Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 395
Filter
Add more filters

Publication year range
1.
Immunity ; 38(2): 237-49, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23313588

ABSTRACT

Interactions with antigen-presenting cells (APCs) interrupt T cell migration through tissues and trigger signaling pathways that converge on the activation of transcriptional regulators, including nuclear factor of activated T cells (NFAT), which control T cell function and differentiation. Both stable and unstable modes of cognate T cell-APC interactions have been observed in vivo, but the functional significance of unstable, serial contacts has remained unclear. Here we used multiphoton intravital microscopy in lymph nodes and tumors to show that while NFAT nuclear import was fast (t(1/2 max)∼1 min), nuclear export was slow (t(1/2)∼20 min) in T cells. During delayed export, nuclear NFAT constituted a short-term imprint of transient TCR signals and remained transcriptionally active for the T cell tolerance gene Egr2, but not for the effector gene Ifng, which required continuous TCR triggering for expression. This provides a potential mechanistic basis for the observation that a predominance of unstable APC interactions correlates with the induction of T cell tolerance.


Subject(s)
Antigen-Presenting Cells/metabolism , Immune Tolerance , Immunologic Memory , Lymph Nodes/metabolism , NFATC Transcription Factors/genetics , T-Lymphocytes/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Cell Communication , Cell Differentiation , Cell Movement , Cell Nucleus/metabolism , Cytosol/metabolism , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/immunology , Gene Expression Regulation , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Microscopy, Fluorescence, Multiphoton , NFATC Transcription Factors/immunology , Protein Transport , Receptors, Antigen, T-Cell , Signal Transduction , T-Lymphocytes/immunology , Tumor Cells, Cultured
2.
J Immunol ; 205(8): 2265-2275, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32917787

ABSTRACT

Loss of immune tolerance to gut microflora is inextricably linked to chronic intestinal inflammation and colitis-associated colorectal cancer (CAC). The LRP5/6 signaling cascade in APCs contributes to immune homeostasis in the gut, but whether this pathway in APCs protects against CAC is not known. In the current study, using a mouse model of CAC, we show that the LRP5/6-ß-catenin-IL-10 signaling axis in intestinal CD11c+ APCs protects mice from CAC by regulating the expression of tumor-promoting inflammatory factors in response to commensal flora. Genetic deletion of LRP5/6 in CD11c+ APCs in mice (LRP5/6ΔCD11c) resulted in enhanced susceptibility to CAC. This is due to a microbiota-dependent increased expression of proinflammatory factors and decreased expression of the immunosuppressive cytokine IL-10. This condition could be improved in LRP5/6ΔCD11c mice by depleting the gut flora, indicating the importance of LRP5/6 in mediating immune tolerance to the gut flora. Moreover, mechanistic studies show that LRP5/6 suppresses the expression of tumor-promoting inflammatory factors in CD11c+ APCs via the ß-catenin-IL-10 axis. Accordingly, conditional activation of ß-catenin specifically in CD11c+ APCs or in vivo administration of IL-10 protected LRP5/6ΔCD11c mice from CAC by suppressing the expression of inflammatory factors. In summary, in this study, we identify a key role for the LRP5/6-ß-catenin-IL-10 signaling pathway in intestinal APCs in resolving chronic intestinal inflammation and protecting against CAC in response to the commensal flora.


Subject(s)
Antigen-Presenting Cells/immunology , Colitis/immunology , Colonic Neoplasms/immunology , Gastrointestinal Microbiome/immunology , Interleukin-10/immunology , Wnt Signaling Pathway/immunology , beta Catenin/immunology , Animals , Antigen-Presenting Cells/pathology , Colitis/complications , Colitis/genetics , Colitis/pathology , Colonic Neoplasms/etiology , Colonic Neoplasms/genetics , Colonic Neoplasms/prevention & control , Gastrointestinal Microbiome/genetics , Interleukin-10/genetics , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics
3.
Proc Natl Acad Sci U S A ; 116(19): 9533-9542, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31019077

ABSTRACT

T cell-invigorating cancer immunotherapies have near-curative potential. However, their clinical benefit is currently limited, as only a fraction of patients respond, suggesting that these regimens may benefit from combination with tumor-targeting treatments. As oncogenic progression is accompanied by alterations in metabolic pathways, tumors often become heavily reliant on antioxidant machinery and may be susceptible to increases in oxidative stress. The cystine-glutamate antiporter xCT is frequently overexpressed in cancer and fuels the production of the antioxidant glutathione; thus, tumors prone to redox stress may be selectively vulnerable to xCT disruption. However, systemic inhibition of xCT may compromise antitumor immunity, as xCT is implicated in supporting antigen-induced T cell proliferation. Therefore, we utilized immune-competent murine tumor models to investigate whether cancer cell expression of xCT was required for tumor growth in vivo and if deletion of host xCT impacted antitumor immune responses. Deletion of xCT in tumor cells led to defective cystine uptake, accumulation of reactive oxygen species, and impaired tumor growth, supporting a cancer cell-autonomous role for xCT. In contrast, we observed that, although T cell proliferation in culture was exquisitely dependent on xCT expression, xCT was dispensable for T cell proliferation in vivo and for the generation of primary and memory immune responses to tumors. These findings prompted the combination of tumor cell xCT deletion with the immunotherapeutic agent anti-CTLA-4, which dramatically increased the frequency and durability of antitumor responses. Together, these results identify a metabolic vulnerability specific to tumors and demonstrate that xCT disruption can expand the efficacy of anticancer immunotherapies.


Subject(s)
Amino Acid Transport System y+/deficiency , Antigen-Presenting Cells/immunology , Cell Proliferation , Immunologic Memory , Neoplasms, Experimental/immunology , T-Lymphocytes/immunology , Amino Acid Transport System y+/immunology , Animals , Antigen-Presenting Cells/pathology , Cell Line , Gene Deletion , Glutathione/genetics , Glutathione/immunology , Immunotherapy , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , T-Lymphocytes/pathology
4.
Proc Natl Acad Sci U S A ; 116(30): 15134-15139, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31285344

ABSTRACT

B cells play important roles in autoimmune diseases through autoantibody production, cytokine secretion, or antigen presentation to T cells. In most cases, the contribution of B cells as antigen-presenting cells is not well understood. We have studied the autoantibody response against the enzyme transglutaminase 2 (TG2) in celiac disease patients by generating recombinant antibodies from single gut plasma cells reactive with discrete antigen domains and by undertaking proteomic analysis of anti-TG2 serum antibodies. The majority of the cells recognized epitopes in the N-terminal domain of TG2. Antibodies recognizing C-terminal epitopes interfered with TG2 cross-linking activity, and B cells specific for C-terminal epitopes were inefficient at taking up TG2-gluten complexes for presentation to gluten-specific T cells. The bias toward N-terminal epitopes hence reflects efficient T-B collaboration. Production of antibodies against N-terminal epitopes coincided with clinical onset of disease, suggesting that TG2-reactive B cells with certain epitope specificities could be the main antigen-presenting cells for pathogenic, gluten-specific T cells. The link between B cell epitopes, antigen presentation, and disease onset provides insight into the pathogenic mechanisms of a T cell-mediated autoimmune condition.


Subject(s)
Antigen-Presenting Cells/immunology , B-Lymphocytes/immunology , Celiac Disease/immunology , Epitopes, B-Lymphocyte/immunology , GTP-Binding Proteins/immunology , T-Lymphocytes/immunology , Transglutaminases/immunology , Age of Onset , Antigen-Presenting Cells/pathology , Autoantibodies/biosynthesis , Autoantibodies/genetics , Autoantigens/genetics , Autoantigens/immunology , B-Lymphocytes/pathology , Celiac Disease/genetics , Celiac Disease/pathology , Duodenum/immunology , Duodenum/pathology , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/genetics , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics , Glutens/chemistry , Glutens/immunology , Humans , Immune Sera/chemistry , Immunoglobulin Light Chains/biosynthesis , Immunoglobulin Light Chains/genetics , Models, Molecular , Protein Binding , Protein Conformation , Protein Glutamine gamma Glutamyltransferase 2 , T-Lymphocytes/pathology , Transglutaminases/chemistry , Transglutaminases/genetics
5.
J Immunol ; 202(9): 2558-2569, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30926644

ABSTRACT

We have reported that the major histocompatibility molecule HLA-DQ2 (DQA1*05:01/DQB1*02:01) (DQ2) is relatively resistant to HLA-DM (DM), a peptide exchange catalyst for MHC class II. In this study, we analyzed the role of DQ2/DM interaction in the generation of DQ2-restricted gliadin epitopes, relevant to celiac disease, or DQ2-restricted viral epitopes, relevant to host defense. We used paired human APC, differing in DM expression (DMnull versus DMhigh) or differing by expression of wild-type DQ2, versus a DM-susceptible, DQ2 point mutant DQ2α+53G. The APC pairs were compared for their ability to stimulate human CD4+ T cell clones. Despite higher DQ2 levels, DMhigh APC attenuated T cell responses compared with DMnull APC after intracellular generation of four tested gliadin epitopes. DMhigh APC expressing the DQ2α+53G mutant further suppressed these gliadin-mediated responses. The gliadin epitopes were found to have moderate affinity for DQ2, and even lower affinity for the DQ2 mutant, consistent with DM suppression of their presentation. In contrast, DMhigh APC significantly promoted the presentation of DQ2-restricted epitopes derived intracellularly from inactivated HSV type 2, influenza hemagglutinin, and human papillomavirus E7 protein. When extracellular peptide epitopes were used as Ag, the DQ2 surface levels and peptide affinity were the major regulators of T cell responses. The differential effect of DM on stimulation of the two groups of T cell clones implies differences in DQ2 presentation pathways associated with nonpathogen- and pathogen-derived Ags in vivo.


Subject(s)
Antigen Presentation , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Celiac Disease/immunology , Epitopes, T-Lymphocyte/immunology , Gliadin/immunology , HLA-DQ Antigens/immunology , Viral Proteins/immunology , Virus Diseases/immunology , Antigen-Presenting Cells/pathology , CD4-Positive T-Lymphocytes/pathology , Celiac Disease/pathology , Cell Line , Humans
6.
Proc Natl Acad Sci U S A ; 115(39): E9153-E9161, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30201728

ABSTRACT

In vivo delivery of antigen-encoding mRNA is a promising approach to personalized cancer treatment. The therapeutic efficacy of mRNA vaccines is contingent on safe and efficient gene delivery, biological stability of the mRNA, and the immunological properties of the vaccine. Here we describe the development and evaluation of a versatile and highly efficient mRNA vaccine-delivery system that employs charge-altering releasable transporters (CARTs) to deliver antigen-coding mRNA to antigen-presenting cells (APCs). We demonstrate in human peripheral blood mononuclear cells that CART vaccines can activate a robust antigen-specific immune response against mRNA-encoded viral epitopes. In an established mouse model, we demonstrate that CARTs preferentially target professional APCs in secondary lymphoid organs upon i.v. injections and target local APCs upon s.c. injection. Finally, we show that CARTs coformulated with mRNA and a Toll-like receptor ligand simultaneously transfect and activate target cells to generate an immune response that can treat and cure mice with large, established tumors.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Immunity, Cellular , Neoplasms, Experimental/therapy , RNA, Messenger/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Vaccination , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Antigens, Neoplasm/genetics , Cancer Vaccines/genetics , Cancer Vaccines/pharmacology , Cell Line, Tumor , Female , HeLa Cells , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , RNA, Messenger/genetics , RNA, Messenger/pharmacology , T-Lymphocytes/pathology
7.
Int J Mol Sci ; 22(21)2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34768993

ABSTRACT

The latest literature demonstrates the predominant role of the programmed cell death axis (PD-1/PD-L1/PD-L2) in ovarian cancer (OC) pathogenesis. However, data concerning this issue is ambiguous. Our research aimed to evaluate the clinical importance of PD-L1/PD-L2 expression in OC environments. We evaluated the role of PD-L1/PD-L2 in OC patients (n = 53). The analysis was performed via flow cytometry on myeloid (mDCs) and plasmacytoid dendritic cells (pDCs) and monocytes/macrophages (MO/MA) in peripheral blood, peritoneal fluid (PF), and tumor tissue (TT). The data were correlated with clinicopathological characteristics and prognosis of OC patients. The concentration of soluble PD-L1 (sPD-L1) and PD-1 in the plasma and PF were determined by ELISA. We established an accumulation of PD-L1+/PD-L2+ mDCs, pDCs, and MA in the tumor microenvironment. We showed an elevated level of sPD-L1 in the PF of OC patients in comparison to plasma and healthy subjects. sPD-L1 levels in PF showed a positive relationship with Ca125 concentration. Moreover, we established an association between higher sPD-L1 levels in PF and shorter survival of OC patients. An accumulation of PD-L1+/PD-L2+ mDCs, pDCs, and MA in the TT and high sPD-L1 levels in PF could represent the hallmark of immune regulation in OC patients.


Subject(s)
Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/pathology , B7-H1 Antigen/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Adult , Aged , Aged, 80 and over , Ascitic Fluid/metabolism , Ascitic Fluid/pathology , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Female , Humans , Middle Aged , Prognosis , Tumor Microenvironment/physiology , Young Adult
8.
Circulation ; 139(22): 2554-2566, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31136220

ABSTRACT

BACKGROUND: Hypercholesterolemic mice lacking factors required for activation of CD4+ T cells are characterized by reduced development of atherosclerosis. Consequently, it has been assumed that atherosclerosis involves loss of tolerance against modified self-antigens generated in response to hypercholesterolemia and that presentation of such antigens on major histocompatibility complex class II (MHCII) leads to activation of proatherogenic Th1 cells. In this study, we wanted to determine the role of antigen presentation on MHCII in atherosclerosis development. METHODS: Apolipoprotein E (ApoE-/-) mice deficient for MHCII (ApoE-/-MHCII-/-) were used to study the role of MHCII in atherosclerosis development. RESULTS: Compared with ApoE-/- mice, ApoE-/-MHCII-/- mice had reduced levels of CD4+ T cells, immunoglobulin G and M levels, and Th1 and Th2 cytokines in plasma. CD8+ T cells were increased and regulatory T cells were reduced both in spleen and in lesions of ApoE-/-MHCII-/- mice. Decreased plasma levels of inflammatory cytokines in ApoE-/-MHCII-/- mice indicated reduced systemic inflammation. Despite this, ApoE-/-MHCII-/- mice had significantly more atherosclerosis as assessed by en face Oil Red O staining of the aorta (4.7±2.9% versus 1.9±1.3%; P<0.01) and cross-sectional area of subvalvular lesions (7.7±2.2×105 µm2 versus 4.6±2.8×105 µm2; P<0.05). Cell transfer and blocking antibody studies suggested that loss of regulatory T cells is the most important cause of aggravated atherosclerosis in ApoE-/-MHCII-/- mice. CONCLUSIONS: Our observations demonstrate that antigen presentation on MHCII has important protective functions in atherosclerosis and that this is primarily the result of activation of regulatory T cells. These findings have implications for understanding the possible risks and benefits of immunosuppressive therapy in patients with cardiovascular disease.


Subject(s)
Antigen-Presenting Cells/immunology , Aorta/immunology , Aortic Diseases/immunology , Atherosclerosis/immunology , Histocompatibility Antigens Class II/immunology , Plaque, Atherosclerotic , T-Lymphocytes, Regulatory/immunology , Animals , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/pathology , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Male , Mice, Knockout, ApoE , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
9.
J Neuroinflammation ; 17(1): 119, 2020 Apr 16.
Article in English | MEDLINE | ID: mdl-32299492

ABSTRACT

BACKGROUND: Many lines of evidence suggest that accumulation of aggregated alpha-synuclein (αSYN) in the Parkinson's disease (PD) brain causes infiltration of T cells. However, in which ways the stationary brain cells interact with the T cells remain elusive. Here, we identify astrocytes as potential antigen-presenting cells capable of activating T cells in the PD brain. Astrocytes are a major component of the nervous system, and accumulating data indicate that astrocytes can play a central role during PD progression. METHODS: To investigate the role of astrocytes in antigen presentation and T-cell activation in the PD brain, we analyzed post mortem brain tissue from PD patients and controls. Moreover, we studied the capacity of cultured human astrocytes and adult human microglia to act as professional antigen-presenting cells following exposure to preformed αSYN fibrils. RESULTS: Our analysis of post mortem brain tissue demonstrated that PD patients express high levels of MHC-II, which correlated with the load of pathological, phosphorylated αSYN. Interestingly, a very high proportion of the MHC-II co-localized with astrocytic markers. Importantly, we found both perivascular and infiltrated CD4+ T cells to be surrounded by MHC-II expressing astrocytes, confirming an astrocyte T cell cross-talk in the PD brain. Moreover, we showed that αSYN accumulation in cultured human astrocytes triggered surface expression of co-stimulatory molecules critical for T-cell activation, while cultured human microglia displayed very poor antigen presentation capacity. Notably, intercellular transfer of αSYN/MHC-II deposits occurred between astrocytes via tunneling nanotubes, indicating spreading of inflammation in addition to toxic protein aggregates. CONCLUSIONS: In conclusion, our data from histology and cell culture studies suggest an important role for astrocytes in antigen presentation and T-cell activation in the PD brain, highlighting astrocytes as a promising therapeutic target in the context of chronic inflammation.


Subject(s)
Antigen-Presenting Cells/metabolism , Astrocytes/metabolism , Brain/metabolism , Microglia/metabolism , Parkinson Disease/metabolism , Aged , Aged, 80 and over , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Astrocytes/immunology , Astrocytes/pathology , Brain/immunology , Brain/pathology , Cells, Cultured , Female , Humans , Male , Microglia/immunology , Microglia/pathology , Middle Aged , Parkinson Disease/immunology , Parkinson Disease/pathology
10.
Trends Immunol ; 38(7): 513-525, 2017 07.
Article in English | MEDLINE | ID: mdl-28511816

ABSTRACT

The mechanisms through which immune responses are generated against solid cancers are well characterized and knowledge of the immune evasion pathways exploited by these malignancies has grown considerably. However, for hematological cancers, which develop and disseminate quite differently than solid tumors, the pathways that regulate immune activation or tolerance are less clear. Growing evidence suggests that, while numerous immune escape pathways are shared between hematological and solid malignancies, several unique pathways are exploited by leukemia and lymphoma. Below we discuss immune evasion mechanisms in leukemia and lymphoma, highlighting key differences from solid tumors. A more complete characterization of the mechanisms of immune tolerance in hematological malignancies is critical to inform the development of future immunotherapeutic approaches.


Subject(s)
Antigen-Presenting Cells/immunology , Immune Evasion , Immune Tolerance , Immunotherapy/methods , Leukemia/immunology , Lymphoma/immunology , Animals , Antigen-Presenting Cells/pathology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Gene Expression Regulation , Humans , Leukemia/genetics , Leukemia/pathology , Leukemia/therapy , Lymphoma/genetics , Lymphoma/pathology , Lymphoma/therapy , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/immunology , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/pathology
11.
J Allergy Clin Immunol ; 144(4S): S46-S54, 2019 10.
Article in English | MEDLINE | ID: mdl-30731122

ABSTRACT

Mast cells are highly versatile in terms of their mode of activation by a host of stimuli and their ability to flexibly release a plethora of biologically highly active mediators. Within the immune system, mast cells can best be designated as an active nexus interlinking innate and adaptive immunity. Here we try to draw an arc from initiation of acute inflammatory reactions to microbial pathogens to development of adaptive immunity and allergies. This multifaceted nature of mast cells is made possible by interaction with multiple cell types of immunologic and nonimmunologic origin. Examples for the former include neutrophils, eosinophils, T cells, and professional antigen-presenting cells. These interactions allow mast cells to orchestrate inflammatory innate reactions and complex adaptive immunity, including the pathogenesis of allergies. Important partners of nonimmunologic origin include cells of the sensory neuronal system. The intimate association between mast cells and sensory nerve fibers allows bidirectional communication, leading to neurogenic inflammation. Evidence is accumulating that this mast cell/nerve crosstalk is of pathophysiologic relevance in patients with allergic diseases, such as asthma.


Subject(s)
Adaptive Immunity , Asthma/immunology , Immunity, Innate , Mast Cells/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Asthma/pathology , Cell Communication/immunology , Eosinophils/immunology , Eosinophils/pathology , Humans , Mast Cells/pathology , Neutrophils/immunology , Neutrophils/pathology , Sensory Receptor Cells/immunology , Sensory Receptor Cells/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
12.
Int Ophthalmol ; 40(6): 1603-1618, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32107692

ABSTRACT

PURPOSE: To review the role of antigen-presenting cells (APC) in the pathogenesis of ocular surface diseases (OSD). METHODS: A thorough literature search was performed in PubMed database. An additional search was made in Google Scholar to complete the collected items. RESULTS: APCs have the ability to initiate and direct immune responses and are found in most lymphoid and non-lymphoid tissues. APCs continuously sample their environment, present antigens to T cells and co-ordinate immune tolerance and responses. Many different types of APCs have been described and there is growing evidence that these cells are involved in the pathogenesis of OSD. OSD is a complex term for a myriad of disorders that are often characterized by ocular surface inflammation, tear film instability and impairment of vision. CONCLUSIONS: This review summarizes the current knowledge concerning the immunotopographical distribution of APCs in the normal ocular surface. APCs appear to play a critical role in the pathology of a number of conditions associated with OSD including infectious keratitis, ocular allergy, dry eye disease and pterygium.


Subject(s)
Antigen-Presenting Cells/immunology , Dry Eye Syndromes/immunology , Immunity, Cellular , Tears/metabolism , Antigen-Presenting Cells/pathology , Dry Eye Syndromes/metabolism , Dry Eye Syndromes/pathology , Humans
13.
Am J Physiol Renal Physiol ; 317(1): F77-F89, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31017008

ABSTRACT

In immune-mediated glomerular diseases like crescentic glomerulonephritis (cGN), inflammatory CD4+ T cells accumulate within the tubulointerstitial compartment in close contact to proximal and distal tubular epithelial cells and drive renal inflammation and tissue damage. However, whether renal epithelial cell populations play a role in the pathogenesis of cGN by modulating CD4+ T cell responses is less clear. In the present study, we aimed to investigate the potential of renal epithelial cells to function as antigen-presenting cells, thereby stimulating CD4+ T cell responses. Using a FACS-based protocol that allowed comparative analysis of cortical epithelial cell populations, we showed that particularly proximal tubular epithelial cells (PTECs) express molecules linked with antigen-presenting cell function, including major histocompatibility complex class II (MHCII), CD74, CD80, and CD86 in homeostasis and nephrotoxic nephritis, a murine model of cGN. Protein expression was visualized at the PTEC single cell level by imaging flow cytometry. Interestingly, we found inflammation-dependent regulation of epithelium-expressed CD74, CD80, and CD86, whereas MHCII expression was not altered. Antigen-specific stimulation of CD4+ T cells by PTECs in vitro supported CD4+ T cell survival and induced CD4+ T cell activation, proliferation, and inflammatory cytokine production. In patients with antineutrophil cytoplasmic antibody-associated glomerulonephritis, MHCII and CD74 were expressed by both proximal and distal tubules, whereas CD86 was predominantly expressed by proximal tubules. Thus, particularly PTECs have the potential to induce an inflammatory phenotype in CD4+ T cells in vitro, which might also play a role in the pathology of immune-mediated kidney disease.


Subject(s)
Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Epithelial Cells/immunology , Glomerulonephritis/immunology , Kidney Tubules, Proximal/immunology , Lymphocyte Activation , Paracrine Communication , Animals , Antibodies, Antineutrophil Cytoplasmic/immunology , Antibodies, Antineutrophil Cytoplasmic/metabolism , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/pathology , Antigens, Differentiation, B-Lymphocyte/immunology , Antigens, Differentiation, B-Lymphocyte/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cells, Cultured , Coculture Techniques , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Mice, Inbred C57BL , Phenotype , Signal Transduction
14.
Eur J Immunol ; 48(3): 546-548, 2018 03.
Article in English | MEDLINE | ID: mdl-29193031

ABSTRACT

Medullary thymic epithelial cell (mTEC)-restricted expression of autoimmune regulator (Aire) is essential for establishment of immune tolerance. Recently, Aire was also shown to be expressed in cells of hematopietic and reproductive lineages. Thus, the generation of Airefl/fl mouse strain enables the investigation of the cell-specific function of Aire.


Subject(s)
Immune Tolerance/genetics , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Animals , Antigen-Presenting Cells/pathology , Cell Lineage/genetics , Cell Lineage/immunology , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Polyendocrinopathies, Autoimmune/pathology , Reproduction/genetics , Reproduction/immunology , AIRE Protein
15.
Immunology ; 153(2): 179-189, 2018 02.
Article in English | MEDLINE | ID: mdl-28898395

ABSTRACT

The global obesity epidemic and its associated co-morbidities, including type 2 diabetes, cardiovascular disease and certain types of cancers, have drawn attention to the pivotal role of adipocytes in health and disease. Besides their 'classical' function in energy storage and release, adipocytes interact with adipose-tissue-resident immune cells, among which are lipid-responsive invariant natural killer T (iNKT) cells. The iNKT cells are activated by lipid antigens presented by antigen-presenting cells as CD1d/lipid complexes. Upon activation, iNKT cells can rapidly secrete soluble mediators that either promote or oppose inflammation. In lean adipose tissue, iNKT cells elicit a predominantly anti-inflammatory immune response, whereas obesity is associated with declining iNKT cell numbers. Recent work showed that adipocytes act as non-professional antigen-presenting cells for lipid antigens. Here, we discuss endogenous lipid antigen processing and presentation by adipocytes, and speculate on how these lipid antigens, together with 'environmental factors' such as tissue/organ environment and co-stimulatory signals, are able to influence the fate of adipose-tissue-resident iNKT cells, and thereby the role of these cells in obesity and its associated pathologies.


Subject(s)
Adipose Tissue/immunology , Antigen Presentation , Antigen-Presenting Cells/immunology , Antigens/immunology , Lipids/immunology , Natural Killer T-Cells/immunology , Obesity/immunology , Adipocytes/immunology , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Antigen-Presenting Cells/pathology , Antigens, CD1d/immunology , Humans , Natural Killer T-Cells/pathology , Obesity/pathology
16.
Blood ; 128(1): 72-81, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27103745

ABSTRACT

Donor T-cell immune responses can eradicate lymphomas after allogeneic hematopoietic stem cell transplantation (AHSCT), but can also damage healthy tissues resulting in harmful graft-versus-host disease (GVHD). Next-generation sequencing has recently identified many new genetic lesions in follicular lymphoma (FL). One such gene, tumor necrosis factor receptor superfamily 14 (TNFRSF14), abnormal in 40% of FL patients, encodes the herpes virus entry mediator (HVEM) which limits T-cell activation via ligation of the B- and T-lymphocyte attenuator. As lymphoma B cells can act as antigen-presenting cells, we hypothesized that TNFRSF14 aberrations that reduce HVEM expression could alter the capacity of FL B cells to stimulate allogeneic T-cell responses and impact the outcome of AHSCT. In an in vitro model of alloreactivity, human lymphoma B cells with TNFRSF14 aberrations had reduced HVEM expression and greater alloantigen-presenting capacity than wild-type lymphoma B cells. The increased immune-stimulatory capacity of lymphoma B cells with TNFRSF14 aberrations had clinical relevance, associating with higher incidence of acute GVHD in patients undergoing AHSCT. FL patients with TNFRSF14 aberrations may benefit from more aggressive immunosuppression to reduce harmful GVHD after transplantation. Importantly, this study is the first to demonstrate the impact of an acquired genetic lesion on the capacity of tumor cells to stimulate allogeneic T-cell immune responses which may have wider consequences for adoptive immunotherapy strategies.


Subject(s)
Graft vs Host Disease/genetics , Hematopoietic Stem Cell Transplantation , Lymphocyte Activation/genetics , Lymphoma, Follicular/genetics , Lymphoma, Follicular/therapy , Receptors, Tumor Necrosis Factor, Member 14/genetics , Adult , Aged , Allografts , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/pathology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Female , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , Humans , Lymphoma, Follicular/pathology , Male , Middle Aged , Receptors, Tumor Necrosis Factor, Member 14/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
17.
Cytokine ; 111: 33-40, 2018 11.
Article in English | MEDLINE | ID: mdl-30114627

ABSTRACT

Acute graft-versus-host disease (aGVHD) remains a major challenging complication of patients receiving allogeneic hematopoietic cell transplantation (allo-HCT). CD4+ effector T cells and their related cytokines mediate pathogenesis of aGVHD, in which donor-T-cell derived interleukin-22 (IL-22) was recently indicated to play a role. The role of recipient-derived IL-22 in aGVHD remains to be elucidated. By applying IL-22 knock out (IL-22KO) mice as recipients of allotransplant, we found recipient derived IL-22 alleviated aGVHD and improved survival of allotransplant recipients. Knock out of IL-22 in recipient increased levels of T-helper (Th1) 1 cells but decreased levels of regulatory T cells (Tregs) in target tissues of aGVHD. Levels of IL-22 increased in aGVHD mice. Recipient antigen presenting cells (APCs) are important sources of IL-22. IL-22 reduced activation of APCs in vitro. Defect of IL-22 in APCs resulted in increased polarization of Th1 cells but decreased level of Tregs in an in vitro co-culture system. Our data highlight an immunoregulatory function of recipient-derived IL-22 in aGVHD.


Subject(s)
Antigen-Presenting Cells/immunology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Interleukins/immunology , Acute Disease , Allografts , Animals , Antigen-Presenting Cells/pathology , Gene Knockdown Techniques , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Interleukins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Interleukin-22
18.
J Immunol ; 197(11): 4274-4282, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27799310

ABSTRACT

Discovery of immune tolerance mechanisms, which inhibit pre-existing autoimmune inflammation, may provide us with new strategies for treating autoimmune diseases. We have identified a CD8αα+MHC class II+ cell with professional APC capacity during our investigation on spontaneous recovery from autoimmune glomerulonephritis in a rat model. This cell actively invades inflamed target tissue and further terminates an ongoing autoimmune inflammation by selective killing of effector autoreactive T cells. In this study, we show that this cell used a cytotoxic machinery of Ly49s+ NK cells in killing of target T cells. Thus, this CD8αα+MHC class II+ cell was a dually functional Ag-presenting NK-like (AP-NK) cell. Following its coupling with target T cells through Ag presentation, killing stimulatory receptor Ly49s6 and coreceptor CD8αα on this cell used rat nonclassic MHC class I C/E16 on the target T cells as a ligand to initiate killing. Thus, activated effector T cells with elevated expression of rat nonclassic MHC class I C/E16 were highly susceptible to the killing by the CD8αα+ AP-NK cell. Granule cytolytic perforin/granzyme C from this cell subsequently mediated cytotoxicity. Thus, inhibition of granzyme C effectively attenuated the killing. As it can recognize and eliminate effector autoreactive T cells in the inflamed target tissue, the CD8αα+ AP-NK cell not only represents a new type of immune cell involved in immune tolerance, but it also is a potential candidate for developing a cell-based therapy for pre-existing autoimmune diseases.


Subject(s)
Antigen-Presenting Cells/immunology , Autoimmune Diseases/immunology , CD8 Antigens/immunology , Glomerulonephritis/immunology , Histocompatibility Antigens Class II/immunology , Killer Cells, Natural/immunology , Animals , Antigen-Presenting Cells/pathology , Autoimmune Diseases/pathology , Female , Glomerulonephritis/pathology , Granzymes/immunology , Histocompatibility Antigens Class I/immunology , Inflammation/immunology , Inflammation/pathology , Killer Cells, Natural/pathology , Rats , Rats, Inbred Lew , Rats, Inbred WKY
19.
Mol Ther ; 25(7): 1676-1685, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28408181

ABSTRACT

Targeted approaches to treat autoimmune diseases would improve upon current therapies that broadly suppress the immune system and lead to detrimental side effects. Antigen-specific tolerance was induced using poly(lactide-co-glycolide) nanoparticles conjugated with disease-relevant antigen to treat a model of multiple sclerosis. Increasing the nanoparticle dose and amount of conjugated antigen both resulted in more durable immune tolerance. To identify active tolerance mechanisms, we investigated downstream cellular and molecular events following nanoparticle internalization by antigen-presenting cells. The initial cell response to nanoparticles indicated suppression of inflammatory signaling pathways. Direct and functional measurement of surface MHC-restricted antigen showed positive correlation with both increasing particle dose from 1 to 100 µg/mL and increasing peptide conjugation by 2-fold. Co-stimulatory analysis of cells expressing MHC-restricted antigen revealed most significant decreases in positive co-stimulatory molecules (CD86, CD80, and CD40) following high doses of nanoparticles with higher peptide conjugation, whereas expression of a negative co-stimulatory molecule (PD-L1) remained high. T cells isolated from mice immunized against myelin proteolipid protein (PLP139-151) were co-cultured with antigen-presenting cells administered PLP139-151-conjugated nanoparticles, which resulted in reduced T cell proliferation, increased T cell apoptosis, and a stronger anti-inflammatory response. These findings indicate several potential mechanisms used by peptide-conjugated nanoparticles to induce antigen-specific tolerance.


Subject(s)
Antigens/pharmacology , Delayed-Action Preparations/chemistry , Encephalomyelitis, Autoimmune, Experimental/therapy , Immunoconjugates/pharmacology , Myelin Proteolipid Protein/pharmacology , Nanoparticles/chemistry , Ovalbumin/pharmacology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Antigens/chemistry , Antigens/immunology , B7-1 Antigen/genetics , B7-1 Antigen/immunology , B7-2 Antigen/genetics , B7-2 Antigen/immunology , CD40 Antigens/genetics , CD40 Antigens/immunology , Delayed-Action Preparations/administration & dosage , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression , Immune Tolerance/drug effects , Immunoconjugates/chemistry , Immunoconjugates/metabolism , Mice , Mice, Inbred C57BL , Myelin Proteolipid Protein/chemistry , Myelin Proteolipid Protein/immunology , Nanoparticles/administration & dosage , Ovalbumin/chemistry , Ovalbumin/immunology , Particle Size , Polyglactin 910/chemistry , Polyglactin 910/metabolism , Primary Cell Culture , Spleen/drug effects , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
20.
Int J Mol Sci ; 19(9)2018 Sep 14.
Article in English | MEDLINE | ID: mdl-30223431

ABSTRACT

This study investigated the relationship between clinical severity and percentage of conjunctival antigen-presenting cells (APCs) in Sjögren's syndrome (SS)-associated keratoconjunctivitis sicca (KCS). KCS clinical severity was based on symptom severity, tear volume, tear break-up time, and ocular surface dye staining. Conjunctival goblet cell density (GCD) was measured in periodic acid Schiff (PAS)-stained membranes. Conjunctival cells obtained by impression cytology were used for flow cytometry to measure percentages of CD45⁺HLA-DR⁺ APCs and mature CD11c⁺CD86⁺ dendritic cells (DCs). Compared to normal conjunctiva, the percentages of HLA-DR⁺ and CD11c⁺CD86⁺ cells were higher in the conjunctiva of the KCS group (p < 0.05). The percentage of CD45⁺HLA-DR⁺ cells positively correlated with clinical severity (r = 0.71, p < 0.05) and negatively correlated with GCD (r = -0.61, p < 0.05). Clinical severity also negatively correlated with GCD (r = -0.54, p < 0.05). These findings indicate that a higher percentage of APCs and mature DCs in the conjunctiva is associated with more severe KCS in SS. These APCs may contribute to the generation of the pathogenic Th1 cells that cause goblet cell loss in KCS.


Subject(s)
Antigen-Presenting Cells/immunology , Keratoconjunctivitis Sicca/diagnosis , Keratoconjunctivitis Sicca/etiology , Sjogren's Syndrome/complications , Sjogren's Syndrome/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/pathology , Biomarkers , Case-Control Studies , Cell Count , Dendritic Cells/immunology , Dendritic Cells/metabolism , Goblet Cells/immunology , Goblet Cells/metabolism , Humans , Immunophenotyping , Severity of Illness Index , Sjogren's Syndrome/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL