Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Cell ; 185(4): 585-602.e29, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35051368

ABSTRACT

The relevance of extracellular magnesium in cellular immunity remains largely unknown. Here, we show that the co-stimulatory cell-surface molecule LFA-1 requires magnesium to adopt its active conformation on CD8+ T cells, thereby augmenting calcium flux, signal transduction, metabolic reprogramming, immune synapse formation, and, as a consequence, specific cytotoxicity. Accordingly, magnesium-sufficiency sensed via LFA-1 translated to the superior performance of pathogen- and tumor-specific T cells, enhanced effectiveness of bi-specific T cell engaging antibodies, and improved CAR T cell function. Clinically, low serum magnesium levels were associated with more rapid disease progression and shorter overall survival in CAR T cell and immune checkpoint antibody-treated patients. LFA-1 thus directly incorporates information on the composition of the microenvironment as a determinant of outside-in signaling activity. These findings conceptually link co-stimulation and nutrient sensing and point to the magnesium-LFA-1 axis as a therapeutically amenable biologic system.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Magnesium/metabolism , Animals , Bacterial Infections/immunology , Caloric Restriction , Cell Line, Tumor , Cytotoxicity, Immunologic , HEK293 Cells , Humans , Immunologic Memory , Immunological Synapses/metabolism , Immunotherapy , Lymphocyte Activation/immunology , MAP Kinase Signaling System , Magnesium/administration & dosage , Male , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism
2.
Cell ; 175(2): 458-471.e19, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30173917

ABSTRACT

Inflammatory disorders of the CNS are frequently accompanied by synaptic loss, which is thought to involve phagocytic microglia and complement components. However, the mechanisms accounting for aberrant synaptic connectivity in the context of CD8+ T cell-driven neuronal damage are poorly understood. Here, we profiled the neuronal translatome in a murine model of encephalitis caused by CD8+ T cells targeting antigenic neurons. Neuronal STAT1 signaling and downstream CCL2 expression were essential for apposition of phagocytes, ensuing synaptic loss and neurological disease. Analogous observations were made in the brains of Rasmussen's encephalitis patients. In this devastating CD8+ T cell-driven autoimmune disease, neuronal STAT1 phosphorylation and CCL2 expression co-clustered with infiltrating CD8+ T cells as well as phagocytes. Taken together, our findings uncover an active role of neurons in coordinating phagocyte-mediated synaptic loss and highlight neuronal STAT1 and CCL2 as critical steps in this process that are amenable to pharmacological interventions.


Subject(s)
Neurons/metabolism , Phagocytosis/physiology , Synapses/physiology , Animals , Brain/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/physiology , Disease Models, Animal , Encephalitis/genetics , Encephalitis/immunology , Encephalitis/physiopathology , Female , Humans , Inflammation/immunology , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Nervous System Diseases/metabolism , Neurons/physiology , Phagocytes/immunology , Phagocytes/metabolism , Phagocytosis/immunology , Phosphorylation , STAT1 Transcription Factor/physiology , Transcriptome/genetics
3.
Immunity ; 48(5): 937-950.e8, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768177

ABSTRACT

Infections are thought to trigger CD8+ cytotoxic T lymphocyte (CTL) responses during autoimmunity. However, the transcriptional programs governing the tissue-destructive potential of CTLs remain poorly defined. In a model of central nervous system (CNS) inflammation, we found that infection with lymphocytic choriomeningitis virus (LCMV), but not Listeria monocytogenes (Lm), drove autoimmunity. The DNA-binding factor TOX was induced in CTLs during LCMV infection and was essential for their encephalitogenic properties, and its expression was inhibited by interleukin-12 during Lm infection. TOX repressed the activity of several transcription factors (including Id2, TCF-1, and Notch) that are known to drive CTL differentiation. TOX also reduced immune checkpoint sensitivity by restraining the expression of the inhibitory checkpoint receptor CD244 on the surface of CTLs, leading to increased CTL-mediated damage in the CNS. Our results identify TOX as a transcriptional regulator of tissue-destructive CTLs in autoimmunity, offering a potential mechanistic link to microbial triggers.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Homeodomain Proteins/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Adult , Aged , Animals , Autoimmunity/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Signaling Lymphocytic Activation Molecule Family/immunology , T-Lymphocytes, Cytotoxic/immunology
4.
Acta Neuropathol ; 145(3): 335-355, 2023 03.
Article in English | MEDLINE | ID: mdl-36695896

