Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Nature ; 611(7935): 405-412, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36323780

ABSTRACT

Solid tumours are innervated by nerve fibres that arise from the autonomic and sensory peripheral nervous systems1-5. Whether the neo-innervation of tumours by pain-initiating sensory neurons affects cancer immunosurveillance remains unclear. Here we show that melanoma cells interact with nociceptor neurons, leading to increases in their neurite outgrowth, responsiveness to noxious ligands and neuropeptide release. Calcitonin gene-related peptide (CGRP)-one such nociceptor-produced neuropeptide-directly increases the exhaustion of cytotoxic CD8+ T cells, which limits their capacity to eliminate melanoma. Genetic ablation of the TRPV1 lineage, local pharmacological silencing of nociceptors and antagonism of the CGRP receptor RAMP1 all reduced the exhaustion of tumour-infiltrating leukocytes and decreased the growth of tumours, nearly tripling the survival rate of mice that were inoculated with B16F10 melanoma cells. Conversely, CD8+ T cell exhaustion was rescued in sensory-neuron-depleted mice that were treated with local recombinant CGRP. As compared with wild-type CD8+ T cells, Ramp1-/- CD8+ T cells were protected against exhaustion when co-transplanted into tumour-bearing Rag1-deficient mice. Single-cell RNA sequencing of biopsies from patients with melanoma revealed that intratumoral RAMP1-expressing CD8+ T cells were more exhausted than their RAMP1-negative counterparts, whereas overexpression of RAMP1 correlated with a poorer clinical prognosis. Overall, our results suggest that reducing the release of CGRP from tumour-innervating nociceptors could be a strategy to improve anti-tumour immunity by eliminating the immunomodulatory effects of CGRP on cytotoxic CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes , Melanoma , Nociceptors , Animals , Mice , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/pharmacology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Melanoma/immunology , Melanoma/pathology , Nociceptors/physiology , Sensory Receptor Cells/metabolism , Neurites/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Survival Rate , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Genes, RAG-1/genetics , Humans , Biopsy , Prognosis
2.
BMC Immunol ; 22(1): 56, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34384367

ABSTRACT

BACKGROUND: Invariant chain (CD74, Ii) is a multifunctional protein expressed in antigen presenting cells. It assists the ER exit of various cargos and serves as a receptor for the macrophage migration inhibitory factor. The newly translated Ii chains trimerize, a structural feature that is not readily understood in the context of its MHCII chaperoning function. Two segments of Ii, the luminal C-terminal region (TRIM) and the transmembrane domain (TM), have been shown to participate in the trimerization process but their relative importance and impact on the assembly with MHCII molecules remains debated. Here, we addressed the requirement of these domains in the trimerization of human Ii as well as in the oligomerization with MHCII molecules. We used site-directed mutagenesis to generate series of Ii and DR mutants. These were transiently transfected in HEK293T cells to test their cell surface expression and analyse their interactions by co-immunoprecipitations. RESULTS: Our results showed that the TRIM domain is not essential for Ii trimerization nor for intracellular trafficking with MHCII molecules. We also gathered evidence that in the absence of TM, TRIM allows the formation of multi-subunit complexes with HLA-DR. Similarly, in the absence of TRIM, Ii can assemble into high-order structures with MHCII molecules. CONCLUSIONS: Altogether, our data show that trimerization of Ii through either TM or TRIM sustains nonameric complex formation with MHCII molecules.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Cell Membrane/metabolism , HLA-DR Antigens/genetics , Histocompatibility Antigens Class II/metabolism , Antigen Presentation , Antigens, Differentiation, B-Lymphocyte/genetics , HEK293 Cells , HLA-A24 Antigen/metabolism , Histocompatibility Antigens Class II/genetics , Humans , Mutagenesis, Site-Directed , Mutation/genetics , Protein Domains/genetics , Protein Multimerization
3.
J Allergy Clin Immunol ; 147(6): 2330-2342, 2021 06.
Article in English | MEDLINE | ID: mdl-33453289

ABSTRACT

BACKGROUND: Lung nociceptor neurons amplify immune cell activity and mucus metaplasia in response to an inhaled allergen challenge in sensitized mice. OBJECTIVE: We sought to identify the cellular mechanisms by which these sensory neurons are activated subsequent to allergen exposure. METHODS: We used calcium microscopy and electrophysiologic recording to assess whether vagal neurons directly respond to the model allergen ovalbumin (OVA). Next, we generated the first nociceptor-specific FcεR1γ knockdown (TRPV1Cre::FcεR1γfl/fl) mice to assess whether this targeted invalidation would affect the severity of allergic inflammation in response to allergen challenges. RESULTS: Lung-innervating jugular nodose complex ganglion neurons express the high-affinity IgE receptor FcεR1, the levels of which increase in OVA-sensitized mice. FcεR1γ-expressing vagal nociceptor neurons respond directly to OVA complexed with IgE with depolarization, action potential firing, calcium influx, and neuropeptide release. Activation of vagal neurons by IgE-allergen immune complexes, through the release of substance P from their peripheral terminals, directly amplifies TH2 cell influx and polarization in the airways. Allergic airway inflammation is decreased in TRPV1Cre::FcεR1γfl/fl mice and in FcεR1α-/- mice into which bone marrow has been transplanted. Finally, increased in vivo circulating levels of IgE following allergen sensitization enhances the responsiveness of FcεR1 to immune complexes in both mouse jugular nodose complex ganglion neurons and human induced pluripotent stem cell-derived nociceptors. CONCLUSIONS: Allergen sensitization triggers a feedforward inflammatory loop between IgE-producing plasma cells, FcεR1-expressing vagal sensory neurons, and TH2 cells, which helps to both initiate and amplify allergic airway inflammation. These data highlight a novel target for reducing allergy, namely, FcεR1γ expressed by nociceptors.


Subject(s)
Gene Expression , Hypersensitivity/immunology , Hypersensitivity/metabolism , Receptors, IgE/genetics , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Allergens/immunology , Animals , Calcium/metabolism , Disease Models, Animal , Disease Susceptibility/immunology , Genetic Predisposition to Disease , Hypersensitivity/genetics , Hypersensitivity/pathology , Mice , Mice, Knockout , Neurons/immunology , Neurons/metabolism , Nociceptors/metabolism , Ovalbumin/adverse effects , Ovalbumin/immunology , Receptors, IgE/metabolism , Respiratory Mucosa/pathology , Substance P/metabolism , Vagus Nerve
4.
Front Immunol ; 11: 1953, 2020.
Article in English | MEDLINE | ID: mdl-32973799

ABSTRACT

Obesity is a major risk factor for the development of insulin resistance and type 2 diabetes. However, the mechanisms that trigger the underlying adipose tissues inflammation are not completely understood. Here, we show that the E3 ubiquitin ligase March1 controls the phenotypic and functional properties of CD8+ T cells in mice white adipose tissue. In a diet-induced obesity model, mice lacking March1 [March1 knockout (KO)] show increased insulin resistance compared to their WT counterparts. Also, in obese March1 KO mice, the proportions of effector/memory (Tem) and resident/memory (Trm) CD8+ T cells were higher in the visceral adipose tissue, but not in the spleen. The effect of March1 on insulin resistance and on the phenotype of adipose tissue CD8+ T cells was independent of major histocompatibility complex class II ubiquitination. Interestingly, we adoptively transferred either WT or March1 KO splenic CD8+ T cells into obese WT chimeras that had been reconstituted with Rag1-deficient bone marrow. We observed an enrichment of Tem and Trm cells and exacerbated insulin resistance in mice that received March1 KO CD8 T cells. Mechanistically, we found that March1 deficiency alters the metabolic activity of CD8+ T cells. Our results provide additional evidence of the involvement of CD8+ T cells in adipose tissue inflammation and suggest that March1 controls the metabolic reprogramming of these cells.


Subject(s)
Adipose Tissue, White/enzymology , CD8-Positive T-Lymphocytes/metabolism , Immunologic Memory , Insulin Resistance , Obesity/enzymology , Ubiquitin-Protein Ligases/deficiency , Adipose Tissue, White/immunology , Adoptive Transfer , Animals , Blood Glucose/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Diet, High-Fat , Disease Models, Animal , Energy Metabolism , Lymphocyte Activation , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/immunology , Phenotype , Spleen/enzymology , Spleen/immunology , Ubiquitin-Protein Ligases/genetics
5.
J Immunol ; 203(11): 2887-2898, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31659013

ABSTRACT

Dendritic cells (DCs) are critical players in skin homeostasis. A subset of mannose receptor (CD206)-expressing monocyte-derived DCs was found in skin, and their migratory counterpart is present in skin-draining lymph nodes (sdLNs). Skin CD206+ DCs were shown to upregulate MHC class II (MHCII) progressively, raising the question of whether this feature affects their biology. In this study, we assessed the role of MHCII regulation in the development and migration of these cells in mouse models expressing differential MHCII levels. Using CD206 as a surrogate marker, we found that skin CD206+ DCs develop in an MHCII-independent manner. However, their migration to sdLNs was affected by overexpression rather than absence or lower expression of MHCII. Accordingly, B16 tumor growth was exacerbated in mice overexpressing MHCII in the absence of ubiquitination. Mechanistically, CD206+ DCs from these mice showed decreased IRF4 and CCR7 expression. LPS, which is known to promote monocyte-derived DC recruitment to sdLNs, partially improved these defects. However, GM-CSF delivery restored CD206+ DC migration by promoting IRF4 expression. Collectively, these data show that MHCII downregulation is crucial for IRF4-dependent migration of CD206+ DCs to sdLNs in health and disease.


Subject(s)
Cell Movement , Dendritic Cells/metabolism , Down-Regulation , Histocompatibility Antigens Class II/metabolism , Lectins, C-Type/metabolism , Lymph Nodes/metabolism , Mannose-Binding Lectins/metabolism , Receptors, Cell Surface/metabolism , Skin/metabolism , Ubiquitination , Animals , Mannose Receptor , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Hum Immunol ; 80(1): 15-31, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30321632

ABSTRACT

Classical HLA class II molecules are highly polymorphic heterodimeric transmembrane proteins encoded by a polygenic cluster on chromosome 6. Polymorphic residues in the membrane-distal domains ensure that a large collection of microbial peptides can be bound in the human population. Still, the HLA-DR, -DP and -DQ isotypes show a high degree of conservation in their overall tertiary and quaternary structures, in line with their common function in T cell receptor activation. Interestingly, the primary structure of the intracellular domains are highly divergent between isotypes and they also show allotypic variations. The functional impact of these differences remains to be fully appreciated. Here, we address the role of the MHC class II cytoplasmic tails in intracellular trafficking. First, the emphasis will be on the interplay between the cytoplasmic domains of classical human MHC class II molecules and those of the invariant chain chaperone (CD74) isoforms. Then, we will examine the importance of the highly conserved ß-chain cytoplasmic lysine residue in the ubiquitin-driven trafficking of MHC class II molecules. These considerations should help understand the potential functional impact of sequence variations that may arise in the cytoplasmic tails and transmembrane domains of MHC class II molecules.


Subject(s)
Amino Acid Substitution , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/genetics , Polymorphism, Single Nucleotide , Amino Acid Sequence , Antigen Presentation , Conserved Sequence , Histocompatibility Antigens Class II/metabolism , Humans , Intracellular Space , Models, Biological , Protein Interaction Domains and Motifs , Protein Transport , Structure-Activity Relationship
7.
J Immunol ; 201(6): 1784-1798, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30097529

ABSTRACT

Ischemic myocardial injury results in sterile cardiac inflammation that leads to tissue repair, two processes controlled by mononuclear phagocytes. Despite global burden of cardiovascular diseases, we do not understand the functional contribution to pathogenesis of specific cardiac mononuclear phagocyte lineages, in particular dendritic cells. To address this limitation, we used detailed lineage tracing and genetic studies to identify bona fide murine and human CD103+ conventional dendritic cell (cDC)1s, CD11b+ cDC2s, and plasmacytoid DCs (pDCs) in the heart of normal mice and immunocompromised NSG mice reconstituted with human CD34+ cells, respectively. After myocardial infarction (MI), the specific depletion of cDCs, but not pDCs, improved cardiac function and prevented adverse cardiac remodeling. Our results showed that fractional shortening measured after MI was not influenced by the absence of pDCs. Interestingly, however, depletion of cDCs significantly improved reduction in fractional shortening. Moreover, fibrosis and cell areas were reduced in infarcted zones. This correlated with reduced numbers of cardiac macrophages, neutrophils, and T cells, indicating a blunted inflammatory response. Accordingly, mRNA levels of proinflammatory cytokines IL-1ß and IFN-γ were reduced. Collectively, our results demonstrate the unequivocal pathological role of cDCs following MI.


Subject(s)
Cell Movement/immunology , Dendritic Cells/immunology , Myocardial Infarction/immunology , Animals , Cell Movement/genetics , Dendritic Cells/pathology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , Monocytes/immunology , Monocytes/pathology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Neutrophils/immunology , Neutrophils/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
8.
J Immunol Res ; 2018: 3823910, 2018.
Article in English | MEDLINE | ID: mdl-29854835

ABSTRACT

Membrane-associated RING-CH-1 (March1) is a member of the March family of E3 ubiquitin ligases. March1 downregulates cell surface expression of MHC II and CD86 by targeting them to lysosomal degradation. Given the key roles of MHC class II and CD86 in T cell activation and to get further insights into the development of allergic inflammation, we asked whether March1 deficiency exacerbates or attenuates features of allergic asthma in mice. Herein, we used an acute model of allergy to compare the asthmatic phenotype of March1-deficient and -sufficient mice immunized with ovalbumin (OVA) and later challenged by intranasal instillation of OVA in the lungs. We found that eosinophilic inflammation in airways and lung tissue was similar between WT and March1-/- allergic mice, whereas neutrophilic inflammation was significant only in March1-/- mice. Airway hyperresponsiveness as well as levels of IFN-γ, IL-13, IL-6, and IL-10 was lower in the lungs of asthmatic March1-/- mice compared to WT, whereas lung levels of TNF-α, IL-4, and IL-5 were not significantly different. Interestingly, in the serum, levels of total and ova-specific IgE were reduced in March1-deficient mice as compared to WT mice. Taken together, our results demonstrate a role of March1 E3 ubiquitin ligase in modulating allergic responses.


Subject(s)
Asthma/immunology , Lung/immunology , Pneumonia/immunology , Ubiquitin-Protein Ligases/metabolism , Allergens/immunology , Animals , Cytokines/metabolism , Disease Models, Animal , Humans , Immunoglobulin E/blood , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Ovalbumin/immunology , Ubiquitin-Protein Ligases/genetics
9.
Front Immunol ; 9: 1199, 2018.
Article in English | MEDLINE | ID: mdl-29899744

ABSTRACT

Streptococcus suis is an important swine pathogen and emerging zoonotic agent. Encapsulated strains of S. suis modulate dendritic cell (DC) functions, leading to poorly activated CD4+ T cells. However, the antigen presentation ability of S. suis-stimulated DCs has not been investigated yet. In this work, we aimed to characterize the antigen presentation profiles of S. suis-stimulated DCs, both in vitro and in vivo. Upon direct activation in vitro, S. suis-stimulated murine bone marrow-derived DCs (bmDCs) preserved their antigen capture/processing capacities. However, they showed delayed kinetics of MHC-II expression compared to lipopolysaccharide-stimulated bmDCs. Meanwhile, splenic DCs from infected mice exhibited a compromised MHC-II expression, despite an appropriate expression of maturation markers. To identify potential interfering mechanisms, Class II Major Histocompatibility Complex Transactivator (CIITA) and membrane-associated RING-CH (MARCH)1/8 transcription were studied. S. suis-stimulated DCs maintained low levels of CIITA at early time points, both in vitro and in vivo, which could limit their ability to increase MHC-II synthesis. S. suis-stimulated DCs also displayed sustained/upregulated levels of MARCH1/8, thus possibly leading to MHC-II lysosomal degradation. The bacterial capsular polysaccharide played a partial role in this modulation. Finally, interleukin (IL)-12p70 production was inhibited in splenic DCs from infected mice, a profile compatible with DC indirect activation by pro-inflammatory compounds. Consequently, these cells induced lower levels of IL-2 and TNF-α in an antigen-specific CD4+ T cell presentation assay and blunted T cell CD25 expression. It remains unclear at this stage whether these phenotypical and transcriptional modulations observed in response to S. suis in in vivo infections are part of a bacterial immune evasion strategy or rather a feature common to systemic inflammatory response-inducing agents. However, it appears that the MHC-II-restricted antigen presentation and Th1-polarizing cytokine production capacities of DCs are impaired during S. suis infection. This study highlights the potential consequences of inflammation on the type and magnitude of the immune response elicited by a pathogen.


Subject(s)
Dendritic Cells/immunology , Interleukin-12/metabolism , Streptococcal Infections/immunology , Streptococcus suis/physiology , Th1 Cells/immunology , Animals , Antigen Presentation , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Female , Histocompatibility Antigens Class II/metabolism , Immune Evasion , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Serogroup , Trans-Activators/genetics , Trans-Activators/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
10.
Front Microbiol ; 8: 1196, 2017.
Article in English | MEDLINE | ID: mdl-28706510

ABSTRACT

Dendritic cells (DCs) and NK cells play a crucial role in the first phase of host defense against infections. Group B Streptococcus (GBS) and Streptococcus suis are encapsulated streptococci causing severe systemic inflammation, leading to septicemia and meningitis. Yet, the involvement of NK cells in the innate immune response to encapsulated bacterial infection is poorly characterized. Here, it was observed that these two streptococcal species rapidly induce the release of IFN-γ and that NK cells are the major cell type responsible for this production during the acute phase of the infection. Albeit S. suis capacity to activate NK cells was lower than that of GBS, these cells partially contribute to S. suis systemic infection; mainly through amplification of the inflammatory loop. In contrast, such a role was not observed during GBS systemic infection. IFN-γ release by NK cells required the presence of DCs, which in turn had a synergistic effect on DC cytokine production. These responses were mainly mediated by direct DC-NK cell contact and partially dependent on soluble factors. Though IL-12 and LFA-1 were shown to be critical in S. suis-mediated activation of the DC-NK cell crosstalk, different or redundant molecular pathways modulate DC-NK interactions during GBS infection. The bacterial capsular polysaccharides also differently modulated NK cell activation. Together, these results demonstrated a role of NK cells in the innate immune response against encapsulated streptococcal infections; yet the molecular pathways governing NK activation seem to differ upon the pathogen and should not be generalized when studying bacterial infections.

11.
J Immunol ; 198(2): 852-861, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27940660

ABSTRACT

Ubiquitination was recently identified as a central process in the pathogenesis and development of numerous inflammatory diseases, such as obesity, atherosclerosis, and asthma. Treatment with proteasomal inhibitors led to severe side effects because ubiquitination is heavily involved in a plethora of cellular functions. Thus, new players regulating ubiquitination processes must be identified to improve therapies for inflammatory diseases. In addition to their role in adaptive immunity, endosomal MHC class II (MHCII) molecules were shown to modulate innate immune responses by fine tuning the TLR4 signaling pathway. However, the role of MHCII ubiquitination by membrane associated ring-CH-type finger 1 (MARCH1) E3 ubiquitin ligase in this process remains to be assessed. In this article, we demonstrate that MARCH1 is a key inhibitor of innate inflammation in response to bacterial endotoxins. The higher mortality of March1-/- mice challenged with a lethal dose of LPS was associated with significantly stronger systemic production of proinflammatory cytokines and splenic NK cell activation; however, we did not find evidence that MARCH1 modulates LPS or IL-10 signaling pathways. Instead, the mechanism by which MARCH1 protects against endotoxic shock rests on its capacity to promote the transition of monocytes from Ly6CHi to Ly6C+/- Moreover, in competitive bone marrow chimeras, March1-/- monocytes and polymorphonuclear neutrophils outcompeted wild-type cells with regard to bone marrow egress and homing to peripheral organs. We conclude that MARCH1 exerts MHCII-independent effects that regulate the innate arm of immunity. Thus, MARCH1 might represent a potential new target for emerging therapies based on ubiquitination reactions in inflammatory diseases.


Subject(s)
Endotoxemia/immunology , Immunity, Innate/immunology , Inflammation/immunology , Monocytes/immunology , Ubiquitin-Protein Ligases/immunology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Histocompatibility Antigens Class II/immunology , Lipopolysaccharides/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Ubiquitination
12.
Sci Rep ; 6: 38061, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27905502

ABSTRACT

The pathogenesis of Streptococcus suis infection, a major swine and human pathogen, is only partially understood and knowledge on the host adaptive immune response is critically scarce. Yet, S. suis virulence factors, particularly its capsular polysaccharide (CPS), enable this bacterium to modulate dendritic cell (DC) functions and potentially impair the immune response. This study aimed to evaluate modulation of T cell activation during S. suis infection and the role of DCs in this response. S. suis-stimulated total mouse splenocytes readily produced TNF-α, IL-6, IFN-γ, CCL3, CXCL9, and IL-10. Ex vivo and in vivo analyses revealed the involvement of CD4+ T cells and a Th1 response. Nevertheless, during S. suis infection, levels of the Th1-derived cytokines TNF-α and IFN-γ were very low. A transient splenic depletion of CD4+ T cells and a poor memory response were also observed. Moreover, CD4+ T cells secreted IL-10 and failed to up-regulate optimal levels of CD40L and CD69 in coculture with DCs. The CPS hampered release of several T cell-derived cytokines in vitro. Finally, a correlation was established between severe clinical signs of S. suis disease and impaired antibody responses. Altogether, these results suggest S. suis interferes with the adaptive immune response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Streptococcal Infections/microbiology , Streptococcus suis/immunology , Animals , Bacterial Capsules/immunology , Disease Models, Animal , Humans , Interferon-gamma/metabolism , Mice , Streptococcal Infections/immunology , Swine , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/metabolism , Virulence Factors
13.
Nat Commun ; 7: 12639, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27577745

ABSTRACT

Insulin resistance is a key driver of type 2 diabetes (T2D) and is characterized by defective insulin receptor (INSR) signalling. Although surface INSR downregulation is a well-established contributor to insulin resistance, the underlying molecular mechanisms remain obscure. Here we show that the E3 ubiquitin ligase MARCH1 impairs cellular insulin action by degrading cell surface INSR. Using a large-scale RNA interference screen, we identify MARCH1 as a negative regulator of INSR signalling. March1 loss-of-function enhances, and March1 overexpression impairs, hepatic insulin sensitivity in mice. MARCH1 ubiquitinates INSR to decrease cell surface INSR levels, but unlike other INSR ubiquitin ligases, MARCH1 acts in the basal state rather than after insulin stimulation. Thus, MARCH1 may help set the basal gain of insulin signalling. MARCH1 expression is increased in white adipose tissue of obese humans, suggesting that MARCH1 contributes to the pathophysiology of T2D and could be a new therapeutic target.


Subject(s)
Antigens, CD/metabolism , Diabetes Mellitus, Type 2/pathology , Insulin Resistance/physiology , Insulin/metabolism , Obesity/pathology , Receptor, Insulin/metabolism , Ubiquitin-Protein Ligases/metabolism , Adipose Tissue, White/pathology , Adolescent , Animals , Antigens, CD/genetics , Biopsy , Cell Line , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/therapy , Diet, High-Fat/adverse effects , Disease Models, Animal , Down-Regulation , Female , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Obesity/blood , Obesity/etiology , Obesity/therapy , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Phosphorylation , RNA, Small Interfering/metabolism , Receptor, Insulin/genetics , Signal Transduction/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Up-Regulation
14.
J Immunol Res ; 2016: 5290604, 2016.
Article in English | MEDLINE | ID: mdl-26989699

ABSTRACT

Group B Streptococcus (GBS) serotype III causes life-threatening infections. Cytokines have emerged as important players for the control of disease, particularly IFN-γ. Although potential sources of this cytokine have been proposed, no specific cell line has ever been described as a leading contributor. In this study, CD4(+) T cell activation profiles in response to GBS were evaluated through in vivo, ex vivo, and in vitro approaches. Total splenocytes readily produce a type 1 proinflammatory response by releasing IFN-γ, TNF-α, and IL-6 and actively recruit T cells via chemokines like CXCL9, CXCL10, and CCL3. Responding CD4(+) T cells differentiate into Th1 cells producing large amounts of IFN-γ, TNF-α, and IL-2. In vitro studies using dendritic cell and CD4(+) T cell cocultures infected with wild-type GBS or a nonencapsulated mutant suggested that GBS capsular polysaccharide, one of the major bacterial virulence factors, differentially modulates surface expression of CD69 and IFN-γ production. Overall, CD4(+) T cells are important producers of IFN-γ and might thus influence the course of GBS infection through the expression balance of this cytokine.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Interferon-gamma/immunology , Polysaccharides, Bacterial/pharmacology , Streptococcal Infections/immunology , Streptococcus agalactiae/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Differentiation/drug effects , Chemokine CCL3/genetics , Chemokine CCL3/immunology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Gene Expression Regulation , Interferon-gamma/genetics , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Mice , Mice, Inbred C57BL , Polysaccharides, Bacterial/biosynthesis , Signal Transduction , Spleen/drug effects , Spleen/immunology , Spleen/pathology , Streptococcal Infections/genetics , Streptococcal Infections/microbiology , Streptococcal Infections/mortality , Streptococcus agalactiae/metabolism , Streptococcus agalactiae/pathogenicity , Survival Analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
15.
Cytokine ; 82: 112-21, 2016 06.
Article in English | MEDLINE | ID: mdl-26854212

ABSTRACT

Type II diabetes regroups different physiological anomalies that ultimately lead to low-grade chronic inflammation, insulin resistance and loss of pancreatic ß-cells. Obesity is one of the best examples of such a condition that can develop into Metabolic Syndrome, causing serious health problems of great socio-economic consequences. The pathological outcome of obesity has a genetic basis and depends on the delicate balance between pro- and anti-inflammatory effectors of the immune system. The causal link between obesity and inflammation is well established. While innate immunity plays a key role in the development of a pro-inflammatory state in obese adipose tissues, it has now become clear that adaptive immune cells are also involved and participate in the cascade of events that lead to metabolic perturbations. The efficacy of some immunotherapeutic protocols in reducing the symptoms of obesity-driven metabolic syndrome in mice implicated all arms of the immune response. Recently, the production of pathogenic immunoglobulins and pro-inflammatory cytokines by B and T lymphocytes suggested an auto-immune basis for the establishment of a non-healthy obese state. Understanding the cellular landscape of obese adipose tissues and how immune cells sustain chronic inflammation holds the key to the development of targeted therapies. In this review, we emphasize the role of antigen-presenting cells and MHC molecules in obese adipose tissue and the general contribution of the adaptive arm of the immune system in inflammation-induced insulin resistance.


Subject(s)
Adipose Tissue/immunology , Antigen Presentation , B-Lymphocytes/immunology , Cytokines/immunology , Obesity/immunology , T-Lymphocytes/immunology , Adipose Tissue/pathology , Animals , B-Lymphocytes/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Obesity/pathology , T-Lymphocytes/pathology
16.
Hum Immunol ; 76(4): 292-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25731712

ABSTRACT

Four invariant chain (Ii) isoforms assist the folding and trafficking of human MHC class II (MHCIIs). The main isoforms, Iip33 and Iip35, assemble in the ER into homo- and/or hetero-trimers. The sequential binding of up to three MHCII αß heterodimers to Ii trimers results in the formation of pentamers, heptamers and nonamers. MHCIIs are required to overcome the p35-encoded di-arginine (RxR) ER retention motif and to allow anterograde trafficking of the complex. Here, we show that inactivation of the RxR motif requires a direct cis interaction between p35 and the MHCII, precluding ER egress of some unsaturated Ii trimers. Interestingly, as opposed to MHCII/p33 complexes, those including p35 remained in the ER when co-expressed with the NleA protein of enterohaemorrhagic Escherichia coli. Taken together, our results demonstrate that p35 influences distinctively MHCII/Ii assembly and trafficking.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Endoplasmic Reticulum/metabolism , Enterohemorrhagic Escherichia coli/immunology , Escherichia coli Proteins/metabolism , Histocompatibility Antigens Class II/metabolism , Virulence Factors/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Proteins/genetics , HEK293 Cells , HLA Antigens/metabolism , Histocompatibility Antigens Class II/genetics , Humans , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization , Protein Sorting Signals/genetics , Protein Transport/drug effects , Protein Transport/genetics , Virulence Factors/genetics
17.
Monoclon Antib Immunodiagn Immunother ; 33(4): 221-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25171001

ABSTRACT

The invariant chain (Ii; CD74) is a multifunctional protein of the immune system and a major player in the presentation of exogenous antigens to T cells. In the endoplasmic reticulum (ER), Ii assists the folding and trafficking of MHC class II molecules. In the present study, we characterized the recently commercialized D-6 monoclonal antibody (MAb) made against a polypeptide spanning the entire sequence of the p33 isoform of human Ii. Using transgenic mice expressing the human p35 isoform, we showed by flow cytometry that D-6 only slightly cross-reacts with mouse Ii in permeabilized splenocytes. Analysis of the human B lymphoblastoid cell line LG2 revealed that D-6 recognizes Ii only upon membrane permeabilization. Variants of Ii bearing specific mutations or deletions were transfected in human cells to map the D-6 epitope. Our results showed that this MAb binds to the N-terminal cytoplasmic domain of Ii and that the epitope was destroyed upon mutagenesis of the two leucine-based endosomal targeting motifs. Thus, D-6 cannot be used for rapid flow cytometric assessment of CD74 cell surface expression and would be ineffective as a drug conjugate for the treatment of hematological malignancies.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , Animals , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal, Murine-Derived/genetics , Antibodies, Monoclonal, Murine-Derived/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , Cell Line , Epitope Mapping , Flow Cytometry , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Humans , Mice , Mice, Transgenic
18.
Immunol Cell Biol ; 92(6): 553-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24638068

ABSTRACT

Four different isoforms of the human invariant chain (Ii) have been described (p33, p35, p41 and p43). These heterotrimerize in the endoplasmic reticulum (ER) before associating with MHC class II molecules (MHCIIs). However, the final stoichiometry of the Ii/MHCII complex remains debated. This is particularly interesting as both p35 and p43 include a di-arginine motif that requires masking by MHCII to allow ER egress. Here, to functionally address the requirement for stoichiometric interactions, we used a recombinant DR heterodimer bearing its own cytoplasmic di-lysine ER-retention motif (DRKKAA). When coexpressed with p33 and a control myc-tagged DR (DRmyc), DRKKAA was retained in the ER but had little impact on surface expression of DRmyc. However, when coexpressed with p35, DRKKAA restricted the surface expression of DRmyc, indicating that Ii trimers can be loaded with more than one MHCII. Similar results were obtained using HLA-DQ instead of DRmyc, showing that a single trimeric Ii scaffold can include distinct MHCII isotypes. Altogether, these results demonstrate that the subunit stoichiometry of oligomeric Ii/MHCII complexes is influenced by p35.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/immunology , Endoplasmic Reticulum/immunology , Gene Expression Regulation/immunology , HLA-DQ Antigens/immunology , HLA-DR Antigens/immunology , Histocompatibility Antigens Class II/immunology , Amino Acid Motifs , Antigens, Differentiation, B-Lymphocyte/genetics , Endoplasmic Reticulum/genetics , Gene Expression Regulation/genetics , HEK293 Cells , HLA-DQ Antigens/genetics , HLA-DR Antigens/genetics , Histocompatibility Antigens Class II/genetics , Humans , Protein Isoforms/immunology
19.
J Immunol ; 192(4): 1896-906, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24453254

ABSTRACT

Mouse mammary tumor virus superantigens (vSAGs) are notorious for defying structural characterization, and a consensus has yet to be reached regarding their ability to bridge the TCR to MHC class II (MHCII). In this study, we determined the topology of the T cell signaling complex by examining the respective relation of vSAG7 with the MHCII molecule, MHCII-associated peptide, and TCR. We used covalently linked peptide/MHCII complexes to demonstrate that vSAG presentation is tolerant to variation in the protruding side chains of the peptide, but can be sensitive to the nature of the protruding N-terminal extension. An original approach in which vSAG was covalently linked to either MHCII chain confirmed that vSAG binds outside the peptide binding groove. Also, whereas the C-terminal vSAG segment binds to the MHCII α-chain in a conformation-sensitive manner, the membrane-proximal N-terminal domain binds the ß-chain. Because both moieties of the mature vSAG remain noncovalently associated after processing, our results suggest that vSAG crosslinks MHCII molecules. Comparing different T cell hybridomas, we identified key residues on the MHCII α-chain that are differentially recognized by the CDR3ß when engaged by vSAG. Finally, we show that the highly conserved tyrosine residue found in the vSAg TGXY motif is required for T cell activation. Our results reveal a novel SAG/MHCII/TCR architecture in which vSAGs coerce a near-canonical docking between MHCII and TCR that allows eschewing of traditional CDR3 binding with the associated peptide in favor of MHCII α-chain binding. Our findings highlight the plasticity of the TCR CDRs.


Subject(s)
Antigens, Viral/immunology , Histocompatibility Antigens Class II/immunology , Mammary Tumor Virus, Mouse/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Superantigens/immunology , Binding Sites/immunology , HEK293 Cells , HeLa Cells , Humans , Jurkat Cells , Lymphocyte Activation/immunology , Protein Binding/immunology , Protein Structure, Tertiary , Signal Transduction/immunology , T-Lymphocytes/immunology
20.
Hum Immunol ; 74(10): 1280-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23756162

ABSTRACT

HLA-DO (H2-O in mice) is an intracellular non-classical MHC class II molecule (MHCII). It forms a stable complex with HLA-DM (H2-M in mice) and shapes the MHC class II-associated peptide repertoire. Here, we tested the impact of HLA-DO and H2-O on the binding of superantigens (SAgs), which has been shown previously to be sensitive to the structural nature of the class II-bound peptides. We found that the binding of staphylococcal enterotoxin (SE) A and B, as well as toxic shock syndrome toxin 1 (TSST-1), was similar on the HLA-DO(+) human B cell lines 721.45 and its HLA-DO(-) counterpart. However, overexpressing HLA-DO in MHC class II(+) HeLa cells (HeLa-CIITA-DO) improved binding of SEA and TSST-1. Accordingly, knocking down HLA-DO expression using specific siRNAs decreased SEA and TSST-1 binding. We tested directly the impact of the class II-associated invariant chain peptide (CLIP), which dissociation from MHC class II molecules is inhibited by overexpressed HLA-DO. Loading of synthetic CLIP on HLA-DR(+) cells increased SEA and TSST-1 binding. Accordingly, knocking down HLA-DM had a similar effect. In mice, H2-O deficiency had no impact on SAgs binding to isolated splenocytes. Altogether, our results demonstrate that the sensitivity of SAgs to the MHCII-associated peptide has physiological basis and that the effect of HLA-DO on SEA and TSST-1 is mediated through the inhibition of CLIP release.


Subject(s)
Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Antigens, Differentiation, B-Lymphocyte/immunology , HLA-D Antigens/immunology , HLA-D Antigens/metabolism , Histocompatibility Antigens Class II/immunology , Superantigens/immunology , Superantigens/metabolism , Animals , Antigens, Differentiation, B-Lymphocyte/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Cell Line , Enterotoxins/immunology , Enterotoxins/metabolism , Gene Expression , Gene Knockout Techniques , HLA-D Antigens/chemistry , HLA-D Antigens/genetics , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/metabolism , Humans , Mice , Protein Binding/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...