Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Immunol ; 208(4): 898-909, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35039332

ABSTRACT

Type 1 diabetes is an autoimmune disease characterized by pancreatic ß cell destruction. It is a complex genetic trait driven by >30 genetic loci with parallels between humans and mice. The NOD mouse spontaneously develops autoimmune diabetes and is widely used to identify insulin-dependent diabetes (Idd) genetic loci linked to diabetes susceptibility. Although many Idd loci have been extensively studied, the impact of the Idd2 locus on autoimmune diabetes susceptibility remains to be defined. To address this, we generated a NOD congenic mouse bearing B10 resistance alleles on chromosome 9 in a locus coinciding with part of the Idd2 locus and found that NOD.B10-Idd2 congenic mice are highly resistant to diabetes. Bone marrow chimera and adoptive transfer experiments showed that the B10 protective alleles provide resistance in an immune cell-intrinsic manner. Although no T cell-intrinsic differences between NOD and NOD.B10-Idd2 mice were observed, we found that the Idd2 resistance alleles limit the formation of spontaneous and induced germinal centers. Comparison of B cell and dendritic cell transcriptome profiles from NOD and NOD.B10-Idd2 mice reveal that resistance alleles at the Idd2 locus affect the expression of specific MHC molecules, a result confirmed by flow cytometry. Altogether, these data demonstrate that resistance alleles at the Idd2 locus impair germinal center formation and influence MHC expression, both of which likely contribute to reduced diabetes incidence.


Subject(s)
Autoimmunity , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/metabolism , Genetic Loci , Genetic Predisposition to Disease , Major Histocompatibility Complex/genetics , Alleles , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Diabetes Mellitus, Type 1/diagnosis , Disease Models, Animal , Disease Resistance/genetics , Genetic Variation , Glucose Tolerance Test , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Mice , Mice, Congenic , Mice, Inbred NOD , Mice, Knockout , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
2.
J Immunol ; 207(4): 1055-1064, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34312259

ABSTRACT

Central tolerance aims to limit the production of T lymphocytes bearing TCR with high affinity for self-peptide presented by MHC molecules. The accumulation of thymocytes with such receptors is limited by negative selection or by diversion into alternative differentiation, including T regulatory cell commitment. A role for the orphan nuclear receptor NR4A3 in negative selection has been suggested, but its function in this process has never been investigated. We find that Nr4a3 transcription is upregulated in postselection double-positive thymocytes, particularly those that have received a strong selecting signal and are destined for negative selection. Indeed, we found an accumulation of cells bearing a negative selection phenotype in NR4A3-deficient mice as compared with wild-type controls, suggesting that Nr4a3 transcriptional induction is necessary to limit accumulation of self-reactive thymocytes. This is consistent with a decrease of cleaved caspase-3+-signaled thymocytes and more T regulatory and CD4+Foxp3-HELIOS+ cells in the NR4A3-deficient thymus. We further tested the role for NR4A3 in negative selection by reconstituting transgenic mice expressing the OVA Ag under the control of the insulin promoter with bone marrow cells from OT-I Nr4a3 +/+ or OT-I Nr4a3 -/- mice. Accumulation of autoreactive CD8 thymocytes and autoimmune diabetes developed only in the absence of NR4A3. Overall, our results demonstrate an important role for NR4A3 in T cell development.


Subject(s)
Diabetes Mellitus, Type 1 , Receptors, Steroid , Animals , DNA-Binding Proteins , Mice , Mice, Transgenic , Nerve Tissue Proteins , Receptors, Thyroid Hormone , Thymocytes , Transcription Factors
3.
Methods Mol Biol ; 2130: 29-51, 2021.
Article in English | MEDLINE | ID: mdl-33284434

ABSTRACT

In recent years, circadian rhythms have been observed in many aspects of the immune system, both for the innate immunity (the first line of defense against pathogens) and the adaptive immunity (a more specific set of responses, which lead to immune memory). Here, to illustrate principles to be taken into account when working on circadian rhythms in immunology experiments, two protocols will be presented. The first one aims to analyze immune parameters in blood sampled from human subjects at different times over the day: counts of different cell types among the peripheral blood mononuclear cells and cytokine secretion by monocytes and T cells after ex vivo stimulation. The second protocol describes how to follow the response of CD8+ T cells after immunization of mice with antigen presenting cells loaded with a peptide antigen. These two protocols are optimized for circadian experiments, and outcome measures are mainly based on flow cytometry, which allows analysis of different parameters in the same cells.


Subject(s)
Circadian Rhythm , Flow Cytometry/methods , Leukocytes, Mononuclear/immunology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Humans , Immunoassay/methods , Mice
4.
Proc Natl Acad Sci U S A ; 117(39): 24392-24402, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32913051

ABSTRACT

Enhancing long-term persistence while simultaneously potentiating the effector response of CD8+ T cells has been a long-standing goal in immunology to produce better vaccines and adoptive cell therapy products. NR4A3 is a transcription factor of the orphan nuclear receptor family. While it is rapidly and transiently expressed following T cell activation, its role in the early stages of T cell response is unknown. We show that NR4A3-deficient murine CD8+ T cells differentiate preferentially into memory precursor and central memory cells, but also produce more cytokines. This is explained by an early influence of NR4A3 deficiency on the memory transcriptional program and on accessibility of chromatin regions with motifs for bZIP transcription factors, which impacts the transcription of Fos/Jun target genes. Our results reveal a unique and early role for NR4A3 in programming CD8+ T cell differentiation and function. Manipulating NR4A3 activity may represent a promising strategy to improve vaccination and T cell therapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/immunology , Nerve Tissue Proteins/immunology , Receptors, Steroid/immunology , Receptors, Thyroid Hormone/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , DNA-Binding Proteins/genetics , Gene Expression Regulation , Immunologic Memory , Mice , Nerve Tissue Proteins/genetics , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Transcription Factors/genetics , Transcription Factors/immunology
5.
Cell Rep ; 30(4): 1129-1140.e5, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31995754

ABSTRACT

Plasma membrane damage and cell death during processes such as necroptosis and apoptosis result from cues originating intracellularly. However, death caused by pore-forming agents, like bacterial toxins or complement, is due to direct external injury to the plasma membrane. To prevent death, the plasma membrane has an intrinsic repair ability. Here, we found that repair triggered by pore-forming agents involved TMEM16F, a calcium-activated lipid scramblase also mutated in Scott's syndrome. Upon pore formation and the subsequent influx of intracellular calcium, TMEM16F induced rapid "lipid scrambling" in the plasma membrane. This response was accompanied by membrane blebbing, extracellular vesicle release, preserved membrane integrity, and increased cell viability. TMEM16F-deficient mice exhibited compromised control of infection by Listeria monocytogenes associated with a greater sensitivity of neutrophils to the pore-forming Listeria toxin listeriolysin O (LLO). Thus, the lipid scramblase TMEM16F is critical for plasma membrane repair after injury by pore-forming agents.


Subject(s)
Anoctamins/metabolism , Bacterial Toxins/toxicity , Cell Membrane/metabolism , Extracellular Vesicles/metabolism , Heat-Shock Proteins/toxicity , Hemolysin Proteins/toxicity , Phosphatidylserines/metabolism , Phospholipid Transfer Proteins/metabolism , Thymocytes/metabolism , Animals , Anoctamins/genetics , Calcium/metabolism , Cell Death/drug effects , Cell Death/genetics , Cell Membrane/drug effects , Extracellular Vesicles/drug effects , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Liver/cytology , Liver/metabolism , Liver/microbiology , Liver/pathology , Membrane Lipids/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/microbiology , Neutrophils/pathology , Phospholipid Transfer Proteins/genetics , Spleen/cytology , Spleen/metabolism , Spleen/microbiology , Spleen/pathology , Thymocytes/drug effects , Thymocytes/ultrastructure
6.
PLoS One ; 14(8): e0221528, 2019.
Article in English | MEDLINE | ID: mdl-31437241

ABSTRACT

Expressed strongly by myeloid cells, damage-associated molecular pattern (DAMP) proteins S100A8 and S100A9 are found in the serum of patients with infectious and autoimmune diseases. Compared to S100A9, the role of S100A8 is controversial. We investigated its biological activity in collagen-induced arthritis using the first known viable and fertile S100a8-deficient (S100a8-/-) mouse. Although comparable to the wild type (WT) in terms of lymphocyte distribution in blood and in the primary and secondary lymphoid organs, S100a8-/- mice had increased numbers of neutrophils, monocytes and dendritic cells in the blood and bone marrow, and these all expressed myeloid markers such as CD11b, Ly6G and CD86 more strongly. Granulocyte-macrophage common precursors were increased in S100a8-/- bone marrow and yielded greater numbers of macrophages and dendritic cells in culture. The animals also developed more severe arthritic disease leading to aggravated osteoclast activity and bone destruction. These findings were correlated with increased inflammatory cell infiltration and cytokine secretion in the paws. This study suggests that S100A8 is an anti-inflammatory DAMP that regulates myeloid cell differentiation, thereby mitigating the development of experimental arthritis.


Subject(s)
Arthritis, Experimental/pathology , Calgranulin A/deficiency , Myelopoiesis , Animals , Arthritis, Experimental/diagnostic imaging , Bone Marrow/pathology , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Calgranulin A/metabolism , Cartilage/pathology , Cell Differentiation , Dendritic Cells/metabolism , Female , Gene Deletion , Mice , Myeloid Cells/pathology
7.
Proc Natl Acad Sci U S A ; 116(30): 15150-15159, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31285338

ABSTRACT

In response to microbial stimulation, monocytes can differentiate into macrophages or monocyte-derived dendritic cells (MoDCs) but the molecular requirements guiding these possible fates are poorly understood. In addition, the physiological importance of MoDCs in the host cellular and immune responses to microbes remains elusive. Here, we demonstrate that the nuclear orphan receptor NR4A3 is required for the proper differentiation of MoDCs but not for other types of DCs. Indeed, the generation of DC-SIGN+ MoDCs in response to LPS was severely impaired in Nr4a3-/- mice, which resulted in the inability to mount optimal CD8+ T cell responses to gram-negative bacteria. Transcriptomic analyses revealed that NR4A3 is required to skew monocyte differentiation toward MoDCs, at the expense of macrophages, and allows the acquisition of migratory characteristics required for MoDC function. Altogether, our data identify that the NR4A3 transcription factor is required to guide the fate of monocytes toward MoDCs.


Subject(s)
Cell Lineage/immunology , DNA-Binding Proteins/genetics , Dendritic Cells/immunology , Lipopolysaccharides/pharmacology , Monocytes/immunology , Nerve Tissue Proteins/genetics , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Differentiation , Cell Lineage/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/immunology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Gene Expression Profiling , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interleukin-4/pharmacology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lymphocyte Activation , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Knockout , Monocytes/cytology , Monocytes/drug effects , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/immunology , Primary Cell Culture , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Steroid/deficiency , Receptors, Steroid/immunology , Receptors, Thyroid Hormone/deficiency , Receptors, Thyroid Hormone/immunology , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
8.
PLoS One ; 14(4): e0215012, 2019.
Article in English | MEDLINE | ID: mdl-30951556

ABSTRACT

During CD8+ T cell response, Notch signaling controls short-lived-effector-cell (SLEC) generation, but the exact mechanisms by which it does so remains unclear. The Notch signaling pathway can act as a key regulator of Akt signaling via direct transcriptional induction of Hes1, which will then repress the transcription of Pten, an inhibitor of Akt signaling. As both Notch and Akt signaling can promote effector CD8+ T cell differentiation, we asked whether Notch signaling influences SLEC differentiation via the HES1-PTEN axis. Here, we demonstrate that HES1 deficiency in murine CD8+ T cells did not impact SLEC differentiation. Moreover, we show that Pten transcriptional repression in effector CD8+ T cells is not mediated by Notch signaling although Akt activation requires Notch signaling. Therefore, HES1 is not an effector of Notch signaling during CD8+ T cell response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Receptors, Notch/immunology , Signal Transduction/immunology , Transcription Factor HES-1/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , Mice , Mice, Knockout , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/immunology , Receptors, Notch/genetics , Signal Transduction/genetics , Transcription Factor HES-1/genetics
9.
Sci Immunol ; 3(21)2018 03 16.
Article in English | MEDLINE | ID: mdl-29549139

ABSTRACT

Obesity gives rise to metabolic complications by mechanisms that are poorly understood. Although chronic inflammatory signaling in adipose tissue is typically associated with metabolic deficiencies linked to excessive weight gain, we identified a subset of neuropilin-1 (NRP1)-expressing myeloid cells that accumulate in adipose tissue and protect against obesity and metabolic syndrome. Ablation of NRP1 in macrophages compromised lipid uptake in these cells, which reduced substrates for fatty acid ß-oxidation and shifted energy metabolism of these macrophages toward a more inflammatory glycolytic metabolism. Conditional deletion of NRP1 in LysM Cre-expressing cells leads to inadequate adipose vascularization, accelerated weight gain, and reduced insulin sensitivity even independent of weight gain. Transfer of NRP1+ hematopoietic cells improved glucose homeostasis, resulting in the reversal of a prediabetic phenotype. Our findings suggest a pivotal role for adipose tissue-resident NRP1+-expressing macrophages in driving healthy weight gain and maintaining glucose tolerance.


Subject(s)
Adipose Tissue/metabolism , Macrophages/metabolism , Neuropilin-1/metabolism , Animals , Metabolic Syndrome/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Obesity/metabolism
10.
J Immunol ; 198(1): 292-299, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27864474

ABSTRACT

The increasing use of plant viruses for the development of new vaccines and immunotherapy approaches poses questions regarding the mechanism by which the mammalian immune system recognizes these viruses. For example, although natural Abs (NA) and complement are key components of the innate immune system involved in the opsonization, phagocytosis, and destruction of microorganisms infecting mammals, their implication in plant virus recognition and immunogenicity is not well defined. In this study, we address the involvement of NA and the complement system in the activation of innate immunity through engagement of TLR7 with papaya mosaic virus (PapMV)-like nanoparticles. We demonstrate that NA, although binding to PapMV, are not involved in its recognition by the immune system. On the other hand, C3 strongly binds to PapMV nanoparticles and its depletion significantly reduces PapMV's interaction with immune cells. Unexpectedly, however, we observed increased immune cell activation following administration of PapMV to complement-depleted mice. TLR7 activation by PapMV in the absence of C3 induced higher IFN-α production, resulting in superior immune cell activation and increased immunotherapeutic properties. In conclusion, in this study we established the involvement of the complement system in the recognition and the phagocytosis of PapMV nanoparticles and identified an unsuspected role for C3 in regulating the production of IFN-α following TLR7 activation.


Subject(s)
Complement C3/immunology , Dendritic Cells/immunology , Interferon-gamma/biosynthesis , Membrane Glycoproteins/immunology , Mosaic Viruses/immunology , Toll-Like Receptor 7/immunology , Animals , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Nanoparticles , Phagocytosis/immunology , Polymerase Chain Reaction , Toll-Like Receptor 7/metabolism
11.
Eur J Immunol ; 47(2): 269-279, 2017 02.
Article in English | MEDLINE | ID: mdl-27873323

ABSTRACT

Vaccination with antigen-pulsed CD40-activated B (CD40-B) cells can efficiently lead to the in vivo differentiation of naive CD8+ T cells into fully functional effectors. In contrast to bone marrow-derived dendritic cell (BMDC) vaccination, CD40-B cell priming does not allow for memory CD8+ T-cell generation but the reason for this deficiency is unknown. Here, we show that compared to BMDCs, murine CD40-B cells induce lower expression of several genes regulated by T-cell receptor signaling, costimulation, and inflammation (signals 1-3) in mouse T cells. The reduced provision of signals 1 and 2 by CD40-B cells can be explained by a reduction in the quality and duration of the interactions with naive CD8+ T cells as compared to BMDCs. Furthermore, CD40-B cells produce less inflammatory mediators, such as IL-12 and type I interferon, and increasing inflammation by coadministration of polyriboinosinic-polyribocytidylic acid with CD40-B-cell immunization allowed for the generation of long-lived and functional CD8+ memory T cells. In conclusion, it is possible to manipulate CD40-B-cell vaccination to promote the formation of long-lived functional CD8+ memory T cells, a key step before translating the use of CD40-B cells for therapeutic vaccination.


Subject(s)
B-Lymphocytes/immunology , Bone Marrow Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Inflammation/immunology , Polynucleotides/administration & dosage , Animals , B-Lymphocytes/transplantation , CD40 Antigens/metabolism , CD40 Ligand/genetics , CD40 Ligand/metabolism , Cell Differentiation , Cells, Cultured , Coculture Techniques , Fibroblasts/immunology , Fibroblasts/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Immunologic Memory , Interleukin-4/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Poly I-C , Vaccination
12.
J Immunol ; 197(9): 3618-3627, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27664281

ABSTRACT

Diverse signals received by CD8+ T cells are integrated to achieve the required magnitude of cell expansion and the appropriate balance of effector/memory CD8+ T cell generation. Notably, the strength and nature of TCR signaling influence the differentiation and functional capacity of effector and memory CD8+ T cells. Dok-1 and Dok-2, the two members of the Dok family expressed in T cells, negatively regulate TCR signaling in vitro. However, the role of Dok proteins in modulating T cell function in vivo has not yet studied. We studied the function of Dok-1 and Dok-2 proteins in the regulation of the CD8+ T cell response to vaccinia virus infection. Comparison of responses to vaccinia virus expressing OVA peptide SIINFEKL by wild-type and Dok-1/2-/- CD8+ OT-I cells showed that the absence of Dok-1 and Dok-2 slightly reduced the magnitude of virus-specific effector CD8+ T cell expansion. This was not due to reduced proliferation or enhanced apoptosis of effector CD8+ T cells. Dok-1/2-deficient effector CD8+ T cells showed increased cell surface TCR expression following virus infection in vivo and increased expression of granzyme B and TNF upon stimulation with peptide Ag ex vivo. Finally, Dok-1/2-deficient effector CD8+ T had a severe defect in survival that resulted in impaired generation of memory CD8+ T cells. These results reveal the critical involvement of Dok-1 and Dok-2 in a negative-feedback loop that prevents overactivation of CD8+ T cells and promotes memory formation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , CD8-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/metabolism , Immunologic Memory , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Vaccinia/immunology , Virus Diseases/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , CD8-Positive T-Lymphocytes/virology , Cell Differentiation , Cell Survival , Cells, Cultured , DNA-Binding Proteins/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/genetics , RNA-Binding Proteins/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction
13.
Invest Ophthalmol Vis Sci ; 57(4): 1530-6, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27035626

ABSTRACT

PURPOSE: Neuropilin-1 (NRP-1) is a transmembrane receptor that is critical for vascular development within the central nervous system (CNS). It binds and influences signaling of several key angiogenic factors, such as VEGF-165, semaphorin 3A, platelet derived growth factor, and more. Neuropilin-1 is expressed by neurons and endothelial cells as well as a subpopulation of proangiogenic macrophages/microglia that are thought to interact with endothelial tip cells to promote vascular anastomosis during brain vascularization. We previously demonstrated a significant role for NRP-1 in macrophage chemotaxis and showed that NRP-1-expressing microglia are major contributors to pathologic retinal angiogenesis. Given this influence on CNS angiogenesis, we now investigated the involvement of microglia-resident NRP-1 in developmental retinal vascularization. METHODS: We followed NRP-1 expressing microglia during retinal development. We used LysM-cre myeloid lineage-driver cre mice to reduce expression of NRP-1 in retinal myeloid-derived cells and performed a comprehensive morphometric analysis of retinal vasculature during development. RESULTS: We provide evidence that NRP-1+ microglia are present throughout the retina during vascular development with a preference for the non-vascularized retina. Using LysM-Cre/Nrp1(fl/fl) mice, we reduced NRP-1 expression by ~65% in retinal microglia and demonstrate that deficiency in NRP-1 in these microglia does not impair retinal angiogenesis. CONCLUSIONS: Our data draw a dichotomous role for NRP-1 in cells of myeloid lineage where it is dispensable for adequate retinal developmental vascularization yet obligate for pathologic retinal angiogenesis.


Subject(s)
Microglia/physiology , Neovascularization, Physiologic/physiology , Neuropilin-1/metabolism , Retinal Vessels/physiology , Animals , Animals, Newborn , Bacterial Proteins/metabolism , Endothelium, Vascular , Female , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes/metabolism , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morphogenesis/physiology , Myeloid Cells/metabolism , Signal Transduction
14.
J Immunol ; 194(12): 5654-62, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25972473

ABSTRACT

Following an infection, naive CD8(+) T cells expand and differentiate into two main populations of effectors: short-lived effector cells (SLECs) and memory precursor effector cells (MPECs). There is limited understanding of the molecular mechanism and cellular processes governing this cell fate. Notch is a key regulator of cell fate decision relevant in many immunological pathways. In this study, we add to the role of Notch in cell fate decision and demonstrate that the Notch signaling pathway controls the MPEC/SLEC differentiation choice following both Listeria infection and dendritic cell immunization of mice. Although fewer SLECs were generated, Notch deficiency did not alter the rate of memory CD8(+) T cell generation. Moreover, we reveal that the Notch signaling pathway plays a context-dependent role for optimal cytokine production by effector CD8(+) T cells. Together, our results unravel critical functions for the Notch signaling pathway during effector CD8(+) T cell differentiation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunologic Memory , Receptors, Notch/metabolism , Signal Transduction , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cytokines/biosynthesis , Gene Expression , Listeria/immunology , Listeriosis/immunology , Listeriosis/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Receptors, Notch/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
15.
Immunology ; 145(1): 161-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25521218

ABSTRACT

Extracellular signal-regulated kinase 3 (ERK3 )is an atypical member of the mitogen-activated protein kinase (MAPK) family. We have previously shown that ERK3 is expressed during thymocyte differentiation and that its expression is induced in mature peripheral T cells following activation of ERK1/2 by T-cell receptor (TCR) signalling. Herein, we have investigated whether ERK3 expression is required for proper T-cell selection. Using a knock-in mouse model in which the coding sequence of ERK3 is replaced by the gene encoding for the ß-galactosidase reporter, we show that ERK3 is expressed by double-positive (DP) thymocytes undergoing positive selection. In ERK3-deficient mice with a polyclonal TCR repertoire, we observe a decrease in positive selection. This reduction in positive selection was also observed when ERK3-deficient mice were backcrossed to class I- and class II-restricted TCR transgenic mice. Furthermore, the response of DP thymocytes to in vitro TCR stimulation was strongly reduced in ERK3-deficient mice. Together, these results show that ERK3 expression following TCR signalling is critical for proper thymic positive selection.


Subject(s)
Clonal Selection, Antigen-Mediated , MAP Kinase Signaling System/immunology , Mitogen-Activated Protein Kinase 6/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/immunology , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Mitogen-Activated Protein Kinase 6/genetics , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/cytology , Thymocytes/cytology , Thymus Gland/cytology
16.
J Clin Invest ; 124(11): 4807-22, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25271625

ABSTRACT

Immunological activity in the CNS is largely dependent on an innate immune response and is heightened in diseases, such as diabetic retinopathy, multiple sclerosis, amyotrophic lateral sclerosis, and Alzheimer's disease. The molecular dynamics governing immune cell recruitment to sites of injury and disease in the CNS during sterile inflammation remain poorly defined. Here, we identified a subset of mononuclear phagocytes (MPs) that responds to local chemotactic cues that are conserved among central neurons, vessels, and immune cells. Patients suffering from late-stage proliferative diabetic retinopathy (PDR) had elevated vitreous semaphorin 3A (SEMA3A). Using a murine model, we found that SEMA3A acts as a potent attractant for neuropilin-1-positive (NRP-1-positive) MPs. These proangiogenic MPs were selectively recruited to sites of pathological neovascularization in response to locally produced SEMA3A as well as VEGF. NRP-1-positive MPs were essential for disease progression, as NRP-1-deficient MPs failed to enter the retina in a murine model of oxygen-induced retinopathy (OIR), a proxy for PDR. OIR mice with NRP-1-deficient MPs exhibited decreased vascular degeneration and diminished pathological preretinal neovascularization. Intravitreal administration of a NRP-1-derived trap effectively mimicked the therapeutic benefits observed in mice lacking NRP-1-expressing MPs. Our findings indicate that NRP-1 is an obligate receptor for MP chemotaxis, bridging neural ischemia to an innate immune response in neovascular retinal disease.


Subject(s)
Chemotaxis , Myeloid Cells/physiology , Neuropilin-1/physiology , Adult , Aged , Aged, 80 and over , Animals , Cell Proliferation , Cells, Cultured , Choroid/immunology , Diabetic Retinopathy/immunology , Diabetic Retinopathy/metabolism , Humans , Immunity, Innate , Mice, Inbred C57BL , Middle Aged , Mononuclear Phagocyte System/immunology , Neovascularization, Physiologic , Neuroimmunomodulation , Semaphorin-3A/metabolism , Tissue Culture Techniques
17.
J Immunol ; 193(4): 1847-54, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25015830

ABSTRACT

During infection or vaccination, only a small proportion of CD8(+) T cells differentiate into memory cells. The mechanisms underlying the differentiation of CD8(+) T cells into short-lived effector cells (SLECs) or memory precursor effector cells are poorly defined. It was recently shown in infectious models that the transcriptional repressor B lymphocyte-induced maturation protein 1 (Blimp-1) enhances the formation of SLECs. The factors controlling Blimp-1 expression leading to the in vivo formation of SLECs are still not known. However, it has been shown that cytokines such as IL-2 induce Blimp-1 expression in vitro. In this study, we took advantage of the low-inflammation model of dendritic cell immunization to study the role of the IL-2/Blimp-1 axis in SLEC differentiation as well as the importance of Blimp-1 expression in memory precursor effector cells for proper CD8(+) memory generation. Our results show that Blimp-1 deficiency affects effector differentiation and function in the absence of inflammation. Unexpectedly, memory generation was not affected in Blimp-1-deficient OT-I cells responding to vaccination. In addition, modulation of the bioavailability of IL-2 by injection either of a blocking Ab or of the cytokine, demonstrates a link between IL-2, Blimp-1 induction, and SLEC formation in wild-type cells. Conversely, injection of IL-2 had less effect on Blimp-1-deficient CD8(+) T cells, indicating that the effect of IL-2 on in vivo SLEC differentiation is mediated by Blimp-1. In conclusion, IL-2 induction of Blimp-1 expression is a key regulator of SLEC differentiation in vivo.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Immunologic Memory , Interleukin-2/immunology , Transcription Factors/biosynthesis , Animals , Antibodies, Blocking/immunology , Apoptosis/immunology , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/immunology , Female , Granzymes/biosynthesis , Inflammation/immunology , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Interleukin-7 Receptor alpha Subunit/biosynthesis , Interleukin-7 Receptor alpha Subunit/genetics , Lectins, C-Type , Listeria monocytogenes/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Positive Regulatory Domain I-Binding Factor 1 , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , Transcription Factors/genetics
18.
Mol Cell Biol ; 34(18): 3374-87, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25002529

ABSTRACT

Extracellular signal-regulated kinase 3 (ERK3) is an atypical member of the mitogen-activated protein kinase (MAPK) family whose function is largely unknown. Given the central role of MAPKs in T cell development, we hypothesized that ERK3 may regulate thymocyte development. Here we have shown that ERK3 deficiency leads to a 50% reduction in CD4(+) CD8(+) (DP) thymocyte number. Analysis of hematopoietic chimeras revealed that the reduction in DP thymocytes is intrinsic to hematopoietic cells. We found that early thymic progenitors seed the Erk3(-/-) thymus and can properly differentiate and proliferate to generate DP thymocytes. However, ERK3 deficiency results in a decrease in the DP thymocyte half-life, associated with a higher level of apoptosis. As a consequence, ERK3-deficient DP thymocytes are impaired in their ability to make successful secondary T cell receptor alpha (TCRα) gene rearrangement. Introduction of an already rearranged TCR transgene restores thymic cell number. We further show that knock-in of a catalytically inactive allele of Erk3 fails to rescue the loss of DP thymocytes. Our results uncover a unique role for ERK3, dependent on its kinase activity, during T cell development and show that this atypical MAPK is essential to sustain DP survival during RAG-mediated rearrangements.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Mitogen-Activated Protein Kinase 6/genetics , Mitogen-Activated Protein Kinase 6/metabolism , Thymocytes/cytology , Thymus Gland/cytology , Animals , Animals, Newborn , CD4-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/enzymology , Catalytic Domain , Cell Differentiation/genetics , Cell Proliferation , Cell Survival , Embryo, Mammalian , Gene Knock-In Techniques , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor , Mice , Mice, Inbred C57BL , Mice, Transgenic , Thymocytes/immunology
19.
PLoS One ; 9(1): e86681, 2014.
Article in English | MEDLINE | ID: mdl-24475167

ABSTRACT

The classical mitogen-activated protein kinases (MAPKs) ERK1 and ERK2 are activated upon stimulation of cells with a broad range of extracellular signals (including antigens) allowing cellular responses to occur. ERK3 is an atypical member of the MAPK family with highest homology to ERK1/2. Therefore, we evaluated the role of ERK3 in mature T cell response. Mouse resting T cells do not transcribe ERK3 but its expression is induced in both CD4⁺ and CD8⁺ T cells following T cell receptor (TCR)-induced T cell activation. This induction of ERK3 expression in T lymphocytes requires activation of the classical MAPK ERK1 and ERK2. Moreover, ERK3 protein is phosphorylated and associates with MK5 in activated primary T cells. We show that ERK3-deficient T cells have a decreased proliferation rate and are impaired in cytokine secretion following in vitro stimulation with low dose of anti-CD3 antibodies. Our findings identify the atypical MAPK ERK3 as a new and important regulator of TCR-induced T cell activation.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Cellular/immunology , Lymphocyte Activation/immunology , Mitogen-Activated Protein Kinase 6/metabolism , T-Lymphocytes/immunology , Animals , Cell Proliferation , Cytokines/metabolism , DNA Primers/genetics , Flow Cytometry , Immunoblotting , Immunoprecipitation , Mice , Mitogen-Activated Protein Kinase 6/deficiency , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , beta-Galactosidase
20.
J Immunol ; 192(3): 1071-8, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24376264

ABSTRACT

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.


Subject(s)
Adjuvants, Immunologic , CD8-Positive T-Lymphocytes/immunology , Listeria monocytogenes/immunology , Listeriosis/prevention & control , Membrane Glycoproteins/agonists , T-Lymphocyte Subsets/immunology , Toll-Like Receptor 7/agonists , Tymovirus/immunology , Vaccination , Adaptive Immunity , Animals , Dendritic Cells/immunology , Drug Evaluation, Preclinical , Female , Immunoglobulin G/biosynthesis , Immunologic Memory , Interferon Type I/immunology , Listeriosis/immunology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/immunology , Nanoparticles , Ovalbumin/immunology , RNA, Viral/immunology , Receptor, Interferon alpha-beta/deficiency , Toll-Like Receptor 7/deficiency , Toll-Like Receptor 7/immunology , Tymovirus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...