Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Intervalo de año de publicación
1.
Front Immunol ; 14: 1143012, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37143657

RESUMEN

Introduction: Plasmodium sporozoites (SPZ) inoculated by Anopheles mosquitoes into the skin of the mammalian host migrate to the liver before infecting hepatocytes. Previous work demonstrated that early production of IL-6 in the liver is detrimental for the parasite growth, contributing to the acquisition of a long-lasting immune protection after immunization with live attenuated parasites. Methods: Considering that IL-6 as a critical pro-inflammatory signal, we explored a novel approach whereby the parasite itself encodes for the murine IL-6 gene. We generated transgenic P. berghei parasites that express murine IL-6 during liver stage development. Results and Discussion: Though IL-6 transgenic SPZ developed into exo-erythrocytic forms in hepatocytes in vitro and in vivo, these parasites were not capable of inducing a blood stage infection in mice. Furthermore, immunization of mice with transgenic IL-6-expressing P. berghei SPZ elicited a long-lasting CD8+ T cell-mediated protective immunity against a subsequent infectious SPZ challenge. Collectively, this study demonstrates that parasite-encoded IL-6 attenuates parasite virulence with abortive liver stage of Plasmodium infection, forming the basis of a novel suicide vaccine strategy to elicit protective antimalarial immunity.


Asunto(s)
Hepatopatías , Vacunas contra la Malaria , Animales , Ratones , Linfocitos T CD8-positivos , Interleucina-6 , Mamíferos , Plasmodium berghei
2.
Eur J Immunol ; 52(10): 1648-1661, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36030374

RESUMEN

Anti-CD20 monoclonal antibodies such as Rituximab, Ofatumumab, and Obinutuzumab are widely used to treat lymphomas and autoimmune diseases. They act by depleting B cells, mainly through Fc-dependent effectors functions. Some patients develop resistance to treatment but the underlying mechanisms are poorly understood. Here, we performed a genome-wide CRISPR/Cas9 screen to identify genes regulating the efficacy of anti-CD20 antibodies. We used as a model the killing of RAJI B cells by Rituximab through complement-dependent-cytotoxicity (CDC). As expected, the screen identified MS4A1, encoding CD20, the target of Rituximab. Among other identified genes, the role of Interferon Regulatory Factor 8 (IRF8) was validated in two B-cell lines. IRF8 KO also decreased the efficacy of antibody-dependent cellular cytotoxicity and phagocytosis (ADCC and ADCP) induced by anti-CD20 antibodies. We further show that IRF8 is necessary for efficient CD20 transcription. Levels of IRF8 and CD20 RNA or proteins correlated in normal B cells and in hundreds of malignant B cells. Therefore, IRF8 regulates CD20 expression and controls the depleting capacity of anti-CD20 antibodies. Our results bring novel insights into the pathways underlying resistance to CD20-targeting immunotherapies.


Asunto(s)
Antígenos CD20 , Antineoplásicos , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , ARN , Rituximab/farmacología , Rituximab/uso terapéutico
3.
Nat Commun ; 13(1): 4123, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35840625

RESUMEN

Plasmodium vivax is the most widespread human malaria parasite. Due to the presence of extravascular reservoirs and relapsing infections from dormant liver stages, P. vivax is particularly difficult to control and eliminate. Experimental research is hampered by the inability to maintain P. vivax cultures in vitro, due to its tropism for immature red blood cells (RBCs). Here, we describe a new humanized mice model that can support efficient human erythropoiesis and maintain long-lasting multiplication of inoculated cryopreserved P. vivax parasites and their sexual differentiation, including in bone marrow. Mature gametocytes were transmitted to Anopheles mosquitoes, which led to the formation of salivary gland sporozoites. Importantly, blood-stage P. vivax parasites were maintained after the secondary transfer of fresh or frozen infected bone marrow cells to naïve chimeras. This model provides a unique tool for investigating, in vivo, the biology of intraerythrocytic P. vivax.


Asunto(s)
Anopheles , Malaria Vivax , Animales , Anopheles/parasitología , Humanos , Malaria Vivax/parasitología , Ratones , Recurrencia Local de Neoplasia , Plasmodium vivax , Esporozoítos
4.
Curr Biol ; 31(10): 2203-2213.e5, 2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-33711249

RESUMEN

The midbody at the center of the intercellular bridge connecting dividing cells recruits the machinery essential for the final steps of cytokinesis.1-5 Successive abscission on both sides of the midbody generates a free midbody remnant (MBR) that can be inherited and accumulated in many cancer, immortalized, and stem cells, both in culture and in vivo.6-12 Strikingly, this organelle was recently shown to contain information that induces cancer cell proliferation, influences cell polarity, and promotes dorso-ventral axis specification upon interaction with recipient cells.13-16 Yet the mechanisms by which the MBR is captured by either a daughter cell or a distant cell are poorly described.10,14 Here, we report that BST2/tetherin, a well-established restriction factor that blocks the release of numerous enveloped viruses from the surface of infected cells,17-20 plays an analogous role in retaining midbody remnants. We found that BST2 is enriched at the midbody during cytokinesis and localizes at the surface of MBRs in a variety of cells. Knocking out BST2 induces the detachment of MBRs from the cell surface, their accumulation in the extracellular medium, and their transfer to distant cells. Mechanistically, the localization of BST2 at the MBR membrane is both necessary and sufficient for the interaction between MBRs and the cell surface. We thus propose that BST2 tethers post-cytokinetic midbody remnants to the cell surface. This finding reveals new parallels between cytokinesis and viral biology21-26 that unexpectedly extend beyond the ESCRT-dependent abscission step.


Asunto(s)
Antígenos CD , Antígeno 2 del Estroma de la Médula Ósea , Citocinesis , Antígenos CD/genética , Antígenos CD/fisiología , Antígeno 2 del Estroma de la Médula Ósea/fisiología , Membrana Celular , Proteínas Ligadas a GPI/fisiología , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Orgánulos
5.
PLoS Genet ; 16(10): e1009022, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33125370

RESUMEN

Adult skeletal muscles are maintained during homeostasis and regenerated upon injury by muscle stem cells (MuSCs). A heterogeneity in self-renewal, differentiation and regeneration properties has been reported for MuSCs based on their anatomical location. Although MuSCs derived from extraocular muscles (EOM) have a higher regenerative capacity than those derived from limb muscles, the molecular determinants that govern these differences remain undefined. Here we show that EOM and limb MuSCs have distinct DNA methylation signatures associated with enhancers of location-specific genes, and that the EOM transcriptome is reprogrammed following transplantation into a limb muscle environment. Notably, EOM MuSCs expressed host-site specific positional Hox codes after engraftment and self-renewal within the host muscle. However, about 10% of EOM-specific genes showed engraftment-resistant expression, pointing to cell-intrinsic molecular determinants of the higher engraftment potential of EOM MuSCs. Our results underscore the molecular diversity of distinct MuSC populations and molecularly define their plasticity in response to microenvironmental cues. These findings provide insights into strategies designed to improve the functional capacity of MuSCs in the context of regenerative medicine.


Asunto(s)
Plasticidad de la Célula/genética , Epigenoma/genética , Trasplante de Células Madre , Transcriptoma/genética , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Proliferación Celular/genética , Extremidades/crecimiento & desarrollo , Variación Genética/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Células Musculares/citología , Fibras Musculares Esqueléticas , Músculo Esquelético/citología , Mioblastos/citología , Regeneración/genética , Células Madre/citología , Células Madre/metabolismo
6.
Cell Microbiol ; 21(7): e13021, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30835870

RESUMEN

Protozoan pathogens secrete nanosized particles called extracellular vesicles (EVs) to facilitate their survival and chronic infection. Here, we show the inhibition by Plasmodium berghei NK65 blood stage-derived EVs of the proliferative response of CD4+ T cells in response to antigen presentation. Importantly, these results were confirmed in vivo by the capacity of EVs to diminish the ovalbumin-specific delayed type hypersensitivity response. We identified two proteins associated with EVs, the histamine releasing factor (HRF) and the elongation factor 1α (EF-1α) that were found to have immunosuppressive activities. Interestingly, in contrast to WT parasites, EVs from genetically HRF- and EF-1α-deficient parasites failed to inhibit T cell responses in vitro and in vivo. At the level of T cells, we demonstrated that EVs from WT parasites dephosphorylate key molecules (PLCγ1, Akt, and ERK) of the T cell receptor signalling cascade. Remarkably, immunisation with EF-1α alone or in combination with HRF conferred a long-lasting antiparasite protection and immune memory. In conclusion, we identified a new mechanism by which P. berghei-derived EVs exert their immunosuppressive functions by altering T cell responses. The identification of two highly conserved immune suppressive factors offers new conceptual strategies to overcome EV-mediated immune suppression in malaria-infected individuals.


Asunto(s)
Biomarcadores de Tumor/genética , Vesículas Extracelulares/inmunología , Malaria/genética , Factor 1 de Elongación Peptídica/genética , Animales , Presentación de Antígeno/inmunología , Antígenos/genética , Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/parasitología , Proliferación Celular/genética , Vesículas Extracelulares/genética , Humanos , Evasión Inmune/genética , Evasión Inmune/inmunología , Malaria/parasitología , Malaria/patología , Plasmodium berghei/genética , Plasmodium berghei/patogenicidad , Linfocitos T/inmunología , Linfocitos T/parasitología , Proteína Tumoral Controlada Traslacionalmente 1
7.
Elife ; 72018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30106373

RESUMEN

Skeletal muscle regeneration depends on satellite cells. After injury these muscle stem cells exit quiescence, proliferate and differentiate to regenerate damaged fibres. We show that this progression is accompanied by metabolic changes leading to increased production of reactive oxygen species (ROS). Using Pitx2/3 single and double mutant mice that provide genetic models of deregulated redox states, we demonstrate that moderate overproduction of ROS results in premature differentiation of satellite cells while high levels lead to their senescence and regenerative failure. Using the ROS scavenger, N-Acetyl-Cysteine (NAC), in primary cultures we show that a physiological increase in ROS is required for satellite cells to exit the cell cycle and initiate differentiation through the redox activation of p38α MAP kinase. Subjecting cultured satellite cells to transient inhibition of P38α MAP kinase in conjunction with NAC treatment leads to their rapid expansion, with striking improvement of their regenerative potential in grafting experiments.


Asunto(s)
Proteínas de Homeodominio/genética , Proteína Quinasa 14 Activada por Mitógenos/genética , Regeneración/genética , Factores de Transcripción/genética , Acetilcisteína/administración & dosificación , Animales , Diferenciación Celular/genética , Senescencia Celular/genética , Ratones , Músculo Esquelético/citología , Mutación , Oxidación-Reducción , Especies Reactivas de Oxígeno , Células Satélite del Músculo Esquelético , Células Madre/citología , Proteína del Homeodomínio PITX2
8.
J Exp Med ; 213(8): 1419-28, 2016 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-27432939

RESUMEN

Although most vaccines against blood stage malaria in development today use subunit preparations, live attenuated parasites confer significantly broader and more lasting protection. In recent years, Plasmodium genetically attenuated parasites (GAPs) have been generated in rodent models that cause self-resolving blood stage infections and induce strong protection. All such GAPs generated so far bear mutations in housekeeping genes important for parasite development in red blood cells. In this study, using a Plasmodium berghei model compatible with tracking anti-blood stage immune responses over time, we report a novel blood stage GAP that lacks a secreted factor related to histamine-releasing factor (HRF). Lack of HRF causes an IL-6 increase, which boosts T and B cell responses to resolve infection and leave a cross-stage, cross-species, and lasting immunity. Mutant-induced protection involves a combination of antiparasite IgG2c antibodies and FcγR(+) CD11b(+) cell phagocytes, especially neutrophils, which are sufficient to confer protection. This immune-boosting GAP highlights an important role of opsonized parasite-mediated phagocytosis, which may be central to protection induced by all self-resolving blood stage GAP infections.


Asunto(s)
Biomarcadores de Tumor/genética , Malaria , Plasmodium berghei , Proteínas Protozoarias , Linfocitos T/inmunología , Animales , Anticuerpos Antiprotozoarios/inmunología , Linfocitos B/inmunología , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina G/inmunología , Interleucina-6/inmunología , Malaria/genética , Malaria/inmunología , Ratones , Neutrófilos/inmunología , Fagocitosis/inmunología , Plasmodium berghei/genética , Plasmodium berghei/inmunología , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Proteína Tumoral Controlada Traslacionalmente 1
9.
PLoS One ; 9(2): e89479, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586810

RESUMEN

Homozygous mutations in EVER genes cause epidermodysplasia verruciformis (EV), characterized by an immune defect and the development of skin cancers associated with ß-human papillomavirus (HPV) infections. The effects of EVER protein loss on the keratinocyte immune response remain unknown. We show here that EVER2 plays a critical role in the interplay between the NF-κB and JNK/AP-1 signaling pathways. EVER2-deficient cells overproduce IL-6 following the upregulation of JNK activation. They respond poorly to phorbol ester and TNF via the NF-κB pathway. They have lower levels of IKKα subunit, potentially accounting for impairments of p100 processing and the alternative NF-κB pathway. The loss of EVER2 is associated with an unusual TRAF protein profile. We demonstrate that EVER2 deficiency sustains TRAF2 ubiquitination and decreases the pool of TRAF2 available in the detergent-soluble fraction of the cell. Finally, we demonstrate that EVER2 loss induces constitutive PKCα-dependent c-jun phosphorylation and facilitates activation of the HPV5 long control region through a JNK-dependent pathway. These findings indicate that defects of the EVER2 gene may create an environment conducive to HPV replication and the persistence of lesions with the potential to develop into skin cancer.


Asunto(s)
Resistencia a la Enfermedad/genética , Queratinocitos/metabolismo , Proteínas de la Membrana/deficiencia , FN-kappa B/metabolismo , Infecciones por Papillomavirus/metabolismo , Transducción de Señal/fisiología , Western Blotting , Cartilla de ADN/genética , Humanos , Quinasa I-kappa B/metabolismo , Inmunoprecipitación , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microscopía Fluorescente , Infecciones por Papillomavirus/genética , Fosforilación , Proteína Quinasa C-alfa/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor 2 Asociado a Receptor de TNF/metabolismo
10.
J Cell Sci ; 126(Pt 19): 4424-35, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23886947

RESUMEN

Cell-to-cell communication is essential in multicellular organisms. Tunneling nanotubes (TNTs) have emerged as a new type of intercellular spreading mechanism allowing the transport of various signals, organelles and pathogens. Here, we study the role of the unconventional molecular motor myosin-X (Myo10) in the formation of functional TNTs within neuronal CAD cells. Myo10 protein expression increases the number of TNTs and the transfer of vesicles between co-cultured cells. We also show that TNT formation requires both the motor and tail domains of the protein, and identify the F2 lobe of the FERM domain within the Myo10 tail as necessary for TNT formation. Taken together, these results indicate that, in neuronal cells, TNTs can arise from a subset of Myo10-driven dorsal filopodia, independent of its binding to integrins and N-cadherins. In addition our data highlight the existence of different mechanisms for the establishment and regulation of TNTs in neuronal cells and other cell types.


Asunto(s)
Comunicación Celular/fisiología , Miosinas/metabolismo , Nanotubos , Neuronas/citología , Neuronas/metabolismo , Animales , Transporte Biológico , Ratones , Seudópodos/metabolismo , Transfección
11.
PLoS Negl Trop Dis ; 7(4): e2154, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23593521

RESUMEN

BACKGROUND/OBJECTIVES: Human leishmaniases are parasitic diseases causing severe morbidity and mortality. No vaccine is available and numerous factors limit the use of current therapies. There is thus an urgent need for innovative initiatives to identify new chemotypes displaying selective activity against intracellular Leishmania amastigotes that develop and proliferate inside macrophages, thereby causing the pathology of leishmaniasis. METHODOLOGY/PRINCIPAL FINDINGS: We have developed a biologically sound High Content Analysis assay, based on the use of homogeneous populations of primary mouse macrophages hosting Leishmania amazonensis amastigotes. In contrast to classical promastigote-based screens, our assay more closely mimics the environment where intracellular amastigotes are growing within acidic parasitophorous vacuoles of their host cells. This multi-parametric assay provides quantitative data that accurately monitors the parasitic load of amastigotes-hosting macrophage cultures for the discovery of leishmanicidal compounds, but also their potential toxic effect on host macrophages. We validated our approach by using a small set of compounds of leishmanicidal drugs and recently published chemical entities. Based on their intramacrophagic leishmanicidal activity and their toxicity against host cells, compounds were classified as irrelevant or relevant for entering the next step in the drug discovery pipeline. CONCLUSIONS/SIGNIFICANCE: Our assay represents a new screening platform that overcomes several limitations in anti-leishmanial drug discovery. First, the ability to detect toxicity on primary macrophages allows for discovery of compounds able to cross the membranes of macrophage, vacuole and amastigote, thereby accelerating the hit to lead development process for compounds selectively targeting intracellular parasites. Second, our assay allows discovery of anti-leishmanials that interfere with biological functions of the macrophage required for parasite development and growth, such as organelle trafficking/acidification or production of microbicidal effectors. These data thus validate a novel phenotypic screening assay using virulent Leishmania amastigotes growing inside primary macrophage to identify new chemical entities with bona fide drug potential.


Asunto(s)
Antiprotozoarios/farmacología , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Leishmania/patogenicidad , Macrófagos/parasitología , Animales , Células Cultivadas , Leishmania/efectos de los fármacos , Leishmaniasis/parasitología , Ratones
12.
J Immunother ; 34(1): 65-75, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21150714

RESUMEN

Dendritic cell-derived exosomes (Dex) are nanovesicles bearing major histocompatibility complexes promoting T-cell-dependent antitumor effects in mice. Two phase I clinical trials aimed at vaccinating cancer patients with peptide-pulsed Dex have shown the feasibility and safety of inoculating clinical-grade Dex, but have failed to show their immunizing capacity. These low immunogenic capacities have led us to develop second-generation Dex with enhanced immunostimulatory properties. Here, we show that interferon-γ is a key cytokine conditioning the dendritic cell to induce the expression of CD40, CD80, CD86, and CD54 on Dex, endowing them with direct and potent peptide-dependent CD8(+) T-cell-triggering potential in vitro and in vivo. In this study, we describe the clinical grade process to manufacture large-scale interferon-γ-Dex vaccines and their quality control parameters currently used in a phase II trial.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer , Células Dendríticas/inmunología , Exosomas/inmunología , Interferón gamma/inmunología , Animales , Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Antígeno B7-1/genética , Antígeno B7-2/genética , Antígenos CD40 , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/metabolismo , Expresión Génica , Humanos , Immunoblotting , Molécula 1 de Adhesión Intercelular/genética , Activación de Linfocitos , Ratones , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA