Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.424
Filter
1.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273326

ABSTRACT

Due to the genetic diversity between the mother and the fetus, heightened control over the immune system during pregnancy is crucial. Immunological parameters determined by clinicians in women with idiopathic recurrent spontaneous abortion (RSA) include the quantity and activity of Natural Killer (NK) and Natural Killer T (NKT) cells, the quantity of regulatory T lymphocytes, and the ratio of pro-inflammatory cytokines, which indicate imbalances in Th1 and Th2 cell response. The processes are controlled by immune checkpoint proteins (ICPs) expressed on the surface of immune cells. We aim to investigate differences in the expression of ICPs on T cells, T regulatory lymphocytes, NK cells, and NKT cells in peripheral blood samples collected from RSA women, pregnant women, and healthy multiparous women. We aim to discover new insights into the role of ICPs involved in recurrent pregnancy loss. Peripheral blood mononuclear cells (PBMCs) were isolated by gradient centrifugation from blood samples obtained from 10 multiparous women, 20 pregnant women (11-14th week of pregnancy), and 20 RSA women, at maximum of 72 h after miscarriage. The PBMCs were stained for flow cytometry analysis. Standard flow cytometry immunophenotyping of PBMCs was performed using antibodies against classical lymphocyte markers, including CD3, CD4, CD8, CD56, CD25, and CD127. Additionally, ICPs were investigated using antibodies against Programmed Death Protein-1 (PD-1, CD279), T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3, CD366), V-domain Ig suppressor of T cell activation (VISTA), T cell immunoglobulin and ITIM domain (TIGIT), and Lymphocyte activation gene 3 (LAG-3). We observed differences in the surface expression of ICPs in the analyzed subpopulations of lymphocytes between early pregnancy and RSA, after miscarriage, and in women. We noted diminished expression of PD-1 on T lymphocytes (p = 0.0046), T helper cells (CD3CD4 positive cells, p = 0.0165), T cytotoxic cells (CD3CD8 positive cells, p = 0.0046), T regulatory lymphocytes (CD3CD4CD25CD127 low positive cells, p = 0.0106), and NKT cells (CD3CD56/CD16 positive cells, p = 0.0438), as well as LAG-3 on lymphocytes T (p = 0.0225) T helper, p = 0.0426), T cytotoxic cells (p = 0.0458) and Treg (p = 0.0293), and cells from RSA women. Impaired expression of TIM-3 (p = 0.0226) and VISTA (p = 0.0039) on CD8 cytotoxic T and NK (TIM3 p = 0.0482; VISTA p = 0.0118) cells was shown, with an accompanying increased expression of TIGIT (p = 0.0211) on NKT cells. The changes in the expression of surface immune checkpoints indicate their involvement in the regulation of pregnancy. The data might be utilized to develop specific therapies for RSA women based on the modulation of ICP expression.


Subject(s)
Abortion, Habitual , Biomarkers , Immune Checkpoint Proteins , Killer Cells, Natural , Humans , Female , Pregnancy , Abortion, Habitual/immunology , Abortion, Habitual/metabolism , Abortion, Habitual/blood , Adult , Biomarkers/blood , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Immune Checkpoint Proteins/metabolism , Immune Checkpoint Proteins/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Immunophenotyping , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Antigens, CD/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Programmed Cell Death 1 Receptor/metabolism
2.
Front Immunol ; 15: 1433028, 2024.
Article in English | MEDLINE | ID: mdl-39281681

ABSTRACT

Objective: Acute respiratory distress syndrome (ARDS) presents a global health challenge, characterized by significant morbidity and mortality. However, the role of natural killer T (NKT) cells in human ARDS remains poorly understood. Therefore, this study explored the numerical and functional status of NKT cells in patients with ARDS, examining their clinical relevance and interactions with macrophages and fibroblasts during various stages of the syndrome. Methods: Peripheral blood from 40 ARDS patients and 30 healthy controls was analyzed, with paired samples of peripheral blood and bronchoalveolar lavage fluid (BALF) from seven ARDS patients. We measured levels of NKT cells, cytokines, CD69, programmed death-1 (PD-1), and annexin-V using flow cytometry, and extracellular matrix (ECM) protein expression using real-time PCR. Results: ARDS patients exhibited decreased circulating NKT cells with elevated CD69 expression and enhanced IL-17 production. The reduction in NKT cells correlated with PaO2/FiO2 ratio, albumin, and C-reactive protein levels. Proliferative responses to α-galactosylceramide (α-GalCer) were impaired, and co-culturing NKT cells with monocytes or T cells from ARDS patients resulted in a reduced α-GalCer response. Increased and activated NKT cells in BALF induced proinflammatory cytokine release by macrophages and ECM protein expression in fibroblasts. Conclusion: ARDS is associated with a numerical deficiency but functional activation of circulating NKT cells, showing impaired responses to α-GalCer and altered interactions with immune cells. The increase in NKT cells within BALF suggests their role in inducing inflammation and remodeling/fibrosis, highlighting the potential of targeting NKT cells as a therapeutic approach for ARDS.


Subject(s)
Natural Killer T-Cells , Respiratory Distress Syndrome , Humans , Respiratory Distress Syndrome/immunology , Male , Female , Middle Aged , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/cytology , Adult , Aged , Macrophages/immunology , Macrophages/metabolism , Cytokines/metabolism , Fibroblasts/metabolism , Fibroblasts/immunology , Lymphocyte Activation/immunology , Antigens, Differentiation, T-Lymphocyte , Antigens, CD , Lectins, C-Type
3.
Front Immunol ; 15: 1457771, 2024.
Article in English | MEDLINE | ID: mdl-39224603

ABSTRACT

Invariant natural killer T (iNKT) cells are a small subset of T lymphocytes that release large amounts of cytokines such as IFN-γ and exhibit cytotoxic activity upon activation, inducing strong anti-tumor effects. Harnessing the anti-tumor properties of iNKT cells, iNKT cell-based immunotherapy has been developed to treat cancer patients. In one of the iNKT cell-based immunotherapies, two approaches are utilized, namely, active immunotherapy or adoptive immunotherapy, the latter involving the ex vivo expansion and subsequent administration of iNKT cells. There are two sources of iNKT cells for adoptive transfer, autologous and allogeneic, each with its own advantages and disadvantages. Here, we assess clinical trials conducted over the last decade that have utilized iNKT cell adoptive transfer as iNKT cell-based immunotherapy, categorizing them into two groups based on the use of autologous iNKT cells or allogeneic iNKT cells.


Subject(s)
Immunotherapy, Adoptive , Natural Killer T-Cells , Neoplasms , Animals , Humans , Allogeneic Cells/immunology , Clinical Trials as Topic , Immunotherapy, Adoptive/methods , Natural Killer T-Cells/immunology , Neoplasms/therapy , Neoplasms/immunology , Transplantation, Autologous , Transplantation, Homologous
4.
Front Immunol ; 15: 1441634, 2024.
Article in English | MEDLINE | ID: mdl-39267746

ABSTRACT

Invariant natural killer T (iNKT) cells are a subset of lipid-reactive, unconventional T cells that have anti-tumor properties that make them a promising target for cancer immunotherapy. Recent studies have deciphered the developmental pathway of human MAIT and Vγ9Vδ2 γδ-T cells as well as murine iNKT cells, yet our understanding of human NKT cell development is limited. Here, we provide an update in our understanding of how NKT cells develop in the human body and how knowledge regarding their development could enhance human treatments by targeting these cells.


Subject(s)
Immunotherapy , Natural Killer T-Cells , Thymus Gland , Humans , Natural Killer T-Cells/immunology , Immunotherapy/methods , Thymus Gland/immunology , Animals , Cell Differentiation/immunology , Neoplasms/therapy , Neoplasms/immunology
5.
Gut Microbes ; 16(1): 2388801, 2024.
Article in English | MEDLINE | ID: mdl-39132842

ABSTRACT

The interaction between the gut microbiota and invariant Natural Killer T (iNKT) cells plays a pivotal role in colorectal cancer (CRC). The pathobiont Fusobacterium nucleatum influences the anti-tumor functions of CRC-infiltrating iNKT cells. However, the impact of other bacteria associated with CRC, like Porphyromonas gingivalis, on their activation status remains unexplored. In this study, we demonstrate that mucosa-associated P. gingivalis induces a protumour phenotype in iNKT cells, subsequently influencing the composition of mononuclear-phagocyte cells within the tumor microenvironment. Mechanistically, in vivo and in vitro experiments showed that P. gingivalis reduces the cytotoxic functions of iNKT cells, hampering the iNKT cell lytic machinery through increased expression of chitinase 3-like-1 protein (CHI3L1). Neutralization of CHI3L1 effectively restores iNKT cell cytotoxic functions suggesting a therapeutic potential to reactivate iNKT cell-mediated antitumour immunity. In conclusion, our data demonstrate how P. gingivalis accelerates CRC progression by inducing the upregulation of CHI3L1 in iNKT cells, thus impairing their cytotoxic functions and promoting host tumor immune evasion.


Subject(s)
Chitinase-3-Like Protein 1 , Colorectal Neoplasms , Natural Killer T-Cells , Porphyromonas gingivalis , Colorectal Neoplasms/immunology , Colorectal Neoplasms/microbiology , Natural Killer T-Cells/immunology , Porphyromonas gingivalis/immunology , Chitinase-3-Like Protein 1/metabolism , Chitinase-3-Like Protein 1/genetics , Humans , Animals , Mice , Tumor Microenvironment/immunology , Immune Evasion , Tumor Escape , Gastrointestinal Microbiome/immunology , Cell Line, Tumor , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/microbiology , Female , Mice, Inbred C57BL , Male
6.
J Diabetes ; 16(8): e13593, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39136533

ABSTRACT

BACKGROUND: We aimed to investigate whether alpha-galactosylceramide (α-GalCer)-producing Bacteroides fragilis could induce natural killer T (NKT) cells in nonobese diabetic (NOD) mice and reduce their diabetes incidence. METHODS: Five-week-old female NOD mice were treated orally with B. fragilis, and islet pathology and diabetes onset were monitored. Immune responses were analyzed by flow cytometry and multiplex technology. Effects of ultraviolet (UV)-killed α-GalCer-producing B. fragilis and their culture medium on invariant NKT (iNKT) cells were tested ex vivo on murine splenocytes, and the immunosuppressive capacity of splenocytes from B. fragilis-treated NOD mice were tested by adoptive transfer to nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. RESULTS: B. fragilis reduced the diabetes incidence from 69% to 33% and the percent of islets with insulitis from 40% to 7%, which doubled the serum insulin level compared with the vehicle-treated control mice. Furthermore, the early treatment reduced proinflammatory mediators in the serum, whereas the proportion of CD4+ NKT cell population was increased by 33%. B. fragilis growth media stimulated iNKT cells and anti-inflammatory M2 macrophages ex vivo in contrast to UV-killed bacteria, which had no effect, strongly indicating an α-GalCer-mediated effect. Adoptive transfer of splenocytes from B. fragilis-treated NOD mice induced a similar diabetes incidence as splenocytes from untreated NOD mice. CONCLUSIONS: B. fragilis induced iNKT cells and M2 macrophages and reduced type 1 diabetes in NOD mice. The protective effect seemed to be more centered on gut-pancreas interactions rather than a systemic immunosuppression. B. fragilis should be considered for probiotic use in individuals at risk of developing type 1 diabetes.


Subject(s)
Bacteroides fragilis , Galactosylceramides , Mice, Inbred NOD , Natural Killer T-Cells , Probiotics , Animals , Female , Galactosylceramides/pharmacology , Probiotics/therapeutic use , Probiotics/pharmacology , Mice , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 1/metabolism , Incidence , Mice, SCID
7.
Genes Genomics ; 46(9): 1097-1106, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39115674

ABSTRACT

BACKGROUND: Granzymes are essential serine proteases in cytotoxic T cells and natural killer (NK) cells, with GZMK's expression being less understood. This study aims to uncover GZMK expression profiles across various immune cell types using single-cell RNA sequencing meta-analysis. OBJECTIVE: This study aims to uncover GZMK expression profiles across various immune cell types using single-cell RNA sequencing meta-analysis. METHODS: We conducted a meta-analysis using cellxgene, an interactive data exploration platform developed by the Chan Zuckerberg Initiative. We focused on mature T cells, NK cells, B cells, and NKT cells. We also checked transcription factor binding sites at the granzyme gene promoter regions using JASPAR. Comparative analysis was also done using mouse single-cell RNA sequencing data. RESULTS: GZMK was the most lowly expressed in NK cells and mature NKT cells in most tissues except for colon and lymph nodes. In mature T cells, GZMK is similarly or more highly expressed than other granzymes. HBCA data revealed weak expression of GZMK in NK cells but strong expression in effector memory CD8-positive, alpha-beta T cells. Combined data shows no significant difference in GZMK expression between cell types. Subtype analysis shows that GZMK expression was higher in CD16-negative, CD56-bright NK cells when compared to CD16-positive, CD56-dim NK cells. We also identified unique transcription factor binding sites for GZMK. While this pattern in mouse data with low Gzmk expression in NK cells and higher T cells was repeated. CONCLUSION: GZMK expression is distinctively regulated among immune cells and tissues, with unique promoter regions and transcription factor binding sites contributing to this differential expression. These insights into GZMK's role in immune function and regulation offer potential therapeutic targets.


Subject(s)
Granzymes , Killer Cells, Natural , Single-Cell Analysis , Granzymes/genetics , Granzymes/metabolism , Animals , Single-Cell Analysis/methods , Mice , Killer Cells, Natural/metabolism , Killer Cells, Natural/immunology , Humans , RNA-Seq/methods , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/immunology , Promoter Regions, Genetic , Binding Sites , Single-Cell Gene Expression Analysis
8.
Cells ; 13(16)2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39195289

ABSTRACT

The adaptive response occurs only after 7-10 days of antigen presentation. Nevertheless, the autoreactive T cells infiltrate the stroke lesion within the first 48 h. Thus, we hypothesized that the unconventional lymphocytes as invariant natural killer T cells (iNKT) and γδT cells that share immediate innate and delayed adaptive response features are involved in acute stroke pathophysiology. We assessed prospectively the quantity of circulating iNKT cells, γδT cells, and NK cells with flow cytometry in 52 subjects within three months after stroke, and we compared the results with those obtained in age-, sex-, and vascular risk factor-matched controls. We studied lymphocyte parameters regarding clinical outcomes, infarct volume, stroke-associated infection (SAI), and burden risk factors. The reduced number of circulating γδT cells and decreased percentage of the Vδ2 subset in the acute phase of stroke correlated with worse neurological status in the recovery phase. In subjects treated with thrombolysis and those who developed SAI, a lower percentage of γδT cells in the 90-day follow-up was observed. An increased percentage of iNKT cells in the acute and subacute phases of stroke was observed, and it was related to the worse clinical status. The circulating NK cells do not change temporarily or affect the outcomes after stroke. It seems that γδT cells play a long-lasting role in ischemic stroke, mainly related to the Vδ2 subset. The role of iNKT cells appears to be detrimental, especially in the acute and subacute phases of stroke. The effect of circulating NK cells on the outcome after stroke seems negligible.


Subject(s)
Ischemic Stroke , Killer Cells, Natural , Natural Killer T-Cells , Humans , Male , Female , Natural Killer T-Cells/immunology , Ischemic Stroke/immunology , Ischemic Stroke/blood , Case-Control Studies , Killer Cells, Natural/immunology , Prospective Studies , Aged , Middle Aged , Treatment Outcome
9.
Proc Natl Acad Sci U S A ; 121(34): e2321686121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39141352

ABSTRACT

To broadly measure the spectrum of cellular self-antigens for natural killer T cells (NKT), we developed a sensitive lipidomics system to analyze lipids trapped between CD1d and NKT T cell receptors (TCRs). We captured diverse antigen complexes formed in cells from natural endogenous lipids, with or without inducing endoplasmic reticulum (ER) stress. After separating protein complexes with no, low, or high CD1d-TCR interaction, we eluted lipids to establish the spectrum of self-lipids that facilitate this interaction. Although this unbiased approach identified fifteen molecules, they clustered into only two related groups: previously known phospholipid antigens and unexpected neutral lipid antigens. Mass spectrometry studies identified the neutral lipids as ceramides, deoxyceramides, and diacylglycerols, which can be considered headless lipids because they lack polar headgroups that usually form the TCR epitope. The crystal structure of the TCR-ceramide-CD1d complex showed how the missing headgroup allowed the TCR to predominantly contact CD1d, supporting a model of CD1d autoreactivity. Ceramide and related headless antigens mediated physiological TCR binding affinity, weak NKT cell responses, and tetramer binding to polyclonal human and mouse NKT cells. Ceramide and sphingomyelin are oppositely regulated components of the "sphingomyelin cycle" that are altered during apoptosis, transformation, and ER stress. Thus, the unique molecular link of ceramide to NKT cell response, along with the recent identification of sphingomyelin blockers of NKT cell activation, provide two mutually reinforcing links for NKT cell response to sterile cellular stress conditions.


Subject(s)
Antigens, CD1d , Lipidomics , Natural Killer T-Cells , Receptors, Antigen, T-Cell , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Animals , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Mice , Lipidomics/methods , Humans , Autoantigens/immunology , Autoantigens/metabolism , Ceramides/metabolism , Ceramides/immunology , Lipids/chemistry , Lipids/immunology , Endoplasmic Reticulum Stress/immunology
10.
Kidney Int ; 106(3): 364-366, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39174198

ABSTRACT

Previous studies of the immune control of cytomegalovirus infection have primarily focused on analysis of the traditional adaptive T-cell response. Donadeu et al. bring a new perspective through evaluation of multiple adaptive and innate immune subtypes in parallel with cytomegalovirus-specific cell-mediated immunity in a prospective cohort of kidney transplant recipients with findings validated in 2 independent studies. Identification of a natural killer T-cell subtype associated with cell-mediated immunity and freedom from cytomegalovirus infection demonstrates the importance of the coordinated innate and adaptive immune response for effective viral control.


Subject(s)
Adaptive Immunity , Cytomegalovirus Infections , Cytomegalovirus , Immunity, Innate , Kidney Transplantation , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Humans , Kidney Transplantation/adverse effects , Cytomegalovirus/immunology , Immunity, Cellular , Natural Killer T-Cells/immunology
11.
Technol Cancer Res Treat ; 23: 15330338241273198, 2024.
Article in English | MEDLINE | ID: mdl-39166278

ABSTRACT

Background: To evaluate the efficacy and safety of allogenic CD8 + natural killer T (CD8+ NKT) immunotherapy combined with gefitinib in the treatment of advanced or metastatic EGFR mutant non-small cell lung cancer (NSCLC). Methods: This study is prospective. The NSCLC patients with exon 19 (Ex19del) or exon 21 L858R point mutations, and response to gefitinib treatment were enrolled into the trial to be randomly assigned into the gefitinib arm and the gefitinib/NKT arm. Allogenic CD8+ NKT cells were cultured in vitro and adaptive transferred into the patients via vein in the gefitinib/NKT arm. The primary endpoint was progression-free survival (PFS). Secondary endpoint analysis included time to disease progression (TTP), overall survival (OS), levels of serum tumour markers for carcinoembryonic antigen (CEA) and alanine aminotransferase (ALT) in the blood, the response rate and safety. From July 2017 to June 2021, 19 patients were randomly assigned to the gefitinib arm (n = 8) and the gefitinib/NKT arm (n = 11). Results: The estimated median survival PFS in the gefitinib/NKT arm was significantly longer than that of the gefitinib arm (12 months vs 7 months). Similar results were also observed for the median TTP. Moreover, the gefitinib/NKT arm had better CEA control than the gefitinib arm. Clinical grade 3 adverse reactions occurred in 64% and 39% of patients in the gefitinib/NKT arm and the gefitinib arm, respectively. The most common grade 3 adverse events in the gefitinib/NKT arm included abnormal liver function in 8 cases (73%) and diarrhoea in 1 case (9%), both of which resolved after drug intervention. Conclusion: The PFS of EGFR-mutated advanced NSCLC treated with allogenic CD8+ NKT cells combined with gefitinib was longer than that of gefitinib alone. No obvious serious adverse reactions occurred, and the patients compliance and survival status were good.


Subject(s)
ErbB Receptors , Lung Neoplasms , Mutation , Natural Killer T-Cells , Humans , Female , ErbB Receptors/genetics , Lung Neoplasms/therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/mortality , Middle Aged , Male , Aged , Natural Killer T-Cells/immunology , Protein Kinase Inhibitors/therapeutic use , Adult , Gefitinib/therapeutic use , Combined Modality Therapy , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Prospective Studies , Immunotherapy/methods , Treatment Outcome , Neoplasm Staging
12.
Int J Med Sci ; 21(10): 1890-1902, 2024.
Article in English | MEDLINE | ID: mdl-39113896

ABSTRACT

Objective: The immune response initiated by SARS-CoV-2 infection in pregnancy is poorly elucidated. We aimed to access and compare the antiviral cellular responses and lymphocytes activation between healthy pregnancies and pregnant women infected with SARS-CoV-2. Methods: We detected the immunological changes of lymphocytes in peripheral blood of healthy non-pregnant women, non-pregnant women with COVID-19, healthy pregnant women, pregnant women with COVID-19 and convalescent group by flow cytometry. In vitro blockade was used to identify NKT-like cell activation through ICOS-ICOSL pathway. Results: We found that CD3+CD56+ NKT-like cells decreased significantly in COVID-19 positive pregnant women compared to healthy pregnant women. NKT-like cells of pregnant women expressed higher level of activating receptors CD69 and NKp46 after SARS-CoV-2 infection. Particularly, they also increased the expression of the co-stimulatory molecule ICOS. NKT-like cells of pregnant women with COVID-19 up-regulated the expression of IFN-γ, CD107a and Ki67. Meanwhile, we found that ICOSL expression was significantly increased on pDCs in pregnant women with COVID-19. Blocking ICOS in vitro significantly decreased the antiviral activity of NKT-like cells in COVID-19 positive pregnant women, suggesting that ICOS-ICOSL may play an important role in the virus clearance by NKT-like cells. Conclusions: During SARS-CoV-2 infection, NKT-like cells of pregnant women activated through ICOS-ICOSL pathway and played an important role in the antiviral response.


Subject(s)
COVID-19 , Inducible T-Cell Co-Stimulator Ligand , Inducible T-Cell Co-Stimulator Protein , Natural Killer T-Cells , Pregnancy Complications, Infectious , SARS-CoV-2 , Humans , Female , Pregnancy , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , COVID-19/immunology , COVID-19/virology , Inducible T-Cell Co-Stimulator Protein/metabolism , Adult , Inducible T-Cell Co-Stimulator Ligand/metabolism , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , SARS-CoV-2/immunology , Lymphocyte Activation/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Signal Transduction/immunology , Interferon-gamma/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Lectins, C-Type/metabolism
13.
J Clin Invest ; 134(18)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39088280

ABSTRACT

NKT cells are innate-like T cells, recruited to the skin during viral infection, yet their contributions to long-term immune memory to viruses are unclear. We identified granzyme K, a product made by cytotoxic cells including NKT cells, as linked to induction of Th1-associated antibodies during primary dengue virus (DENV) infection in humans. We examined the role of NKT cells in vivo using DENV-infected mice lacking CD1d-dependent (CD1ddep) NKT cells. In CD1d-KO mice, Th1-polarized immunity and infection resolution were impaired, which was dependent on intrinsic NKT cell production of IFN-γ, since it was restored by adoptive transfer of WT but not IFN-γ-KO NKT cells. Furthermore, NKT cell deficiency triggered immune bias, resulting in higher levels of Th2-associated IgG1 than Th1-associated IgG2a, which failed to protect against a homologous DENV rechallenge and promoted antibody-dependent enhanced disease during secondary heterologous infections. Similarly, Th2 immunity, typified by a higher IgG4/IgG3 ratio, was associated with worsened human disease severity during secondary infections. Thus, CD1ddep NKT cells establish Th1 polarity during the early innate response to DENV, which promotes infection resolution, memory formation, and long-term protection from secondary homologous and heterologous infections in mice, with consistent associations observed in humans. These observations illustrate how early innate immune responses during primary infections can influence secondary infection outcomes.


Subject(s)
Antibodies, Viral , Antigens, CD1d , Dengue Virus , Dengue , Interferon-gamma , Mice, Knockout , Natural Killer T-Cells , Th1 Cells , Animals , Dengue Virus/immunology , Th1 Cells/immunology , Natural Killer T-Cells/immunology , Mice , Dengue/immunology , Antigens, CD1d/immunology , Antigens, CD1d/genetics , Antibodies, Viral/immunology , Antibodies, Viral/blood , Humans , Interferon-gamma/immunology , Immunoglobulin G/immunology , Granzymes/immunology , Th2 Cells/immunology , Immunologic Memory
14.
J Immunol ; 213(7): 941-951, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39158281

ABSTRACT

Invariant NKT (iNKT) cells are a group of innate-like T cells that plays important roles in immune homeostasis and activation. We found that iNKT cells, compared with CD4+ T cells, have significantly higher levels of lipid peroxidation in both mice and humans. Proteomic analysis also demonstrated that iNKT cells express higher levels of phospholipid hydroperoxidase glutathione peroxidase 4 (Gpx4), a major antioxidant enzyme that reduces lipid peroxidation and prevents ferroptosis. T cell-specific deletion of Gpx4 reduces iNKT cell population, most prominently the IFN-γ-producing NKT1 subset. RNA-sequencing analysis revealed that IFN-γ signaling, cell cycle regulation, and mitochondrial function are perturbed by Gpx4 deletion in iNKT cells. Consistently, we detected impaired cytokine production, elevated cell proliferation and cell death, and accumulation of lipid peroxides and mitochondrial reactive oxygen species in Gpx4 knockout iNKT cells. Ferroptosis inhibitors, iron chelators, vitamin E, and vitamin K2 can prevent ferroptosis induced by Gpx4 deficiency in iNKT cells and ameliorate the impaired function of iNKT cells due to Gpx4 inhibition. Last, vitamin E rescues iNKT cell population in Gpx4 knockout mice. Altogether, our findings reveal the critical role of Gpx4 in regulating iNKT cell homeostasis and function, through controlling lipid peroxidation and ferroptosis.


Subject(s)
Ferroptosis , Homeostasis , Lipid Peroxidation , Mice, Knockout , Natural Killer T-Cells , Phospholipid Hydroperoxide Glutathione Peroxidase , Ferroptosis/immunology , Ferroptosis/physiology , Animals , Lipid Peroxidation/immunology , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Mice , Homeostasis/immunology , Humans , Natural Killer T-Cells/immunology , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Male , Female , Mitochondria/metabolism , Interferon-gamma/metabolism
15.
Front Immunol ; 15: 1436968, 2024.
Article in English | MEDLINE | ID: mdl-39170618

ABSTRACT

Invariant natural killer T (iNKT) cells are immune cells that harness properties of both the innate and adaptive immune system and exert multiple functions critical for the control of various diseases. Prevention of graft-versus-host disease (GVHD) by iNKT cells has been demonstrated in mouse models and in correlative human studies in which high iNKT cell content in the donor graft is associated with reduced GVHD in the setting of allogeneic hematopoietic stem cell transplants. This suggests that approaches to increase the number of iNKT cells in the setting of an allogeneic transplant may reduce GVHD. iNKT cells can also induce cytolysis of tumor cells, and murine experiments demonstrate that activating iNKT cells in vivo or treating mice with ex vivo expanded iNKT cells can reduce tumor burden. More recently, research has focused on testing anti-tumor efficacy of iNKT cells genetically modified to express a chimeric antigen receptor (CAR) protein (CAR-iNKT) cells to enhance iNKT cell tumor killing. Further, several of these approaches are now being tested in clinical trials, with strong safety signals demonstrated, though efficacy remains to be established following these early phase clinical trials. Here we review the progress in the field relating to role of iNKT cells in GVHD prevention and anti- cancer efficacy. Although the iNKT field is progressing at an exciting rate, there is much to learn regarding iNKT cell subset immunophenotype and functional relationships, optimal ex vivo expansion approaches, ideal treatment protocols, need for cytokine support, and rejection risk of iNKT cells in the allogeneic setting.


Subject(s)
Graft vs Host Disease , Natural Killer T-Cells , Natural Killer T-Cells/immunology , Humans , Animals , Graft vs Host Disease/immunology , Graft vs Host Disease/therapy , Graft vs Host Disease/prevention & control , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/adverse effects , Neoplasms/therapy , Neoplasms/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Translational Research, Biomedical , Mice , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics
16.
Adv Exp Med Biol ; 1448: 75-101, 2024.
Article in English | MEDLINE | ID: mdl-39117809

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) constitutes a rare, potentially life-threatening hyperinflammatory immune dysregulation syndrome that can present with a variety of clinical signs and symptoms, including fever, hepatosplenomegaly, and abnormal laboratory and immunological findings such as cytopenias, hyperferritinemia, hypofibrinogenemia, hypertriglyceridemia, elevated blood levels of soluble CD25 (interleukin (IL)-2 receptor α-chain), or diminished natural killer (NK)-cell cytotoxicity (reviewed in detail in Chapter 11 of this book). While HLH can be triggered by an inciting event (e.g., infections), certain monogenic causes have been associated with a significantly elevated risk of development of HLH, or recurrence of HLH in patients who have recovered from their disease episode. These monogenic predisposition syndromes are variably referred to as "familial" (FHL) or "primary" HLH (henceforth referred to as "pHLH") and are the focus of this chapter. Conversely, secondary HLH (sHLH) often occurs in the absence of monogenic etiologies that are commonly associated with pHLH and can be triggered by infections, malignancies, or rheumatological diseases; these triggers and the genetics associated with sHLH are discussed in more detail in other chapters in this book.


Subject(s)
Lymphohistiocytosis, Hemophagocytic , Animals , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/immunology , Recurrence , Risk Factors , Genetic Predisposition to Disease , Interleukin-2 Receptor alpha Subunit/immunology , Natural Killer T-Cells/immunology , Cytotoxicity, Immunologic
17.
Vaccine ; 42(21): 126145, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39034218

ABSTRACT

Protein-based subunit vaccines like RBD-Fc are promising tools to fight COVID-19. RBD-Fc fuses the receptor-binding domain (RBD) of the SARS-CoV-2 virus spike protein with the Fc region of human IgG1, making it more immunogenic than RBD alone. Earlier work showed that combining RBD-Fc with iNKT cell agonists as adjuvants improved neutralizing antibodies but did not sufficiently enhance T cell responses, a limitation RBD-Fc vaccines share with common adjuvants. Here we demonstrate that aluminum hydroxide combined with α-C-GC, a C-glycoside iNKT cell agonist, significantly improved the RBD-Fc vaccine's induction of RBD-specific T-cell responses. Additionally, aluminum hydroxide with α-GC-CPOEt, a phosphonate diester derivative, synergistically elicited more robust neutralizing antibodies. Remarkably, modifying αGC with phosphate (OPO3H2) or phosphonate (CPO3H2) to potentially enhance aluminum hydroxide interaction did not improve efficacy over unmodified αGC with aluminum hydroxide. These findings underscore the straightforward yet potent potential of this approach in advancing COVID-19 vaccine development and provide insights for iNKT cell-based immunotherapy.


Subject(s)
Adjuvants, Immunologic , Aluminum Hydroxide , Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Vaccines, Subunit , COVID-19 Vaccines/immunology , Aluminum Hydroxide/immunology , Aluminum Hydroxide/pharmacology , Aluminum Hydroxide/chemistry , Antibodies, Neutralizing/immunology , Vaccines, Subunit/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Viral/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , COVID-19/prevention & control , COVID-19/immunology , Mice , Immunogenicity, Vaccine , Humans , Natural Killer T-Cells/immunology , Glycolipids/immunology , Glycolipids/chemistry , Female , Adjuvants, Vaccine , Mice, Inbred BALB C
18.
Inflamm Res ; 73(9): 1511-1527, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39028491

ABSTRACT

BACKGROUND: Invariant natural killer T (iNKT) cells play protective or pathogenic roles in a variety of immune and inflammatory diseases. However, whether iNKT cells contribute to the progression of acute neuroinflammation remains unclear. Thus, we addressed this question with a mouse model of lipopolysaccharide (LPS)-induced acute neuroinflammation. METHODS: For induction of acute neuroinflammation, wild-type (WT) C57BL/6 (B6) mice were injected intraperitoneally (i.p.) with LPS for either three or five consecutive days, and then these mice were analyzed for brain-infiltrating leukocytes or mouse behaviors, respectively. To examine the role of iNKT cell activation in LPS-induced neuroinflammation, mice were injected i.p. with the iNKT cell agonist α-galactosylceramide (α-GalCer) seven days prior to LPS treatment. Immune cells infiltrated into the brain during LPS-induced neuroinflammation were determined by flow cytometry. In addition, LPS-induced clinical behavior symptoms such as depressive-like behavior and memory impairment in mice were evaluated by the open field and Y-maze tests, respectively. RESULTS: We found that iNKT cell-deficient Jα18 mutant mice display delayed disease progression and decreased leukocyte infiltration into the brain compared with WT mice, indicating that iNKT cells contribute to the pathogenesis of LPS-induced neuroinflammation. Since it has been reported that pre-treatment with α-GalCer, an iNKT cell agonist, can convert iNKT cells towards anti-inflammatory phenotypes, we next explored whether pre-activation of iNKT cells with α-GalCer can regulate LPS-induced neuroinflammation. Strikingly, we found that α-GalCer pre-treatment significantly delays the onset of clinical symptoms, including depression-like behavior and memory impairment, while decreasing brain infiltration of pro-inflammatory natural killer cells and neutrophils, in this model of LPS-induced neuroinflammation. Such anti-inflammatory effects of α-GalCer pre-treatment closely correlated with iNKT cell polarization towards IL4- and IL10-producing phenotypes. Furthermore, α-GalCer pre-treatment restored the expression of suppressive markers on brain regulatory T cells during LPS-induced neuroinflammation. CONCLUSION: Our findings provide strong evidence that α-GalCer-induced pre-activation of iNKT cells expands iNKT10 cells, mitigating depressive-like behaviors and brain infiltration of inflammatory immune cells induced by LPS-induced acute neuroinflammation. Thus, we suggest the prophylactic potential of iNKT cells and α-GalCer against acute neuroinflammation.


Subject(s)
Brain , Galactosylceramides , Lipopolysaccharides , Mice, Inbred C57BL , Natural Killer T-Cells , Neuroinflammatory Diseases , Animals , Galactosylceramides/pharmacology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/drug effects , Brain/pathology , Brain/drug effects , Brain/immunology , Male , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/chemically induced , Mice , Cytokines/metabolism
19.
Biomed Pharmacother ; 177: 117040, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38959605

ABSTRACT

Invariant natural killer T cell (iNKT) cells produce large amounts of cytokines in response to α-Galactosylceramide (α-GalCer) stimulation. An analog containing two phenyl rings on the acyl chain, C34, was previously found to be more Th1-biased than α-GalCer and triggered greater anticancer activities against breast cancer, melanoma and lung cancer in mice. Since liver is enriched in iNKT cells, we investigated anticancer efficacy of C34 on neuroblastoma with hepatic metastasis. C34 induced Th1-biased cytokine secretions in the liver, significantly suppressed neuroblastoma growth/metastasis and prolonged mouse survival. The anti-tumor efficacy might be attributed to greater expansions of hepatic NKT, NK, CD4+ T, and CD8+ T cells as well as reduction of the number of SSCloGr1intCD11b+ subset of myeloid-derived suppressor cells (MDSCs) in the liver of tumor-bearing mice, as compared to DMSO control group. C34 also upregulated expression of CD1d and CD11c, especially in the SSCloGr1intCD11b+ subset of MDSCs, which might be killed by C34-activated NKT cells, attributing to their reduced number. In addition, C34 also induced expansion of CD4+ T, CD8+ T, and NK cells, which might eliminate neuroblastoma cells. These immune-modulating effects of C34 might act in concert in the local milieu of liver to suppress the tumor growth. Further analysis of database of neuroblastoma revealed that patients with high CD11c expression in the monocytic MDSCs in the tumor had longer survival, suggesting the potential clinical application of C34 for treatment of neuroblastoma.


Subject(s)
Glycolipids , Liver Neoplasms , Natural Killer T-Cells , Neuroblastoma , Animals , Neuroblastoma/pathology , Neuroblastoma/drug therapy , Neuroblastoma/immunology , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Cell Line, Tumor , Mice , Glycolipids/pharmacology , Humans , Female , Cytokines/metabolism , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Antineoplastic Agents/pharmacology , Galactosylceramides/pharmacology
20.
J Med Chem ; 67(15): 12819-12834, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39031770

ABSTRACT

Synthetic α-galactosylceramide (αGalCer) and its analogues as powerful agonists for natural killer T (NKT) cell manipulation have received significant attention in immunotherapy and adjuvant development. However, identifying new potent NKT cell agonists, especially those with Th1 selectivity that promote anticancer effects, remains a challenging task. In this work, we introduced a sulfonamide group into the acyl chain of αGalCer to form additional hydrogen bonds to intensify the glycolipid/CD1d interaction. Two compounds GCS-11 and GCS-12 demonstrated remarkable potency while exhibiting different cytokine induction patterns. Compared to αGalCer, the Th1-biased GCS-11 exhibited a 6-fold increase in IFN-γ but not IL-4, while the Th1/2-balanced GCS-12 elicited 7- and 5-fold increase in IFN-γ and IL-4, respectively, in vivo. These findings place them among the most potent NKT cell agonists, with superior antitumor effects. Therefore, hydrogen-bond-involved derivatization could be a powerful strategy to develop potent and polarized NKT cell agonists for various immunotherapies.


Subject(s)
Antigens, CD1d , Cytokines , Drug Design , Galactosylceramides , Hydrogen Bonding , Natural Killer T-Cells , Natural Killer T-Cells/immunology , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/metabolism , Animals , Galactosylceramides/chemistry , Galactosylceramides/pharmacology , Galactosylceramides/chemical synthesis , Mice , Cytokines/metabolism , Antigens, CD1d/metabolism , Antigens, CD1d/chemistry , Humans , Interleukin-4/metabolism , Structure-Activity Relationship , Mice, Inbred C57BL , Interferon-gamma/metabolism , Sulfonamides/chemistry , Sulfonamides/pharmacology , Sulfonamides/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor
SELECTION OF CITATIONS
SEARCH DETAIL