Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Immunity ; 53(4): 824-839.e10, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33053331

RESUMEN

CD8+ T cells within the tumor microenvironment (TME) are exposed to various signals that ultimately determine functional outcomes. Here, we examined the role of the co-activating receptor CD226 (DNAM-1) in CD8+ T cell function. The absence of CD226 expression identified a subset of dysfunctional CD8+ T cells present in peripheral blood of healthy individuals. These cells exhibited reduced LFA-1 activation, altered TCR signaling, and a distinct transcriptomic program upon stimulation. CD226neg CD8+ T cells accumulated in human and mouse tumors of diverse origin through an antigen-specific mechanism involving the transcriptional regulator Eomesodermin (Eomes). Despite similar expression of co-inhibitory receptors, CD8+ tumor-infiltrating lymphocyte failed to respond to anti-PD-1 in the absence of CD226. Immune checkpoint blockade efficacy was hampered in Cd226-/- mice. Anti-CD137 (4-1BB) agonists also stimulated Eomes-dependent CD226 loss that limited the anti-tumor efficacy of this treatment. Thus, CD226 loss restrains CD8+ T cell function and limits the efficacy of cancer immunotherapy.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias/inmunología , Proteínas de Dominio T Box/inmunología , Animales , Humanos , Inhibidores de Puntos de Control Inmunológico/inmunología , Inmunoterapia/métodos , Ratones , Ratones Endogámicos C57BL , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Transcriptoma/inmunología , Microambiente Tumoral/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
2.
Blood ; 139(8): 1160-1176, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35201323

RESUMEN

Anti-CD38 monoclonal antibodies (mAbs) represent a breakthrough in the treatment of multiple myeloma (MM), yet some patients fail to respond or progress quickly with this therapy, highlighting the need for novel approaches. In this study we compared the preclinical efficacy of SAR442085, a next-generation anti-CD38 mAb with enhanced affinity for activating Fcγ receptors (FcγR), with first-generation anti-CD38 mAb daratumumab and isatuximab. In surface plasmon resonance and cellular binding assays, we found that SAR442085 had higher binding affinity than daratumumab and isatuximab for FcγRIIa (CD32a) and FcγRIIIa (CD16a). SAR442085 also exhibited better in vitro antibody-dependent cellular cytotoxicity (ADCC) against a panel of MM cells expressing variable CD38 receptor densities including MM patients' primary plasma cells. The enhanced ADCC of SAR442085 was confirmed using NK-92 cells bearing low and high affinity FcγRIIIa (CD16a)-158F/V variants. Using MM patients' primary bone marrow cells, we confirmed that SAR442085 had an increased ability to engage FcγRIIIa, resulting in higher natural killer (NK) cell activation and degranulation against primary plasma cells than preexisting Fc wild-type anti-CD38 mAbs. Finally, using huFcgR transgenic mice that express human Fcγ receptors under the control of their human regulatory elements, we demonstrated that SAR442085 had higher NK cell-dependent in vivo antitumor efficacy and better survival than daratumumab and isatuximab against EL4 thymoma or VK*MYC myeloma cells overexpressing human CD38. These results highlight the preclinical efficacy of SAR442085 and support the current evaluation of this next-generation anti-CD38 antibody in phase I clinical development in patients with relapsed/refractory MM.


Asunto(s)
ADP-Ribosil Ciclasa 1/antagonistas & inhibidores , Antineoplásicos Inmunológicos/farmacología , Células de la Médula Ósea , Glicoproteínas de Membrana/antagonistas & inhibidores , Mieloma Múltiple , Proteínas de Neoplasias/antagonistas & inhibidores , ADP-Ribosil Ciclasa 1/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Línea Celular Tumoral , Células HEK293 , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones Transgénicos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Proteínas de Neoplasias/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33766913

RESUMEN

CD4+Foxp3+ regulatory T (Treg) cells are central modulators of autoimmune diseases. However, the timing and location of Treg cell-mediated suppression of tissue-specific autoimmunity remain undefined. Here, we addressed these questions by investigating the role of tumor necrosis factor (TNF) receptor 2 (TNFR2) signaling in Treg cells during experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis. We found that TNFR2-expressing Treg cells were critical to suppress EAE at peak disease in the central nervous system but had no impact on T cell priming in lymphoid tissues at disease onset. Mechanistically, TNFR2 signaling maintained functional Treg cells with sustained expression of CTLA-4 and Blimp-1, allowing active suppression of pathogenic T cells in the inflamed central nervous system. This late effect of Treg cells was further confirmed by treating mice with TNF and TNFR2 agonists and antagonists. Our findings show that endogenous Treg cells specifically suppress an autoimmune disease by acting in the target tissue during overt inflammation. Moreover, they bring a mechanistic insight to some of the adverse effects of anti-TNF therapy in patients.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Médula Ósea/patología , Antígeno CTLA-4/metabolismo , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Humanos , Ratones , Ratones Noqueados , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/agonistas , Receptores Tipo II del Factor de Necrosis Tumoral/antagonistas & inhibidores , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Transducción de Señal/inmunología , Linfocitos T Reguladores/metabolismo
4.
Blood ; 132(16): 1689-1694, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-29986909

RESUMEN

Immune-based therapies hold promise for the treatment of multiple myeloma (MM), but so far, immune checkpoint blockade targeting programmed cell death protein 1 has not proven effective as single agent in this disease. T-cell immunoglobulin and ITIM domains (TIGIT) is another immune checkpoint receptor known to negatively regulate T-cell functions. In this study, we investigated the therapeutic potential of TIGIT blockade to unleash immune responses against MM. We observed that, in both mice and humans, MM progression was associated with high levels of TIGIT expression on CD8+ T cells. TIGIT+ CD8+ T cells from MM patients exhibited a dysfunctional phenotype characterized by decreased proliferation and inability to produce cytokines in response to anti-CD3/CD28/CD2 or myeloma antigen stimulation. Moreover, when challenged with Vk*MYC mouse MM cells, TIGIT-deficient mice showed decreased serum monoclonal immunoglobulin protein levels associated with reduced tumor burden and prolonged survival, indicating that TIGIT limits antimyeloma immune responses. Importantly, blocking TIGIT using monoclonal antibodies increased the effector function of MM patient CD8+ T cells and suppressed MM development. Altogether our data provide evidence for an immune-inhibitory role of TIGIT in MM and support the development of TIGIT-blocking strategies for the treatment of MM patients.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Linfocitos T CD8-positivos/inmunología , Mieloma Múltiple/prevención & control , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptores Inmunológicos/antagonistas & inhibidores , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Células Cultivadas , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mieloma Múltiple/etiología , Mieloma Múltiple/patología , Receptor de Muerte Celular Programada 1/inmunología , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/fisiología
5.
PLoS Genet ; 12(7): e1006185, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27438086

RESUMEN

The guanine nucleotide exchange factor Vav1 is essential for transducing T cell antigen receptor signals and therefore plays an important role in T cell development and activation. Our previous genetic studies identified a locus on rat chromosome 9 that controls the susceptibility to neuroinflammation and contains a non-synonymous polymorphism in the major candidate gene Vav1. To formally demonstrate the causal implication of this polymorphism, we generated a knock-in mouse bearing this polymorphism (Vav1R63W). Using this model, we show that Vav1R63W mice display reduced susceptibility to experimental autoimmune encephalomyelitis (EAE) induced by MOG35-55 peptide immunization. This is associated with a lower production of effector cytokines (IFN-γ, IL-17 and GM-CSF) by autoreactive CD4 T cells. Despite increased proportion of Foxp3+ regulatory T cells in Vav1R63W mice, we show that this lowered cytokine production is intrinsic to effector CD4 T cells and that Treg depletion has no impact on EAE development. Finally, we provide a mechanism for the above phenotype by showing that the Vav1R63W variant has normal enzymatic activity but reduced adaptor functions. Together, these data highlight the importance of Vav1 adaptor functions in the production of inflammatory cytokines by effector T cells and in the susceptibility to neuroinflammation.


Asunto(s)
Encefalomielitis Autoinmune Experimental/genética , Variación Genética , Proteínas Proto-Oncogénicas c-vav/genética , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T Reguladores/citología , Animales , Calcio/metabolismo , Sistema Nervioso Central/fisiopatología , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Femenino , Factores de Transcripción Forkhead/metabolismo , Predisposición Genética a la Enfermedad , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Polimorfismo Genético , Ratas , Transducción de Señal , Timo/metabolismo
6.
J Immunol ; 195(4): 1608-16, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26163585

RESUMEN

The development of inflammatory diseases depends on complex interactions between several genes and various environmental factors. Discovering new genetic risk factors and understanding the mechanisms whereby they influence disease development is of paramount importance. We previously reported that deficiency in Themis1, a new actor of TCR signaling, impairs regulatory T cell (Treg) function and predisposes Brown-Norway (BN) rats to spontaneous inflammatory bowel disease (IBD). In this study, we reveal that the epistasis between Themis1 and Vav1 controls the occurrence of these phenotypes. Indeed, by contrast with BN rats, Themis1 deficiency in Lewis rats neither impairs Treg suppressive functions nor induces pathological manifestations. By using congenic lines on the BN genomic background, we show that the impact of Themis1 deficiency on Treg suppressive functions depends on a 117-kb interval coding for a R63W polymorphism that impacts Vav1 expression and functions. Indeed, the introduction of a 117-kb interval containing the Lewis Vav1-R63 variant restores Treg function and protects Themis1-deficient BN rats from spontaneous IBD development. We further show that Themis1 binds more efficiently to the BN Vav1-W63 variant and is required to stabilize its recruitment to the transmembrane adaptor LAT and to fully promote the activation of Erk kinases. Together, these results highlight the importance of the signaling pathway involving epistasis between Themis1 and Vav1 in the control of Treg suppressive function and susceptibility to IBD development.


Asunto(s)
Epistasis Genética , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Proto-Oncogénicas c-vav/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Mutación , Proteínas Proto-Oncogénicas c-vav/metabolismo , Ratas , Ratas Transgénicas , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Timocitos/inmunología , Timocitos/metabolismo
7.
Clin J Pain ; 37(12): 904-907, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34757342

RESUMEN

OBJECTIVES: Evaluation of pain in critically ill intubated patients is difficult and subjective. This study aimed to evaluate the accuracy of oximetry-derived peripheral perfusion index (PPI) in pain assessment in critically ill intubated patients using the behavioral pain scale (BPS) as a reference. MATERIALS AND METHODS: This prospective observational study included 35 adult mechanically ventilated surgical patients during their first 2 postoperative days in the intensive care unit. Values of PPI, BPS, Richmond Agitation Sedation Scale (RASS), heart rate, and blood pressure were obtained before and after a standard painful stimulus (changing the patient position) and the ratio between the second and the first reading was calculated to determine the change (Δ) in all variables. The outcomes were the correlation between ΔBPS and ΔPPI as well as other hemodynamic parameters. The ability of the PPI to detect pain (defined as BPS ≥6) was analyzed using the area under receiver operating characteristic curve. RESULTS: Paired readings were obtained from 35 patients. After the standard painful stimulus, the PPI decreased while the BPS and the Richmond agitation sedation scale increased. The Spearman correlation coefficient (95% confidence interval) between Δ PPI and Δ BPS was 0.41 (0.09-0.65). PPI values showed poor accuracy in detecting pain with area under receiver operating characteristic curve (95% confidence interval): 0.65 (0.53-0.76), with best cutoff value of ≤2.7. CONCLUSION: The PPI decreased after application of a standard painful stimulus in critically ill intubated patients. ∆PPI showed a low correlation with ∆BPS, and a PPI of ≤2.7 showed a low ability to detect BPS ≥6. Therefore, PPI should not be used for pain evaluation in critically ill intubated surgical patients.


Asunto(s)
Enfermedad Crítica , Índice de Perfusión , Adulto , Humanos , Oximetría , Dolor , Dimensión del Dolor
8.
mBio ; 11(2)2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32265335

RESUMEN

Coinfections shape immunity and influence the development of inflammatory diseases, resulting in detrimental or beneficial outcome. Coinfections with concurrent Plasmodium species can alter malaria clinical evolution, and malaria infection itself can modulate autoimmune reactions. Yet, the underlying mechanisms remain ill defined. Here, we demonstrate that the protective effects of some rodent malaria strains on T cell-mediated inflammatory pathologies are due to an RNA virus cohosted in malaria-parasitized blood. We show that live and extracts of blood parasitized by Plasmodium berghei K173 or Plasmodium yoelii 17X YM, protect against P. berghei ANKA-induced experimental cerebral malaria (ECM) and myelin oligodendrocyte glycoprotein (MOG)/complete Freund's adjuvant (CFA)-induced experimental autoimmune encephalomyelitis (EAE), and that protection is associated with a strong type I interferon (IFN-I) signature. We detected the presence of the RNA virus lactate dehydrogenase-elevating virus (LDV) in the protective Plasmodium stabilates and we established that LDV infection alone was necessary and sufficient to recapitulate the protective effects on ECM and EAE. In ECM, protection resulted from an IFN-I-mediated reduction in the abundance of splenic conventional dendritic cell and impairment of their ability to produce interleukin (IL)-12p70, leading to a decrease in pathogenic CD4+ Th1 responses. In EAE, LDV infection induced IFN-I-mediated abrogation of IL-23, thereby preventing the differentiation of granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing encephalitogenic CD4+ T cells. Our work identifies a virus cohosted in several Plasmodium stabilates across the community and deciphers its major consequences on the host immune system. More generally, our data emphasize the importance of considering contemporaneous infections for the understanding of malaria-associated and autoimmune diseases.IMPORTANCE Any infection modifies the host immune status, potentially ameliorating or aggravating the pathophysiology of a simultaneous inflammatory condition. In the course of investigating how malaria infection modulates the severity of contemporaneous inflammatory diseases, we identified a nonpathogenic mouse virus in stabilates of two widely used rodent parasite lines: Plasmodium berghei K173 and Plasmodium yoelii 17X YM. We established that the protective effects of these Plasmodium lines on cerebral malaria and multiple sclerosis are exclusively due to this virus. The virus induces a massive type I interferon (IFN-I) response and causes quantitative and qualitative defects in the ability of dendritic cells to promote pathogenic T cell responses. Beyond revealing a possible confounding factor in rodent malaria models, our work uncovers some bases by which a seemingly innocuous viral (co)infection profoundly changes the immunopathophysiology of inflammatory diseases.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Inflamación/inmunología , Interferón Tipo I/inmunología , Virus Elevador de Lactato Deshidrogenasa/inmunología , Malaria Cerebral/inmunología , Animales , Coinfección/inmunología , Coinfección/parasitología , Coinfección/virología , Citocinas/inmunología , Células Dendríticas/inmunología , Inflamación/fisiopatología , Interferón gamma/inmunología , Malaria Cerebral/sangre , Malaria Cerebral/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei , Plasmodium yoelii , Bazo/citología , Bazo/inmunología
9.
Front Immunol ; 9: 2399, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30410484

RESUMEN

The guanine nucleotide exchange factor Vav1 is essential for transducing T cell receptor (TCR) signals and plays an important role in T cell development and activation. Previous genetic studies identified a natural variant of Vav1 characterized by the substitution of an arginine (R) residue by a tryptophane (W) at position 63 (Vav1R63W). This variant impacts Vav1 adaptor functions and controls susceptibility to T cell-mediated neuroinflammation. To assess the implication of this Vav1 variant on the susceptibility to antibody-mediated diseases, we used the animal model of myasthenia gravis, experimental autoimmune myasthenia gravis (EAMG). To this end, we generated a knock-in (KI) mouse model bearing a R to W substitution in the Vav1 gene (Vav1R63W) and immunized it with either torpedo acetylcholine receptor (tAChR) or the α146-162 immunodominant peptide. We observed that the Vav1R63W conferred increased susceptibility to EAMG, revealed by a higher AChR loss together with an increased production of effector cytokines (IFN-γ, IL-17A, GM-CSF) by antigen-specific CD4+ T cells, as well as an increased frequency of antigen-specific CD4+ T cells. This correlated with the emergence of a dominant antigen-specific T cell clone in KI mice that was not present in wild-type mice, suggesting an impact on thymic selection and/or a different clonal selection threshold following antigen encounter. Our results highlight the key role of Vav1 in the pathophysiology of EAMG and this was associated with an impact on the TCR repertoire of AChR reactive T lymphocytes.


Asunto(s)
Variación Genética , Miastenia Gravis Autoinmune Experimental/etiología , Miastenia Gravis Autoinmune Experimental/metabolismo , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Ratones , Miastenia Gravis Autoinmune Experimental/patología , Fenotipo , Receptores Nicotínicos/metabolismo , Especificidad del Receptor de Antígeno de Linfocitos T
10.
Sci Signal ; 11(538)2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29991650

RESUMEN

The activation of T cells requires the guanine nucleotide exchange factor VAV1. Using mice in which a tag for affinity purification was attached to endogenous VAV1 molecules, we analyzed by quantitative mass spectrometry the signaling complex that assembles around activated VAV1. Fifty VAV1-binding partners were identified, most of which had not been previously reported to participate in VAV1 signaling. Among these was CD226, a costimulatory molecule of immune cells. Engagement of CD226 induced the tyrosine phosphorylation of VAV1 and synergized with T cell receptor (TCR) signals to specifically enhance the production of interleukin-17 (IL-17) by primary human CD4+ T cells. Moreover, co-engagement of the TCR and a risk variant of CD226 that is associated with autoimmunity (rs763361) further enhanced VAV1 activation and IL-17 production. Thus, our study reveals that a VAV1-based, synergistic cross-talk exists between the TCR and CD226 during both physiological and pathological T cell responses and provides a rational basis for targeting CD226 for the management of autoimmune diseases.

11.
Cancer Cell ; 33(4): 634-648.e5, 2018 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-29551594

RESUMEN

Tumor-promoting inflammation and avoiding immune destruction are hallmarks of cancer. Here, we demonstrate that the pro-inflammatory cytokine interleukin (IL)-18 is critically involved in these hallmarks in multiple myeloma (MM). Mice deficient for IL-18 were remarkably protected from Vk∗MYC MM progression in a CD8+ T cell-dependent manner. The MM-niche-derived IL-18 drove generation of myeloid-derived suppressor cells (MDSCs), leading to accelerated disease progression. A global transcriptome analysis of the immune microenvironment in 73 MM patients strongly supported the negative impact of IL-18-driven MDSCs on T cell responses. Strikingly, high levels of bone marrow plasma IL-18 were associated with poor overall survival in MM patients. Furthermore, our preclinical studies suggested that IL-18 could be a potential therapeutic target in MM.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interleucina-18/metabolismo , Mieloma Múltiple/patología , Células Supresoras de Origen Mieloide/inmunología , Regulación hacia Arriba , Animales , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Tolerancia Inmunológica , Interleucina-18/sangre , Masculino , Ratones , Mieloma Múltiple/genética , Mieloma Múltiple/inmunología , Células Supresoras de Origen Mieloide/patología , Trasplante de Neoplasias , Pronóstico , Proteínas Proto-Oncogénicas c-myc/genética , Análisis de Supervivencia , Microambiente Tumoral
12.
Front Immunol ; 8: 1096, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28959254

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) with an increasing incidence in developed countries. Recent reports suggest that modulation of the gut microbiota might be one promising therapy for MS. Here, we investigated whether the probiotic Escherichia coli strain Nissle 1917 (ECN) could modulate the outcome of experimental autoimmune encephalomyelitis (EAE), a murine model of MS. We evidenced that daily oral treatment with ECN, but not with the archetypal K12 E. coli strain MG1655, reduced the severity of EAE induced by immunization with the MOG35-55 peptide. This beneficial effect was associated with a decreased secretion of inflammatory cytokines and an increased production of the anti-inflammatory cytokine IL-10 by autoreactive CD4 T cells, both in peripheral lymph nodes and CNS. Interestingly, ECN-treated mice exhibited increased numbers of MOG-specific CD4+ T cells in the periphery contrasting with severely reduced numbers in the CNS, suggesting that ECN might affect T cell migration from the periphery to the CNS through a modulation of their activation and/or differentiation. In addition, we demonstrated that EAE is associated with a profound defect in the intestinal barrier function and that treatment with ECN, but not with MG1655, repaired intestinal permeability dysfunction. Collectively, our data reveal that EAE induces a disruption of the intestinal homeostasis and that ECN protects from disease and restores the intestinal barrier function.

13.
Artículo en Inglés | MEDLINE | ID: mdl-24825161

RESUMEN

Rho-GTPases belong to the Ras superfamily and are crucial signal transducing proteins downstream of many receptors. In general, the Rho-GTPases function as molecular switches, cycling between inactive (GDP-bound) and active (GTP-bound) states. The activated GTP bound Rho-GTPases interact with a broad spectrum of effectors to regulate a plethora of biological pathways including cytoskeletal dynamics, motility, cytokinesis, cell growth, apoptosis, transcriptional activity and nuclear signaling. Recently, gene targeting in mice allowed the selective inactivation of different Rho-GTPases and has advanced our understanding of the physiological role of these proteins, particularly in the immune system. Particularly, these proteins are key signaling molecules in T lymphocytes, which are generated in the thymus and are major players in the immune system. The scope of this review is to discuss recent data obtained in Rho-GTPases deficient mice by focusing on the role-played by Rho-GTPases in T-lymphocyte development, migration, activation and differentiation.


Asunto(s)
Linfocitos T/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Diferenciación Celular , Movimiento Celular , Citocinesis , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Sinapsis/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Timo/citología
14.
J Exp Med ; 211(9): 1779-92, 2014 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-25073791

RESUMEN

Interventional strategies to treat atherosclerosis, such as transluminal angioplasty and stent implantation, often cause vascular injury. This leads to intimal hyperplasia (IH) formation that induces inflammatory and fibroproliferative processes and ultimately restenosis. We show that phosphoinositide 3-kinase γ (PI3Kγ) is a key player in IH formation and is a valid therapeutic target in its prevention/treatment. PI3Kγ-deficient mice and mice expressing catalytically inactive PI3Kγ (PI3Kγ KD) showed reduced arterial occlusion and accumulation of monocytes and T cells around sites of vascular lesion. The transfer of PI3Kγ KD CD4(+) T cells into Rag2-deficient mice greatly reduced vascular occlusion compared with WT cells, clearly demonstrating the involvement of PI3Kγ in CD4(+) T cells during IH formation. In addition we found that IH is associated with increased levels of Th1 and Th17 cytokines. A specific decrease in the Th1 response was observed in the absence of PI3Kγ activity, leading to decreased CXCL10 and RANTES production by smooth muscle cells. Finally, we show that treatment with the PI3Kγ inhibitor AS-605240 is sufficient to decrease IH in both mouse and rat models, reinforcing the therapeutic potential of PI3Kγ inhibition. Altogether, these findings demonstrate a new role for PI3Kγ activity in Th1-controlled IH development.


Asunto(s)
Neointima/enzimología , Neointima/inmunología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Células TH1/efectos de los fármacos , Células TH1/inmunología , Animales , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Traumatismos de las Arterias Carótidas/enzimología , Traumatismos de las Arterias Carótidas/inmunología , Fosfatidilinositol 3-Quinasa Clase Ib/deficiencia , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Inhibidores Enzimáticos/farmacología , Arteria Femoral/enzimología , Arteria Femoral/inmunología , Arteria Femoral/lesiones , Marcación de Gen , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neointima/tratamiento farmacológico , Quinoxalinas/farmacología , Ratas , Ratas Sprague-Dawley , Tiazolidinedionas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA