Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
J Allergy Clin Immunol ; 153(4): 913-923, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38365015

ABSTRACT

The immune system classically consists of 2 lines of defense, innate and adaptive, both of which interact with one another effectively to protect us against any pathogenic threats. Importantly, there is a diverse subset of cells known as innate-like T cells that act as a bridge between the innate and adaptive immune systems and are pivotal players in eliciting inflammatory immune responses. A growing body of evidence has demonstrated the regulatory impact of these innate-like T cells in central nervous system (CNS) diseases and that such immune cells can traffic into the brain in multiple pathological conditions, which can be typically attributed to the breakdown of the blood-brain barrier. However, until now, it has been poorly understood whether innate-like T cells have direct protective or causative properties, particularly in CNS diseases. Therefore, in this review, our attention is focused on discussing the critical roles of 3 unique subsets of unconventional T cells, namely, natural killer T cells, γδ T cells, and mucosal-associated invariant T cells, in the context of CNS diseases, disorders, and injuries and how the interplay of these immune cells modulates CNS pathology, in an attempt to gain a better understanding of their complex functions.


Subject(s)
Central Nervous System Diseases , Mucosal-Associated Invariant T Cells , Natural Killer T-Cells , Humans , Central Nervous System Diseases/metabolism , Immunity, Innate
2.
Brain Behav Immun ; 116: 321-328, 2024 02.
Article in English | MEDLINE | ID: mdl-38157945

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are a subpopulation of innate-like T cells that can be found throughout the body, predominantly in mucosal sites, the lungs and in the peripheral blood. MAIT cells recognize microbial-derived vitamin B (e.g., riboflavin) metabolite antigens that are presented by the major histocompatibility complex class I-like protein, MR1, found on a variety of cell types in the periphery and the CNS. Since their original discovery, MAIT cells have been studied predominantly in their roles in diseases in the periphery; however, it was not until the early 2000s that these cells were first examined for their contributions to disorders of the CNS, with the bulk of the work being done within the past few years. Currently, the MR1/MAIT cell axis has been investigated in only a few neurological diseases including, multiple sclerosis and experimental autoimmune encephalomyelitis, brain cancer/tumors, ischemia, cerebral palsy, general aging and, most recently, Alzheimer's disease. Each of these diseases demonstrates a role for this under-studied innate immune axis in its neuropathology. Together, they highlight the importance of studying the MR1/MAIT cell axis in CNS disorders. Here, we review the contributions of the MR1/MAIT cell axis in the progression or remission of these neurological diseases. This work has shed some light in terms of potentially exploiting the MR1/MAIT cell axis in novel therapeutic applications.


Subject(s)
Central Nervous System Diseases , Mucosal-Associated Invariant T Cells , Humans , Minor Histocompatibility Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Riboflavin/metabolism , Central Nervous System Diseases/metabolism
3.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Article in English | MEDLINE | ID: mdl-34740973

ABSTRACT

Tumor antigen heterogeneity, a severely immunosuppressive tumor microenvironment (TME) and lymphopenia resulting in inadequate immune intratumoral trafficking, have rendered glioblastoma (GBM) highly resistant to therapy. To address these obstacles, here we describe a unique, sophisticated combinatorial platform for GBM: a cooperative multifunctional immunotherapy based on genetically engineered human natural killer (NK) cells bearing multiple antitumor functions including local tumor responsiveness that addresses key drivers of GBM resistance to therapy: antigen escape, immunometabolic reprogramming of immune responses, and poor immune cell homing. We engineered dual-specific chimeric antigen receptor (CAR) NK cells to bear a third functional moiety that is activated in the GBM TME and addresses immunometabolic suppression of NK cell function: a tumor-specific, locally released antibody fragment which can inhibit the activity of CD73 independently of CAR signaling and decrease the local concentration of adenosine. The multifunctional human NK cells targeted patient-derived GBM xenografts, demonstrated local tumor site-specific activity in the tissue, and potently suppressed adenosine production. We also unveil a complex reorganization of the immunological profile of GBM induced by inhibiting autophagy. Pharmacologic impairment of the autophagic process not only sensitized GBM to antigenic targeting by NK cells but promoted a chemotactic profile favorable to NK infiltration. Taken together, our study demonstrates a promising NK cell-based combinatorial strategy that can target multiple clinically recognized mechanisms of GBM progression simultaneously.


