD8(+) T-cell immune response to liver antigens is often functionally diminished or absent. This may occur via deletion of these autoaggressive T-cells, through the acquisition of an anergic phenotype, or via active suppression mediated by other cell populations. We generated a double transgenic model in which mice express CD8(+) T-cells specific for the lymphocytic choriomeningitis virus nucleoprotein (LCMV-NP) and LCMV-NP as a hepatic neo-autoantigen, to study the immunological response of potentially liver antigen autoaggressive CD8(+) T-cells. Autoreactive transgenic CD8(+) T-cells were analyzed for functionality and cytotoxic effector status. Despite severe peripheral deletion of liver-specific CD8(+) T-cells, a fraction of autoreactive NP-specific CD8(+) T-cells accumulate in liver, resulting in hepatocyte injury and production of auto-antibodies in both male and female mice. NP-specific intrahepatic T-cells showed capacity to proliferate, produce cytokines and up-regulate activation markers. These data provide in vivo evidence that autoreactive CD8(+) T-cells are activated in the liver and developed an inflammatory process, but require additional factors to cause severe autoimmune destruction of hepatocytes. Our new model will provide a valuable tool for further exploration of the immunological response involved in inflammatory liver diseases, including autoimmune hepatitis.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Hepatocitos/inmunología , Hígado/inmunología , Nucleoproteínas/metabolismo , Animales , Antígenos Virales/genética , Antígenos Virales/inmunología , Apoptosis/genética , Apoptosis/inmunología , Células Cultivadas , Citotoxicidad Inmunológica/genética , Modelos Animales de Enfermedad , Femenino , Hepatitis Autoinmune/inmunología , Hepatocitos/patología , Virus de la Coriomeningitis Linfocítica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nucleoproteínas/genética , Nucleoproteínas/inmunología , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Transgenes/genética
16.
J Biol Chem ; 286(48): 41135-41152, 2011 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-21976681

RESUMEN

Erythropoietin-producing hepatocellular kinases (Eph kinases) constitute the largest family of cell membrane receptor tyrosine kinases, and their ligand ephrins are also cell surface molecules. Because of promiscuous interaction between Ephs and ephrins, there is considerable redundancy in this system, reflecting the essential roles of these molecules in the biological system through evolution. In this study, both Efnb1 and Efnb2 were null-mutated in the T cell compartment of mice through loxP-mediated gene deletion. Mice with this double conditional mutation (double KO mice) showed reduced thymus and spleen size and cellularity. There was a significant decrease in the DN4, double positive, and single positive thymocyte subpopulations and mature CD4 and CD8 cells in the periphery. dKO thymocytes and peripheral T cells failed to compete with their WT counterparts in irradiated recipients, and the T cells showed compromised ability of homeostatic expansion. dKO naive T cells were inferior in differentiating into Th1 and Th17 effectors in vitro. The dKO mice showed diminished immune response against LCMV infection. Mechanistic studies revealed that IL-6 signaling in dKO T cells was compromised, in terms of abated induction of STAT3 phosphorylation upon IL-6 stimulation. This defect likely contributed to the observed in vitro and in vivo phenotype in dKO mice. This study revealed novel roles of Efnb1 and Efnb2 in T cell development and function.


Asunto(s)
Diferenciación Celular/inmunología , Efrina-B1/inmunología , Inmunidad Innata , Interleucina-6/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Transducción de Señal/inmunología , Células TH1/inmunología , Células Th17/inmunología , Timocitos/inmunología , Animales , Linfocitos T CD8-positivos , Diferenciación Celular/genética , Efrina-B1/genética , Interleucina-6/genética , Coriomeningitis Linfocítica/genética , Ratones , Ratones Noqueados , Tamaño de los Órganos/genética , Tamaño de los Órganos/inmunología , Fosforilación/genética , Fosforilación/inmunología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Transducción de Señal/genética , Bazo/inmunología , Bazo/patología , Bazo/virología , Células TH1/patología , Células Th17/patología , Timocitos/patología , Timocitos/virología , Timo/inmunología , Timo/patología , Timo/virología
17.
J Virol ; 85(13): 6513-20, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21561919

RESUMEN

Vesicular stomatitis virus (VSV) has been widely used to characterize cellular processes, viral resistance, and cytopathogenicity. Recently, VSV has also been used for oncolytic virotherapy due to its capacity to selectively lyse tumor cells. Mutants of the matrix (M) protein of VSV have generally been preferred to the wild-type virus for oncolysis because of their ability to induce type I interferon (IFN) despite causing weaker cytopathic effects. However, due to the large variability of tumor types, it is quite clear that various approaches and combinations of multiple oncolytic viruses will be needed to effectively treat most cancers. With this in mind, our work focused on characterizing the cytopathogenic profiles of four replicative envelope glycoprotein (G) VSV mutants. In contrast to the prototypic M mutant, VSV G mutants are as efficient as wild-type virus at inhibiting cellular transcription and host protein translation. Despite being highly cytopathic, the mutant G(6R) triggers type I interferon secretion as efficiently as the M mutant. Importantly, most VSV G mutants are more effective at killing B16 and MC57 tumor cells in vitro than the M mutant or wild-type virus through apoptosis induction. Taken together, our results demonstrate that VSV G mutants retain the high cytopathogenicity of wild-type VSV, with G(6R) inducing type I IFN secretion at levels similar to that of the M mutant. VSV G protein mutants could therefore prove to be highly valuable for the development of novel oncolytic virotherapy strategies that are both safe and efficient for the treatment of various types of cancer.


Asunto(s)
Glicoproteínas de Membrana/genética , Mutación , Virus Oncolíticos/patogenicidad , Virus de la Estomatitis Vesicular Indiana/patogenicidad , Proteínas del Envoltorio Viral/genética , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Efecto Citopatogénico Viral , Fibroblastos/virología , Humanos , Células L , Ratones , Datos de Secuencia Molecular , Viroterapia Oncolítica , Virus Oncolíticos/genética , Virus Oncolíticos/metabolismo , Análisis de Secuencia de ADN , Células Vero , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/metabolismo
18.
J Exp Med ; 207(12): 2751-65, 2010 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-21041454

RESUMEN

Activation-induced deaminase (AID) is the mutator enzyme that initiates somatic hypermutation and isotype switching of the antibody genes in B lymphocytes. Undesired byproducts of AID function are oncogenic mutations. AID expression levels seem to correlate with the extent of its physiological and pathological functions. In this study, we identify AID as a novel Hsp90 (heat shock protein 90 kD) client. We find that cytoplasmic AID is in a dynamic equilibrium regulated by Hsp90. Hsp90 stabilizes cytoplasmic AID, as specific Hsp90 inhibition leads to cytoplasmic polyubiquitination and proteasomal degradation of AID. Consequently, Hsp90 inhibition results in a proportional reduction in antibody gene diversification and off-target mutation. This evolutionarily conserved regulatory mechanism determines the functional steady-state levels of AID in normal B cells and B cell lymphoma lines. Thus, Hsp90 assists AID-mediated antibody diversification by stabilizing AID. Hsp90 inhibition provides the first pharmacological means to down-regulate AID expression and activity, which could be relevant for therapy of some lymphomas and leukemias.


Asunto(s)
Diversidad de Anticuerpos , Citidina Desaminasa/metabolismo , Proteínas HSP90 de Choque Térmico/fisiología , Animales , Benzoquinonas/farmacología , Estabilidad de Enzimas , Proteínas de Fusión bcr-abl/análisis , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Células HeLa , Humanos , Cambio de Clase de Inmunoglobulina , Lactamas Macrocíclicas/farmacología , Ratones , Ubiquitina-Proteína Ligasas/fisiología
19.
Eur J Immunol ; 40(8): 2190-9, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20518030

RESUMEN

The initiation of CD8(+) T cell (CTL) immune responses can occur via cross-priming. Recent data suggested a relationship between cross-presentation and immunodominance of epitope-specific T cells. To test this association, we evaluated the efficacy of cross-presentation for several virus epitopes in vitro and examined if this can be extrapolated in vivo. Employing lymphocytic choriomeningitis virus (LCMV), we demonstrate that the cross-presentation and cross-priming of LCMV antigens were dominated by NP396, but not NP205 when analyzing the LCMV-NP. Although with LCMV-GP, cross-presentation was dominated by GP276, and cross-priming was dominated by GP33. Importantly, although NP396 was significantly more efficient than GP33 in cross-presentation, cross-priming of their specific CTL was comparable. In a subsequent virus challenge after cross-priming, GP33-specific CTL dominated the response. Accordingly, based on our data, the ability of viral epitopes to be cross-presented in vitro does not entirely reflect what would occur in cross-priming. Thus, weak cross-presenting antigens may still cross-prime an efficient CTL response depending on other in vivo elements such as the naïve T-cell precursor frequencies.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Reactividad Cruzada , Células Dendríticas/metabolismo , Virus de la Coriomeningitis Linfocítica/inmunología , Macrófagos/metabolismo , Linfocitos T Citotóxicos/metabolismo , Animales , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Línea Celular , Proliferación Celular , Células Dendríticas/inmunología , Células Dendríticas/virología , Humanos , Epítopos Inmunodominantes/inmunología , Epítopos Inmunodominantes/metabolismo , Activación de Linfocitos , Virus de la Coriomeningitis Linfocítica/patogenicidad , Macrófagos/inmunología , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología
20.
Antiviral Res ; 84(1): 14-22, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19622372

RESUMEN

In the absence of an immunocompetent mouse model for HCV replication, we developed a convenient xenograft mouse model that produces infectious viral particles. For this purpose, HCV-permissive tumors were generated in SCID/beige mice using a tumorigenic population of the human hepatocarcinoma-derived Huh7 cell line. Following infection, HCV RNA increased in the mouse sera and the human tumor by up to 10(5)GE/ml and 10(7)GE/microg of RNA, respectively. Immunohistochemistry analysis revealed that active viral replication had taken place within the tumor. Moreover, virus recovered from infected mice sera was readily infectious in cell culture. Finally, we showed that interferon-alpha and the protease inhibitor BILN-2061 inhibited the cell culture HCVcc strain JFH1 replication in vivo. In conclusion, we developed a simple and inexpensive mouse model that allows the production of infectious HCV particles in vivo. Such a model will be an extremely valuable tool for the characterization of promising drug candidates.


Asunto(s)
Carcinoma Hepatocelular/virología , Hepacivirus/fisiología , Hepatitis C/virología , Neoplasias Hepáticas Experimentales/virología , Modelos Animales , Cultivo de Virus/métodos , Replicación Viral , Animales , Antivirales/farmacología , Línea Celular Tumoral , Femenino , Hepacivirus/efectos de los fármacos , Hepacivirus/genética , Humanos , Interferón-alfa/farmacología , Ratones , Ratones Desnudos , Ratones SCID , Trasplante Heterólogo , Cultivo de Virus/economía , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA