1
|
Jennings-Gee JE, Daly CA, Bray AS, Dyevoich AM, Spurrier MA, Haas KM. B cell-expressed CD1d promotes MPL/TDCM lipid emulsion adjuvant effects in polysaccharide vaccines. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf074. [PMID: 40280183 DOI: 10.1093/jimmun/vkaf074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 03/09/2025] [Indexed: 04/29/2025]
Abstract
T cell-independent type 2 antigens (TI-2 Ags), such as pneumococcal polysaccharides, elicit weak immunoglobulin G (IgG) responses and are refractive to boosting. Overcoming this challenge is critical for improving vaccines. Previously, we demonstrated a lipid-based adjuvant composed of monophosphoryl lipid A, synthetic cord factor, and squalene significantly boosts primary and secondary IgM and IgG production against polysaccharide Ags. Herein, we show beta-2 microglobulin, but not MHC class II, is essential for adjuvant-induced increases in polysaccharide-specific IgG levels. Furthermore, we demonstrate CD1d expression is essential for optimal adjuvant-induced increases in IgG, but is not required for IgG responses to TI-2 Ags administered without adjuvant, with the exception of the bacterial cell wall polysaccharide component, phosphorylcholine. Adoptive transfer of splenic and peritoneal cells from VHB1-8 transgenic mice into CD1d-/- mice revealed adjuvant-induced increases in NP-Ficoll-specific IgG production by CD1d+/+ transgenic B cells, but not recipient B cells, suggesting B cell-expressed CD1d is critical for adjuvant-induced effects on TI-2 antibody responses. Consistent with this, bone marrow chimera mice with selective CD1d deficiency in B cells demonstrated B cell-expressed CD1d was dispensable for iNKT cell development and maintenance but was required for adjuvant-induced increases in protective levels of polysaccharide- and phosphorylcholine-specific IgG. Notably, both iNKT cells and CD1d crosslinking significantly increased IgG production by B cells coactivated with TI-2 Ag and adjuvant, suggesting iNKT-induced CD1d signaling may promote increased IgG production. In summary, our study reveals B cell-dependent CD1d expression is critical for effectiveness of a potent lipid-based adjuvant in augmenting polysaccharide- and phosphorylcholine-specific IgG responses.
Collapse
Affiliation(s)
- Jamie E Jennings-Gee
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Christina A Daly
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Andrew S Bray
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Allison M Dyevoich
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - M Ariel Spurrier
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Karen M Haas
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
2
|
Qiao J, Gao Z, Zhang C, Hennigs, Wu B, Jing L, Gao R, Yang Y. Structural characterization and immune modulation activities of Chinese Angelica polysaccharide (CAP) and selenizing CAP (sCAP) on dendritic cells. Int J Biol Macromol 2024; 277:132628. [PMID: 38797292 DOI: 10.1016/j.ijbiomac.2024.132628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
sCAP was obtained by the nitrate‑sodium selenite method. SEM, molecular weight evaluation, monosaccharide composition, FT-IR and NMR of sCAP were carried out. Compared with CAP, sCAP had a relatively smooth and lamellar sheet morphology with edge folds on the surface, presented molecular weights in range of 0.90-97.08 KDa, and was mainly composed of GalA, Ara and Gal. sCAP had both α and β configurations of the pyranose ring, the characteristic vibrational peak of Se-O-C and the signal of galacturonic acid residue. The phagocytic activity of immature BMDCs, the expression of CD40, CD80, CD86, and MHCII on BMDCs were detected by flow cytometry, the ability of sCAP-treated BMDCs to stimulate the proliferation of allogeneic lymphocytes, presentation of antigens, cytokines in the supernatants and the protein in MyD88/NF-κB signaling pathway were detected. The results showed that the phagocytic activity of immature BMDCs was significantly enhanced when sCAP was at 3.92-1.96 μg·mL-1. The levels of IL-6, TGF-β1, INF-γ, and TNF-α were significantly elevated, IL-1β and MIP-1α were significantly reduced. These results indicate that sCAP could be as a new immunopotentiator by increasing MyD88/NF-κB signaling pathway. This study provides a reference for the research and development of new dosage forms of polysaccharide.
Collapse
Affiliation(s)
- Jie Qiao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, PR China
| | - Zhenzhen Gao
- College of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, Jiangsu 212499, PR China; Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, PR China.
| | - Chao Zhang
- College of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, Jiangsu 212499, PR China
| | - Hennigs
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, PR China
| | - Bo Wu
- Ordos Vocational College of Agriculture and Forestry, Ordos, Inner Mongolia 017010, PR China
| | - Lirong Jing
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, PR China
| | - Ruifeng Gao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, PR China
| | - Ying Yang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, PR China
| |
Collapse
|
3
|
Evans L, Barral P. CD1 molecules: Beyond antigen presentation. Mol Immunol 2024; 170:1-8. [PMID: 38579449 PMCID: PMC11481681 DOI: 10.1016/j.molimm.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/18/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024]
Abstract
CD1 molecules are well known for their role in binding and presenting lipid antigens to mediate the activation of CD1-restricted T cells. However, much less appreciated is the fact that CD1 molecules can have additional "unconventional" roles which impact the activation and functions of CD1-expressing cells, ultimately controlling tissue homeostasis as well as the progression of inflammatory and infectious diseases. Some of these roles are mediated by so-called reverse signalling, by which crosslinking of CD1 molecules at the cell surface initiates intracellular signalling. On the other hand, CD1 molecules can also control metabolic and inflammatory pathways in CD1-expressing cells through cell-intrinsic mechanisms independent of CD1 ligation. Here, we review the evidence for "unconventional" functions of CD1 molecules and the outcomes of such roles for health and disease.
Collapse
Affiliation(s)
- Lauren Evans
- The Peter Gorer Department of Immunobiology. King's College London, London, UK; The Francis Crick Institute, London, UK
| | - Patricia Barral
- The Peter Gorer Department of Immunobiology. King's College London, London, UK; The Francis Crick Institute, London, UK.
| |
Collapse
|
4
|
Lei Z, Yu J, Wu Y, Shen J, Lin S, Xue W, Mao C, Tang R, Sun H, Qi X, Wang X, Xu L, Wei C, Wang X, Chen H, Hao P, Yin W, Zhu J, Li Y, Wu Y, Liu S, Liang H, Chen X, Su C, Zhou S. CD1d protects against hepatocyte apoptosis in non-alcoholic steatohepatitis. J Hepatol 2024; 80:194-208. [PMID: 38438948 DOI: 10.1016/j.jhep.2023.10.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 10/02/2023] [Accepted: 10/17/2023] [Indexed: 03/06/2024]
Abstract
BACKGROUND & AIMS Hepatocyte apoptosis, a well-defined form of cell death in non-alcoholic steatohepatitis (NASH), is considered the primary cause of liver inflammation and fibrosis. However, the mechanisms underlying the regulation of hepatocyte apoptosis in NASH remain largely unclear. We explored the anti-apoptotic effect of hepatocyte CD1d in NASH. METHODS Hepatocyte CD1d expression was analyzed in patients with NASH and mouse models. Hepatocyte-specific gene overexpression or knockdown and anti-CD1d crosslinking were used to investigate the anti-apoptotic effect of hepatocyte CD1d on lipotoxicity-, Fas-, and concanavalin (ConA)-mediated liver injuries. A high-fat diet, a methionine-choline-deficient diet, a Fas agonist, and ConA were used to induce lipotoxic and/or apoptotic liver injuries. Palmitic acid was used to mimic lipotoxicity-induced apoptosis in vitro. RESULTS We identified a dramatic decrease in CD1d expression in hepatocytes of patients with NASH and mouse models. Hepatocyte-specific CD1d overexpression and knockdown experiments collectively demonstrated that hepatocyte CD1d protected against hepatocyte apoptosis and alleviated hepatic inflammation and injuries in NASH mice. Furthermore, decreased JAK2-STAT3 signaling was observed in NASH patient livers. Mechanistically, anti-CD1d crosslinking on hepatocytes induced tyrosine phosphorylation of the CD1d cytoplasmic tail, leading to the recruitment and phosphorylation of JAK2. Phosphorylated JAK2 activated STAT3 and subsequently reduced apoptosis in hepatocytes, which was associated with an increase in anti-apoptotic effectors (Bcl-xL and Mcl-1) and a decrease in pro-apoptotic effectors (cleaved-caspase 3/7). Moreover, anti-CD1d crosslinking effectively protected against Fas- or ConA-mediated hepatocyte apoptosis and liver injury in mice. CONCLUSIONS Our study uncovered a previously unrecognized anti-apoptotic CD1d-JAK2-STAT3 axis in hepatocytes that conferred hepatoprotection and highlighted the potential of hepatocyte CD1d-directed therapy for liver injury and fibrosis in NASH, as well as in other liver diseases associated with hepatocyte apoptosis. IMPACT AND IMPLICATIONS Excessive and/or sustained hepatocyte apoptosis is critical in driving liver inflammation and injury. The mechanisms underlying the regulation of hepatocyte apoptosis in non-alcoholic steatohepatitis (NASH) remain largely unclear. Here, we found that CD1d expression in hepatocytes substantially decreases and negatively correlates with the severity of liver injury in patients with NASH. We further revealed a previously unrecognized anti-apoptotic CD1d-JAK2-STAT3 signaling axis in hepatocytes, which confers significant protection against liver injury in NASH and acute liver diseases. Thus, hepatocyte CD1d-targeted therapy could be a promising strategy to manipulate liver injury in both NASH and other hepatocyte apoptosis-related liver diseases.
Collapse
Affiliation(s)
- Zhigang Lei
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiaojiao Yu
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yu Wu
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junyao Shen
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shibo Lin
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weijie Xue
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenxu Mao
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Tang
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haoran Sun
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Qi
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaohong Wang
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Xu
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuan Wei
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaowei Wang
- Department of Blood Transfusion, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongbing Chen
- Department of Clinical Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ping Hao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Wen Yin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Jifeng Zhu
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yalin Li
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Shouguo Liu
- Center for Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hui Liang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaojun Chen
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Chuan Su
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Sha Zhou
- Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
5
|
Orlova E, Loginova O, Shirshev S. Leptin regulates thymic plasmacytoid dendritic cell ability to influence the thymocyte distribution in vitro. Int Immunopharmacol 2023; 117:109912. [PMID: 36857934 DOI: 10.1016/j.intimp.2023.109912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 03/03/2023]
Abstract
Leptin, the adipocyte-derived hormone, involved in regulating food intake and body weight, plays an important role in immunity and reproduction. Leptin signals via the specific membrane receptors expressed in most types of immune cells including dendritic cells (DCs) and thymocytes. Leptin enhances thymopoiesis and modulates T-cell-mediated immunity. Thymic plasmacytoid DCs (pDCs) are predominated in the thymus. They play an important role in thymocyte differentiation. We have analyzed whether leptin mediates its effects on human thymocytes by influencing on pDCs. We used leptin at concentration corresponding to its level during II-III trimesters of physiological pregnancy. We cultivated leptin-primed pDCs with autologous thymocytes and estimated the main thymocyte subsets expressing αβ chains of the T-cell receptor (αβTCR), natural regulatory T-cells (tTreg), natural T-helpers producing interleukin-17 (nTh17) and invariant natural killer T-cells (iNKT) in vitro. We have shown that leptin augmented CD86, CD276 expressions and depressed IL-10 productions by pDCs. Leptin-primed pDCs decreased the percentage of CD4+CD8+αβTCR+ thymocytes, increased CD4hiCD8-/loαβTCR+ cells. pDCs cultivated with leptin decreased the number of iNKT precursors, and did not change the number of tTreg and nTh17 precursors. Thus, leptin's important role in regulation of thymic pDC abilities to influence on the thymocyte distribution was indicated in vitro.
Collapse
Affiliation(s)
- Ekaterina Orlova
- Laboratory of Immunoregulation, Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, 13A Lenina str., 614015 Perm, Russia.
| | - Olga Loginova
- Laboratory of Immunoregulation, Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, 13A Lenina str., 614015 Perm, Russia.
| | - Sergei Shirshev
- Laboratory of Immunoregulation, Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, 13A Lenina str., 614015 Perm, Russia.
| |
Collapse
|
6
|
Xiao R, Mansour AG, Huang W, Hassan QN, Wilkins RK, Komatineni SV, Bates R, Ali S, Chrislip LA, Queen NJ, Ma S, Yu J, Lordo MR, Mundy-Bosse BL, Caligiuri MA, Cao L. Adipocyte CD1d Gene Transfer Induces T Cell Expansion and Adipocyte Inflammation in CD1d Knockout Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2109-2121. [PMID: 35418470 PMCID: PMC9050908 DOI: 10.4049/jimmunol.2100313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 02/15/2022] [Indexed: 05/03/2023]
Abstract
CD1d, a lipid Ag-presenting molecule for invariant NKT (iNKT) cells, is abundantly expressed on adipocytes and regulates adipose homeostasis through iNKT cells. CD1d gene expression was restored in visceral adipose tissue adipocytes of CD1d knockout (KO) mice to investigate the interactions between adipocytes and immune cells within adipose tissue. We developed an adipocyte-specific targeting recombinant adeno-associated viral vector, with minimal off-target transgene expression in the liver, to rescue CD1d gene expression in visceral adipose tissue adipocytes of CD1d KO mice, followed by assessment of immune cell alternations in adipose tissue and elucidation of the underlying mechanisms of alteration. We report that adeno-associated virus-mediated gene transfer of CD1d to adipocytes in CD1d KO mice fails to rescue iNKT cells but leads to massive and selective expansion of T cells within adipose tissue, particularly CD8+ T effector cells, that is associated with adipocyte NLRP3 inflammasome activation, dysregulation of adipocyte functional genes, and upregulation of apoptotic pathway proteins. An NLRP3 inhibitor has no effect on T cell phenotypes whereas depletion of CD8+ T cells significantly attenuates inflammasome activation and abolishes the dysregulation of adipocyte functional genes induced by adipocyte CD1d. In contrast, adipocyte overexpression of CD1d fails to induce T cell activation in wild-type mice or in invariant TCR α-chain Jα18 KO mice that have a normal lymphocyte repertoire except for iNKT cells. Our studies uncover an adipocyte CD1d → CD8+ T cell → adipocyte inflammasome cascade, in which CD8+ T cells function as a key mediator of adipocyte inflammation likely induced by an allogeneic response against the CD1d molecule.
Collapse
Affiliation(s)
- Run Xiao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Anthony G Mansour
- Department of Hematological Malignancies and Stem Cell Transplantation, City of Hope National Medical Center and the Beckman Research Institute, Los Angeles, CA
| | - Wei Huang
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Quais N Hassan
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH
- Medical Scientist Training Program, The Ohio State University, Columbus, OH; and
| | - Ryan K Wilkins
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Suraj V Komatineni
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Rhiannon Bates
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Seemaab Ali
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH
- Medical Scientist Training Program, The Ohio State University, Columbus, OH; and
| | - Logan A Chrislip
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Nicholas J Queen
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Shoubao Ma
- Department of Hematological Malignancies and Stem Cell Transplantation, City of Hope National Medical Center and the Beckman Research Institute, Los Angeles, CA
| | - Jianhua Yu
- Department of Hematological Malignancies and Stem Cell Transplantation, City of Hope National Medical Center and the Beckman Research Institute, Los Angeles, CA
| | - Matthew R Lordo
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH
- Medical Scientist Training Program, The Ohio State University, Columbus, OH; and
| | - Bethany L Mundy-Bosse
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH
- Division of Hematology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Michael A Caligiuri
- Department of Hematological Malignancies and Stem Cell Transplantation, City of Hope National Medical Center and the Beckman Research Institute, Los Angeles, CA;
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH;
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH
| |
Collapse
|
7
|
Khan MA, Malik A, Alruwetei A, Alzohairy MA, Alhatlani BY, Al Rugaie O, Alhumaydhi FA, Khan A. Delivery of MERS antigen encapsulated in α-GalCer-bearing liposomes elicits stronger antigen-specific immune responses. J Drug Target 2022; 30:884-893. [PMID: 35418263 DOI: 10.1080/1061186x.2022.2066681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alpha-Galactosylceramide (α-GalCer) effectively activates the natural killer T (NKT) cells to secrete remarkable amounts of Th1 and Th2 cytokines and therefore, acts as a potential immunoadjuvant in vaccine formulation. In the present study, we prepared α-GalCer-bearing or α-GalCer-free liposomes and loaded them with Middle East Respiratory Syndrome Corona virus papain-like protease (α-GalCer-Lip-MERS-CoV PLpro or Lip- MERS-CoV PLpro). These formulations were injected in mice to investigate the antigen-specific humoral and cell-mediated immune responses. The immunization with α-GalCer-Lip-MERS-CoV PLpro or Lip- MERS-CoV PLpro did not induce any notable toxicity in immunized mice. The results demonstrated that mice immunized with α-GalCer-Lip-MERS-CoV PLpro showed greater antigen-specific antibody titer, switching of IgG isotyping to IgG2a subclass and higher lymphocyte proliferation. Moreover, the splenocytes from α-GalCer-Lip-MERS-CoV PLpro immunized mice secreted greater levels of IFN-γ, IL-4, IL-2 and IL-12. Interestingly, a booster dose induced stronger memory immune responses in mice previously immunized with α-GalCer-Lip-MERS-CoV PLpro. In summary, α-GalCer-Lip-MERS-CoV PLpro may prove to be a promising vaccine formulation to protect the individuals against MERS-CoV infection.
Collapse
Affiliation(s)
- Masood Alam Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Ajamaluddin Malik
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abdulmohsen Alruwetei
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Mohammad A Alzohairy
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Bader Y Alhatlani
- Department of Applied Medical Sciences, Applied College in Unayzah, Qassim University, Unayzah, Saudi Arabia
| | - Osamah Al Rugaie
- Department of Basic Medical Sciences, College of Medicine and Medical Sciences, Qassim University, Unayzah, Saudi Arabia
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Arif Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
8
|
Yang J, Chang T, Tang L, Deng H, Chen D, Luo J, Wu H, Tang T, Zhang C, Li Z, Dong L, Yang XP, Tang ZH. Increased Expression of Tim-3 Is Associated With Depletion of NKT Cells In SARS-CoV-2 Infection. Front Immunol 2022; 13:796682. [PMID: 35250975 PMCID: PMC8889099 DOI: 10.3389/fimmu.2022.796682] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 01/28/2022] [Indexed: 12/14/2022] Open
Abstract
In the ongoing coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), natural killer T (NKT) cells act as primary initiators of immune responses. However, a decrease of circulating NKT cells has been observed in COVID-19 different stages, of which the underlying mechanism remains to be elucidated. Here, by performing single-cell RNA sequencing analysis in three large cohorts of COVID-19 patients, we found that increased expression of Tim-3 promotes depletion of NKT cells during the progression stage of COVID-19, which is associated with disease severity and outcome of patients with COVID-19. Tim-3+ NKT cells also expressed high levels of CD147 and CD26, which are potential SARS-CoV-2 spike binding receptors. In the study, Tim-3+ NKT cells showed high enrichment of apoptosis, higher expression levels of mitochondrial genes and caspase genes, with a larger pseudo time value. In addition, Tim-3+ NKT cells in COVID-19 presented a stronger capacity to secrete IFN-γ, IL-4 and IL-10 compared with healthy individuals, they also demonstrated high expression of co-inhibitory receptors such as PD-1, CTLA-4, and LAG-3. Moreover, we found that IL-12 secreted by dendritic cells (DCs) was positively correlated with up-regulated expression of Tim-3 in NKT cells in COVID-19 patients. Overall, this study describes a novel mechanism by which up-regulated Tim-3 expression induced the depletion and dysfunction of NKT cells in COVID-19 patients. These findings not only have possible implications for the prediction of severity and prognosis in COVID-19 but also provide a link between NKT cells and future new therapeutic strategies in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jingzhi Yang
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Wuhan, China
| | - Teding Chang
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Wuhan, China
| | - Liangsheng Tang
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Wuhan, China
| | - Hai Deng
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Wuhan, China
| | - Deng Chen
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Wuhan, China
| | - Jialiu Luo
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Wuhan, China
| | - Han Wu
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Wuhan, China
| | - TingXuan Tang
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Cong Zhang
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Wuhan, China
| | - Zhenwen Li
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Wuhan, China
| | - Liming Dong
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Wuhan, China
| | - Xiang-Ping Yang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao-Hui Tang
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Wuhan, China
| |
Collapse
|
9
|
Cui S, Wang C, Bai W, Li J, Pan Y, Huang X, Yang H, Feng Z, Xiang Q, Fei L, Zheng L, Huang J, Zhang Q, Wu Y, Chen Y. CD1d1 intrinsic signaling in macrophages controls NLRP3 inflammasome expression during inflammation. SCIENCE ADVANCES 2020; 6:6/43/eaaz7290. [PMID: 33087357 PMCID: PMC7577718 DOI: 10.1126/sciadv.aaz7290] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 09/08/2020] [Indexed: 06/11/2023]
Abstract
Dysregulation of immune responses in the gut often associates with inflammatory bowel diseases (IBD). Mouse CD1d1, an ortholog of human CD1d mainly participating in lipid-antigen presentation to NKT cells, is able to generate intrinsic signals upon stimulation. Mice with macrophage-specific CD1d1 deficiency (LymCD1d1-/- ) acquire resistance to dextran sodium sulfate (DSS)-induced colitis, attributing to the transcriptional inhibition of NLRP3 inflammasome components. The hyperactivation of NLRP3 inflammasome accounts for gut epithelial proliferation and intestine-blood barrier integrity. Mechanistically, occupancy by the natural ligand glycosphingolipid iGb3, CD1d1 responds with intracellular Ser330 dephosphorylation thus to reduce the Peroxiredoxin 1 (PRDX1)-associated AKT-STAT1 phosphorylation and subsequent NF-κB activation, eventually causing transcriptional down-regulation of Nlrp3 and its immediate substrates Il1b and Il18 in macrophages. Therefore, the counterbalancing role of CD1d1 in macrophages appears to determine severity of DSS-mediated colitis in mice. These findings propose new intervention strategies for treating IBD and other inflammatory disorders.
Collapse
Affiliation(s)
- Shan Cui
- Yanbian University Hospital, Yanbian University, Jilin Province 133000, People's Republic of China
| | - Chenhui Wang
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Weizhi Bai
- Department of Emergency, Chongqing University Center Hospital, Chongqing Emergency Medical Center, Chongqing 400038, People's Republic of China
| | - Jiao Li
- School of Medicine, Yanbian University, Jilin Province 133000, People's Republic of China
| | - Yue Pan
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Xiaoyong Huang
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Han Yang
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Zeqing Feng
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Qun Xiang
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Lei Fei
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, People's Republic of China
| | - Lixin Zheng
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Jian Huang
- Department of Emergency, Chongqing University Center Hospital, Chongqing Emergency Medical Center, Chongqing 400038, People's Republic of China.
| | - Qinggao Zhang
- School of Medicine, Yanbian University, Jilin Province 133000, People's Republic of China.
| | - Yuzhang Wu
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, People's Republic of China.
| | - Yongwen Chen
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, People's Republic of China.
| |
Collapse
|
10
|
Glycolipid iGb3 feedback amplifies innate immune responses via CD1d reverse signaling. Cell Res 2018; 29:42-53. [PMID: 30514903 DOI: 10.1038/s41422-018-0122-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 11/14/2018] [Indexed: 02/07/2023] Open
Abstract
The cross-talk between cellular lipid metabolism and the innate immune responses remains obscure. In addition to presenting lipid antigens to Natural Killer T-cells (NKT cells), the Cluster of Differentiation 1D Glycoprotein (CD1d) might mediate reverse signaling in antigen-presenting cells (APCs). Here we found CD1d deficiency attenuated Toll-like receptor (TLR)-triggered inflammatory innate responses in macrophages and dendritic cells, protecting mice from endotoxin shock. TLR activation in macrophages induced metabolic changes of glycosphingolipids (GSLs), among which glycolipid isoglobotrihexosylceramide (iGb3) was rapidly produced. The endogenously generated iGb3 bound CD1d in endosomal compartments and then synergized with the initially activated TLR signal to induce Tyr332 phosphorylation of CD1d intracellular domain. This led to the recruitment and activation of proline-rich tyrosine kinase 2 (Pyk2). Pyk2 interacted with IκB kinase β (IKKβ) and TANK-binding kinase 1 (TBK1), and enhanced tyrosine phosphorylation of Tyr188/199 of IKKβ and Tyr179 of TBK1 and thus, their activation to promote full activation of TLR signaling. Thus, intracellular CD1d reverse signaling, triggered by endogenous iGb3, amplifies inflammatory innate responses in APCs. Our findings identify a non-canonical function of CD1d reverse signaling activated by lipid metabolite in the innate immune response.
Collapse
|
11
|
Hapil FZ, Wingender G. The interaction between invariant Natural Killer T cells and the mucosal microbiota. Immunology 2018; 155:164-175. [PMID: 29893412 DOI: 10.1111/imm.12958] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 05/21/2018] [Indexed: 02/06/2023] Open
Abstract
The surface of mammalian bodies is colonized by a multitude of microbial organisms, which under normal conditions support the host and are considered beneficial commensals. This requires, however, that the composition of the commensal microbiota is tightly controlled and regulated. The host immune system plays an important role in the maintenance of this microbiota composition. Here we focus on the contribution of one particular immune cell type, invariant Natural Killer T (iNKT) cells, in this process. The iNKT cells are a unique subset of T cells characterized by two main features. First, they express an invariant T-cell receptor that recognizes glycolipid antigens presented by CD1d, a non-polymorphic major histocompatibility complex class I-like molecule. Second, iNKT cells develop as effector/memory cells and swiftly exert effector functions, like cytokine production and cytotoxicity, after activation. We outline the influence that the mucosal microbiota can have on iNKT cells, and how iNKT cells contribute to the maintenance of the microbiota composition.
Collapse
Affiliation(s)
| | - Gerhard Wingender
- Izmir Biomedicine and Genome Center, Balcova/Izmir, Turkey.,Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Balcova/Izmir, Turkey
| |
Collapse
|
12
|
Saez de Guinoa J, Jimeno R, Farhadi N, Jervis PJ, Cox LR, Besra GS, Barral P. CD1d-mediated activation of group 3 innate lymphoid cells drives IL-22 production. EMBO Rep 2017; 18:39-47. [PMID: 27799287 PMCID: PMC5210076 DOI: 10.15252/embr.201642412] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 09/07/2016] [Accepted: 10/04/2016] [Indexed: 12/21/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a heterogeneous family of immune cells that play a critical role in a variety of immune processes including host defence against infection, wound healing and tissue repair. Whether these cells are involved in lipid-dependent immunity remains unexplored. Here we show that murine ILCs from a variety of tissues express the lipid-presenting molecule CD1d, with group 3 ILCs (ILC3s) showing the highest level of expression. Within the ILC3 family, natural cytotoxicity triggering receptor (NCR)-CCR6+ cells displayed the highest levels of CD1d. Expression of CD1d on ILCs is functionally relevant as ILC3s can acquire lipids in vitro and in vivo and load lipids on CD1d to mediate presentation to the T-cell receptor of invariant natural killer T (iNKT) cells. Conversely, engagement of CD1d in vitro and administration of lipid antigen in vivo induce ILC3 activation and production of IL-22. Taken together, our data expose a previously unappreciated role for ILCs in CD1d-mediated immunity, which can modulate tissue homeostasis and inflammatory responses.
Collapse
Affiliation(s)
| | - Rebeca Jimeno
- The Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Nazanin Farhadi
- The Peter Gorer Department of Immunobiology, King's College London, London, UK
| | - Peter J Jervis
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Liam R Cox
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, UK
| | - Gurdyal S Besra
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Patricia Barral
- The Peter Gorer Department of Immunobiology, King's College London, London, UK
| |
Collapse
|
13
|
Julian MW, Strange HR, Ballinger MN, Hotchkiss RS, Papenfuss TL, Crouser ED. Tolerance and Cross-Tolerance following Toll-Like Receptor (TLR)-4 and -9 Activation Are Mediated by IRAK-M and Modulated by IL-7 in Murine Splenocytes. PLoS One 2015. [PMID: 26218271 PMCID: PMC4517781 DOI: 10.1371/journal.pone.0132921] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objective Immune suppression during critical illness predisposes to serious infections. We sought to determine the mechanisms regulating tolerance and cross-tolerance to common pro-inflammatory danger signals in a model that recapitulates the intact in vivo immune response. Materials and Methods Flt3-expanded splenocytes obtained from wild-type or matching IRAK-M knockout (IRAK-M-/-), C57BL/6, male mice (8–10 weeks old) were treated repeatedly or alternately with either LPS or CpGA DNA, agonists of Toll-like receptor (TLR)-4 and -9, respectively, over successive 24-hour periods. Supernatants were collected following each 24-hour period with cytokine release (ELISA) and splenocyte IRAK-M expression (Western blot) determined. Tolerance and cross-tolerance were assessed in the absence or presence of programmed death receptor (PD)-1 blocking antibody or IL-7 pre-treatment. Main Results Splenocytes notably exhibited both tolerance and cross-tolerance to subsequent treatments with either LPS or CpGA DNA. The character of tolerance and cross-tolerance in this model was distinct following initial LPS or CpGA treatment in that TNFα and IFNγ release (not IL-10) were suppressed following LPS; whereas, initial CpGA treatment suppressed TNFα, IFNγ and IL-10 release in response to subsequent stimulation (LPS or CpGA). Tolerance and cross-tolerance were unrelated to IL-10 release or PD-1 but were attenuated in IRAK-M-/- splenocytes. IL-7 significantly suppressed IRAK-M expression and restored TNFα and IFNγ production without influencing IL-10 release. Conclusions In summary, acute immune tolerance and cross-tolerance in response to LPS or CpGA were distinct in that LPS selectively suppressed pro-inflammatory cytokine responses; whereas, CpGA suppressed both pro- and anti-inflammatory responses. The induction of tolerance and cross-tolerance in response to common danger signals was mechanistically unrelated to IL-10 or PD-1 but was directly influenced by IRAK-M expression. IL-7 reduced IRAK-M expression and attenuated immune tolerance induced by either LPS or CpGA, and thus may be useful for reversal of immune tolerance in the setting of critical illness.
Collapse
Affiliation(s)
- Mark W. Julian
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary Allergy, Critical Care, and Sleep Medicine, Wexner Medical Center, Columbus, OH, United States of America
| | - Heather R. Strange
- College of Veterinary Medicine, Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States of America
| | - Megan N. Ballinger
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary Allergy, Critical Care, and Sleep Medicine, Wexner Medical Center, Columbus, OH, United States of America
| | - Richard S. Hotchkiss
- Departments of Anesthesiology, Medicine and Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Tracey L. Papenfuss
- College of Veterinary Medicine, Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States of America
| | - Elliott D. Crouser
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary Allergy, Critical Care, and Sleep Medicine, Wexner Medical Center, Columbus, OH, United States of America
- * E-mail:
| |
Collapse
|
14
|
Loffredo S, Staiano RI, Granata F, Costantino V, Borriello F, Frattini A, Lepore MT, Mangoni A, Marone G, Triggiani M. Simplexide induces CD1d-dependent cytokine and chemokine production from human monocytes. PLoS One 2014; 9:e111326. [PMID: 25390653 PMCID: PMC4229102 DOI: 10.1371/journal.pone.0111326] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 09/29/2014] [Indexed: 12/24/2022] Open
Abstract
Monocytes are major effector cells of innate immunity and recognize several endogenous and exogenous molecules due to the expression of wide spectrum of receptors. Among them, the MHC class I-like molecule CD1d interacts with glycolipids and presents them to iNKT cells, mediating their activation. Simplexide belongs to a novel class of glycolipids isolated from marine sponges and is structurally distinct from other immunologically active glycolipids. In this study we have examined the effects of simplexide on cytokine and chemokine release from human monocytes. Simplexide induces a concentration- and time-dependent release of IL-6, CXCL8, TNF-α and IL-10 and increases the expression of IL6, CXCL8 and IL10 mRNA. Cytokine and chemokine release induced by simplexide from monocytes is dependent on CD1d since: i) a CD1d antagonist, 1,2-bis (diphenylphosphino) ethane [DPPE]-polyethylene glycolmonomethylether [PEG], specifically blocks simplexide-induced activation of monocytes; ii) CD1d knockdown inhibits monocyte activation by simplexide and iii) simplexide induces cytokine production from CD1d-transfected but not parental C1R cell line. Finally, we have shown that simplexide also induces iNKT cell expansion in vitro. Our results demonstrate that simplexide, apart from activating iNKT cells, induces the production of cytokines and chemokines from human monocytes by direct interaction with CD1d.
Collapse
Affiliation(s)
- Stefania Loffredo
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Rosaria I. Staiano
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Francescopaolo Granata
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Valeria Costantino
- The NeaNAT group - Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Francesco Borriello
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Annunziata Frattini
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Maria Teresa Lepore
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Alfonso Mangoni
- The NeaNAT group - Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- * E-mail:
| | - Massimo Triggiani
- Division of Allergy and Clinical Immunology, University of Salerno, Salerno, Italy
| |
Collapse
|
15
|
Lameris R, Schneiders FL, de Gruijl TD, van der Vliet HJ. Exploiting the CD1d-iNKT cell axis for potentiation of DC-based cancer vaccines. Methods Mol Biol 2014; 1139:155-65. [PMID: 24619678 DOI: 10.1007/978-1-4939-0345-0_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Invariant natural killer T cells (iNKT) and dendritic cells (DC) play a central role in tumor immunity through downstream activation of immune effector cells by pro-inflammatory cytokines. Evidence is accumulating that the CD1d-iNKT cell axis can be effectively used to potentiate DC-based cancer vaccines. Here, we provide a detailed methodology for the generation of (CD1d-expressing) monocyte-derived DC (moDC) and their subsequent loading with the iNKT cell agonist α-galactosylceramide (α-GalCer) or their direct ligation by agonistic anti-CD1d monoclonal antibodies.
Collapse
Affiliation(s)
- Roeland Lameris
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
16
|
Izhak L, Ambrosino E, Kato S, Parish ST, O’Konek JJ, Weber H, Xia Z, Venzon D, Berzofsky JA, Terabe M. Delicate balance among three types of T cells in concurrent regulation of tumor immunity. Cancer Res 2013; 73:1514-23. [PMID: 23319803 PMCID: PMC3622595 DOI: 10.1158/0008-5472.can-12-2567] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The nature of the regulatory cell types that dominate in any given tumor is not understood at present. Here, we addressed this question for regulatory T cells (Treg) and type II natural killer T (NKT) cells in syngeneic models of colorectal and renal cancer. In mice with both type I and II NKT cells, or in mice with neither type of NKT cell, Treg depletion was sufficient to protect against tumor outgrowth. Surprisingly, in mice lacking only type I NKT cells, Treg blockade was insufficient for protection. Thus, we hypothesized that type II NKT cells may be neutralized by type I NKT cells, leaving Tregs as the primary suppressor, whereas in mice lacking type I NKT cells, unopposed type II NKT cells could suppress tumor immunity even when Tregs were blocked. We confirmed this hypothesis in 3 ways by reconstituting type I NKT cells as well as selectively blocking or activating type II NKT cells with antibody or the agonist sulfatide, respectively. In this manner, we showed that blockade of both type II NKT cells and Tregs is necessary to abrogate suppression of tumor immunity, but a third cell, the type I NKT cell, determines the balance between these regulatory mechanisms. As patients with cancer often have deficient type I NKT cell function, managing this delicate balance among 3 T-cell subsets may be critical for the success of immunotherapy for human cancer.
Collapse
Affiliation(s)
- Liat Izhak
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Elena Ambrosino
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Shingo Kato
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Stanley T. Parish
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Jessica J. O’Konek
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Hannah Weber
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Zheng Xia
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - David Venzon
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Jay A. Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Masaki Terabe
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| |
Collapse
|
17
|
Shah HB, Devera TS, Rampuria P, Lang GA, Lang ML. Type II NKT cells facilitate Alum-sensing and humoral immunity. J Leukoc Biol 2012; 92:883-93. [PMID: 22798686 DOI: 10.1189/jlb.0412177] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Alum-based adjuvants facilitate vaccine-driven humoral immunity, but their mechanism of action remains poorly understood. Herein, we report that lack of type II NKT cells is associated with intact, mature B cells but dampened humoral immunity following immunization with Alum-adsorbed T-dependent antigen. Type II NKT cells facilitated production of IL-4, IL-5, IL-10, IL-13, and antibody by LN and splenocyte cultures following Alum/antigen administration in vivo and antigen restimulation in vitro. Addition of IL-4 and IL-5 to type II NKT-deficient cultures restored in vitro antibody production. Intracellular staining revealed that Alum-primed type II NKT cells coordinated IL-4 secretion by T cells. Alum did not significantly affect CD1d expression in vivo, but addition of CD1d-blocking mAb diminished cytokine production and in vitro antibody production. Type II NKT cells therefore function as part of the Alum-sensing apparatus and in a CD1d-dependent manner, facilitate T(H)2-driven humoral immunity. This may have important consequences for understanding the mechanism of action of Alum-containing vaccines.
Collapse
Affiliation(s)
- Hemangi B Shah
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | | | | | |
Collapse
|
18
|
Vivier E, Ugolini S, Blaise D, Chabannon C, Brossay L. Targeting natural killer cells and natural killer T cells in cancer. Nat Rev Immunol 2012; 12:239-52. [PMID: 22437937 DOI: 10.1038/nri3174] [Citation(s) in RCA: 631] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Natural killer (NK) cells and natural killer T (NKT) cells are subsets of lymphocytes that share some phenotypical and functional similarities. Both cell types can rapidly respond to the presence of tumour cells and participate in antitumour immune responses. This has prompted interest in the development of innovative cancer therapies that are based on the manipulation of NK and NKT cells. Recent studies have highlighted how the immune reactivity of NK and NKT cells is shaped by the environment in which they develop. The rational use of these cells in cancer immunotherapies awaits a better understanding of their effector functions, migratory patterns and survival properties in humans.
Collapse
Affiliation(s)
- Eric Vivier
- Centre d'Immunologie de Marseille-Luminy, Université d'Aix-Marseille, Marseille, France.
| | | | | | | | | |
Collapse
|
19
|
Kotas ME, Lee HY, Gillum MP, Annicelli C, Guigni BA, Shulman GI, Medzhitov R. Impact of CD1d deficiency on metabolism. PLoS One 2011; 6:e25478. [PMID: 21980475 PMCID: PMC3183002 DOI: 10.1371/journal.pone.0025478] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 09/05/2011] [Indexed: 01/29/2023] Open
Abstract
Invariant natural killer T cells (iNKTs) are innate-like T cells that are highly concentrated in the liver and recognize lipids presented on the MHC-like molecule CD1d. Although capable of a myriad of responses, few essential functions have been described for iNKTs. Among the many cell types of the immune system implicated in metabolic control and disease, iNKTs seem ideally poised for such a role, yet little has been done to elucidate such a possible function. We hypothesized that lipid presentation by CD1d could report on metabolic status and engage iNKTs to regulate cellular lipid content through their various effector mechanisms. To test this hypothesis, we examined CD1d deficient mice in a variety of metabolically stressed paradigms including high fat feeding, choline-deficient feeding, fasting, and acute inflammation. CD1d deficiency led to a mild exacerbation of steatosis during high fat or choline-deficient feeding, accompanied by impaired hepatic glucose tolerance. Surprisingly, however, this phenotype was not observed in Jα18⁻/⁻ mice, which are deficient in iNKTs but express CD1d. Thus, CD1d appears to modulate some metabolic functions through an iNKT-independent mechanism.
Collapse
Affiliation(s)
- Maya E Kotas
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America.
| | | | | | | | | | | | | |
Collapse
|
20
|
Exley MA, Lynch L, Varghese B, Nowak M, Alatrakchi N, Balk SP. Developing understanding of the roles of CD1d-restricted T cell subsets in cancer: reversing tumor-induced defects. Clin Immunol 2011; 140:184-95. [PMID: 21646050 PMCID: PMC3143311 DOI: 10.1016/j.clim.2011.04.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 04/20/2011] [Accepted: 04/21/2011] [Indexed: 12/22/2022]
Abstract
Invariant natural killer T-cells ('iNKT') are the best-known CD1d-restricted T-cells, with recently-defined roles in controlling adaptive immunity. CD1d-restricted T-cells can rapidly produce large amounts of Th1 and/or Th2//Treg/Th17-type cytokines, thereby regulating immunity. iNKT can stimulate potent anti-tumor immune responses via production of Th1 cytokines, direct cytotoxicity, and activation of effectors. However, Th2//Treg-type iNKT can inhibit anti-tumor activity. Furthermore, iNKT are decreased and/or reversibly functionally impaired in many advanced cancers. In some cases, CD1d-restricted T-cell cancer defects can be traced to CD1d(+) tumor interactions, since hematopoietic, prostate, and some other tumors can express CD1d. Ligand and IL-12 can reverse iNKT defects and therapeutic opportunities exist in correcting such defects alone and in combination. Early stage clinical trials have shown potential for reconstitution of iNKT IFN-gamma responses and evidence of activity in a subset of patients, with rational new approaches to capitalize on this progress ongoing, as will be discussed here.
Collapse
Affiliation(s)
- Mark A Exley
- Department of Medicine, Division of Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Arrenberg P, Maricic I, Kumar V. Sulfatide-mediated activation of type II natural killer T cells prevents hepatic ischemic reperfusion injury in mice. Gastroenterology 2011; 140:646-55. [PMID: 20950612 PMCID: PMC3114423 DOI: 10.1053/j.gastro.2010.10.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Revised: 09/09/2010] [Accepted: 10/01/2010] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Hepatic ischemic reperfusion injury (IRI) is a major complication of liver transplantation and resectional hepatic surgeries. Natural killer T (NKT) cells predominate in liver, where they recognize lipid antigens bound to CD1d molecules. Type I NKT cells use a semi-invariant T-cell receptor and react with α-galactosylceramide; type II NKT cells use diverse T-cell receptors. Some type II NKT cells recognize the self-glycolipid sulfatide. It is not clear whether or how these distinct NKT cell subsets mediate hepatocellular damage after IRI. METHODS We examined the roles of type I and type II NKT cells in mice with partial hepatic, warm ischemia, and reperfusion injury. RESULTS Mice that lack type I NKT cells (Jα18-/-) were protected from hepatic IRI, indicated by reduced hepatocellular necrosis and serum levels of alanine aminotransferase. Sulfatide-mediated activation of type II NKT cells reduced interferon-γ secretion by type I NKT cells and prevented IRI. Protection from hepatic IRI by sulfatide-mediated inactivation of type I NKT cells was associated with significant reductions in hepatic recruitment of myeloid cell subsets, especially the CD11b(+)Gr-1(int), Gr-1(-), and NK cells. CONCLUSIONS In mice, subsets of NKT cells have opposing roles in hepatic IRI: type I NKT cells promote injury whereas sulfatide-reactive type II NKT cells protect against injury. CD1d activation of NKT cells is conserved from mice to human beings, so strategies to modify these processes might be developed to treat patients with hepatic reperfusion injury.
Collapse
Affiliation(s)
- Philomena Arrenberg
- Laboratory of Autoimmunity, Torrey Pines Institute for Molecular Studies, San Diego, California 92121, USA
| | | | | |
Collapse
|
22
|
Abstract
Sepsis is currently a leading cause of death in hospital intensive care units. Previous studies suggest that the pathophysiology of sepsis involves the hyperactivation of complex proinflammatory cascades that include the activation of various immune cells and the exuberant secretion of proinflammatory cytokines by these cells. Natural killer T-cells (NKTs) are a sublineage of T cells that share characteristics of conventional T cells and NK cells and bridge innate and adaptive immunity. More recently, NKT cells have been implicated in microbial immunity, including the onset of sepsis. Moreover, apolipoprotein E (apoE), a component of triglyceride-rich lipoproteins, has been shown to be protective in endotoxemia and gram-negative infections in addition to its well-known role in lipid metabolism. Here, we will review the role of NKT cells in sepsis and septic shock, the immunoregulatory role of apoE in the host immune response to infection, and propose a mechanism for this immunoregulation.
Collapse
|