Subject(s)
Genetic Engineering , Glioblastoma/therapy , Immunotherapy, Adoptive , Killer Cells, Natural , Tumor Microenvironment/immunology , Animals , Autophagy , Glioblastoma/immunology , Humans , Mice , Xenograft Model Antitumor Assays
4.
J Neuroinflammation ; 20(1): 78, 2023 Mar 21.
Article in English | MEDLINE | ID: mdl-36944969

ABSTRACT

BACKGROUND: Neuroinflammation is an important feature of Alzheimer's disease (AD). Understanding which aspects of the immune system are important in AD may lead to new therapeutic approaches. We study the major histocompatibility complex class I-related immune molecule, MR1, which is recognized by an innate-like T cell population called mucosal-associated invariant T (MAIT) cells. METHODS: Having found that MR1 gene expression is elevated in the brain tissue of AD patients by mining the Agora database, we sought to examine the role of the MR1/MAIT cell axis in AD pathology. Brain tissue from AD patients and the 5XFAD mouse model of AD were used to analyze MR1 expression through qPCR, immunofluorescence, and flow cytometry. Furthermore, mice deficient in MR1 and MAIT cells were crossed with the 5XFAD mice to produce a model to study how the loss of this innate immune axis alters AD progression. Moreover, 5XFAD mice were also used to study brain-resident MAIT cells over time. RESULTS: In tissue samples from AD patients and 5XFAD mice, MR1 expression was substantially elevated in the microglia surrounding plaques vs. those that are further away (human AD: P < 0.05; 5XFAD: P < 0.001). In 5XFAD mice lacking the MR1/MAIT cell axis, the development of amyloid-beta plaque pathology occurred at a significantly slower rate than in those mice with MR1 and MAIT cells. Furthermore, in brain tissue from 5XFAD mice, there was a temporal increase in MAIT cell numbers (P < 0.01) and their activation state, the latter determined by detecting an upregulation of both CD69 (P < 0.05) and the interleukin-2 receptor alpha chain (P < 0.05) via flow cytometry. CONCLUSIONS: Together, these data reveal a previously unknown role for the MR1/MAIT cell innate immune axis in AD pathology and its potential utility as a novel therapeutic target.


Subject(s)
Alzheimer Disease , Mucosal-Associated Invariant T Cells , Humans , Mice , Animals , Mucosal-Associated Invariant T Cells/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Histocompatibility Antigens Class I/genetics , Minor Histocompatibility Antigens/metabolism
5.
Eur J Immunol ; 49(2): 255-265, 2019 02.
Article in English | MEDLINE | ID: mdl-30467836

ABSTRACT

Invariant natural killer T (iNKT) cells play critical roles in autoimmune, anti-tumor, and anti-microbial immune responses, and are activated by glycolipids presented by the MHC class I-like molecule, CD1d. How the activation of signaling pathways impacts antigen (Ag)-dependent iNKT cell activation is not well-known. In the current study, we found that the MAPK JNK2 not only negatively regulates CD1d-mediated Ag presentation in APCs, but also contributes to CD1d-independent iNKT cell activation. A deficiency in the JNK2 (but not JNK1) isoform enhanced Ag presentation by CD1d. Using a vaccinia virus (VV) infection model known to cause a loss in iNKT cells in a CD1d-independent, but IL-12-dependent manner, we found the virus-induced loss of iNKT cells in JNK2 KO mice was substantially lower than that observed in JNK1 KO or wild-type (WT) mice. Importantly, compared to WT mice, JNK2 KO mouse iNKT cells were found to express less surface IL-12 receptors. As with a VV infection, an IL-12 injection also resulted in a smaller decrease in JNK2 KO iNKT cells as compared to WT mice. Overall, our work strongly suggests JNK2 is a negative regulator of CD1d-mediated Ag presentation and contributes to IL-12-induced iNKT cell activation and loss during viral infections.


Subject(s)
Antigens, CD1d/immunology , Lymphocyte Activation , Mitogen-Activated Protein Kinase 9/immunology , Natural Killer T-Cells/immunology , Animals , Antigens, CD1d/genetics , Female , Interleukin-12/genetics , Interleukin-12/immunology , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 9/genetics , Receptors, Interleukin-12/genetics , Receptors, Interleukin-12/immunology , Virus Diseases/genetics , Virus Diseases/immunology
7.
J Immunol ; 197(8): 2971-2979, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27824592

ABSTRACT

Cell signaling pathways regulate much in the life of a cell: from shuttling cargo through intracellular compartments and onto the cell surface, how it should respond to stress, protecting itself from harm (environmental insults or infections), to ultimately, death by apoptosis. These signaling pathways are important for various aspects of the immune response as well. However, not much is known in terms of the participation of cell signaling pathways in Ag presentation, a necessary first step in the activation of innate and adaptive T cells. In this brief review, I discuss the known signaling molecules (and pathways) that regulate how Ags are presented to T cells and the mechanism(s), if identified. Studies in this area have important implications in vaccine development and new treatment paradigms against infectious diseases, autoimmunity, and cancer.


Subject(s)
Antigen Presentation , Autoimmune Diseases/immunology , Infections/immunology , Neoplasms/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Vaccines/immunology , Adaptive Immunity , Animals , Humans , Immunity, Innate , Infections/therapy , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasms/therapy
8.
Immunology ; 152(2): 232-242, 2017 10.
Article in English | MEDLINE | ID: mdl-28518215

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are conserved T cells that express a semi-invariant T-cell receptor (Vα7.2 in humans and Vα19 in mice). The development of MAIT cells requires the antigen-presenting MHC-related protein 1 (MR1), as well as commensal bacteria. The mechanisms that regulate the functional expression of MR1 molecules and their loading with bacterial antigen in antigen-presenting cells are largely unknown. We have found that treating B cells with the Toll-like receptor 9 (TLR9) agonist CpG increases MR1 surface expression. Interestingly, activation of TLR9 by CpG-A (but not CpG-B) enhances MR1 surface expression. This is limited to B cells and not other types of cells such as monocytes, T or natural killer cells. Knocking-down TLR9 expression by short hairpin RNA reduces MR1 surface expression and MR1-mediated bacterial antigen presentation. CpG-A triggers early endosomal TLR9 activation, whereas CpG-B is responsible for late endosomal/lysosomal activation of TLR9. Consistently, blocking endoplasmic reticulum to Golgi protein transport, rather than lysosomal acidification, suppressed MR1 antigen presentation. Overall, our results indicate that early endosomal TLR9 activation is important for MR1-mediated bacterial antigen presentation.


Subject(s)
Antigen Presentation , Antigens, Bacterial/immunology , B-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Lymphocyte Activation , Minor Histocompatibility Antigens/immunology , Toll-Like Receptor 9/immunology , Antigens, Bacterial/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/microbiology , Cell Line, Tumor , CpG Islands , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Endosomes/immunology , Endosomes/metabolism , Golgi Apparatus/immunology , Golgi Apparatus/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , Hydrogen-Ion Concentration , Lysosomes/immunology , Lysosomes/metabolism , Minor Histocompatibility Antigens/metabolism , Oligonucleotides/pharmacology , Protein Transport , RNA Interference , Signal Transduction , Time Factors , Toll-Like Receptor 9/drug effects , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism , Transfection
9.
Immunology ; 146(3): 444-55, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26260288

ABSTRACT

Cytokines that regulate the immune response signal through the Janus kinase / signal transducer and activation of transcription (JAK/STAT) pathway, but whether this pathway can regulate CD1d-mediated lipid antigen presentation to natural killer T (NKT) cells is unknown. Here, we found that STAT3 promotes antigen presentation by CD1d. Antigen-presenting cells (APCs) in which STAT3 expression was inhibited exhibited markedly reduced endogenous lipid antigen presentation to NKT cells without an impact on exogenous lipid antigen presentation by CD1d. Consistent with this observation, in APCs where STAT3 was knocked down, dramatically decreased levels of UDP glucose ceramide glucosyltransferase (UGCG), an enzyme involved in the first step of glycosphingolipid biosynthesis, were observed. Impaired lipid antigen presentation was reversed by ectopic expression of UGCG in STAT3-silenced CD1d(+) APCs. Hence, by controlling a fundamental step in CD1d-mediated lipid antigen presentation, STAT3 signalling promotes innate immune responses driven by CD1d.


Subject(s)
Antigens, CD1d/metabolism , Glycosphingolipids/biosynthesis , Lipids/immunology , STAT3 Transcription Factor/immunology , Animals , Antigen Presentation , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cell Line , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Glucosyltransferases/metabolism , HEK293 Cells , Histocompatibility Antigens Class II/metabolism , Humans , Immunity, Innate , Mice , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Signal Transduction/immunology
10.
Blood ; 121(17): 3473-83, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23426944

ABSTRACT

Efferocytosis of apoptotic neutrophils by macrophages following tissue injury is fundamental to the resolution of inflammation and initiation of tissue repair. Using a sterile peritonitis model in mice, we identified interleukin (IL)-4-producing efferocytosing macrophages in the peritoneum that activate invariant natural killer T (iNKT) cells to produce cytokines including IL-4, IL-13, and interferon-γ. Importantly, IL-4 from macrophages contributes to alternative activation of peritoneal exudate macrophages and augments type 2 cytokine production from NKT cells to suppress inflammation. The increased peritonitis in mice deficient in IL-4, NKT cells, or IL-4Rα expression on myeloid cells suggested that each is a key component for resolution of sterile inflammation. The reduced NAD phosphate oxidase is also critical for this model, because in mice with X-linked chronic granulomatous disease (X-CGD) that lack oxidase subunits, activation of iNKT cells by X-CGD peritoneal exudate macrophages was impaired during sterile peritonitis, resulting in enhanced and prolonged inflammation in these mice. Therefore, efferocytosis-induced IL-4 production and activation of IL-4-producing iNKT cells by macrophages are immunomodulatory events in an innate immune circuit required to resolve sterile inflammation and promote tissue repair.


Subject(s)
Genetic Diseases, X-Linked/immunology , Granulomatous Disease, Chronic/immunology , Inflammation/prevention & control , Interleukin-4/physiology , Macrophages/immunology , Natural Killer T-Cells/immunology , Phagocytosis/physiology , Animals , Cytokines/metabolism , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/pathology , Granulomatous Disease, Chronic/metabolism , Granulomatous Disease, Chronic/pathology , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology , Peritoneal Diseases/immunology , Peritoneal Diseases/pathology , Peritoneal Diseases/prevention & control , Receptors, Cell Surface/physiology
11.
Immunology ; 143(4): 679-91, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24990409

ABSTRACT

CD1d-mediated lipid antigen presentation activates a subset of innate immune lymphocytes called invariant natural killer T (NKT) cells that, by virtue of their potent cytokine production, bridge the innate and adaptive immune systems. Transforming growth factor (TGF-ß) is a known immune modulator that can activate the mitogen-activated protein kinase p38; we have previously shown that p38 is a negative regulator of CD1d-mediated antigen presentation. Several studies implicate a role for TGF-ß in the activation of p38. Therefore, we hypothesized that TGF-ß would impair antigen presentation by CD1d. Indeed, a dose-dependent decrease in CD1d-mediated antigen presentation and impairment of lipid antigen processing was observed in response to TGF-ß treatment. However, it was found that this inhibition was not through p38 activation. Instead, Smads 2, 3 and 4, downstream elements of the TGF-ß canonical signalling pathway, contributed to the observed effects. In marked contrast to that observed with CD1d, TGF-ß was found to enhance MHC class II-mediated antigen presentation. Overall, these results suggest that the canonical TGF-ß/Smad pathway negatively regulates an important arm of the host's innate immune responses - CD1d-mediated lipid antigen presentation to NKT cells.


Subject(s)
Antigen Presentation/immunology , Antigens, CD1d/metabolism , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antigen Presentation/drug effects , Cell Line , Female , Histocompatibility Antigens Class II/immunology , Lipids/immunology , Lysosomes/metabolism , Mice , Models, Immunological , Protein Transport , Transforming Growth Factor beta/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Immunology ; 141(4): 531-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24205828

ABSTRACT

Vaccinia virus (VV) has been used globally as a vaccine to eradicate smallpox. Widespread use of this viral vaccine has been tempered in recent years because of its immuno-evasive properties, with restrictions prohibiting VV inoculation of individuals with immune deficiencies or atopic skin diseases. VV infection is known to perturb several pathways for immune recognition including MHC class II (MHCII) and CD1d-restricted antigen presentation. MHCII and CD1d molecules associate with a conserved intracellular chaperone, CD74, also known as invariant chain. Upon VV infection, cellular CD74 levels are significantly reduced in antigen-presenting cells, consistent with the observed destabilization of MHCII molecules. In the current study, the ability of sustained CD74 expression to overcome VV-induced suppression of antigen presentation was investigated. Viral inhibition of MHCII antigen presentation could be partially ameliorated by ectopic expression of CD74 or by infection of cells with a recombinant VV encoding murine CD74 (mCD74-VV). In contrast, virus-induced disruptions in CD1d-mediated antigen presentation persisted even with sustained CD74 expression. Mice immunized with the recombinant mCD74-VV displayed greater protection during VV challenge and more robust anti-VV antibody responses. Together, these observations suggest that recombinant VV vaccines encoding CD74 may be useful tools to improve CD4⁺ T-cell responses to viral and tumour antigens.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , Smallpox Vaccine/immunology , Vaccinia virus/immunology , Vaccinia/prevention & control , Viral Proteins/immunology , Animals , Antibodies, Viral/blood , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , Antigens, Viral/immunology , Antigens, Viral/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Line , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Humans , Mice , Mice, Inbred C57BL , Smallpox Vaccine/administration & dosage , Smallpox Vaccine/genetics , Smallpox Vaccine/metabolism , Time Factors , Transfection , Vaccination , Vaccines, Synthetic/immunology , Vaccinia/immunology , Vaccinia/metabolism , Vaccinia/virology , Vaccinia virus/genetics , Vaccinia virus/metabolism , Viral Proteins/genetics
13.
Blood ; 119(24): 5731-41, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22535665

ABSTRACT

In the present study, surface CD1d, which is involved in immune cell interactions, was assessed for effects on hematopoiesis. Mouse BM hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) express CD1d. The numbers and cycling status of HPCs in the BM and spleen of different strains of cd1d(-/-) mice were enhanced significantly, suggesting that CD1d is a negative regulator of HPCs. In support of this, CD1d was required for the SCF and Flt3 ligand synergistic enhancement of CSF induction of HPC colony formation and for HPC response to myelosuppressive chemokines. Colony formation by immature subsets of HPCs was greatly enhanced when normal, but not cd1d(-/-), BM cells were pretreated with CD1d Abs in vitro. These effects required the full CD1d cytoplasmic tail. In contrast, long-term, but not short-term, repopulating HSC engraftment was impaired significantly, an effect that was minimally influenced by the presence of a truncated CD1d cytoplasmic tail. Pretreatment of normal BM cells with CD1d Abs greatly enhanced their engraftment of HSCs. The results of the present study implicate CD1d in a previously unrecognized regulatory role of normal and stressed hematopoiesis.


Subject(s)
Antigens, CD1d/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Antibodies/pharmacology , Antigens, CD1d/chemistry , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Count , Cell Proliferation/drug effects , Chemokines/pharmacology , Colony-Forming Units Assay , Galactosylceramides/pharmacology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Interferon-gamma/pharmacology , Membrane Proteins/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Phenotype , Protein Structure, Tertiary , Stem Cell Factor/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
14.
J Immunol ; 189(4): 1689-98, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22798677

ABSTRACT

CD1d molecules are MHC class I-like molecules that present lipid Ags to NKT cells. Although we have previously shown that several different cell signaling molecules can play a role in the control of Ag presentation by CD1d, a defined mechanism by which a cell signaling pathway regulates CD1d function has been unclear. In the current study, we have found that the Rho kinases, Rho-associated, coiled-coil containing protein kinase (ROCK)1 and ROCK2, negatively regulate both human and mouse CD1d-mediated Ag presentation. Inhibition of ROCK pharmacologically, through specific ROCK1 and ROCK2 short hairpin RNA, or by using dendritic cells generated from ROCK1-deficient mice all resulted in enhanced CD1d-mediated Ag presentation compared with controls. ROCK regulates the actin cytoskeleton by phosphorylating LIM kinase, which, in turn, phosphorylates cofilin, prohibiting actin fiber depolymerization. Treatment of APCs with the actin filament depolymerizing agent, cytochalasin D, as well as knockdown of LIM kinase by short hairpin RNA, resulted in enhanced Ag presentation to NKT cells by CD1d, consistent with our ROCK inhibition data. Therefore, our overall results reveal a model whereby CD1d-mediated Ag presentation is negatively regulated by ROCK via its effects on the actin cytoskeleton.


Subject(s)
Actin Cytoskeleton/immunology , Antigen Presentation/immunology , Antigens, CD1d/immunology , rho-Associated Kinases/immunology , Actin Cytoskeleton/metabolism , Animals , Antigens, CD1d/metabolism , Blotting, Western , Cell Line , Humans , Mice , Mice, Knockout , Microscopy, Confocal , rho-Associated Kinases/genetics
15.
J Neuroimmunol ; 390: 578332, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38537322

ABSTRACT

Emerging evidence has supported a role for the immune system and liver in Alzheimer's disease (AD). However, our understanding of how hepatic immune cells are altered in AD is limited. We previously found that brain mucosal-associated invariant T (MAIT) cell numbers are increased in AD. Furthermore, loss of MAIT cells and their antigen-presenting molecule, MR1, reduced amyloid-ß accumulation in the brain. MAIT cells are also significantly present in the liver. Therefore, we sought to analyze MAIT and other immune cells in the AD liver. Increased frequency of activated MAIT cells (but not conventional T cells) were found in 8-month-old 5XFAD mouse livers. Therefore, these data raise the possibility that there is a role for peripheral MAIT cells in AD pathology.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Liver , Mice, Transgenic , Mucosal-Associated Invariant T Cells , Animals , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Mice , Mucosal-Associated Invariant T Cells/immunology , Mucosal-Associated Invariant T Cells/metabolism , Liver/pathology , Liver/immunology , Liver/metabolism , Mice, Inbred C57BL , Humans , Female , Male
16.
Immunology ; 140(2): 191-201, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23710894

ABSTRACT

CD1d-restricted T (natural killer T; NKT) cells are important for controlling herpesvirus infections. Interestingly, herpes simplex virus (HSV) can down-regulate CD1d-mediated activation of NKT cells. We have previously shown that the Thr322 residue in the cytoplasmic tail of human CD1d is important for its intracellular trafficking and functional expression. We proposed that the phosphorylation of T322 is a signal for CD1d lysosomal targeting and subsequent degradation. In the current study, we generated dual mutants by substituting the T322 and S323 residues of wild-type (WT) CD1d with Ala (non-phosphorylatable) or Asp (mimicking phosphorylation) and ectopically expressed them in human embryonic kidney 293 cells. We found that the surface expression levels of the CD1d mutants was in this order: T322AS323A > WT > T322A > S323A > S323D > T322D > T322DS323D. Our results therefore suggest that mimicking the phosphorylation of both T322 and S323 has a cumulative negative effect on the functional expression of CD1d. As previously reported, we also found that upon an HSV infection, antigen presentation by WT CD1d is reduced and the CD1d molecule is degraded. Interestingly, the T322A/S323A double mutation inhibited CD1d degradation and rescued CD1d-mediated antigen presentation following an HSV-1 infection. This suggests that the T322/S323 dyad may be phosphorylated, which then targets CD1d for lysosomal degradation post-infection as a means of immune evasion, explaining (at least in part) the reduced antigen presentation observed. Hence, our findings strongly suggest that T322 and S323 form a dual residue motif that can regulate the functional expression of CD1d during a viral infection.


Subject(s)
Antigen Presentation/physiology , Antigens, CD1d/metabolism , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Natural Killer T-Cells/immunology , Amino Acid Motifs , Antigens, CD1d/chemistry , Down-Regulation , Flow Cytometry , HEK293 Cells , Humans , Lymphocyte Activation/immunology , Microscopy, Confocal , Mutagenesis, Site-Directed , Natural Killer T-Cells/virology , Phosphorylation , Protein Transport/physiology
17.
J Exp Med ; 204(9): 2145-57, 2007 Sep 03.
Article in English | MEDLINE | ID: mdl-17724129

ABSTRACT

Recently, a new developmental pathway for CD4 T cells that is mediated by major histocompatibility complex class II-positive thymocytes was identified (Choi, E.Y., K.C. Jung, H.J. Park, D.H. Chung, J.S. Song, S.D. Yang, E. Simpson, and S.H. Park. 2005. Immunity. 23:387-396; Li, W., M.G. Kim, T.S. Gourley, B.P. McCarthy, D.B. Sant'angelo, and C.H. Chang. 2005. Immunity. 23:375-386). We demonstrate that thymocyte-selected CD4 (T-CD4) T cells can rapidly produce interferon gamma and interleukin (IL) 4 upon in vivo and in vitro T cell receptor stimulation. These T-CD4 T cells appear to be effector cells producing both T helper type 1 (Th1) and Th2 cytokines, and they maintain a potential to produce Th2 cytokines under Th1-skewing conditions in a signal transducer and activator of transcription 6-independent manner. The IL-4 mRNA level is high in CD4 single-positive thymocytes if they are selected on thymocytes, which is at least partly caused by enhanced histone acetylation of the IL-4 locus. However, mice that can generate T-CD4 T cells showed attenuated immune responses in an allergen-induced airway inflammation model, suggesting a protective role for T-CD4 T cells during an airway challenge. Our results imply that this thymic selection pathway plays an important role in determining the effector function of the resulting CD4 cells and in regulating immune response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Selection, Genetic , Thymus Gland/immunology , Allergens , Animals , Antigen Presentation/immunology , Epithelial Cells/immunology , Gene Expression Regulation , Histocompatibility Antigens Class II , Humans , Inflammation , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Interleukin-4/genetics , Killer Cells, Natural/immunology , Mice , Mice, Transgenic , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Respiratory System/pathology , STAT6 Transcription Factor/metabolism , Th1 Cells/metabolism , Th2 Cells/metabolism
18.
Blood ; 117(23): 6198-201, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21505191

ABSTRACT

The contribution of specific cell types to the production of cytokines that regulate hematopoiesis is still not well defined. We have previously identified T cell-dependent regulation of hematopoietic progenitor cell (HPC) numbers and cycling. In this report, we demonstrated that HPC activity is decreased in mice with STAT3-deficient T cells, a phenotype that is not because of decreased expression of IL-17 or RORγt. STAT3 expression in T cells was required for IL-21 production by multiple T helper subsets, and neutralization of IL-21 resulted in decreased HPC activity identical to that in mice with STAT3-deficient T cells. Importantly, injection of IL-21 rescued HPC activity in mice with STAT3-deficient T cells. Thus, STAT3-dependent IL-21 production in T cells is required for HPC homeostasis.


Subject(s)
Gene Expression Regulation/immunology , Hematopoietic Stem Cells/immunology , Homeostasis/immunology , Interleukins/immunology , STAT3 Transcription Factor/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Gene Expression Regulation/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeostasis/genetics , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukins/biosynthesis , Interleukins/genetics , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism
19.
bioRxiv ; 2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37905092

ABSTRACT

Tissue damage resulting from a spinal cord injury (SCI) is primarily driven by a robust neuroimmune/neuroinflammatory response. This intricate process is mainly governed by a multitude of cytokines and cell surface proteins in the central nervous system (CNS). However, the critical components of the neuroimmune/neuroinflammatory response during SCI are still not well-defined. In this study, we investigated the impact of CD1d, an MHC class I-like molecule mostly known for presenting lipid antigens to natural killer T (NKT) cells and regulating immune/inflammatory responses, on neuroimmune/neuroinflammatory responses induced by SCI. We observed an increased expression of CD1d on various cell types within the spinal cord, including microglia/macrophages, oligodendrocytes (ODCs), and endothelial cells (DCs), but not on neurons or astrocytes post-SCI. In comparison to wildtype (WT) mice, a T10 contusive SCI in CD1d knockout (CD1dKO or Cd1d -/- ) mice resulted in markedly reduced proinflammatory cytokine release, microglia/macrophage activation and proliferation. Following SCI, the levels of inflammatory cytokines and activation/proliferation of microglia/macrophages were dramatically reduced, while anti-inflammatory cytokines such as IL-4 and growth factors like VEGF were substantially increased in the spinal cord tissues of CD1dKO mice when compared to WT mice. In the post-acute phase of SCI (day 7 post-SCI), CD1dKO mice had a significantly higher frequency of tissue-repairing macrophages, but not other types of immune cells, in the injured spinal cord tissues compared to WT mice. Moreover, CD1d-deficiency protected spinal cord neuronal cells and tissue, promoting functional recovery after a SCI. However, the neuroinflammation in WT mouse spinal cords was independent of the canonical CD1d/NKT cell axis. Finally, treatment of injured mice with a CD1d-specific monoclonal antibody significantly enhanced neuroprotection and improved functional recovery. Therefore, CD1d promotes the proinflammatory response following a SCI and represents a potential therapeutic target for spinal cord repair. Significance Statement: The cell surface molecule, CD1d, is known to be recognized by cells of the immune system. To our knowledge, this is the first observation that the CD1d molecule significantly contributes to neuroinflammation following a spinal cord injury (SCI) in a manner independent of the CD1d/NKT cell axis. This is important, because this work reveals CD1d as a potential therapeutic target following an acute SCI for which there are currently no effective treatments.

20.
J Immunol ; 184(9): 4973-81, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20368272

ABSTRACT

CD1d molecules are MHC class I-like molecules that present lipids to a unique subpopulation of T cells called NKT cells. The cytoplasmic tail of human CD1d possesses a tyrosine-based endosomal targeting motif (YXXZ). As such, these molecules traffic through the endocytic pathway, where it is believed that they are loaded with the antigenic lipid that stimulates NKT cells. In the current study, it was found that the T322 residue in the human CD1d tail is a major signal controlling transport to the cell surface and thus its functional expression. Mimicking the phosphorylation of this residue or removal of the entire cytoplasmic tail negates its ability to regulate CD1d trafficking, resulting in lysosomal targeting and degradation. These results demonstrate an important role of a heretofore unknown signal in the cytoplasmic tail of CD1d that may have relevance to other type I integral membrane proteins that traverse through the endocytic pathway.


Subject(s)
Antigens, CD1d/physiology , Cytoplasm/immunology , Gene Expression Regulation/immunology , Signal Transduction/immunology , Threonine/physiology , Amino Acid Motifs/genetics , Amino Acid Motifs/immunology , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Antigens, CD1d/biosynthesis , Antigens, CD1d/genetics , Cell Line , Cell Line, Transformed , Cells, Cultured , Coculture Techniques , Cytoplasm/chemistry , Cytoplasm/genetics , Endocytosis/genetics , Endocytosis/immunology , Gene Targeting , Humans , Membrane Proteins/chemistry , Membrane Proteins/classification , Membrane Proteins/physiology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Protein Structure, Tertiary/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Signal Transduction/genetics , Threonine/chemistry , Threonine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL