Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
1.
J Leukoc Biol ; 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38489541

RESUMEN

Toll-like receptors are pattern recognition receptors that bridge the innate and adaptive immune responses and are critical for host defense. Most studies of Toll-like receptors have focused upon their roles in myeloid cells. B lymphocytes express most Toll-like receptors and are responsive to Toll-like receptor ligands, yet Toll-like receptor-mediated signaling in B cells is relatively understudied. This is an important knowledge gap, as Toll-like receptor functions can be cell type specific. In striking contrast to myeloid cells, TRAF3 inhibits TLR-mediated functions in B cells. TRAF3-deficient B cells display enhanced IRF3 and NFκB activation, cytokine production, immunoglobulin isotype switching, and antibody production in response to Toll-like receptors 3, 4, 7, and 9. Here, we address the question of how TRAF3 impacts initial B-cell Toll-like receptor signals to regulate downstream activation. We found that TRAF3 in B cells associated with proximal Toll-like receptor 4 and 7 signaling proteins, including MyD88, TRAF6, and the tyrosine kinase Syk. In the absence of TRAF3, TRAF6 showed a greater association with several Toll-like receptor signaling proteins, suggesting that TRAF3 may inhibit TRAF6 access to Toll-like receptor signaling complexes and thus early Toll-like receptor signaling. In addition, our results highlight a key role for Syk in Toll-like receptor signaling in B cells. In the absence of TRAF3, Syk activation was enhanced in response to ligands for Toll-like receptors 4 and 7, and Syk inhibition reduced downstream Toll-like receptor-mediated NFκB activation and proinflammatory cytokine production. This study reveals multiple mechanisms by which TRAF3 serves as a key negative regulator of early Toll-like receptor signaling events in B cells.

2.
Nat Commun ; 15(1): 615, 2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38242888

RESUMEN

Anti-multiple myeloma B cell maturation antigen (BCMA)-specific chimeric antigen receptor (CAR) T-cell therapies represent a promising treatment strategy with high response rates in myeloma. However, durable cures following anti-BCMA CAR-T cell treatment of myeloma are rare. One potential reason is that a small subset of minimal residual myeloma cells seeds relapse. Residual myeloma cells following BCMA-CAR-T-mediated treatment show less-differentiated features and express stem-like genes, including CD24. CD24-positive myeloma cells represent a large fraction of residual myeloma cells after BCMA-CAR-T therapy. In this work, we develop CD24-CAR-T cells and test their ability to eliminate myeloma cells. We find that CD24-CAR-T cells block the CD24-Siglec-10 pathway, thereby enhancing macrophage phagocytic clearance of myeloma cells. Additionally, CD24-CAR-T cells polarize macrophages to a M1-like phenotype. A dual-targeted BCMA-CD24-CAR-T exhibits improved efficacy compared to monospecific BCMA-CAR-T-cell therapy. This work presents an immunotherapeutic approach that targets myeloma cells and promotes tumor cell clearance by macrophages.


Asunto(s)
Mieloma Múltiple , Receptores Quiméricos de Antígenos , Humanos , Mieloma Múltiple/patología , Linfocitos T , Antígeno de Maduración de Linfocitos B/genética , Recurrencia Local de Neoplasia , Anticuerpos , Antígeno CD24
3.
J Immunol ; 212(1): 1-2, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-38118105
4.
PLoS Pathog ; 19(10): e1011720, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37824591

RESUMEN

Sepsis, an amplified immune response to systemic infection, is characterized by a transient cytokine storm followed by chronic immune dysfunction. Consequently, sepsis survivors are highly susceptible to newly introduced infections, suggesting sepsis can influence the function and composition of the naïve CD8 T cell pool and resulting pathogen-induced primary CD8 T cell responses. Here, we explored the extent to which sepsis induces phenotypic and functional changes within the naïve CD8 T cell pool. To interrogate this, the cecal ligation and puncture (CLP) mouse model of polymicrobial sepsis was used. In normal, non-septic mice, we show type-I interferon (IFN I)-mediated signaling plays an important role in driving the phenotypic and functional heterogeneity in the naïve CD8 T cell compartment leading to increased representation of Ly6C+ naïve CD8 T cells. In response to viral infection after sepsis resolution, naïve Ly6C+ CD8 T cells generated more primary effector and memory CD8 T cells with slower conversion to a central memory CD8 T cell phenotype (Tcm) than Ly6C- naïve CD8 T cells. Importantly, as a potent inducer of cytokine storm and IFN I production, sepsis leads to increased representation of Ly6C+ naïve CD8 T cells that maintained their heightened ability to respond (i.e., effector and memory CD8 T cell accumulation and cytokine production) to primary LCMV infection. Lastly, longitudinal analyses of peripheral blood samples obtained from septic patients revealed profound changes in CD8 T cell subset composition and frequency compared to healthy controls. Thus, sepsis has the capacity to alter the composition of naïve CD8 T cells, directly influencing primary CD8 T cell responses to newly introduced infections.


Asunto(s)
Síndrome de Liberación de Citoquinas , Sepsis , Humanos , Ratones , Animales , Linfocitos T CD8-positivos , Inmunidad Innata , Fenotipo , Ratones Endogámicos C57BL , Memoria Inmunológica
5.
Viruses ; 15(6)2023 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-37376652

RESUMEN

Macrophages are critical in the pathogenesis of a diverse group of viral pathogens, both as targets of infection and for eliciting primary defense mechanisms. Our prior in vitro work identified that CD40 signaling in murine peritoneal macrophages protects against several RNA viruses by eliciting IL-12, which stimulates the production of interferon gamma (IFN-γ). Here, we examine the role of CD40 signaling in vivo. We show that CD40 signaling is a critical, but currently poorly appreciated, component of the innate immune response using two distinct infectious agents: mouse-adapted influenza A virus (IAV, PR8) and recombinant VSV encoding the Ebola virus glycoprotein (rVSV-EBOV GP). We find that stimulation of CD40 signaling decreases early IAV titers, whereas loss of CD40 elevated early titers and compromised lung function by day 3 of infection. Protection conferred by CD40 signaling against IAV is dependent on IFN-γ production, consistent with our in vitro studies. Using rVSV-EBOV GP that serves as a low-biocontainment model of filovirus infection, we demonstrate that macrophages are a CD40-expressing population critical for protection within the peritoneum and T-cells are the key source of CD40L (CD154). These experiments reveal the in vivo mechanisms by which CD40 signaling in macrophages regulates the early host responses to RNA virus infection and highlight how CD40 agonists currently under investigation for clinical use may function as a novel class of broad antiviral treatments.


Asunto(s)
Antígenos CD40 , Infecciones por Virus ARN , Virus ARN , Animales , Ratones , Antígenos CD40/metabolismo , Interferón gamma , Macrófagos , Infecciones por Virus ARN/inmunología
6.
Front Immunol ; 14: 1129251, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36814922

RESUMEN

Tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3) is an adapter protein with many context-specific functions. Early studies of lymphocyte TRAF3 hinted at TRAF3's importance for T cell function, but elucidation of specific mechanisms was delayed by early lethality of globally TRAF3-/- mice. Development of a conditional TRAF3-deficient mouse enabled important descriptive and mechanistic insights into how TRAF3 promotes optimal T cell function. Signaling through the T cell antigen receptor (TCR) fails to induce normal proliferation and survival in TRAF3 -/- T cells, and TCR-activated cells in vitro and in vivo have deficient cytokine production. These defects can be traced to incorrect localization and function of negative regulatory phosphatases acting at different parts of the signaling cascade, as can dysregulated effector responses and memory T cell homeostasis in vivo and an enlarged regulatory T cell (Treg) compartment. The important regulatory activity of TRAF3 is also evident at members of the TNFR superfamily and cytokine receptors. Here, we review significant advances in mechanistic understanding of how TRAF3 regulates T cell differentiation and function, through modulation of signaling through the TCR, costimulatory receptors, and cytokine receptors. Finally, we briefly discuss the recent identification of families carrying single allele loss-of-function mutations in TRAF3, and compare the findings in their T cells with the T cell defects identified in mice whose T cells completely lack T cell TRAF3. Together, the body of work describing TRAF3-mediated regulation of T cell effector function and differentiation frame TRAF3 as an important modulator of T cell signal integration.


Asunto(s)
Transducción de Señal , Linfocitos T Reguladores , Factor 3 Asociado a Receptor de TNF , Animales , Ratones , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Citocinas/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo
7.
Cancers (Basel) ; 14(20)2022 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-36291813

RESUMEN

TNF receptor-associated factor 3 (TRAF3) is an adapter protein that inhibits many signals that promote B cell survival and activation. Mice with a B cell-specific TRAF3 deficiency and humans with a rare haploinsufficiency in TRAF3 have enhanced development of BCLs as they age. Loss-of-function mutations in TRAF3 are common in B cell malignancies. Recent studies show that pharmacological inhibition of the enzyme glycogen synthase kinase 3 (GSK3), which regulates cellular growth, survival, and metabolism, inhibits growth and survival of BCL-derived B cells. In this study, we found that TRAF3 and GSK3 associated in B cells. The relative levels of TRAF3 in BCL cell lines correlated positively with the ratio of inactive to total GSK3ß, and negatively correlated with susceptibility to GSK3 inhibition by the GSK3 inhibitory drug 9-ING-41, currently in clinical trials. Uniquely in BCLs with low TRAF3, GSK3 inhibition caused increased loss of the TRAF3-regulated, anti-apoptotic protein Mcl-1. GSK3 inhibition also blocked hyperresponsiveness to IL-6 receptor signaling in TRAF3-deficient BCL cells. Together, these results support the utility of 9-ING-41 as a treatment for BCL, and suggest that a decrease or loss of TRAF3 in BCLs could act as a biomarker for increased susceptibility to GSK3 inhibitor treatment.

8.
Sci Signal ; 15(753): eabn5507, 2022 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-36166512

RESUMEN

Type I interferons (IFNs) are among the most powerful tools that host cells deploy against intracellular pathogens. Their effectiveness is due both to the rapid, directly antiviral effects of IFN-stimulated gene products and to the effects of type I IFN on responding immune cells. Type I IFN signaling through its receptor, IFNAR, is tightly regulated at multiple steps in the signaling cascade, including at the level of IFNAR downstream effectors, which include the kinase JAK1 and the transcriptional regulator STAT1. Here, we found that tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3) enhanced the activation of JAK1 and STAT1 specifically in CD4+ T cells by preventing recruitment of the negative regulatory phosphatase PTPN22 to the IFNAR complex. The balance between signals through IFNAR and other cytokine receptors influences CD4+ T cell differentiation and function during infections. Our work reveals TRAF3 and PTPN22 as key regulators of CD4+ T cell activation by type I IFNs.


Asunto(s)
Interferón Tipo I , Factor 3 Asociado a Receptor de TNF , Antivirales/metabolismo , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Receptor de Interferón alfa y beta/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Linfocitos T/metabolismo , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/metabolismo
9.
Sci Immunol ; 7(74): eabn3800, 2022 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-35960817

RESUMEN

Tumor necrosis factor receptor-associated factor 3 (TRAF3) is a central regulator of immunity. TRAF3 is often somatically mutated in B cell malignancies, but its role in human immunity is not defined. Here, in five unrelated families, we describe an immune dysregulation syndrome of recurrent bacterial infections, autoimmunity, systemic inflammation, B cell lymphoproliferation, and hypergammaglobulinemia. Affected individuals each had monoallelic mutations in TRAF3 that reduced TRAF3 expression. Immunophenotyping showed that patients' B cells were dysregulated, exhibiting increased nuclear factor-κB 2 activation, elevated mitochondrial respiration, and heightened inflammatory responses. Patients had mild CD4+ T cell lymphopenia, with a reduced proportion of naïve T cells but increased regulatory T cells and circulating T follicular helper cells. Guided by this clinical phenotype, targeted analyses demonstrated that common genetic variants, which also reduce TRAF3 expression, are associated with an increased risk of B cell malignancies, systemic lupus erythematosus, higher immunoglobulin levels, and bacterial infections in the wider population. Reduced TRAF3 conveys disease risks by driving B cell hyperactivity via intrinsic activation of multiple intracellular proinflammatory pathways and increased mitochondrial respiration, with a likely contribution from dysregulated T cell help. Thus, we define monogenic TRAF3 haploinsufficiency syndrome and demonstrate how common TRAF3 variants affect a range of human diseases.


Asunto(s)
Neoplasias , Factor 3 Asociado a Receptor de TNF , Autoinmunidad/genética , Linfocitos B , Humanos , Mutación , Neoplasias/patología , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/metabolismo
10.
Cells ; 10(8)2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34440839

RESUMEN

Uncontrolled inflammation is associated with neurodegenerative conditions in central nervous system tissues, including the retina and brain. We previously found that the neural retina (NR) plays an important role in retinal immunity. Tumor necrosis factor Receptor-Associated Factor 3 (TRAF3) is a known immune regulator expressed in the retina; however, whether TRAF3 regulates retinal immunity is unknown. We have generated the first conditional NR-Traf3 knockout mouse model (Chx10-Cre/Traf3f/f) to enable studies of neuronal TRAF3 function. Here, we evaluated NR-Traf3 depletion effects on whole retinal TRAF3 protein expression, visual acuity, and retinal structure and function. Additionally, to determine if NR-Traf3 plays a role in retinal immune regulation, we used flow cytometry to assess immune cell infiltration following acute local lipopolysaccharide (LPS) administration. Our results show that TRAF3 protein is highly expressed in the NR and establish that NR-Traf3 depletion does not affect basal retinal structure or function. Importantly, NR-Traf3 promoted LPS-stimulated retinal immune infiltration. Thus, our findings propose NR-Traf3 as a positive regulator of retinal immunity. Further, the NR-Traf3 mouse provides a tool for investigations of neuronal TRAF3 as a novel potential target for therapeutic interventions aimed at suppressing retinal inflammatory disease and may also inform treatment approaches for inflammatory neurodegenerative brain conditions.


Asunto(s)
Proteínas de Homeodominio/genética , Neuronas/metabolismo , Retina/metabolismo , Factor 3 Asociado a Receptor de TNF/genética , Factores de Transcripción/genética , Animales , Modelos Animales de Enfermedad , Electrorretinografía , Inmunidad/efectos de los fármacos , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Neuronas/inmunología , Receptores CCR2/genética , Receptores CCR2/metabolismo , Retina/fisiología , Factor 3 Asociado a Receptor de TNF/deficiencia , Factor 3 Asociado a Receptor de TNF/metabolismo , Factores de Transcripción/deficiencia , Uveítis/etiología , Uveítis/inmunología , Uveítis/metabolismo , Agudeza Visual
11.
J Biol Chem ; 297(3): 101097, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34418432

RESUMEN

Tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3) plays context-specific roles in multiple receptor-mediated signaling pathways in different cell types. Mice lacking TRAF3 in T cells display defective T-cell-mediated immune responses to immunization and infection and demonstrate defective early signaling via the TCR complex. However, the role of TRAF3 in the function of GITR/TNFRSF18, an important costimulatory member of the TNFR superfamily, is unclear. Here we investigated the impact of T cell TRAF3 status on both GITR expression and activation of specific kinases in the GITR signaling pathway in T cells. Our results indicate that TRAF3 negatively regulates GITR functions in several ways. First, expression of GITR protein was elevated in TRAF3-deficient T cells, resulting from both transcriptional and posttranslational regulation that led to greater GITR transcript levels, as well as enhanced GITR protein stability. TRAF3 associated with T cell GITR in a manner dependent upon GITR ligation. TRAF3 also inhibited several events of the GITR mediated early signaling cascade, in a manner independent of recruitment of phosphatases, a mechanism by which TRAF3 inhibits signaling through several other cytokine receptors. These results add new information to our understanding of GITR signaling and function in T cells, which is relevant to the potential use of GITR to enhance immune therapies.


Asunto(s)
Receptores Coestimuladores e Inhibidores de Linfocitos T/metabolismo , Proteína Relacionada con TNFR Inducida por Glucocorticoide/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Animales , Femenino , Proteína Relacionada con TNFR Inducida por Glucocorticoide/fisiología , Interleucina-2/metabolismo , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/inmunología , Linfocitos T/metabolismo , Factor 3 Asociado a Receptor de TNF/fisiología
12.
J Immunol ; 207(1): 322-332, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34145060

RESUMEN

The adaptor protein TNFR-associated factor 3 (TRAF3) is required for in vivo T cell effector functions and for normal TCR/CD28 signaling. TRAF3-mediated enhancement of TCR function requires engagement of both CD3 and CD28, but the molecular mechanisms underlying how TRAF3 interacts with and impacts TCR/CD28-mediated complexes to enhance their signaling remains an important knowledge gap. We investigated how TRAF3 is recruited to, and regulates, CD28 as a TCR costimulator. Direct association with known signaling motifs in CD28 was dispensable for TRAF3 recruitment; rather, TRAF3 associated with the CD28-interacting protein linker of activated T cells (LAT) in human and mouse T cells. TRAF3-LAT association required the TRAF3 TRAF-C domain and a newly identified TRAF2/3 binding motif in LAT. TRAF3 inhibited function of the LAT-associated negative regulatory protein Dok1, which is phosphorylated at an inhibitory tyrosine residue by the tyrosine kinase breast tumor kinase (Brk/PTK6). TRAF3 regulated Brk activation in T cells, limiting the association of protein tyrosine phosphatase 1B (PTP1B) with the LAT complex. In TRAF3-deficient cells, LAT complex-associated PTP1B was associated with dephosphorylation of Brk at an activating tyrosine residue, potentially reducing its ability to inhibit Dok1. Consistent with these findings, inhibiting PTP1B activity in TRAF3-deficient T cells rescued basal and TCR/CD28-mediated activation of Src family kinases. These results reveal a new mechanism for promotion of TCR/CD28-mediated signaling through restraint of negative regulation of LAT by TRAF3, enhancing the understanding of regulation of the TCR complex.


Asunto(s)
Antígenos CD28/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Factor 3 Asociado a Receptor de TNF/inmunología , Animales , Células Cultivadas , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal/inmunología , Factor 3 Asociado a Receptor de TNF/deficiencia , Factor 3 Asociado a Receptor de TNF/genética
13.
Cancer Immunol Immunother ; 70(11): 3093-3103, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33765210

RESUMEN

Cancer vaccines that utilize patient antigen-presenting cells to fight their own tumors have shown exciting promise in many preclinical studies, but have proven quite challenging to translate to clinical feasibility. Dendritic cells have typically been the cell of choice for such vaccine platforms, due to their ability to endocytose antigens nonspecifically, and their expression of multiple surface molecules that enhance antigen presentation. However, dendritic cells are present in low numbers in human peripheral blood and must be matured in culture before use in vaccines. Mature B lymphocytes, in contrast, are relatively abundant in peripheral blood, and can be quickly activated and expanded in overnight cultures. We devised an optimal stimulation cocktail that engages the B cell antigen receptor, CD40, TLR4 and TLR7, to activate B cells to present antigens from lysates of the recipient's tumor cells, precluding the need for known tumor antigens. This B cell vaccine (Bvac) improved overall survival from B16F1 melanoma challenge, as well as reduced tumor size and increased time to tumor appearance. Bvac upregulated B cell antigen presentation molecules, stimulated activation of both CD4+ and CD8+ T cells, and induced T cell migration. Bvac provides an alternative cellular vaccine strategy that has considerable practical advantages for translation to clinical settings.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos B/inmunología , Vacunas contra el Cáncer/inmunología , Melanoma Experimental/inmunología , Neoplasias Cutáneas/inmunología , Animales , Presentación de Antígeno/inmunología , Vacunas contra el Cáncer/farmacología , Quimiotaxis de Leucocito/inmunología , Femenino , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T/inmunología
14.
J Biol Chem ; 296: 100465, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33639170

RESUMEN

TRAF3 has diverse signaling functions, which vary by cell type. Uniquely in B lymphocytes, TRAF3 inhibits homeostatic survival. Highlighting the role of TRAF3 as a tumor suppressor, loss-of-function TRAF3 mutations are associated with human B-cell malignancies, while B-cell-specific deletion of TRAF3 in mice leads to autoimmunity and lymphoma development. The role of TRAF3 in inhibiting noncanonical NF-κB activation, CD40 and BAFF-R signaling to B cells is well documented. In contrast, TRAF3 enhances many T-cell effector functions, through associating with and enhancing signaling by the T-cell receptor (TCR)-CD28 complex. The present study was designed to determine the role of TRAF3 in signaling via the B-cell antigen receptor (BCR). The BCR is crucial for antigen recognition, survival, proliferation, and antibody production, and defects in BCR signaling can promote abnormal survival of malignant B cells. Here, we show that TRAF3 is associated with both CD79B and the BCR-activated kinases Syk and Btk following BCR stimulation. BCR-induced phosphorylation of Syk and additional downstream kinases was increased in TRAF3-/- B cells, with regulation observed in both follicular and marginal zone B-cell subsets. BCR stimulation of TRAF3-/- B cells resulted in increased surface expression of MHC-II, CD80, and CD86 molecules. Interestingly, increased survival of TRAF3-/- primary B cells was resistant to inhibition of Btk, while TRAF3-deficient malignant B-cell lines showed enhanced sensitivity. TRAF3 serves to restrain normal and malignant BCR signaling, with important implications for its role in normal B-cell biology and abnormal survival of malignant B cells.


Asunto(s)
Receptores de Antígenos de Linfocitos B/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Agammaglobulinemia Tirosina Quinasa/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Antígenos CD79/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Receptores de Antígenos de Linfocitos B/genética , Transducción de Señal/genética , Quinasa Syk/metabolismo , Linfocitos T/metabolismo , Factor 2 Asociado a Receptor de TNF/metabolismo , Factor 3 Asociado a Receptor de TNF/genética
15.
Front Immunol ; 12: 745873, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35046933

RESUMEN

B cells can act as potent suppressors of anti-tumor T cell immunity, presenting a mechanism of resistance to immunotherapy. In pancreatic ductal adenocarcinoma, B cells can display a T cell-suppressive or regulatory phenotype centered on the expression of the cytokine Interleukin 35 (IL-35). While B cell-mediated immunosuppression presents a barrier to anti-tumorigenic T cell function, it is not clear how regulatory B cell function could be targeted, and the signals that promote this suppressive phenotype in B cells are not well understood. Here we use a novel IL-35 reporter model to understand which signaling pathways are important for immunosuppressive properties in B cells. In vitro analysis of IL-35 reporter B cells revealed a synergy between the BCR and TLR4 signaling pathways is sufficient to induce IL-35 expression. However, in vivo, B cell receptor activation, as opposed to MyD88 signaling in B cells, is central to B cell-mediated suppression and promotion of pancreatic cancer growth. Further analysis identified protein kinase D2 (PKD2) as being a key downstream regulator of IL-35 expression in B cells. Regulatory B cells with an inactivating mutation in PKD2 failed to produce IL-35 or fully suppress effector T cell function in vitro. Furthermore, inhibition of PKD in B cells decreased tumor growth and promoted effector T cell function upon adoptive transfer into B cell-deficient mice. Collectively, these data provide insight into how regulatory B cell function is promoted in pancreatic cancer and identify potential therapeutic targets to restrain this function.


Asunto(s)
Linfocitos B Reguladores/inmunología , Carcinoma Ductal Pancreático/inmunología , Neoplasias Pancreáticas/inmunología , Proteína Quinasa D2/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Animales , Interleucinas/biosíntesis , Interleucinas/inmunología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología
16.
J Leukoc Biol ; 109(2): 309-325, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32441445

RESUMEN

Many acute viral infections target tissue Mϕs, yet the mechanisms of Mϕ-mediated control of viruses are poorly understood. Here, we report that CD40 expressed by peritoneal Mϕs restricts early infection of a broad range of RNA viruses. Loss of CD40 expression enhanced virus replication as early as 12-24 h of infection and, conversely, stimulation of CD40 signaling with an agonistic Ab blocked infection. With peritoneal cell populations infected with the filovirus, wild-type (WT) Ebola virus (EBOV), or a BSL2 model virus, recombinant vesicular stomatitis virus encoding Ebola virus glycoprotein (rVSV/EBOV GP), we examined the mechanism conferring protection. Here, we demonstrate that restricted virus replication in Mϕs required CD154/CD40 interactions that stimulated IL-12 production through TRAF6-dependent signaling. In turn, IL-12 production resulted in IFN-γ production, which induced proinflammatory polarization of Mϕs, protecting the cells from infection. These CD40-dependent events protected mice against virus challenge. CD40-/- mice were exquisitely sensitive to intraperitoneal challenge with a dose of rVSV/EBOV GP that was sublethal to CD40+/+ mice, exhibiting viremia within 12 h of infection and rapidly succumbing to infection. This study identifies a previously unappreciated role for Mϕ-intrinsic CD40 signaling in controlling acute virus infection.


Asunto(s)
Antígenos CD40/metabolismo , Inmunidad Innata , Macrófagos/inmunología , Macrófagos/virología , Virus ARN/fisiología , Transducción de Señal , Virosis/inmunología , Replicación Viral/fisiología , Enfermedad Aguda , Animales , Ligando de CD40/metabolismo , Ebolavirus/fisiología , Glicoproteínas/inmunología , Humanos , Interferón gamma/metabolismo , Interleucina-12/biosíntesis , Ratones Endogámicos C57BL , Modelos Biológicos , Peritoneo/patología , Peritoneo/virología , Factor 6 Asociado a Receptor de TNF/metabolismo , Virosis/virología
17.
Front Immunol ; 11: 606936, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33613527

RESUMEN

Systemic lupus erythematosus (SLE) and other autoimmune diseases are propelled by immune dysregulation and pathogenic, disease-specific autoantibodies. Autoimmunity against the lupus autoantigen Sm is associated with cross-reactivity to Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA-1). Additionally, EBV latent membrane protein-1 (LMP1), initially noted for its oncogenic activity, is an aberrantly active functional mimic of the B cell co-stimulatory molecule CD40. Mice expressing a transgene (Tg) for the mCD40-LMP1 hybrid molecule (containing the cytoplasmic tail of LMP1) have mild autoantibody production and other features of immune dysregulation by 2-3 months of age, but no overt autoimmune disease. This study evaluates whether exposure to the EBV molecular mimic, EBNA-1, stimulates antigen-specific and concurrently-reactive humoral and cellular immunity, as well as lupus-like features. After immunization with EBNA-1, mCD40-LMP1 Tg mice exhibited enhanced, antigen-specific, cellular and humoral responses compared to immunized WT congenic mice. EBNA-1 specific proliferative and inflammatory cytokine responses, including IL-17 and IFN-γ, were significantly increased (p<0.0001) in mCD40-LMP1 Tg mice, as well as antibody responses to amino- and carboxy-domains of EBNA-1. Of particular interest was the ability of mCD40-LMP1 to drive EBNA-1 associated molecular mimicry with the lupus-associated autoantigen, Sm. EBNA-1 immunized mCD40-LMP1 Tg mice exhibited enhanced proliferative and cytokine cellular responses (p<0.0001) to the EBNA-1 homologous epitope PPPGRRP and the Sm B/B' cross-reactive sequence PPPGMRPP. When immunized with the SLE autoantigen Sm, mCD40-LMP1 Tg mice again exhibited enhanced cellular and humoral immune responses to both Sm and EBNA-1. Cellular immune dysregulation with EBNA-1 immunization in mCD40-LMP1 Tg mice was accompanied by enhanced splenomegaly, increased serum blood urea nitrogen (BUN) and creatinine levels, and elevated anti-dsDNA and antinuclear antibody (ANA) levels (p<0.0001 compared to mCD40 WT mice). However, no evidence of immune-complex glomerulonephritis pathology was noted, suggesting that a combination of EBV and genetic factors may be required to drive lupus-associated renal disease. These data support that the expression of LMP1 in the context of EBNA-1 may interact to increase immune dysregulation that leads to pathogenic, autoantigen-specific lupus inflammation.


Asunto(s)
Autoantígenos/inmunología , Autoinmunidad , Antígenos Nucleares del Virus de Epstein-Barr/inmunología , Inmunidad Celular , Inmunidad Humoral , Lupus Eritematoso Sistémico/inmunología , Imitación Molecular , Proteínas de la Matriz Viral/inmunología , Proteínas Nucleares snRNP/inmunología , Animales , Anticuerpos Antinucleares/sangre , Autoantígenos/administración & dosificación , Antígenos CD40/genética , Antígenos CD40/inmunología , Antígenos CD40/metabolismo , Reacciones Cruzadas , Epítopos , Antígenos Nucleares del Virus de Epstein-Barr/administración & dosificación , Inmunización , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Proteínas Nucleares snRNP/administración & dosificación
18.
J Natl Cancer Inst ; 112(5): 507-515, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31406992

RESUMEN

BACKGROUND: Treatment failures in cancers, including multiple myeloma (MM), are most likely due to the persistence of a minor population of tumor-initiating cells (TICs), which are noncycling or slowly cycling and very drug resistant. METHODS: Gene expression profiling and real-time quantitative reverse transcription polymerase chain reaction were employed to define genes differentially expressed between the side-population cells, which contain the TICs, and the main population of MM cells derived from 11 MM patient samples. Self-renewal potential was analyzed by clonogenicity and drug resistance of CD24+ MM cells. Flow cytometry (n = 60) and immunofluorescence (n = 66) were applied on MM patient samples to determine CD24 expression. Therapeutic effects of CD24 antibodies were tested in xenograft MM mouse models containing three to six mice per group. RESULTS: CD24 was highly expressed in the side-population cells, and CD24+ MM cells exhibited high expression of induced pluripotent or embryonic stem cell genes. CD24+ MM cells showed increased clonogenicity, drug resistance, and tumorigenicity. Only 10 CD24+ MM cells were required to develop plasmacytomas in mice (n = three of five mice after 27 days). The frequency of CD24+ MM cells was highly variable in primary MM samples, but the average of CD24+ MM cells was 8.3% after chemotherapy and in complete-remission MM samples with persistent minimal residual disease compared with 1.0% CD24+ MM cells in newly diagnosed MM samples (n = 26). MM patients with a high initial percentage of CD24+ MM cells had inferior progression-free survival (hazard ratio [HR] = 3.81, 95% confidence interval [CI] = 5.66 to 18.34, P < .001) and overall survival (HR = 3.87, 95% CI = 16.61 to 34.39, P = .002). A CD24 antibody inhibited MM cell growth and prevented tumor progression in vivo. CONCLUSION: Our studies demonstrate that CD24+ MM cells maintain the TIC features of self-renewal and drug resistance and provide a target for myeloma therapy.


Asunto(s)
Mieloma Múltiple/patología , Células Madre Neoplásicas/patología , Animales , Antígeno CD24/biosíntesis , Antígeno CD24/inmunología , Carcinogénesis , Autorrenovación de las Células/fisiología , Resistencia a Antineoplásicos , Femenino , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/inmunología , Células Madre Neoplásicas/inmunología
19.
J Leukoc Biol ; 107(6): 907-915, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31749173

RESUMEN

The TNFR superfamily of receptors, the major focus of the recent TNFR Superfamily Conference held in June 2019, employ the TNFR-associated factor (TRAF) family of adaptor proteins in key aspects of their signaling pathways. Although many early studies investigated TRAF functions via exogenous overexpression in nonhematopoietic cell lines, it has subsequently become clear that whereas TRAFs share some overlap in function, each also plays unique biologic roles, that can be highly context dependent. This brief review summarizes the current state of knowledge of functions of each of the TRAF molecules that mediate important functions in T lymphocytes: TRAFs 1, 2, 3, 5, and 6. Due to our current appreciation of the contextual nature of TRAF function, our focus is upon findings made specifically in T lymphocytes. Key T cell functions for each TRAF are detailed, as well as future knowledge gaps of interest and importance.


Asunto(s)
Síndromes de Inmunodeficiencia/genética , Receptores del Factor de Necrosis Tumoral/genética , Transducción de Señal/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Factor de Necrosis Tumoral alfa/genética , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/patología , Memoria Inmunológica , Ratones , Ratones Noqueados , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Transducción de Señal/inmunología , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/deficiencia , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/inmunología , Factor de Necrosis Tumoral alfa/inmunología
20.
Sci Rep ; 9(1): 17502, 2019 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-31745166

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...