ABSTRACT

B cells contribute to the pathogenesis of both cellular- and humoral-mediated central nervous system (CNS) inflammatory diseases through a variety of mechanisms. In such conditions, B cells may enter the CNS parenchyma and contribute to local tissue destruction. It remains unexplored, however, how infection and autoimmunity drive transcriptional phenotypes, repertoire features, and antibody functionality. Here, we profiled B cells from the CNS of murine models of intracranial (i.c.) viral infections and autoimmunity. We identified a population of clonally expanded, antibody-secreting cells (ASCs) that had undergone class-switch recombination and extensive somatic hypermutation following i.c. infection with attenuated lymphocytic choriomeningitis virus (rLCMV). Recombinant expression and characterisation of these antibodies revealed specificity to viral antigens (LCMV glycoprotein GP), correlating with ASC persistence in the brain weeks after resolved infection. Furthermore, these virus-specific ASCs upregulated proliferation and expansion programs in response to the conditional and transient induction of the LCMV GP as a neo-self antigen by astrocytes. This class-switched, clonally expanded, and mutated population persisted and was even more pronounced when peripheral B cells were depleted prior to autoantigen induction in the CNS. In contrast, the most expanded B cell clones in mice with persistent expression of LCMV GP in the CNS did not exhibit neo-self antigen specificity, potentially a consequence of local tolerance induction. Finally, a comparable population of clonally expanded, class-switched, and proliferating ASCs was detected in the cerebrospinal fluid of relapsing multiple sclerosis (RMS) patients. Taken together, our findings support the existence of B cells that populate the CNS and are capable of responding to locally encountered autoantigens.


Subject(s)
Antibody-Producing Cells , Autoantigens , Mice , Animals , B-Lymphocytes , Lymphocytic choriomeningitis virus , Brain
6.
Clin Nephrol ; 99(5): 260-264, 2023 May.
Article in English | MEDLINE | ID: mdl-36928260

ABSTRACT

In an era of increased accessibility to genetic testing, nephrologists may be able to better understand pathophysiologic mechanisms by which their patients develop specific conditions. In this study, we describe clinical and genetic findings of two patients with kidney cysts, who were found to have variants in HOGA1, a mitochondrial 4-hydroxy-2-oxoglutarate aldolase enzyme associated with primary hyperoxaluria type 3 and the development of oxalate-containing kidney stones. We describe possible mechanisms by which mutations in this enzyme could result in the kidney cyst formation seen in our two patients. We propose that patients with mutations in HOGA1 are predisposed to crystal or stone deposition, tubule dilation, and inflammasome activation, which can result in kidney cyst formation.


Subject(s)
Cysts , Hyperoxaluria, Primary , Kidney Calculi , Oxo-Acid-Lyases , Humans , Hyperoxaluria, Primary/genetics , Kidney , Oxo-Acid-Lyases/chemistry , Oxo-Acid-Lyases/genetics
7.
Genes Immun ; 23(6): 183-195, 2022 09.
Article in English | MEDLINE | ID: mdl-36028771

ABSTRACT

Adaptive immune repertoires are composed by the ensemble of B and T-cell receptors within an individual, reflecting both past and current immune responses. Recent advances in single-cell sequencing enable recovery of the complete adaptive immune receptor sequences in addition to transcriptional information. Here, we recovered transcriptome and immune repertoire information for polyclonal T follicular helper cells following lymphocytic choriomeningitis virus (LCMV) infection, CD8+ T cells with binding specificity restricted to two distinct LCMV peptides, and B and T cells isolated from the nervous system in the context of experimental autoimmune encephalomyelitis. We could relate clonal expansion, germline gene usage, and clonal convergence to cell phenotypes spanning activation, memory, naive, antibody secretion, T-cell inflation, and regulation. Together, this dataset provides a resource for immunologists that can be integrated with future single-cell immune repertoire and transcriptome sequencing datasets.


Subject(s)
Autoimmunity , Lymphocytic Choriomeningitis , Animals , CD8-Positive T-Lymphocytes , Disease Models, Animal , Lymphocytic Choriomeningitis/genetics , Mice , Mice, Inbred C57BL , Peptides , Receptors, Antigen, T-Cell/genetics
8.
J Immunol ; 203(9): 2377-2387, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31548330

ABSTRACT

Flotillin-1 (Flot1) is an evolutionary conserved, ubiquitously expressed lipid raft-associated scaffolding protein. Migration of Flot1-deficient neutrophils is impaired because of a decrease in myosin II-mediated contractility. Flot1 also accumulates in the uropod of polarized T cells, suggesting an analogous role in T cell migration. In this study, we analyzed morphology and migration parameters of murine wild-type and Flot1-/- CD8+ T cells using in vitro assays and intravital two-photon microscopy of lymphoid and nonlymphoid tissues. Flot1-/- CD8+ T cells displayed significant alterations in cell shape and motility parameters in vivo but showed comparable homing to lymphoid organs and intact in vitro migration to chemokines. Furthermore, their clonal expansion and infiltration into nonlymphoid tissues during primary and secondary antiviral immune responses was comparable to wild-type CD8+ T cells. Taken together, Flot1 plays a detectable but unexpectedly minor role for CD8+ T cell behavior under physiological conditions.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Membrane Proteins/physiology , Animals , CD8-Positive T-Lymphocytes/physiology , Cell Movement , Epidermis/immunology , Female , Immunologic Memory , Lymphocyte Activation , Male , Membrane Microdomains/physiology , Mice , Mice, Inbred C57BL
9.
Nat Commun ; 14(1): 3106, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37253744

ABSTRACT

Blood-brain barrier (BBB) breakdown and immune cell infiltration into the central nervous system (CNS) are early hallmarks of multiple sclerosis (MS). High numbers of CD8+ T cells are found in MS lesions, and antigen (Ag) presentation at the BBB has been proposed to promote CD8+ T cell entry into the CNS. Here, we show that brain endothelial cells process and cross-present Ag, leading to effector CD8+ T cell differentiation. Under physiological flow in vitro, endothelial Ag presentation prevented CD8+ T cell crawling and diapedesis resulting in brain endothelial cell apoptosis and BBB breakdown. Brain endothelial Ag presentation in vivo was limited due to Ag uptake by CNS-resident macrophages but still reduced motility of Ag-specific CD8+ T cells within CNS microvessels. MHC class I-restricted Ag presentation at the BBB during neuroinflammation thus prohibits CD8+ T cell entry into the CNS and triggers CD8+ T cell-mediated focal BBB breakdown.


Subject(s)
Blood-Brain Barrier , Multiple Sclerosis , Humans , Blood-Brain Barrier/metabolism , CD8-Positive T-Lymphocytes , Endothelial Cells/metabolism , Central Nervous System/metabolism , Histocompatibility Antigens Class I/metabolism
10.
Front Immunol ; 14: 1085895, 2023.
Article in English | MEDLINE | ID: mdl-37153600

ABSTRACT

Members of the Regulator of G-protein signaling (Rgs) family regulate the extent and timing of G protein signaling by increasing the GTPase activity of Gα protein subunits. The Rgs family member Rgs1 is one of the most up-regulated genes in tissue-resident memory (TRM) T cells when compared to their circulating T cell counterparts. Functionally, Rgs1 preferentially deactivates Gαq, and Gαi protein subunits and can therefore also attenuate chemokine receptor-mediated immune cell trafficking. The impact of Rgs1 expression on tissue-resident T cell generation, their maintenance, and the immunosurveillance of barrier tissues, however, is only incompletely understood. Here we report that Rgs1 expression is readily induced in naïve OT-I T cells in vivo following intestinal infection with Listeria monocytogenes-OVA. In bone marrow chimeras, Rgs1 -/- and Rgs1 +/+ T cells were generally present in comparable frequencies in distinct T cell subsets of the intestinal mucosa, mesenteric lymph nodes, and spleen. After intestinal infection with Listeria monocytogenes-OVA, however, OT-I Rgs1 +/+ T cells outnumbered the co-transferred OT-I Rgs1- /- T cells in the small intestinal mucosa already early after infection. The underrepresentation of the OT-I Rgs1 -/- T cells persisted to become even more pronounced during the memory phase (d30 post-infection). Remarkably, upon intestinal reinfection, mice with intestinal OT-I Rgs1 +/+ TRM cells were able to prevent the systemic dissemination of the pathogen more efficiently than those with OT-I Rgs1 -/- TRM cells. While the underlying mechanisms are not fully elucidated yet, these data thus identify Rgs1 as a critical regulator for the generation and maintenance of tissue-resident CD8+ T cells as a prerequisite for efficient local immunosurveillance in barrier tissues in case of reinfections with potential pathogens.


Subject(s)
CD8-Positive T-Lymphocytes , GTP-Binding Proteins , Listeria monocytogenes , Animals , Mice , GTP-Binding Proteins/metabolism , Protein Subunits/metabolism , T-Lymphocyte Subsets
11.
J Autoimmun ; 39(3): 143-53, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22727561

ABSTRACT

After a long period where the potential of therapeutic peptides was let into oblivion and even dismissed, there is a revival of interest in peptides as potential drug candidates. Novel strategies for limiting metabolism and improve their bioavailability, and alternative routes of administration have emerged. This resulted in a large number of peptide-based drugs that are now being marketed in different indications. Regarding autoimmunity, successful data have been reported in numerous mouse models of autoimmune inflammation, yet relatively few clinical trials based on synthetic peptides are currently underway. This review reports on peptides that show much promises in appropriate mouse models of autoimmunity and describes in more detail clinical trials based on peptides for treating autoimmune patients. A particular emphasis is given to the 21-mer peptide P140/Lupuzor that has completed successfully phase I, phase IIa and phase IIb clinical trials for systemic lupus erythematosus.


Subject(s)
Autoimmune Diseases/drug therapy , Immunologic Factors/pharmacokinetics , Lupus Erythematosus, Systemic/drug therapy , Peptide Fragments/pharmacokinetics , Amino Acid Sequence , Animals , Autoimmune Diseases/immunology , Autoimmunity/drug effects , Biological Availability , Clinical Trials as Topic , Drug Administration Routes , Drug Administration Schedule , Humans , Immunologic Factors/administration & dosage , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/mortality , Mice , Molecular Sequence Data , Peptide Fragments/administration & dosage , Survival Rate
12.
Sci Adv ; 8(23): eabl5162, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35675399

ABSTRACT

Tumor-associated lymphatic vessels promote metastasis and regulate antitumor immune responses. Here, we assessed the impact of cytotoxic T cells on the local lymphatic vasculature and concomitant tumor dissemination during an antitumor response. Interferon-γ (IFN-γ) released by effector T cells enhanced the expression of immunosuppressive markers by tumor-associated lymphatic endothelial cells (LECs). However, at higher effector T cell densities within the tumor, T cell-based immunotherapies induced LEC apoptosis and decreased tumor lymphatic vessel density. As a consequence, lymphatic flow was impaired, and lymph node metastasis was reduced. Mechanistically, T cell-mediated tumor cell death induced the release of tumor antigens and cross-presentation by tumor LECs, resulting in antigen-specific LEC killing by T cells. When LECs lacked the IFN-γ receptor expression, LEC killing was abrogated, indicating that IFN-γ is indispensable for reducing tumor-associated lymphatic vessel density and drainage. This study provides insight into how cytotoxic T cells modulate tumor lymphatic vessels and may help to improve immunotherapeutic protocols.


Subject(s)
Endothelial Cells , Interferon-gamma , Antigens, Neoplasm , Cross-Priming , Endothelial Cells/metabolism , Humans , Interferon-gamma/metabolism , Lymphatic Metastasis
13.
Sci Transl Med ; 14(640): eabl6058, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35417190

ABSTRACT

In chronic inflammatory diseases of the central nervous system (CNS), immune cells persisting behind the blood-brain barrier are supposed to promulgate local tissue destruction. The drivers of such compartmentalized inflammation remain unclear, but tissue-resident memory T cells (TRM) represent a potentially important cellular player in this process. Here, we investigated whether resting CD8+ TRM persisting after cleared infection with attenuated lymphocytic choriomeningitis virus (LCMV) can initiate immune responses directed against cognate self-antigen in the CNS. We demonstrated that time-delayed conditional expression of the LCMV glycoprotein as neo-self-antigen by glia cells reactivated CD8+ TRM. Subsequently, CD8+ TRM expanded and initiated CNS inflammation and immunopathology in an organ-autonomous manner independently of circulating CD8+ T cells. However, in the absence of CD4+ T cells, TCF-1+ CD8+ TRM failed to expand and differentiate into terminal effectors. Similarly, in human demyelinating CNS autoimmune lesions, we found CD8+ T cells expressing TCF-1 that predominantly exhibited a TRM-like phenotype. Together, our study provides evidence for CD8+ TRM-driven CNS immunopathology and sheds light on why inflammatory processes may evade current immunomodulatory treatments in chronic autoimmune CNS conditions.


Subject(s)
CD8-Positive T-Lymphocytes , Immunologic Memory , Autoantigens , CD4-Positive T-Lymphocytes , Central Nervous System , Humans , Inflammation , Lymphocytic choriomeningitis virus
14.
Ann Rheum Dis ; 70(5): 837-43, 2011 May.
Article in English | MEDLINE | ID: mdl-21173017

ABSTRACT

BACKGROUND: The P140 phosphopeptide issued from the spliceosomal U1-70K small nuclear ribonucleoprotein protein displays protective properties in MRL/lpr lupus-prone mice. It binds both major histocompatibility class II (MHCII) and HSC70/Hsp73 molecules. P140 peptide increases MRL/lpr peripheral blood lymphocyte apoptosis and decreases autoepitope recognition by T cells. OBJECTIVE: To explore further the mode of action of P140 peptide on HSC70+ antigen-presenting cells. METHODS: P140 biodistribution was monitored in real time using an imaging system and by fluorescence and electron microscopy. Fluorescence activated cell sorting and Western blotting experiments were used to evaluate the P140 effects on autophagic flux markers. RESULTS: P140 fluorescence accumulated especially in the lungs and spleen. P140 peptide reduced the number of peripheral and splenic T and B cells without affecting these cells in normal mice. Remaining MRL/lpr B cells responded normally to mitogens. P140 peptide decreased the expression levels of HSC70/Hsp73 chaperone and stable MHCII dimers, which are both increased in MRL/lpr splenic B cells. It impaired refolding properties of chaperone HSC70. In MRL/lpr B cells, it increased the accumulation of the autophagy markers p62/SQSTM1 and LC3-II, consistent with a downregulated lysosomal degradation during autophagic flux. CONCLUSION: The study results suggest that after P140 peptide binding to HSC70, the endogenous (auto)antigen processing might be greatly affected in MRL/lpr antigen-presenting B cells, leading to the observed decrease of autoreactive T-cell priming and signalling via a mechanism involving a lysosomal degradation pathway. This unexpected mechanism might explain the beneficial effect of P140 peptide in treated MRL/lpr mice.


Subject(s)
Autophagy/drug effects , HSC70 Heat-Shock Proteins/antagonists & inhibitors , Histocompatibility Antigens Class II/drug effects , Lupus Erythematosus, Systemic/pathology , Peptide Fragments/pharmacology , Animals , Antigen Presentation/drug effects , B-Lymphocytes/pathology , HSC70 Heat-Shock Proteins/metabolism , Histocompatibility Antigens Class II/metabolism , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred MRL lpr , Peptide Fragments/pharmacokinetics , Peptide Fragments/therapeutic use , Signal Transduction/drug effects , Tissue Distribution
15.
Nat Commun ; 12(1): 1009, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33579927

ABSTRACT

Self-reactive CD8+ T cells are important mediators of progressive tissue damage in autoimmune diseases, but the molecular program underlying these cells' functional adaptation is unclear. Here we characterize the transcriptional and epigenetic landscape of self-reactive CD8+ T cells in a mouse model of protracted central nervous system (CNS) autoimmunity and compare it to populations of CNS-resident memory CD8+ T cells emerging from acute viral infection. We find that autoimmune CD8+ T cells persisting at sites of self-antigen exhibit characteristic transcriptional regulation together with distinct epigenetic remodeling. This self-reactive CD8+ T cell fate depends on the transcriptional regulation by the DNA-binding HMG-box protein TOX which remodels more than 400 genomic regions including loci such as Tcf7, which is central to stemness of CD8+ T cells. Continuous exposure to CNS self-antigen sustains TOX levels in self-reactive CD8+ T cells, whereas genetic ablation of TOX in CD8+ T cells results in shortened persistence of self-reactive CD8+ T cells in the inflamed CNS. Our study establishes and characterizes the genetic differentiation program enabling chronic T cell-driven immunopathology in CNS autoimmunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Central Nervous System/metabolism , Chromatin Assembly and Disassembly , Animals , Autoimmunity/immunology , Female , Gene Expression Regulation , Immunologic Memory , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Cell Metab ; 33(11): 2231-2246.e8, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34687652

ABSTRACT

Autoimmunity is energetically costly, but the impact of a metabolically active state on immunity and immune-mediated diseases is unclear. Ly6Chi monocytes are key effectors in CNS autoimmunity with an elusive role in priming naive autoreactive T cells. Here, we provide unbiased analysis of the immune changes in various compartments during cold exposure and show that this energetically costly stimulus markedly ameliorates active experimental autoimmune encephalomyelitis (EAE). Cold exposure decreases MHCII on monocytes at steady state and in various inflammatory mouse models and suppresses T cell priming and pathogenicity through the modulation of monocytes. Genetic or antibody-mediated monocyte depletion or adoptive transfer of Th1- or Th17-polarized cells for EAE abolishes the cold-induced effects on T cells or EAE, respectively. These findings provide a mechanistic link between environmental temperature and neuroinflammation and suggest competition between cold-induced metabolic adaptations and autoimmunity as energetic trade-off beneficial for the immune-mediated diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Neuroinflammatory Diseases , Adoptive Transfer , Animals , Autoimmunity , Mice , Mice, Inbred C57BL , Th17 Cells
17.
Sci Immunol ; 5(46)2020 04 03.
Article in English | MEDLINE | ID: mdl-32245888

ABSTRACT

It is well established that tissue macrophages and tissue-resident memory CD8+ T cells (TRM) play important roles for pathogen sensing and rapid protection of barrier tissues. In contrast, the mechanisms by which these two cell types cooperate for homeostatic organ surveillance after clearance of infections is poorly understood. Here, we used intravital imaging to show that TRM dynamically followed tissue macrophage topology in noninflamed murine submandibular salivary glands (SMGs). Depletion of tissue macrophages interfered with SMG TRM motility and caused a reduction of interepithelial T cell crossing. In the absence of macrophages, SMG TRM failed to cluster in response to local inflammatory chemokines. A detailed analysis of the SMG microarchitecture uncovered discontinuous attachment of tissue macrophages to neighboring epithelial cells, with occasional macrophage protrusions bridging adjacent acini and ducts. When dissecting the molecular mechanisms that drive homeostatic SMG TRM motility, we found that these cells exhibit a wide range of migration modes: In addition to chemokine- and adhesion receptor-driven motility, resting SMG TRM displayed a remarkable capacity for autonomous motility in the absence of chemoattractants and adhesive ligands. Autonomous SMG TRM motility was mediated by friction and insertion of protrusions into gaps offered by the surrounding microenvironment. In sum, SMG TRM display a unique continuum of migration modes, which are supported in vivo by tissue macrophages to allow homeostatic patrolling of the complex SMG architecture.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Homeostasis/immunology , Macrophages/immunology , Salivary Glands/immunology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Specificity/immunology , Surveys and Questionnaires
18.
J Cell Biol ; 156(5): 829-41, 2002 Mar 04.
Article in English | MEDLINE | ID: mdl-11877459

ABSTRACT

In Saccharomyces cerevisiae, Bud8p and Bud9p are homologous plasma membrane glycoproteins that appear to mark the distal and proximal cell poles, respectively, as potential sites for budding in the bipolar pattern. Here we provide evidence that Bud8p is delivered to the presumptive bud site (and thence to the distal pole of the bud) just before bud emergence, and that Bud9p is delivered to the bud side of the mother-bud neck (and thence to the proximal pole of the daughter cell) after activation of the mitotic exit network, just before cytokinesis. Like the delivery of Bud8p, that of Bud9p is actin dependent; unlike the delivery of Bud8p, that of Bud9p is also septin dependent. Interestingly, although the transcription of BUD8 and BUD9 appears to be cell cycle regulated, the abundance of BUD8 mRNA peaks in G2/M and that of BUD9 mRNA peaks in late G1, suggesting that the translation and/or delivery to the cell surface of each protein is delayed and presumably also cell cycle regulated. The importance of time of transcription in localization is supported by promoter-swap experiments: expression of Bud8p from the BUD9 promoter leads to its localization predominantly to the sites typical for Bud9p, and vice versa. Moreover, expression of Bud8p from the BUD9 promoter fails to rescue the budding-pattern defect of a bud8 mutant but fully rescues that of a bud9 mutant. However, although expression of Bud9p from the BUD8 promoter fails to rescue a bud9 mutant, it also rescues only partially the budding-pattern defect of a bud8 mutant, suggesting that some feature(s) of the Bud8p protein is also important for Bud8p function. Experiments with chimeric proteins suggest that the critical element(s) is somewhere in the extracytoplasmic domain of Bud8p.


Subject(s)
Cell Cycle/physiology , Cell Polarity/physiology , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal/physiology , Membrane Glycoproteins , Protein Transport/physiology , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/metabolism , Actins/metabolism , Cell Membrane/metabolism , Cytoplasm/metabolism , Fungal Proteins/genetics , Phenotype , RNA, Messenger/metabolism , Saccharomyces cerevisiae/genetics
19.
Front Immunol ; 10: 2086, 2019.
Article in English | MEDLINE | ID: mdl-31552034

ABSTRACT

T cell activation in lymphoid tissue occurs through interactions with cognate peptide-major histocompatibility complex (pMHC)-presenting dendritic cells (DCs). Intravital imaging studies using ex vivo peptide-pulsed DCs have uncovered that cognate pMHC levels imprint a wide range of dynamic contacts between these two cell types. T cell-DC interactions vary between transient, "kinapse-like" contacts at low to moderate pMHC levels to immediate "synapse-like" arrest at DCs displaying high pMHC levels. To date, it remains unclear whether this pattern is recapitulated when the immune system faces a replicative agent, such as a virus, at low and high inoculum. Here, we locally administered low and high inoculum of lymphocytic choriomeningitis virus (LCMV) in mice to follow activation parameters of Ag-specific CD4+ and CD8+ T cells in draining lymph nodes (LNs) during the first 72 h post infection. We correlated these data with kinapse- and synapse-like motility patterns of Ag-specific T cells obtained by intravital imaging of draining LNs. Our data show that initial viral inoculum controls immediate synapse-like T cell arrest vs. continuous kinapse-like motility. This remains the case when the viral inoculum and thus the inflammatory microenvironment in draining LNs remains identical but cognate pMHC levels vary. Our data imply that the Ag-processing capacity of draining LNs is equipped to rapidly present high levels of cognate pMHC when antigenic material is abundant. Our findings further suggest that widespread T cell arrest during the first 72 h of an antimicrobial immune responses is not required to trigger proliferation. In sum, T cells adapt their scanning behavior according to available antigen levels during viral infections, with dynamic changes in motility occurring before detectable expression of early activation markers.


Subject(s)
Cell Movement/immunology , Immunological Synapses/immunology , Lymph Nodes/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , Virus Diseases/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lymphocyte Activation/genetics , Lymphocytic choriomeningitis virus/immunology , Mice , T-Lymphocyte Subsets/metabolism , Virus Diseases/metabolism , Virus Diseases/virology
20.
Cell Rep ; 28(1): 21-29.e6, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31269441

ABSTRACT

Regulatory T cells (Tregs) play a crucial role in controlling autoimmune and inflammatory responses. Recent studies have demonstrated that dendritic cells (DCs) contribute to the homeostasis of peripheral Tregs. Autophagy, a critical pathway for cellular homeostasis, is active in DCs and is upregulated in different inflammatory conditions. We have shown that Tregs are expanded and have phenotypic alterations and impaired suppressive functions in mice with autophagy-deficient DCs. RNA profiling of Tregs revealed that autophagy in DCs is required to stabilize Treg expression signatures. This phenotype is linked to the downregulation of ICOS-Ligand expression in autophagy-deficient DCs, a consequence of the accumulation of ADAM10, the metalloproteinase responsible for its cleavage. Upon inflammation, in antigen-induced arthritis, mice with autophagy-deficient DCs exhibit increased synovial inflammation and cartilage and bone erosion correlating with Treg-to-Th17 conversion. Our data reveal a mechanism that couples autophagy deficiency in DCs to the function, homeostasis, and stability of Tregs.


Subject(s)
Autophagy-Related Protein 5/metabolism , Dendritic Cells/immunology , Macroautophagy/genetics , T-Lymphocytes, Regulatory/immunology , Animals , Autophagy-Related Protein 5/genetics , Dendritic Cells/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeostasis/immunology , Inducible T-Cell Co-Stimulator Ligand/genetics , Inducible T-Cell Co-Stimulator Ligand/metabolism , Inflammation/genetics , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL