1
|
Mooney AH, Draper SL, Burn OK, Anderson RJ, Compton BJ, Tang C, Farrand KJ, Di Lucia P, Ravà M, Fumagalli V, Giustini L, Bono E, Godfrey DI, Heath WR, Yuan W, Chisari FV, Guidotti LG, Iannacone M, Sidney J, Sette A, Gulab SA, Painter GF, Hermans IF. Preclinical evaluation of therapeutic vaccines for chronic hepatitis B that stimulate antiviral activities of T cells and NKT cells. JHEP Rep 2024; 6:101038. [PMID: 38694959 PMCID: PMC11061331 DOI: 10.1016/j.jhepr.2024.101038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 05/04/2024] Open
Abstract
Background & Aims Liver diseases resulting from chronic HBV infection are a significant cause of morbidity and mortality. Vaccines that elicit T-cell responses capable of controlling the virus represent a treatment strategy with potential for long-term effects. Here, we evaluated vaccines that induce the activity of type I natural killer T (NKT) cells to limit viral replication and license stimulation of conventional antiviral T-cells. Methods Vaccines were prepared by conjugating peptide epitopes to an NKT-cell agonist to promote co-delivery to antigen-presenting cells, encouraging NKT-cell licensing and stimulation of T cells. Activity of the conjugate vaccines was assessed in transgenic mice expressing the complete HBV genome, administered intravenously to maximise access to NKT cell-rich tissues. Results The vaccines induced only limited antiviral activity in unmanipulated transgenic hosts, likely attributable to NKT-cell activation as T-cell tolerance to viral antigens is strong. However, in a model of chronic hepatitis B involving transfer of naive HBcAg-specific CD8+ T cells into the transgenic mice, which typically results in specific T-cell dysfunction without virus control, vaccines containing the targeted HBcAg epitope induced prolonged antiviral activity because of qualitatively improved T-cell stimulation. In a step towards a clinical product, vaccines were prepared using synthetic long peptides covering clusters of known HLA-binding epitopes and shown to be immunogenic in HLA transgenic mice. Predictions based on HLA distribution suggest a product containing three selected SLP-based vaccines could give >90 % worldwide coverage, with an average of 3.38 epitopes targeted per individual. Conclusions The novel vaccines described show promise for further clinical development as a treatment for chronic hepatitis B. Impact and Implications Although there are effective prophylactic vaccines for HBV infection, it is estimated that 350-400 million people worldwide have chronic hepatitis B, putting these individuals at significant risk of life-threatening liver diseases. Therapeutic vaccination aimed at activating or boosting HBV-specific T-cell responses holds potential as a strategy for treating chronic infection, but has so far met with limited success. Here, we show that a glycolipid-peptide conjugate vaccine designed to coordinate activity of type I NKT cells alongside conventional antiviral T cells has antiviral activity in a mouse model of chronic infection. It is anticipated that a product based on a combination of three such conjugates, each prepared using long peptides covering clusters of known HLA-binding epitopes, could be developed further as a treatment for chronic hepatitis B with broad global HLA coverage.
Collapse
Affiliation(s)
- Anna H. Mooney
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Sarah L. Draper
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Olivia K. Burn
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Regan J. Anderson
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Benjamin J. Compton
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Chingwen Tang
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Pietro Di Lucia
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Micol Ravà
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Valeria Fumagalli
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Leonardo Giustini
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Bono
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Australia
| | - William R. Heath
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia
| | - Weiming Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Francis V. Chisari
- Department of Immunology & Microbial Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Luca G. Guidotti
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Shivali A. Gulab
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
- Avalia Immunotherapies Limited, Wellington, New Zealand
| | - Gavin F. Painter
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Ian F. Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
2
|
Zhang H, Tang Q, Miao Y, Wang J, Yuan Z, Huang X, Zhu Y, Nong C, Li G, Cui R, Huang X, Zhang L, Yu Q, Jiang Z. Group 1 innate lymphoid cell activation via recognition of NKG2D and liver resident macrophage MULT-1: Collaborated roles in triptolide induced hepatic immunotoxicity in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116072. [PMID: 38342011 DOI: 10.1016/j.ecoenv.2024.116072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/22/2024] [Accepted: 02/01/2024] [Indexed: 02/13/2024]
Abstract
Triptolide (TP) is the major bioactive component of traditional Chinese medicine Tripterygium wilfordii Hook. F., a traditional Chinese medicinal plant categorized within the Tripterygium genus of the Celastraceae family. It is recognized for its therapeutic potential in addressing a multitude of diseases. Nonetheless, TP is known to exhibit multi-organ toxicity, notably hepatotoxicity, which poses a significant concern for the well-being of patients undergoing treatment. The precise mechanisms responsible for TP-induced hepatotoxicity remain unresolved. In our previous investigation, it was determined that TP induces heightened hepatic responsiveness to exogenous lipopolysaccharide (LPS). Additionally, natural killer (NK) cells were identified as a crucial effector responsible for mediating hepatocellular damage in this context. However, associated activating receptors and the underlying mechanisms governing NK cell represented innate lymphoid cell (ILC) activation remained subjects of inquiry and were not yet investigated. Herein, activating receptor Killer cell lectin like receptor K1 (NKG2D) of group 1 ILCs was specifically upregulated in TP- and LPS-induced acute liver failure (ALF), and in vivo blockade of NKG2D significantly reduced group 1 ILC mediated cytotoxicity and mitigated TP- and LPS-induced ALF. NKG2D ligand UL16-binding protein-like transcript 1 (MULT-1) was found upregulated in liver resident macrophages (LRMs) after TP administration, and LRMs did exhibit NK cell activating effect. Furthermore, M1 polarization of LRMs cells was observed, along with an elevation in intracellular tumor necrosis factor (TNF)-α levels. In vivo neutralization of TNF-α significantly alleviated TP- and LPS-induced ALF. In conclusion, the collaborative role of group 1 ILCs and LRMs in mediating hepatotoxicity was confirmed in TP- and LPS-induced ALF. TP-induced MULT-1 expression in LRMs was the crucial mechanism in the activation of group 1 ILCs via MULT-1-NKG2D signal upon LPS stimulation, emphasizing the importance of infection control after TP administration.
Collapse
Affiliation(s)
- Haoran Zhang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Qianhui Tang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yingying Miao
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jie Wang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Zihang Yuan
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Xinliang Huang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Ying Zhu
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Cheng Nong
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Guoqing Li
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ruyu Cui
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Xin Huang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Luyong Zhang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Qinwei Yu
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Zhenzhou Jiang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Animal Experimental Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
3
|
Hebbandi Nanjundappa R, Shao K, Krishnamurthy P, Gershwin ME, Leung PSC, Sokke Umeshappa C. Invariant natural killer T cells in autoimmune cholangiopathies: Mechanistic insights and therapeutic implications. Autoimmun Rev 2024; 23:103485. [PMID: 38040101 DOI: 10.1016/j.autrev.2023.103485] [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: 11/07/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023]
Abstract
Invariant natural killer T cells (iNKT cells) constitute a specialized subset of lymphocytes that bridges innate and adaptive immunity through a combination of traits characteristic of both conventional T cells and innate immune cells. iNKT cells are characterized by their invariant T cell receptors and discerning recognition of lipid antigens, which are presented by the non-classical MHC molecule, CD1d. Within the hepatic milieu, iNKT cells hold heightened prominence, contributing significantly to the orchestration of organ homeostasis. Their unique positioning to interact with diverse cellular entities, ranging from epithelial constituents like hepatocytes and cholangiocytes to immunocytes including Kupffer cells, B cells, T cells, and dendritic cells, imparts them with potent immunoregulatory abilities. Emergering knowledge of liver iNKT cells subsets enable to explore their therapeutic potential in autoimmne liver diseases. This comprehensive review navigates the landscape of iNKT cell investigations in immune-mediated cholangiopathies, with a particular focus on primary biliary cholangitis and primary sclerosing cholangitis, across murine models and human subjects to unravel the intricate involvements of iNKT cells in liver autoimmunity. Additionally, we also highlight the prospectives of iNKT cells as therapeutic targets in cholangiopathies. Modulation of the equilibrium between regulatory and proinflammatory iNKT subsets can be defining determinant in the dynamics of hepatic autoimmunity. This discernment not only enriches our foundational comprehension but also lays the groundwork for pioneering strategies to navigate the multifaceted landscape of liver autoimmunity.
Collapse
Affiliation(s)
| | - Kun Shao
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116023, China
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States.
| | - Patrick S C Leung
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Channakeshava Sokke Umeshappa
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada; Department of Pediatrics, IWK Research Center, Halifax, NS, Canada.
| |
Collapse
|
4
|
Heuser-Loy C, Baumgart AK, Hackstein CP, Courrèges CJF, Philipp MS, Thaiss CA, Holland T, Evaristo C, Garbi N, Kurts C. Conditional NKT Cell Depletion in Mice Reveals a Negative Feedback Loop That Regulates CTL Cross-Priming. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:35-42. [PMID: 38019126 DOI: 10.4049/jimmunol.2300662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/29/2023] [Indexed: 11/30/2023]
Abstract
NKT cells are unconventional T cells whose biological role is incompletely understood. Similar to TH cells, activated NKT cells can cause dendritic cell (DC) maturation, which is required for effective CTL responses. However, it is unclear whether and how NKT cells affect CTLs downstream of the DC maturation phase. This is partially due to the lack of techniques to conditionally deplete NKT cells in vivo. To overcome this problem, we have developed two approaches for this purpose in mice: the first is based on mixed bone marrow chimeras where Jα18 knockout and depletable CD90 congenic bone marrow is combined, and the second used PLZFCre × iDTR bone marrow chimeras, which target innate-like T cells. Using these tools, we found that NKT cell depletion at 20 h, that is, after initial DC activation, did not render CTLs helpless, as CD40L signaling by non-NKT cells sufficed. Instead, NKT cell depletion even augmented CD8 T cell expansion and cytotoxicity by mechanisms distinct from reduced STAT6 signaling. These findings revealed a negative feedback loop by which NKT cells control CTL cross-priming downstream of DC maturation. The techniques described in this study expand the toolbox to study NKT cells and other unconventional T cell subsets in vivo and uncovered a hidden immunoregulatory mechanism.
Collapse
Affiliation(s)
- Christoph Heuser-Loy
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Ann-Kathrin Baumgart
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Carl-Philipp Hackstein
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Christina J F Courrèges
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Marie-Sophie Philipp
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Christoph A Thaiss
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Tristan Holland
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - César Evaristo
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Natalio Garbi
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
| | - Christian Kurts
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rhenish Friedrich Wilhelm University, Bonn, Germany
- The Peter Doherty Institute of Infection and Immunology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
5
|
Heffernan DS, Chung CS, Ayala A. Severity of critical illness correlates with CD3-low expression on iNKT-cells among septic surgical patients. Inflamm Res 2024; 73:1-4. [PMID: 38147124 PMCID: PMC11963472 DOI: 10.1007/s00011-023-01795-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/01/2023] [Accepted: 09/09/2023] [Indexed: 12/27/2023] Open
Abstract
BACKGROUND iNKT-cells are innate regulatory lymphocytes capable of directing immune and inflammatory responses to sepsis. Repeat stimulation of iNKT-cells leads to the induction of anergy with the emergence of a hyporesponsive CD3low iNKT-cell subpopulation. METHODS iNKT-cells were isolated from critical ill surgical patients with sepsis and phenotyped for CD3 expression. This was correlated with degree of severity of illness, as denoted by APACHE-II score. RESULTS Comparing healthy volunteers to critically ill septic patients, it was noted that increasing severity of sepsis was associated with increasing frequency of circulating CD3low-iNKT-cell populations. CONCLUSION The emergence of CD3low -iNKT-cells may serve as a clinically translatable marker of degree of sepsis-induced immune dysfunction.
Collapse
Affiliation(s)
- Daithi S Heffernan
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Brown University School of Medicine, 593 Eddy Street, Providence, Rhode Island, 02903, USA.
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Brown University School of Medicine, Room 205 Middle House, Providence, Rhode Island, 02903, USA.
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Brown University School of Medicine, 593 Eddy Street, Providence, Rhode Island, 02903, USA
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Brown University School of Medicine, 593 Eddy Street, Providence, Rhode Island, 02903, USA
| |
Collapse
|
6
|
Chakrabarti R, Duddu S, Tiwari A, Naidu KT, Sharma P, Chakravorty N, Shukla PC. Natural Killer T cells and the invariant subset promote atherosclerosis: A meta-analysis. Life Sci 2023; 321:121620. [PMID: 37011534 DOI: 10.1016/j.lfs.2023.121620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 04/04/2023]
Abstract
AIMS Natural Killer T (NKT) cells are reported to be both pro- and anti-atherosclerotic. With this meta-analysis, we evaluated the NKT population and their subsets in regulating the atherosclerotic disease in mice. MAIN METHODS Eighteen pre-clinical (mice, n = 1276) and 6 clinical observational studies (humans, n = 116) met the eligibility criteria for inclusion. Random effects model was used and standard mean difference (SMD) was calculated for the cell counts and aortic lesion area. KEY FINDINGS Lesion area decreased in the absence of whole NKT cell population (-1.33[95%CI, -2.14, -0.52]), and in the absence of only iNKT subset (-0.66[95%CI, -1.69, 0.37]). However, lesion area increased after over-expression/activation of iNKTs (1.40[95%CI, 0.28, 2.52]). Atherogenic diet (AD) or high fat diet (HFD) increased the number of NKT cells (2.51[95%CI, 1.42, 3.61]), whereas the iNKT cell numbers and iNKT cell-specific gene expression decreased in mice (-2.04[95%CI, -3.34, -0.75]) and atherosclerotic patients (-1.81[95 % CI, -2.89, -0.74]). SIGNIFICANCE Here we show that, NKT and iNKT cells promote atherosclerosis. In general, NKT cell population increases with the progression of the plaque in mice and the numbers of iNKT cells reduce once the disease is established both in mice and humans.
Collapse
|
7
|
Zhang H, Yuan Z, Wang J, Tang Q, Miao Y, Yuan Z, Huang X, Zhu Y, Nong C, Zhang L, Jiang Z, Yu Q. Triptolide leads to hepatic intolerance to exogenous lipopolysaccharide and natural-killer-cell mediated hepatocellular damage by inhibiting MHC class I molecules. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154621. [PMID: 36610139 DOI: 10.1016/j.phymed.2022.154621] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/09/2022] [Accepted: 12/19/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Tripterygium wilfordii Hook. F (TWHF) is used as a traditional Chinese medicine, called thunder god vine, based on its efficacy for treating inflammatory diseases. However, its hepatotoxicity has limited its clinical application. Triptolide (TP) is the major active and toxic component of TWHF. Previous studies reported that a toxic pretreatment dose of TP leads to hepatic intolerance to exogenous lipopolysaccharide (LPS) stimulation, and to acute liver failure, in mice, but the immune mechanisms of TP-sensitised hepatocytes and the TP-induced excessive immune response to LPS stimulation are unknown. PURPOSE To identify both the key immune cell population and mechanism involved in TP-induced hepatic intolerance of exogenous LPS. STUDY DESIGN In vitro and in vivo experiments were conducted to investigate the inhibitory signal of natural killer (NK) cells maintained in hepatocytes, and the ability of TP to impair that signal. METHODS Flow cytometry was performed to determine NK cell activity and hepatocyte histocompatibility complex (MHC) class I molecules expression; the severity of liver injury was determined based on blood chemistry values, and drug- or cell-mediated hepatocellular damage, by measuring lactate dehydrogenase (LDH) release. In vivo H-2Kb transduction was carried out using an adeno-associated viral vector. RESULTS Interferon (IFN)-γ-mediated necroptosis occurred in C57BL/6N mice treated with 500 μg TP/kg and 0.1 mg LPS/kg to induce fulminant hepatitis. Primary hepatocytes pretreated with TP were more prone to necroptosis when exposed to recombinant murine IFN-γ. In mice administered TP and LPS, the intracellular IFN-γ levels of NK cells increased significantly. Subsequent study confirmed that NK cells were activated and resulted in potent hepatocellular toxicity. In vivo and in vitro TP administration significantly inhibited MHC class I molecules in murine hepatocytes. An in vitro analysis demonstrated the susceptibility of TP-pretreated hepatocytes to NK-cell-mediated cytotoxicity, an effect that was significantly attenuated by the induction of hepatocyte MHC-I molecules by IFN-α. In vivo induction or overexpression of hepatocyte MHC-I also protected mouse liver against TP and LPS-induced injury. CONCLUSION The TP-induced inhibition of hepatocyte MHC-I molecules expression leads to hepatic intolerance to exogenous LPS and NK-cell mediated cytotoxicity against self-hepatocytes. These findings shed light on the toxicity of traditional Chinese medicines administered for their immunomodulatory effects.
Collapse
Affiliation(s)
- Haoran Zhang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Zihang Yuan
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Jie Wang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Qianhui Tang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Yingying Miao
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Ziqiao Yuan
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Xinliang Huang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Ying Zhu
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Cheng Nong
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Luyong Zhang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Zhenzhou Jiang
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China.
| | - Qinwei Yu
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
8
|
Kane H, LaMarche NM, Ní Scannail Á, Garza AE, Koay HF, Azad AI, Kunkemoeller B, Stevens B, Brenner MB, Lynch L. Longitudinal analysis of invariant natural killer T cell activation reveals a cMAF-associated transcriptional state of NKT10 cells. eLife 2022; 11:e76586. [PMID: 36458691 PMCID: PMC9831610 DOI: 10.7554/elife.76586] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 11/30/2022] [Indexed: 12/05/2022] Open
Abstract
Innate T cells, including CD1d-restricted invariant natural killer T (iNKT) cells, are characterized by their rapid activation in response to non-peptide antigens, such as lipids. While the transcriptional profiles of naive, effector, and memory adaptive T cells have been well studied, less is known about the transcriptional regulation of different iNKT cell activation states. Here, using single-cell RNA-sequencing, we performed longitudinal profiling of activated murine iNKT cells, generating a transcriptomic atlas of iNKT cell activation states. We found that transcriptional signatures of activation are highly conserved among heterogeneous iNKT cell populations, including NKT1, NKT2, and NKT17 subsets, and human iNKT cells. Strikingly, we found that regulatory iNKT cells, such as adipose iNKT cells, undergo blunted activation and display constitutive enrichment of memory-like cMAF+ and KLRG1+ populations. Moreover, we identify a conserved cMAF-associated transcriptional network among NKT10 cells, providing novel insights into the biology of regulatory and antigen-experienced iNKT cells.
Collapse
Affiliation(s)
- Harry Kane
- Trinity Biomedical Science Institute, Trinity College DublinDublinIreland
| | - Nelson M LaMarche
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
| | - Áine Ní Scannail
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
| | - Amanda E Garza
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
| | - Hui-Fern Koay
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of MelbourneMelbourneAustralia
| | - Adiba I Azad
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
| | - Britta Kunkemoeller
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
| | - Brenneth Stevens
- Trinity Biomedical Science Institute, Trinity College DublinDublinIreland
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
| | - Michael B Brenner
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
| | - Lydia Lynch
- Trinity Biomedical Science Institute, Trinity College DublinDublinIreland
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
9
|
Lin Q, Kuypers M, Liu Z, Copeland JK, Chan D, Robertson SJ, Kontogiannis J, Guttman DS, Banks EK, Philpott DJ, Mallevaey T. Invariant natural killer T cells minimally influence gut microbiota composition in mice. Gut Microbes 2022; 14:2104087. [PMID: 35912530 PMCID: PMC9348128 DOI: 10.1080/19490976.2022.2104087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Invariant Natural Killer T (iNKT) cells are unconventional T cells that respond to glycolipid antigens found in microbes in a CD1d-dependent manner. iNKT cells exert innate-like functions and produce copious amounts of cytokines, chemokines and cytotoxic molecules within only minutes of activation. As such, iNKT cells can fuel or dampen inflammation in a context-dependent manner. In addition, iNKT cells provide potent immunity against bacteria, viruses, parasites and fungi. Although microbiota-iNKT cell interactions are not well-characterized, mounting evidence suggests that microbiota colonization early in life impacts iNKT cell homeostasis and functions in disease. In this study, we showed that CD1d-/- and Vα14 Tg mice, which lack and have increased numbers of iNKT cells, respectively, had no significant alterations in gut microbiota composition compared to their littermate controls. Furthermore, specific iNKT cell activation by glycolipid antigens only resulted in a transient and minimal shift in microbiota composition when compared to the natural drift found in our colony. Our findings demonstrate that iNKT cells have little to no influence in regulating commensal bacteria at steady state.Abbreviations: iNKT: invariant Natural Killer T cell; αGC: α-galactosylceramide.
Collapse
Affiliation(s)
- Qiaochu Lin
- Department of Immunology, University of Toronto, Toronto, ON, Canada,CONTACT Thierry Mallevaey University of Toronto, Department of Immunology, Medical Sciences Building, Room 7334,1 King’s College Circle, Toronto, OntarioM5S 1A8, Canada
| | - Meggie Kuypers
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Zhewei Liu
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Julia K Copeland
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Donny Chan
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada
| | - Susan J Robertson
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Jean Kontogiannis
- Division of Comparative Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - David S Guttman
- Centre for the Analysis of Genome Evolution & Function, University of Toronto, Toronto, Ontario, Canada,Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - E. Kate Banks
- Division of Comparative Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Thierry Mallevaey
- Department of Immunology, University of Toronto, Toronto, ON, Canada,Institute of Biomaterials & Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
10
|
Farrand K, Holz LE, Ferrer-Font L, Wilson MD, Ganley M, Minnell JJ, Tang CW, Painter GF, Heath WR, Hermans IF, Burn OK. Using Full-Spectrum Flow Cytometry to Phenotype Memory T and NKT Cell Subsets with Optimized Tissue-Specific Preparation Protocols. Curr Protoc 2022; 2:e482. [PMID: 35819836 DOI: 10.1002/cpz1.482] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Full-spectrum flow cytometry is now routinely used in many laboratories internationally, and the demand for this technology is rapidly increasing. With capacity to use larger and more complex staining panels, standardized protocols are required for optimal panel design and analysis. Importantly, for ex vivo analysis, tissue preparation methods also need to be optimized to ensure samples are truly representative of tissues in situ. This is particularly relevant given the recent interest in adaptive immune cells that form residency in specific organs. Here we provide optimized protocols for tissue processing and phenotyping of memory T cells and natural killer T (NKT) cell subsets from liver, lung, spleen, and lymph node using full-spectrum flow cytometry. We provide a 21-color antibody panel for identification of different memory subsets, including tissue-resident memory T (TRM ) cells, which are increasingly regarded as important effectors in adaptive immunity. We show that processing procedures can affect outcomes, with liver TRM cells particularly sensitive to heat, such that accurate evaluation requires fast processing at defined temperatures. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Processing mouse liver for flow cytometric analysis of memory T and NKT cell subsets Basic Protocol 2: Processing mouse spleen for flow cytometric analysis of memory T and NKT cell subsets Basic Protocol 3: Processing mouse lungs for flow cytometric analysis of memory T and NKT cell subsets Basic Protocol 4: Processing mouse lymph nodes for flow cytometric analysis of memory T and NKT cell subsets Basic Protocol 5: Staining and flow cytometric analysis of samples for memory T and NKT cell subsets Support Protocol: Obtaining cell counts from flow cytometry data.
Collapse
Affiliation(s)
- Kathryn Farrand
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Lauren E Holz
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Victoria, Australia
| | - Laura Ferrer-Font
- Malaghan Institute of Medical Research, Wellington, New Zealand
- Maurice Wilkins Centre, Auckland, New Zealand
| | | | - Mitch Ganley
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | | | - Ching-Wen Tang
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Gavin F Painter
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - William R Heath
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Victoria, Australia
| | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand
- Maurice Wilkins Centre, Auckland, New Zealand
| | - Olivia K Burn
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
11
|
Single-cell transcriptomics reveal a unique memory-like NK cell subset that accumulates with ageing and correlates with disease severity in COVID-19. Genome Med 2022; 14:46. [PMID: 35501841 PMCID: PMC9060844 DOI: 10.1186/s13073-022-01049-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 04/15/2022] [Indexed: 12/22/2022] Open
Abstract
Background Natural killer (NK) cells are innate lymphoid cells that mediate antitumour and antiviral responses. However, very little is known about how ageing influences human NK cells, especially at the single-cell level. Methods We applied single-cell sequencing (scRNA-seq) to human lymphocytes and NK cells from 4 young and 4 elderly individuals and then analysed the transcriptome data using Seurat. We detected the proportion and phenotype of NK cell subsets in peripheral blood samples from a total of 62 young and 52 elderly healthy donors by flow cytometry. We also used flow cytometry to examine the effector functions of NK cell subsets upon IFN-α/IL-12+IL-15/K562/IL-2 stimulation in vitro in peripheral blood samples from a total of 64 young and 63 elderly healthy donors. We finally studied and integrated single-cell transcriptomes of NK cells from 15 young and 41 elderly COVID-19 patients with those from 12 young and 6 elderly healthy control individuals to investigate the impacts of ageing on NK cell subsets in COVID-19 disease. Results We discovered a memory-like NK subpopulation (NK2) exhibiting the largest distribution change between elderly and young individuals among lymphocytes. Notably, we discovered a unique NK subset that was predominantly CD52+ NK2 cells (NK2.1). These memory-like NK2.1 cells accumulated with age, exhibited proinflammatory characteristics, and displayed a type I interferon response state. Integrative analyses of a large-cohort COVID-19 dataset and our datasets revealed that NK2.1 cells from elderly COVID-19 patients are enriched for type I interferon signalling, which is positively correlated with disease severity in COVID-19. Conclusions We identified a unique memory-like NK cell subset that accumulates with ageing and correlates with disease severity in COVID-19. Our results identify memory-like NK2.1 cells as a potential target for developing immunotherapies for infectious diseases and for addressing age-related dysfunctions of the immune system. Supplementary Information The online version contains supplementary material available at 10.1186/s13073-022-01049-3.
Collapse
|
12
|
Zhao L, Yang X. Cross Talk Between Natural Killer T and Dendritic Cells and Its Impact on T Cell Responses in Infections. Front Immunol 2022; 13:837767. [PMID: 35185930 PMCID: PMC8850912 DOI: 10.3389/fimmu.2022.837767] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/19/2022] [Indexed: 11/16/2022] Open
Abstract
Both innate and adaptive immunity is vital for host defense against infections. Dendritic cells (DCs) are critical for initiating and modulating adaptive immunity, especially for T-cell responses. Natural killer T (NKT) cells are a small population of innate-like T cells distributed in multiple organs. Many studies have suggested that the cross-talk between these two immune cells is critical for immunobiology and host defense mechanisms. Not only can DCs influence the activation/function of NKT cells, but NKT cells can feedback on DCs also, thus modulating the phenotype and function of DCs and DC subsets. This functional feedback of NKT cells on DCs, especially the preferential promoting effect on CD8α+ and CD103+ DC subsets in lymphoid and non-lymphoid tissues, significantly impacts the systemic and local adaptive CD4 and CD8 T cell responses in infections. This review focuses on the two-way interaction between NKT cells and DCs, emphasizing the importance of NKT cell feedback on DCs in bridging innate and adaptive immune responses for host defense purposes.
Collapse
Affiliation(s)
- Lei Zhao
- Departments of Immunology and Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Xi Yang
- Departments of Immunology and Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
13
|
Johnson DN, Ruan Z, Petley EV, Devi S, Holz LE, Uldrich AP, Mak JYW, Hor JL, Mueller SN, McCluskey J, Fairlie DP, Darcy PK, Beavis PA, Heath WR, Godfrey DI. Differential location of NKT and MAIT cells within lymphoid tissue. Sci Rep 2022; 12:4034. [PMID: 35260653 PMCID: PMC8904549 DOI: 10.1038/s41598-022-07704-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 01/17/2022] [Indexed: 11/17/2022] Open
Abstract
Natural Killer T (NKT) cells and Mucosal-Associated Invariant T (MAIT) cells are innate-like T cells that express semi-invariant αβ T cell receptors (TCRs) through which they recognise CD1d and MR1 molecules, respectively, in complex with specific ligands. These cells play important roles in health and disease in many organs, but their precise intra-organ location is not well established. Here, using CD1d and MR1 tetramer staining techniques, we describe the precise location of NKT and MAIT cells in lymphoid and peripheral organs. Within the thymus, NKT cells were concentrated in the medullary side of the corticomedullary junction. In spleen and lymph nodes, NKT cells were mainly localised within T cell zones, although following in vivo activation with the potent NKT-cell ligand α-GalCer, they expanded throughout the spleen. MAIT cells were clearly detectable in Vα19 TCR transgenic mice and were rare but detectable in lymphoid tissue of non-transgenic mice. In contrast to NKT cells, MAIT cells were more closely associated with the B cell zone and red pulp of the spleen. Accordingly, we have provided an extensive analysis of the in situ localisation of NKT and MAIT cells and suggest differences between the intra-organ location of these two cell types.
Collapse
Affiliation(s)
- Darryl N Johnson
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Zheng Ruan
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Emma V Petley
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Sapna Devi
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Lauren E Holz
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Adam P Uldrich
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jeffrey Y W Mak
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Jyh Liang Hor
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - James McCluskey
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - David P Fairlie
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - William R Heath
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia.,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3010, Australia. .,Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
14
|
Savid-Frontera C, Viano ME, Baez NS, Reynolds D, Matellon M, Young HA, Rodriguez-Galan MC. Safety levels of systemic IL-12 induced by cDNA expression as a cancer therapeutic. Immunotherapy 2022; 14:115-133. [PMID: 34783257 PMCID: PMC8739399 DOI: 10.2217/imt-2021-0080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 10/14/2021] [Indexed: 02/03/2023] Open
Abstract
Aim: The aim of this work is to utilize a gene expression procedure to safely express systemic IL-12 and evaluate its effects in mouse tumor models. Materials & methods: Secondary lymphoid organs and tumors from EL4 and B16 tumor-bearing mice were analyzed by supervised and unsupervised methods. Results: IL-12 cDNA induced systemic IL-12 protein levels lower than the tolerated dose in patients. Control of tumor growth was observed in subcutaneous B16 and EL4 tumors. Systemic IL-12 expression induced a higher frequency of both total tumor-infiltrated CD45+ cells and proliferative IFN-γ+CD8+ T cells along with a lower frequency of CD4+FOXP3+ and CD11b+Gr-1+ cells. Conclusion: This approach characterizes the systemic effects of IL-12, helping to improve treatment of metastases or solid tumors.
Collapse
Affiliation(s)
- Constanza Savid-Frontera
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, 5000, Argentina
| | - Maria E Viano
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, 5000, Argentina
| | - Natalia S Baez
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, 5000, Argentina
| | - Della Reynolds
- Cancer & Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201 USA
| | - Mariana Matellon
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, 5000, Argentina
| | - Howard A Young
- Cancer & Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702-1201 USA
| | - Maria C Rodriguez-Galan
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, 5000, Argentina
| |
Collapse
|
15
|
Goto T, Ito Y, Satoh M, Nakamoto S, Nishizawa N, Hosono K, Naitoh T, Eshima K, Iwabuchi K, Hiki N, Amano H. Activation of iNKT Cells Facilitates Liver Repair After Hepatic Ischemia Reperfusion Injury Through Acceleration of Macrophage Polarization. Front Immunol 2021; 12:754106. [PMID: 34691073 PMCID: PMC8526965 DOI: 10.3389/fimmu.2021.754106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/20/2021] [Indexed: 12/15/2022] Open
Abstract
Macrophage polarization is critical for liver tissue repair following acute liver injury. However, the underlying mechanisms of macrophage phenotype switching are not well defined. Invariant natural killer T (iNKT) cells orchestrate tissue inflammation and tissue repair by regulating cytokine production. Herein, we examined whether iNKT cells played an important role in liver repair after hepatic ischemia-reperfusion (I/R) injury by affecting macrophage polarization. To this end, we subjected male C57BL/6 mice to hepatic I/R injury, and mice received an intraperitoneal (ip) injection of α-galactosylceramide (α-GalCer) or vehicle. Compared with that of the vehicle, α-GalCer administration resulted in the promotion of liver repair accompanied by acceleration of macrophage differentiation and by increases in the numbers of Ly6Chigh pro-inflammatory macrophages and Ly6Clow reparative macrophages. iNKT cells activated with α-GalCer produced interleukin (IL)-4 and interferon (IFN)-γ. Treatment with anti-IL-4 antibodies delayed liver repair, which was associated with an increased number of Ly6Chigh macrophages and a decreased number of Ly6Clow macrophages. Treatment with anti-IFN-γ antibodies promoted liver repair, associated with reduced the number of Ly6Chigh macrophages, but did not change the number of Ly6Clow macrophages. Bone marrow-derived macrophages up-regulated the expression of genes related to both a pro-inflammatory and a reparative phenotype when co-cultured with activated iNKT cells. Anti-IL-4 antibodies increased the levels of pro-inflammatory macrophage-related genes and decreased those of reparative macrophage-related genes in cultured macrophages, while anti-IFN-γ antibodies reversed the polarization of macrophages. Cd1d-deficient mice showed delayed liver repair and suppressed macrophage switching, compared with that in wild-type mice. These results suggest that the activation of iNKT cells by α-GalCer facilitated liver repair after hepatic I/R injury by both IL-4-and IFN-γ-mediated acceleration of macrophage polarization. Therefore, the activation of iNKT cells may represent a therapeutic tool for liver repair after hepatic I/R injury.
Collapse
Affiliation(s)
- Takuya Goto
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan.,Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Japan.,Department of Lower Gastrointestinal Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yoshiya Ito
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan.,Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Masashi Satoh
- Department of Immunology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Shuji Nakamoto
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan.,Department of General Pediatric Hepatobiliary Pancreatic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Nobuyuki Nishizawa
- Department of General Pediatric Hepatobiliary Pancreatic Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kanako Hosono
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan.,Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Takeshi Naitoh
- Department of Lower Gastrointestinal Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Koji Eshima
- Department of Immunology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kazuya Iwabuchi
- Department of Immunology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Naoki Hiki
- Department of Upper Gastrointestinal Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Hideki Amano
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan.,Department of Pharmacology, Kitasato University School of Medicine, Sagamihara, Japan
| |
Collapse
|
16
|
Slepicka PF, Yazdanifar M, Bertaina A. Harnessing Mechanisms of Immune Tolerance to Improve Outcomes in Solid Organ Transplantation: A Review. Front Immunol 2021; 12:688460. [PMID: 34177941 PMCID: PMC8222735 DOI: 10.3389/fimmu.2021.688460] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/24/2021] [Indexed: 12/15/2022] Open
Abstract
Survival after solid organ transplantation (SOT) is limited by chronic rejection as well as the need for lifelong immunosuppression and its associated toxicities. Several preclinical and clinical studies have tested methods designed to induce transplantation tolerance without lifelong immune suppression. The limited success of these strategies has led to the development of clinical protocols that combine SOT with other approaches, such as allogeneic hematopoietic stem cell transplantation (HSCT). HSCT prior to SOT facilitates engraftment of donor cells that can drive immune tolerance. Recent innovations in graft manipulation strategies and post-HSCT immune therapy provide further advances in promoting tolerance and improving clinical outcomes. In this review, we discuss conventional and unconventional immunological mechanisms underlying the development of immune tolerance in SOT recipients and how they can inform clinical advances. Specifically, we review the most recent mechanistic studies elucidating which immune regulatory cells dampen cytotoxic immune reactivity while fostering a tolerogenic environment. We further discuss how this understanding of regulatory cells can shape graft engineering and other therapeutic strategies to improve long-term outcomes for patients receiving HSCT and SOT.
Collapse
Affiliation(s)
- Priscila Ferreira Slepicka
- Division of Hematology, Oncology and Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Mahboubeh Yazdanifar
- Division of Hematology, Oncology and Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Alice Bertaina
- Division of Hematology, Oncology and Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
17
|
Kharkwal SS, Johndrow CT, Veerapen N, Kharkwal H, Saavedra-Avila NA, Carreño LJ, Rothberg S, Zhang J, Garforth SJ, Jervis PJ, Zhang L, Donda A, Besra AK, Cox LR, Almo SC, Howell A, Evans EE, Zauderer M, Besra GS, Porcelli SA. Serial Stimulation of Invariant Natural Killer T Cells with Covalently Stabilized Bispecific T-cell Engagers Generates Antitumor Immunity While Avoiding Anergy. Cancer Res 2021; 81:1788-1801. [PMID: 33483371 PMCID: PMC8137529 DOI: 10.1158/0008-5472.can-20-2219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/15/2020] [Accepted: 01/15/2021] [Indexed: 11/16/2022]
Abstract
CD1d-restricted invariant natural killer T cells (iNKT cells) mediate strong antitumor immunity when stimulated by glycolipid agonists. However, attempts to develop effective iNKT cell agonists for clinical applications have been thwarted by potential problems with dose-limiting toxicity and by activation-induced iNKT cell anergy, which limits the efficacy of repeated administration. To overcome these issues, we developed a unique bispecific T-cell engager (BiTE) based on covalent conjugates of soluble CD1d with photoreactive analogues of the glycolipid α-galactosylceramide. Here we characterize the in vivo activities of iNKT cell-specific BiTEs and assess their efficacy for cancer immunotherapy in mouse models using transplantable colorectal cancer or melanoma tumor lines engineered to express human Her2 as a tumor-associated antigen. Systemic administration of conjugated BiTEs stimulated multiple iNKT cell effector functions including cytokine release, secondary activation of NK cells, and induction of dendritic cell maturation and also initiated epitope spreading for tumor-specific CD8+ cytolytic T-cell responses. The antitumor effects of iNKT-cell activation with conjugated BiTEs were further enhanced by simultaneous checkpoint blockade with antibodies to CTLA-4, providing a potential approach for combination immunotherapy. Multiple injections of covalently stabilized iNKT cell-specific BiTEs activated iNKT cells without causing iNKT cell anergy or exhaustion, thus enabling repeated administration for effective and nontoxic cancer immunotherapy regimens. SIGNIFICANCE: Covalently stabilized conjugates that engage the antigen receptors of iNKT cells and target a tumor antigen activate potent antitumor immunity without induction of anergy or depletion of the responding iNKT cells.
Collapse
Affiliation(s)
- Shalu Sharma Kharkwal
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
- Elstar Therapeutics, Cambridge, Massachusetts
| | - Christopher T Johndrow
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Natacha Veerapen
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Himanshu Kharkwal
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
- Department of Clinical Oncology, Montefiore Medical Centre, Bronx, New York
| | - Noemi A Saavedra-Avila
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Leandro J Carreño
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
- Millennium Institute on Immunology and Immunotherapy, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Samantha Rothberg
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Jinghang Zhang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Scott J Garforth
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Peter J Jervis
- Centre of Chemistry, University of Minho, Braga, Portugal
| | - Lianjun Zhang
- Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, China
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Alena Donda
- Department of Oncology and Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Amareeta K Besra
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Liam R Cox
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | | | | | | | | | - Gurdyal S Besra
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Steven A Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
18
|
Mitochondrial metabolism is essential for invariant natural killer T cell development and function. Proc Natl Acad Sci U S A 2021; 118:2021385118. [PMID: 33753493 PMCID: PMC8020658 DOI: 10.1073/pnas.2021385118] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We show CD1d-restricted natural killer (NK)T cells have distinct metabolic profiles compared with CD4+ conventional T cells. Mature NKT cells have poor fatty acid oxidation and exhibit reduced mitochondrial respiratory reserve in the steady state. In addition, NKT cell development is more sensitive to alterations in mitochondrial electron transport chain function than conventional T cells. Using T cell-specific mitochondrial complex III ablation in mice, we further demonstrate that mitochondrial metabolism plays a crucial role in NKT cell development and function by modulating T cell receptor/interleukin-15 signaling and NFAT activity. Collectively, our data provide evidence for a critical role of mitochondrial metabolism in NKT cell development and activation, opening a new avenue for NKT cell-based immunotherapy by manipulating NKT cell metabolism. Conventional T cell fate and function are determined by coordination between cellular signaling and mitochondrial metabolism. Invariant natural killer T (iNKT) cells are an important subset of “innate-like” T cells that exist in a preactivated effector state, and their dependence on mitochondrial metabolism has not been previously defined genetically or in vivo. Here, we show that mature iNKT cells have reduced mitochondrial respiratory reserve and iNKT cell development was highly sensitive to perturbation of mitochondrial function. Mice with T cell-specific ablation of Rieske iron-sulfur protein (RISP; T-Uqcrfs1−/−), an essential subunit of mitochondrial complex III, had a dramatic reduction of iNKT cells in the thymus and periphery, but no significant perturbation on the development of conventional T cells. The impaired development observed in T-Uqcrfs1−/− mice stems from a cell-autonomous defect in iNKT cells, resulting in a differentiation block at the early stages of iNKT cell development. Residual iNKT cells in T-Uqcrfs1−/− mice displayed increased apoptosis but retained the ability to proliferate in vivo, suggesting that their bioenergetic and biosynthetic demands were not compromised. However, they exhibited reduced expression of activation markers, decreased T cell receptor (TCR) signaling and impaired responses to TCR and interleukin-15 stimulation. Furthermore, knocking down RISP in mature iNKT cells diminished their cytokine production, correlating with reduced NFATc2 activity. Collectively, our data provide evidence for a critical role of mitochondrial metabolism in iNKT cell development and activation outside of its traditional role in supporting cellular bioenergetic demands.
Collapse
|
19
|
Gálvez NMS, Bohmwald K, Pacheco GA, Andrade CA, Carreño LJ, Kalergis AM. Type I Natural Killer T Cells as Key Regulators of the Immune Response to Infectious Diseases. Clin Microbiol Rev 2021; 34:e00232-20. [PMID: 33361143 PMCID: PMC7950362 DOI: 10.1128/cmr.00232-20] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The immune system must work in an orchestrated way to achieve an optimal response upon detection of antigens. The cells comprising the immune response are traditionally divided into two major subsets, innate and adaptive, with particular characteristics for each type. Type I natural killer T (iNKT) cells are defined as innate-like T cells sharing features with both traditional adaptive and innate cells, such as the expression of an invariant T cell receptor (TCR) and several NK receptors. The invariant TCR in iNKT cells interacts with CD1d, a major histocompatibility complex class I (MHC-I)-like molecule. CD1d can bind and present antigens of lipid nature and induce the activation of iNKT cells, leading to the secretion of various cytokines, such as gamma interferon (IFN-γ) and interleukin 4 (IL-4). These cytokines will aid in the activation of other immune cells following stimulation of iNKT cells. Several molecules with the capacity to bind to CD1d have been discovered, including α-galactosylceramide. Likewise, several molecules have been synthesized that are capable of polarizing iNKT cells into different profiles, either pro- or anti-inflammatory. This versatility allows NKT cells to either aid or impair the clearance of pathogens or to even control or increase the symptoms associated with pathogenic infections. Such diverse contributions of NKT cells to infectious diseases are supported by several publications showing either a beneficial or detrimental role of these cells during diseases. In this article, we discuss current data relative to iNKT cells and their features, with an emphasis on their driving role in diseases produced by pathogenic agents in an organ-oriented fashion.
Collapse
Affiliation(s)
- Nicolás M S Gálvez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gaspar A Pacheco
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A Andrade
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leandro J Carreño
- Millennium Institute on Immunology and Immunotherapy, Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
20
|
Amiya T, Nakamoto N, Irie J, Taniki N, Chu PS, Koda Y, Miyamoto K, Yamaguchi A, Shiba S, Morikawa R, Itoh H, Kanai T. C-C motif chemokine receptor 9 regulates obesity-induced insulin resistance via inflammation of the small intestine in mice. Diabetologia 2021; 64:603-617. [PMID: 33399911 DOI: 10.1007/s00125-020-05349-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 10/13/2020] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS Accumulation of adipose tissue macrophages is considered pivotal in the development of obesity-associated inflammation and insulin resistance. In addition, recent studies suggest an involvement of the intestine as the primary organ in inducing hyperglycaemia and insulin resistance. We have reported that the C-C motif chemokine receptor (CCR) CCR9 is associated with intestinal immunity and has a pathogenic role in various liver diseases. However, its contribution to type 2 diabetes is unknown. In the current study, we aimed to clarify the involvement of CCR9 in the pathology of type 2 diabetes and the potential underlying mechanisms. METHODS To elucidate how CCR9 affects the development of metabolic phenotypes, we examined the impact of CCR9 deficiency on the pathogenesis of type 2 diabetes using male C57BL/6J (wild-type [WT]) and CCR9-deficient (CCR9 knockout [KO]) mice fed a 60% high-fat diet (HFD) for 12 weeks. RESULTS WT and Ccr9KO mice fed an HFD exhibited a comparable weight gain; however, glucose tolerance and insulin resistance were significantly improved in Ccr9KO mice. Moreover, visceral adipose tissue (VAT) and the liver of Ccr9KO mice presented with less inflammation and increased expression of glucose metabolism-related genes than WT mice. Ccr9 and Ccl25 expression were specifically higher in the small intestine but was not altered by HFD feeding and type 2 diabetes development. Accumulation of IFN-γ-producing CD4+ T lymphocytes and increased intestinal permeability in the small intestine was observed in WT mice following HFD feeding, but these changes were suppressed in HFD-fed Ccr9KO mice. Adoptive transfer of gut-tropic CCR9-expressing T lymphocytes partially reversed the favourable glucose tolerance found in Ccr9KO mice via exacerbated inflammation in the small intestine and VAT. CONCLUSIONS/INTERPRETATION CCR9 plays a central role in the pathogenesis of type 2 diabetes by inducing an inflammatory shift in the small intestine. Our findings support CCR9 as a new therapeutic target for type 2 diabetes via the gut-VAT-liver axis.
Collapse
MESH Headings
- Animals
- Blood Glucose/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Chemokines, CC/genetics
- Chemokines, CC/metabolism
- Chemotaxis, Leukocyte
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/metabolism
- Diet, High-Fat
- Disease Models, Animal
- Enteritis/etiology
- Enteritis/immunology
- Enteritis/metabolism
- Inflammation Mediators/metabolism
- Insulin/blood
- Insulin Resistance
- Interferon-gamma/metabolism
- Intestine, Small/immunology
- Intestine, Small/metabolism
- Intra-Abdominal Fat/immunology
- Intra-Abdominal Fat/metabolism
- Liver/immunology
- Liver/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Obesity/complications
- Obesity/immunology
- Obesity/metabolism
- Receptors, CCR/genetics
- Receptors, CCR/metabolism
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- Takeru Amiya
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- Research Unit/Immunology & Inflammation, Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Kanagawa, Japan
| | - Nobuhiro Nakamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Junichiro Irie
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Nobuhito Taniki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Po-Sung Chu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yuzo Koda
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- Research Unit/Immunology & Inflammation, Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Kanagawa, Japan
| | - Kentaro Miyamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akihiro Yamaguchi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shunsuke Shiba
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Rei Morikawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Itoh
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
21
|
Gu W, Madrid DMD, Yang G, Artiaga BL, Loeb JC, Castleman WL, Richt JA, Lednicky JA, Driver JP. Unaltered influenza disease outcomes in swine prophylactically treated with α-galactosylceramide. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 114:103843. [PMID: 32871161 PMCID: PMC8119227 DOI: 10.1016/j.dci.2020.103843] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/22/2020] [Accepted: 08/22/2020] [Indexed: 05/10/2023]
Abstract
Influenza A viruses (IAV) are a major cause of respiratory diseases in pigs. Invariant natural killer T (iNKT) cells are an innate-like T cell subset that contribute significantly to IAV resistance in mice. In the current work, we explored whether expanding and activating iNKT cells with the iNKT cell superagonist α-galactosylceramide (α-GalCer) would change the course of an IAV infection in pigs. In one study, α-GalCer was administered to pigs intramuscularly (i.m.) 9 days before infection, which systemically expanded iNKT cells. In another study, α-GalCer was administered intranasally (i.n.) 2 days before virus infection to activate mucosal iNKT cells. Despite a synergistic increase in iNKT cells when α-GalCer i.m. treated pigs were infected with IAV, neither approach reduced disease signs, lung pathology, or virus replication. Our results indicate that prophylactic use of iNKT cell agonists to prevent IAV infection is ineffective in pigs. This is significant because this type of approach has been considered for humans whose iNKT cell levels and IAV infections are more similar to those of pigs than mice.
Collapse
Affiliation(s)
- Weihong Gu
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
| | | | - Guan Yang
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
| | - Bianca L Artiaga
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA
| | - Julia C Loeb
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | | | - Jürgen A Richt
- Department of Diagnostic Medicine/Pathobiology and Center of Excellence for Emerging and Zoonotic Animal Diseases (CEEZAD), College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - John A Lednicky
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA; Department of Environmental and Global Health, University of Florida, Gainesville, FL, USA
| | - John P Driver
- Department of Animal Sciences, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
22
|
Anderson CK, Reilly SP, Brossay L. The Invariant NKT Cell Response Has Differential Signaling Requirements during Antigen-Dependent and Antigen-Independent Activation. THE JOURNAL OF IMMUNOLOGY 2020; 206:132-140. [PMID: 33229442 DOI: 10.4049/jimmunol.2000870] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/26/2020] [Indexed: 12/16/2022]
Abstract
Invariant NKT (iNKT) cells are an innate-like population characterized by their recognition of glycolipid Ags and rapid cytokine production upon activation. Unlike conventional T cells, which require TCR ligation, iNKT cells can also be stimulated independently of their TCR. This feature allows iNKT cells to respond even in the absence of glycolipid Ags, for example, during viral infections. Although the TCR-dependent and -independent activation of iNKT cells have been relatively well established, the exact contributions of IL-12, IL-18, and TLRs remain unclear for these two activation pathways. To definitively investigate how these components affect the direct and indirect stimulation of iNKT cells, we used mice deficient for either MyD88 or the IL-12Rβ2 in the T cell lineage. Using these tools, we demonstrate that IL-12, IL-18, and TLRs are completely dispensable for the TCR activation pathway when a strong agonist is used. In contrast, during murine CMV infection, when the TCR is not engaged, IL-12 signaling is essential, and TLR signaling is expendable. Importantly, to our knowledge, we discovered an intrinsic requirement for IL-18 signaling by splenic iNKT cells but not liver iNKT cells, suggesting that there might be diversity, even within the NKT1 population.
Collapse
Affiliation(s)
- Courtney K Anderson
- Department of Molecular Microbiology and Immunology, Division of Biology and Medicine, Brown University, Providence, RI 02906
| | - Shanelle P Reilly
- Department of Molecular Microbiology and Immunology, Division of Biology and Medicine, Brown University, Providence, RI 02906
| | - Laurent Brossay
- Department of Molecular Microbiology and Immunology, Division of Biology and Medicine, Brown University, Providence, RI 02906
| |
Collapse
|
23
|
Driver JP, de Carvalho Madrid DM, Gu W, Artiaga BL, Richt JA. Modulation of Immune Responses to Influenza A Virus Vaccines by Natural Killer T Cells. Front Immunol 2020; 11:2172. [PMID: 33193296 PMCID: PMC7606973 DOI: 10.3389/fimmu.2020.02172] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/10/2020] [Indexed: 12/20/2022] Open
Abstract
Influenza A viruses (IAVs) circulate widely among different mammalian and avian hosts and sometimes give rise to zoonotic infections. Vaccination is a mainstay of IAV prevention and control. However, the efficacy of IAV vaccines is often suboptimal because of insufficient cross-protection among different IAV genotypes and subtypes as well as the inability to keep up with the rapid molecular evolution of IAV strains. Much attention is focused on improving IAV vaccine efficiency using adjuvants, which are substances that can modulate and enhance immune responses to co-administered antigens. The current review is focused on a non-traditional approach of adjuvanting IAV vaccines by therapeutically targeting the immunomodulatory functions of a rare population of innate-like T lymphocytes called invariant natural killer T (iNKT) cells. These cells bridge the innate and adaptive immune systems and are capable of stimulating a wide array of immune cells that enhance vaccine-mediated immune responses. Here we discuss the factors that influence the adjuvant effects of iNKT cells for influenza vaccines as well as the obstacles that must be overcome before this novel adjuvant approach can be considered for human or veterinary use.
Collapse
Affiliation(s)
- John P Driver
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | | | - Weihong Gu
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Bianca L Artiaga
- Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| | - Jürgen A Richt
- Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
24
|
LaMarche NM, Kane H, Kohlgruber AC, Dong H, Lynch L, Brenner MB. Distinct iNKT Cell Populations Use IFNγ or ER Stress-Induced IL-10 to Control Adipose Tissue Homeostasis. Cell Metab 2020; 32:243-258.e6. [PMID: 32516575 PMCID: PMC8234787 DOI: 10.1016/j.cmet.2020.05.017] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/30/2020] [Accepted: 05/15/2020] [Indexed: 01/22/2023]
Abstract
Adipose tissue invariant natural killer T (iNKT) cells are phenotypically different from other iNKT cells because they produce IL-10 and control metabolic homeostasis. Why that is the case is unclear. Here, using single-cell RNA sequencing, we found several adipose iNKT clusters, which we grouped into two functional populations based on NK1.1 expression. NK1.1NEG cells almost exclusively produced IL-10 and other regulatory cytokines, while NK1.1POS iNKT cells predominantly produced IFNγ. Mechanistically, biochemical fractionation revealed that free fatty acids drive IL-10 production primarily in NK1.1NEG iNKT cells via the IRE1α-XBP1s arm of the unfolded protein response. Correspondingly, adoptive transfer of adipose tissue NK1.1NEG iNKT cells selectively restored metabolic function in obese mice. Further, we found an unexpected role for NK1.1POS iNKT cells in lean adipose tissue, as IFNγ licenses natural killer cell-mediated macrophage killing to limit pathological macrophage expansion. Together, these two iNKT cell populations utilize non-redundant pathways to preserve metabolic integrity.
Collapse
Affiliation(s)
- Nelson M LaMarche
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA; Program in Immunology, Harvard Medical School, Boston, MA, USA
| | - Harry Kane
- Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | | | - Han Dong
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Microbiology and Immunology, Harvard Medical School, Boston, MA, USA
| | - Lydia Lynch
- Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland; Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael B Brenner
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA; Program in Immunology, Harvard Medical School, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
25
|
Adar T, Shankar Lankalapalli R, Bittman R, Ilan Y. The assembly of glycosphingolipid determines their immunomodulatory effect: A novel method for structure-based design of immunotherapy. Cell Immunol 2020; 355:104157. [PMID: 32659503 DOI: 10.1016/j.cellimm.2020.104157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/27/2020] [Accepted: 07/01/2020] [Indexed: 11/18/2022]
Abstract
Structure-activity relationships provide insight into the binding interactions of beta-glycosphingolipids (GSLs) with both the TCR and the CD1d molecules, as well as the subsequent immunologic response of regulatory NKT cells. AIM To determine the effects of synthetic GSL structures on their immune modulatory functions. METHODS GSLs of various structures were tested in vitro and in an animal model of Concanavalin A (ConA) immune-mediated hepatitis. RESULTS In vitro, using SV40 binding to live monkey CV1 cells, the l-threo stereoisomer of C8-β-LacCer inhibits caveolar internalization, reducing viral binding to the cell surface. In vivo, in the ConA model, LR172, which has a saturated C8 chain, and LR178, which has a trans double bond at C-2 in the C8 chain, suppressed the immune-mediated liver inflammation and reduced IFNγ levels in a dose dependent manner. The beneficial effects of LR172 and of LR178 are associated with suppression of liver apoptosis, increased phosphorylated STAT3 expression in the liver, and an increase in the NKT liver/spleen ratio. SUMMARY The assembly of GSLs determines their immunomodulatory effect and can serve as a method for structure-based design of immunotherapy.
Collapse
Affiliation(s)
- Tomer Adar
- Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Ravi Shankar Lankalapalli
- Department of Chemistry & Biochemistry, Queens College of the City University of New York, United States; Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Thiruvananthapuram 695019, Kerala, India
| | - Robert Bittman
- Department of Chemistry & Biochemistry, Queens College of the City University of New York, United States
| | - Yaron Ilan
- Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
26
|
Kulicke CA, Lewinsohn DA, Lewinsohn DM. Clonal enrichments of Vδ2- γδ T cells in Mycobacterium tuberculosis-infected human lungs. J Clin Invest 2020; 130:68-70. [PMID: 31763996 DOI: 10.1172/jci133119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Unconventional T cell subsets, including donor-unrestricted T cells (DURTs) and γδ T cells, are promising new players in the treatment and prevention of infectious diseases. In this issue of the JCI, Ogongo et al. used T cell receptor (TCR) sequencing to characterize unconventional T cell subsets in surgical lung resections and blood from Mycobacterium tuberculosis-infected (Mtb-infected) individuals with and without HIV coinfection. The study revealed highly localized expansions of γδ T cell clonotypes not previously associated with the immune response to Mtb and demonstrates the power of high-throughput analysis of the TCR repertoire directly from infected tissue. The findings contribute to our understanding of tuberculosis control and have implications for the development of both therapeutic and vaccination strategies.
Collapse
Affiliation(s)
- Corinna A Kulicke
- Department of Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Deborah A Lewinsohn
- Department of Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - David M Lewinsohn
- Department of Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, Oregon, USA.,Research and Development, VA Portland Health Care System, , Portland, Oregon, USA
| |
Collapse
|
27
|
Sharma M, Zhang S, Niu L, Lewinsohn DM, Zhang X, Huang S. Mucosal-Associated Invariant T Cells Develop an Innate-Like Transcriptomic Program in Anti-mycobacterial Responses. Front Immunol 2020; 11:1136. [PMID: 32582206 PMCID: PMC7295940 DOI: 10.3389/fimmu.2020.01136] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
Conventional T cells exhibit a delayed response to the initial priming of peptide antigens presented by major histocompatibility complex (MHC) proteins. Unlike conventional T cells, mucosal-associated invariant T (MAIT) cells quickly respond to non-peptidic metabolite antigens presented by MHC-related protein 1 (MR1). To elucidate the MR1-dependent activation program of MAIT cells in response to mycobacterial infections, we determined the surface markers, transcriptomic profiles, and effector responses of activated human MAIT cells. Results revealed that mycobacterial-incubated antigen-presenting cells stimulated abundant human CD8+ MAIT cells to upregulate the co-expression of CD69 and CD26, as a combinatorial activation marker. Further transcriptomic analyses demonstrated that CD69+CD26++ CD8+MAIT cells highly expressed numerous genes for mediating anti-mycobacterial immune responses, including pro-inflammatory cytokines, cytolytic molecules, NK cell receptors, and transcription factors, in contrast to inactivated counterparts CD69+/−CD26+/− CD8+MAIT cells. Gene co-expression, enrichment, and pathway analyses yielded high statistical significance to strongly support that activated CD8+ MAIT cells shared gene expression and numerous pathways with NK and CD8+ T cells in activation, cytokine production, cytokine signaling, and effector functions. Flow cytometry detected that activated CD8+MAIT cells produced TNFα, IFNγ, and granulysin to inhibit mycobacterial growth and fight mycobacterial infection. Together, results strongly support that the combinatorial activation marker CD69+CD26++ labels the activated CD8+MAIT cells that develop an innate-like activation program in anti-mycobacterial immune responses. We speculate that the rapid production of anti-mycobacterial effector molecules facilitates MAIT cells to fight early mycobacterial infection in humans.
Collapse
Affiliation(s)
- Manju Sharma
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Shuangmin Zhang
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Liang Niu
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - David M Lewinsohn
- Pulmonary & Critical Care Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Xiang Zhang
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Genomics, Epigenomics and Sequencing Core, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Shouxiong Huang
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Immunobiology Graduate Program, Cincinnati Children's Hospital, Cincinnati, OH, United States
| |
Collapse
|
28
|
Diversity of peripheral blood human NK cells identified by single-cell RNA sequencing. Blood Adv 2020; 4:1388-1406. [PMID: 32271902 PMCID: PMC7160259 DOI: 10.1182/bloodadvances.2019000699] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023] Open
Abstract
Human natural killer (NK) cells in peripheral blood perform many functions, and classification of specific subsets has been a longstanding goal. We report single-cell RNA sequencing of NK cells, comparing gene expression in unstimulated and interleukin (IL)-2-activated cells from healthy cytomegalovirus (CMV)-negative donors. Three NK cell subsets resembled well-described populations; CD56brightCD16-, CD56dimCD16+CD57-, and CD56dimCD16+CD57+. CD56dimCD16+CD57- cells subdivided to include a population with higher chemokine mRNA and increased frequency of killer-cell immunoglobulin-like receptor expression. Three novel human blood NK cell populations were identified: a population of type I interferon-responding NK cells that were CD56neg; a population exhibiting a cytokine-induced memory-like phenotype, including increased granzyme B mRNA in response to IL-2; and finally, a small population, with low ribosomal expression, downregulation of oxidative phosphorylation, and high levels of immediate early response genes indicative of cellular activation. Analysis of CMV+ donors established that CMV altered the proportion of NK cells in each subset, especially an increase in adaptive NK cells, as well as gene regulation within each subset. Together, these data establish an unexpected diversity in blood NK cells and provide a new framework for analyzing NK cell responses in health and disease.
Collapse
|
29
|
Dölen Y, Valente M, Tagit O, Jäger E, Van Dinther EAW, van Riessen NK, Hruby M, Gileadi U, Cerundolo V, Figdor CG. Nanovaccine administration route is critical to obtain pertinent iNKt cell help for robust anti-tumor T and B cell responses. Oncoimmunology 2020; 9:1738813. [PMID: 33457086 PMCID: PMC7790498 DOI: 10.1080/2162402x.2020.1738813] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Nanovaccines, co-delivering antigen and invariant natural killer T (iNKT) cell agonists, proved to be very effective in inducing anti-tumor T cell responses due to their exceptional helper function. However, it is known that iNKT cells are not equally present in all lymphoid organs and nanoparticles do not get evenly distributed to all immune compartments. In this study, we evaluated the effect of the vaccination route on iNKT cell help to T and B cell responses for the first time in an antigen and agonist co-delivery setting. Intravenous administration of PLGA nanoparticles was mainly targeting liver and spleen where iNKT1 cells are abundant and induced the highest serum IFN-y levels, T cell cytotoxicity, and Th-1 type antibody responses. In comparison, after subcutaneous or intranodal injections, nanoparticles mostly drained or remained in regional lymph nodes where iNKT17 cells were abundant. After subcutaneous and intranodal injections, antigen-specific IgG2 c production was hampered and IFN-y production, as well as cytotoxic T cell responses, depended on sporadic systemic drainage. Therapeutic anti-tumor experiments also demonstrated a clear advantage of intravenous injection over intranodal or subcutaneous vaccinations. Moreover, tumor control could be further improved by PD-1 immune checkpoint blockade after intravenous vaccination, but not by intranodal vaccination. Anti PD-1 antibody combination mainly exerts its effect by prolonging the cytotoxicity of T cells. Nanovaccines also demonstrated synergism with anti-4-1BB agonistic antibody treatment in controlling tumor growth. We conclude that nanovaccines containing iNKT cell agonists shall be preferentially administered intravenously, to optimally reach cellular partners for inducing effective anti-tumor immune responses.
Collapse
Affiliation(s)
- Yusuf Dölen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center & Oncode Institute, Nijmegen, The Netherlands
| | - Michael Valente
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center & Oncode Institute, Nijmegen, The Netherlands
| | - Oya Tagit
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center & Oncode Institute, Nijmegen, The Netherlands
| | - Eliezer Jäger
- Institute of Macromolecular Chemistry V.v.i., Academy of Sciences of the Czech Republic, Prague 6, Czech Republic
| | - Eric A W Van Dinther
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center & Oncode Institute, Nijmegen, The Netherlands
| | - N Koen van Riessen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center & Oncode Institute, Nijmegen, The Netherlands
| | - Martin Hruby
- Institute of Macromolecular Chemistry V.v.i., Academy of Sciences of the Czech Republic, Prague 6, Czech Republic
| | - Uzi Gileadi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center & Oncode Institute, Nijmegen, The Netherlands
| |
Collapse
|
30
|
Elieh Ali Komi D, Ribatti D. Mast cell-mediated mechanistic pathways in organ transplantation. Eur J Pharmacol 2019; 857:172458. [PMID: 31202799 DOI: 10.1016/j.ejphar.2019.172458] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 02/02/2023]
Abstract
Adaptive immunity has gained importance in transplant immunology for years, based on models in which T-cells orchestrate the immune responses during rejection. Most recently, researches revealed that innate immune cells, including mast cells (MCs) also play a pivotal role in allograft rejection. MC mediated immunoregulatory responses influence the innate and adaptive immune responses. Their capability to produce an array of both pro-inflammatory and anti-inflammatory mediators, expressing a wide range of costimulatory molecules in addition to acting as antigen-presenting cells (APCs), make them effective immune cells far beyond their classical role as primary orchestrator cells of allergy. Activated regulatory Tcells (Treg) cells contribute to MC recruitment into grafts by releasing interleukin (IL)-9. Tregs are capable of stabilizing MCs and suppressing IgE mediated degranulation through interaction of Treg expressing OX40 with MCs expressing OX40L. MCs in turn release transforming growth factor (TGF)-β and IL-10 which possess suppressive properties. Thus, these cells can suppress the proliferation of T-cells and support the generation of Tregs. MCs in addition to orchestrating immune responses in grafts by cell-to-cell interactions with variety of immune cells, cause histologic changes, mainly fibrosis by releasing mediators such as histamine, fibroblast growth factor-2 (FGF-2), TGF-β, chymase, and cathepsin G. The role of MCs in transplant rejection remains controversial. The accumulation of MCs in rejected grafts suggests that they play a role in preventing graft tolerance, and contribute to the progression of chronic rejection of allografts. However, high expression of MC-related gene products in tolerant grafts and their known interaction with Tregs on the other hand, support the notion that they are an integral component in achieving peripheral tolerance.
Collapse
Affiliation(s)
- Daniel Elieh Ali Komi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
31
|
Song B, Aoki S, Liu C, Ito K. A toll-like receptor 9 agonist sensitizes mice to mitochondrial dysfunction-induced hepatic apoptosis via the Fas/FasL pathway. Arch Toxicol 2019; 93:1573-1584. [PMID: 30993380 DOI: 10.1007/s00204-019-02454-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/09/2019] [Indexed: 01/17/2023]
Abstract
Early hepatocyte death occurs in most liver injury cases and triggers liver inflammation, which in combination with other risk factors leads to the development of liver disease. However, the pathogenesis of early phase hepatocyte death remains poorly understood. Here, C57BL/6J mice were treated with the hepatotoxic drug flucloxacillin (FLUX) and the toll-like receptor 9 agonist CpG oligodeoxynucleotide (ODN) to reproduce the early phase of drug-induced hepatotoxicity and investigate its pathogenesis. C57BL/6J mice were treated with FLUX (100 mg/kg, gavage) alone or in combination with ODN (40 μg/mouse, intraperitoneally). Plasma alanine aminotransferase (ALT) level was measured as a marker of hepatotoxicity. FLUX or ODN alone was insufficient to induce ALT elevation, whereas combination treatment with FLUX and ODN increased ALT levels 24 h after FLUX treatment and upregulated Fas ligand in natural killer T (NKT) cells and Fas in hepatocytes. FLUX induced mitochondrial permeability transition (MPT), and pretreatment with ODN sensitized mitochondria to FLUX-induced MPT. The increase in ALT levels induced by ODN and FLUX co-treatment was suppressed in Fas ligand (gld/gld)-deficient mice and in mice deficient in a component of MPT pore opening (cyclophilin D-knockout mice). These results suggested that ODN activated the Fas/Fas ligand-mediated pathway in NKT cells and hepatocytes, which may predispose to FLUX-induced mitochondrial dysfunction and lead to early phase hepatocyte apoptosis. Taken together, these findings elucidate a potentially novel mechanism underlying drug-induced early phase hepatocyte death related to the Fas/Fas ligand death receptor pathway and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Binbin Song
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8675, Japan
| | - Shigeki Aoki
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8675, Japan
| | - Cong Liu
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8675, Japan
| | - Kousei Ito
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8675, Japan.
| |
Collapse
|
32
|
Verbeke R, Lentacker I, Breckpot K, Janssens J, Van Calenbergh S, De Smedt SC, Dewitte H. Broadening the Message: A Nanovaccine Co-loaded with Messenger RNA and α-GalCer Induces Antitumor Immunity through Conventional and Natural Killer T Cells. ACS NANO 2019; 13:1655-1669. [PMID: 30742405 DOI: 10.1021/acsnano.8b07660] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Messenger RNA encoding tumor antigens has the potential to evoke effective antitumor immunity. This study reports on a nanoparticle platform, named mRNA Galsomes, that successfully co-delivers nucleoside-modified antigen-encoding mRNA and the glycolipid antigen and immunopotentiator α-galactosylceramide (α-GC) to antigen-presenting cells after intravenous administration. By co-formulating low doses of α-GC, mRNA Galsomes induce a pluripotent innate and adaptive tumor-specific immune response in mice, with invariant natural killer T cells (iNKT) as a driving force. In comparison, mRNA Galsomes exhibit advantages over the state-of-the-art cancer vaccines using unmodified ovalbumin (OVA)-encoding mRNA, as we observed up to seven times more tumor-infiltrating antigen-specific cytotoxic T cells, combined with a strong iNKT cell and NK cell activation. In addition, the presence of suppressive myeloid cells (myeloid-derived suppressor cells and tumor-associated macrophages) in the tumor microenvironment was significantly lowered. Owing to these antitumor effects, OVA mRNA Galsomes significantly reduced tumor growth in established E.G7-OVA lymphoma, with a complete tumor rejection in 40% of the animals. Moreover, therapeutic vaccination with mRNA Galsomes enhanced the responsiveness to treatment with a PD-L1 checkpoint inhibitor in B16-OVA melanoma, as evidenced by a synergistic reduction of tumor outgrowth and a significantly prolonged median survival. Taken together, these data show that intravenously administered mRNA Galsomes can provide controllable, multifaceted, and effective antitumor immunity, especially when combined with checkpoint inhibition.
Collapse
Affiliation(s)
- Rein Verbeke
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy , Ghent University , Ghent 9000 , Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University Hospital , Ghent University , Ghent 9000 , Belgium
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy , Ghent University , Ghent 9000 , Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University Hospital , Ghent University , Ghent 9000 , Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences , Vrije Universiteit Brussel (VUB) , Jette 1090 , Belgium
| | - Jonas Janssens
- Laboratory for Medicinal Chemistry, Faculty of Pharmacy , Ghent University , Ghent 9000 , Belgium
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry, Faculty of Pharmacy , Ghent University , Ghent 9000 , Belgium
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy , Ghent University , Ghent 9000 , Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University Hospital , Ghent University , Ghent 9000 , Belgium
| | - Heleen Dewitte
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy , Ghent University , Ghent 9000 , Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University Hospital , Ghent University , Ghent 9000 , Belgium
- Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences , Vrije Universiteit Brussel (VUB) , Jette 1090 , Belgium
| |
Collapse
|
33
|
Chen L, Gu J, Qian Y, Li M, Qian Y, Xu M, Li J, Wen Y, Xia L, Li J, Xia Q, Kong X, Wu H. Deletion of C-C Motif Chemokine Ligand 5 Worsens Invariant Natural Killer T-Cell-Mediated Hepatitis via Compensatory Up-regulation of CXCR2-Related Chemokine Activity. Cell Mol Gastroenterol Hepatol 2019; 7:623-639. [PMID: 30630119 PMCID: PMC6434496 DOI: 10.1016/j.jcmgh.2018.12.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 12/29/2018] [Accepted: 12/31/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Chemokine-mediated immune cell recruitment plays pivotal roles in liver inflammation. C-C motif chemokine ligand 5 (CCL5) has been shown to be responsible for the recruitment of monocytes/macrophages and has been implicated in various liver diseases, including nonalcoholic fatty liver disease, fibrosis, and hepatocellular carcinoma. Previous studies have also shown that inhibition of CCL5 appears to be a promising therapeutic approach for several chronic liver diseases. However, whether blocking CCL5 could benefit immune cell-mediated hepatitis remains largely elusive. METHODS By adopting a specific agonist, alpha-galactosylceramide (α-Galcer), of invariant natural killer T cells (iNKTs), we investigated the function and mechanism of CCL5 in the iNKT induced murine hepatitis model. RESULTS We found significantly increased CCL5 expression in α-Galcer-induced hepatitis murine model. Such an increase in CCL5 is mainly enriched in non-parenchymal cells such as macrophages and iNKTs but not in hepatocytes. Surprisingly, CCL5 blockage by genetic deletion of Ccl5 does not affect the α-Galcer-induced iNKT activation but greatly worsens α-Galcer-induced liver injury accompanied by an increased hepatic neutrophil infiltration. Mechanistically, we demonstrated that greater neutrophil accumulation in the liver is responsible for the enhanced liver injury in Ccl5-/- mice. Such an increased hepatic neutrophil infiltration is mainly caused by an enhanced CXCL1-CXCR2 signal in Ccl5-/- mice. Therapeutically, either antibody-mediated neutrophil depletion or a CXCR2 antagonist, SB225002, mediated CXCR2 signaling blockage significantly ameliorated α-Galcer-induced liver injury in Ccl5-/- mice. CONCLUSIONS Our present study demonstrates that (1) α-Galcer-induced murine hepatitis could greatly induce CCL5 production in macrophages and iNKT cells; (2) loss of CCL5 could enhance CXCL1 expression in hepatocytes and activate CXCL1-CXCR2 axis in neutrophils to augment their hepatic infiltration; and (3) neutrophil depletion or blockage of CXCL1-CXCR2 axis greatly improves α-Galcer-induced liver injury in Ccl5-/- mice. This study suggests that clinical utilization of CCL5 blockage may compensatorily induce the activation of other chemokine pathways to enhance neutrophil recruitment and liver injury in hepatitis.
Collapse
Affiliation(s)
- Lili Chen
- Shanghai Key Laboratory for Molecular Imaging, Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, China; Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jinyang Gu
- Department of Transplantation, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yihan Qian
- School of Pharmacy, Fudan University, Shanghai, China
| | - Meng Li
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongbing Qian
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Min Xu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jichang Li
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yankai Wen
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiaxin Li
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoni Kong
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Hailong Wu
- Shanghai Key Laboratory for Molecular Imaging, Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, China.
| |
Collapse
|
34
|
Baez NS, Cerbán F, Savid-Frontera C, Hodge DL, Tosello J, Acosta-Rodriguez E, Almada L, Gruppi A, Viano ME, Young HA, Rodriguez-Galan MC. Thymic expression of IL-4 and IL-15 after systemic inflammatory or infectious Th1 disease processes induce the acquisition of "innate" characteristics during CD8+ T cell development. PLoS Pathog 2019; 15:e1007456. [PMID: 30608984 PMCID: PMC6319713 DOI: 10.1371/journal.ppat.1007456] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 11/05/2018] [Indexed: 01/28/2023] Open
Abstract
Innate CD8+ T cells express a memory-like phenotype and demonstrate a strong cytotoxic capacity that is critical during the early phase of the host response to certain bacterial and viral infections. These cells arise in the thymus and depend on IL-4 and IL-15 for their development. Even though innate CD8+ T cells exist in the thymus of WT mice in low numbers, they are highly enriched in KO mice that lack certain kinases, leading to an increase in IL-4 production by thymic NKT cells. Our work describes that in C57BL/6 WT mice undergoing a Th1 biased infectious disease, the thymus experiences an enrichment of single positive CD8 (SP8) thymocytes that share all the established phenotypical and functional characteristics of innate CD8+ T cells. Moreover, through in vivo experiments, we demonstrate a significant increase in survival and a lower parasitemia in mice adoptively transferred with SP8 thymocytes from OT I—T. cruzi-infected mice, demonstrating that innate CD8+ thymocytes are able to protect against a lethal T. cruzi infection in an Ag-independent manner. Interestingly, we obtained similar results when using thymocytes from systemic IL-12 + IL-18-treated mice. This data indicates that cytokines triggered during the acute stage of a Th1 infectious process induce thymic production of IL-4 along with IL-15 expression resulting in an adequate niche for development of innate CD8+ T cells as early as the double positive (DP) stage. Our data demonstrate that the thymus can sense systemic inflammatory situations and alter its conventional CD8 developmental pathway when a rapid innate immune response is required to control different types of pathogens. Murine innate CD8+ T cells demonstrate strong cytotoxic capacity during the early phase of certain bacterial and viral infections. Such cells have been reported to be present in both mice and humans but many questions remain as to their differentiation and maturation process. Innate CD8+ T cells arise in the thymus and depend on IL-4 and IL-15 for their development. A description of the cellular and molecular mechanisms involved during their thymic development has been obtained from KO mice that lack kinases and transcription factors important for TCR signaling. In these mice, SP8 thymocytes with an innate phenotype are highly enriched over the conventional SP8 cells. Our work describes, for the first time, that in WT mice, thymic IL-4 and IL-15 expression triggered by Th1 infectious processes induce an adequate niche for development of innate rather than conventional CD8+ T cells. Our data show that the thymus is able to sense a systemic inflammatory response (probably mediated by systemic IL-12 and IL-18 production) and alter its ontogeny when pathogen control is needed.
Collapse
Affiliation(s)
- Natalia S. Baez
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Fabio Cerbán
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Constanza Savid-Frontera
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Deborah L. Hodge
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States of America
| | - Jimena Tosello
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Eva Acosta-Rodriguez
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Laura Almada
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Adriana Gruppi
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maria Estefania Viano
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Howard A. Young
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States of America
| | - Maria Cecilia Rodriguez-Galan
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- * E-mail:
| |
Collapse
|
35
|
Activation of natural killer T cells contributes to triptolide-induced liver injury in mice. Acta Pharmacol Sin 2018; 39:1847-1854. [PMID: 30013034 DOI: 10.1038/s41401-018-0084-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 06/19/2018] [Indexed: 12/24/2022]
Abstract
Triptolide (TP) is the main active ingredient of Tripterygium wilfordii Hook.f, which has attracted great interest due to its promising efficacy for autoimmune diseases and tumors. However, severe adverse reactions, especially hepatotoxicity, have restricted its approval in the market. In the present study we explored the role of hepatic natural killer T (NKT) cells in the pathogenesis of TP-induced liver injury in mice. TP (600 μg/kg/day, i.g.) was administered to female mice for 1, 3, or 5 days. We found that administration of TP dose-dependently induced hepatotoxicity, evidenced by the body weight reduction, elevated serum ALT and AST levels, as well as significant histopathological changes in the livers. However, the mice were resistant to the development of TP-induced liver injury when their NKT cells were depleted by injection of anti-NK1.1 mAb (200 μg, i.p.) on days -2 and -1 before TP administration. We further revealed that TP administration activated NKT cells, dominantly releasing Th1 cytokine IFN-γ, recruiting neutrophils and macrophages, and leading to liver damage. After anti-NK1.1 injection, however, the mice mainly secreted Th2 cytokine IL-4 in the livers and exhibited a significantly lower percentage of hepatic infiltrating neutrophils and macrophages upon TP challenge. The activation of NKT cells was associated with the upregulation of Toll-like receptor (TLR) signaling pathway. Collectively, these results demonstrate a novel role of NKT cells contributing to the mechanisms of TP-induced liver injury. More importantly, the regulation of NKT cells may promote effective measures that control drug-induced liver injury.
Collapse
|
36
|
Maricic I, Marrero I, Eguchi A, Nakamura R, Johnson CD, Dasgupta S, Hernandez CD, Nguyen PS, Swafford AD, Knight R, Feldstein AE, Loomba R, Kumar V. Differential Activation of Hepatic Invariant NKT Cell Subsets Plays a Key Role in Progression of Nonalcoholic Steatohepatitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:3017-3035. [PMID: 30322964 PMCID: PMC6219905 DOI: 10.4049/jimmunol.1800614] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/07/2018] [Indexed: 02/07/2023]
Abstract
Innate immune mechanisms play an important role in inflammatory chronic liver diseases. In this study, we investigated the role of type I or invariant NKT (iNKT) cell subsets in the progression of nonalcoholic steatohepatitis (NASH). We used α-galactosylceramide/CD1d tetramers and clonotypic mAb together with intracytoplasmic cytokine staining to analyze iNKT cells in choline-deficient l-amino acid-defined (CDAA)-induced murine NASH model and in human PBMCs, respectively. Cytokine secretion of hepatic iNKT cells in CDAA-fed C57BL/6 mice altered from predominantly IL-17+ to IFN-γ+ and IL-4+ during NASH progression along with the downmodulation of TCR and NK1.1 expression. Importantly, steatosis, steatohepatitis, and fibrosis were dependent upon the presence of iNKT cells. Hepatic stellate cell activation and infiltration of neutrophils, Kupffer cells, and CD8+ T cells as well as expression of key proinflammatory and fibrogenic genes were significantly blunted in Jα18-/- mice and in C57BL/6 mice treated with an iNKT-inhibitory RAR-γ agonist. Gut microbial diversity was significantly impacted in Jα18-/- and in CDAA diet-fed mice. An increased frequency of CXCR3+IFN-γ+T-bet+ and IL-17A+ iNKT cells was found in PBMC from NASH patients in comparison with nonalcoholic fatty liver patients or healthy controls. Consistent with their in vivo activation, iNKT cells from NASH patients remained hyporesponsive to ex-vivo stimulation with α-galactosylceramide. Accumulation of plasmacytoid dendritic cells in both mice and NASH patients suggest their role in activation of iNKT cells. In summary, our findings indicate that the differential activation of iNKT cells play a key role in mediating diet-induced hepatic steatosis and fibrosis in mice and its potential involvement in NASH progression in humans.
Collapse
Affiliation(s)
- Igor Maricic
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Idania Marrero
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Akiko Eguchi
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093
| | - Ryota Nakamura
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093
| | - Casey D Johnson
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093
| | - Suryasarathi Dasgupta
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Carolyn D Hernandez
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Phirum Sam Nguyen
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Austin D Swafford
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA 92093
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA 92093
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA 92093; and
| | - Ariel E Feldstein
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093
- Nonalcoholic Fatty Liver Disease Research Center, University of California San Diego, La Jolla, CA 92093
| | - Rohit Loomba
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA 92093
- Nonalcoholic Fatty Liver Disease Research Center, University of California San Diego, La Jolla, CA 92093
| | - Vipin Kumar
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093;
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA 92093
- Nonalcoholic Fatty Liver Disease Research Center, University of California San Diego, La Jolla, CA 92093
| |
Collapse
|
37
|
Huang W, He W, Shi X, He X, Dou L, Gao Y. The Role of CD1d and MR1 Restricted T Cells in the Liver. Front Immunol 2018; 9:2424. [PMID: 30425710 PMCID: PMC6218621 DOI: 10.3389/fimmu.2018.02424] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/01/2018] [Indexed: 12/17/2022] Open
Abstract
The liver is one of the most important immunological organs that remains tolerogenic in homeostasis yet promotes rapid responses to pathogens in the presence of a systemic infection. The composition of leucocytes in the liver is highly distinct from that of the blood and other lymphoid organs, particularly with respect to enrichment of innate T cells, i.e., invariant NKT cells (iNKT cells) and Mucosal-Associated Invariant T cells (MAIT cells). In recent years, studies have revealed insights into their biology and potential roles in maintaining the immune-environment in the liver. As the primary liver-resident immune cells, they are emerging as significant players in the human immune system and are associated with an increasing number of clinical diseases. As such, innate T cells are promising targets for modifying host defense and inflammation of various liver diseases, including viral, autoimmune, and those of tumor origin. In this review, we emphasize and discuss some of the recent discoveries and advances in the biology of innate T cells, their recruitment and diversity in the liver, and their role in various liver diseases, postulating on their potential application in immunotherapy.
Collapse
Affiliation(s)
- Wenyong Huang
- Organ Transplantation Unit, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenjing He
- Organ Transplantation Unit, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaomin Shi
- Organ Transplantation Unit, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoshun He
- Organ Transplantation Unit, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lang Dou
- Organ Transplantation Unit, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yifang Gao
- Organ Transplantation Unit, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
38
|
Regulation of the terminal maturation of iNKT cells by mediator complex subunit 23. Nat Commun 2018; 9:3875. [PMID: 30250136 PMCID: PMC6155209 DOI: 10.1038/s41467-018-06372-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 08/31/2018] [Indexed: 11/21/2022] Open
Abstract
Invariant natural killer T cells (iNKT cells) are a specific subset of T cells that recognize glycolipid antigens and upon activation rapidly exert effector functions. This unique function is established during iNKT cell development; the detailed mechanisms of this process, however, remain to be elucidated. Here the authors show that deletion of the mediator subunit Med23 in CD4+CD8+ double positive (DP) thymocytes completely blocks iNKT cell development at stage 2. This dysregulation is accompanied by a bias in the expression of genes related to the regulation of transcription and metabolism, and functional impairment of the cells including the loss of NK cell characteristics, reduced ability to secrete cytokines and attenuated recruitment capacity upon activation. Moreover, Med23-deficient iNKT cells exhibit impaired anti-tumor activity. Our study identifies Med23 as an essential transcriptional regulator that controls iNKT cell differentiation and terminal maturation. Invariant Natural Killer T cells (iNKT) rapidly exert effector functions upon activation, but the mechanisms of their functional maturation remain to be determined. Here, Xu and colleagues show that the mediator subunit Med23 is a transcriptional regulator controlling iNKT cell terminal maturation.
Collapse
|
39
|
Uchida T, Nakashima H, Ito S, Ishikiriyama T, Nakashima M, Seki S, Kumagai H, Oshima N. Activated natural killer T cells in mice induce acute kidney injury with hematuria through possibly common mechanisms shared by human CD56 + T cells. Am J Physiol Renal Physiol 2018; 315:F618-F627. [PMID: 29993279 DOI: 10.1152/ajprenal.00160.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Although activation of mouse natural killer T (NKT) cells by α-galactosylceramide (α-GalCer) causes failure of multiple organs, including the kidneys, the precise mechanisms underlying kidney injury remain unclear. Here, we showed that α-GalCer-activated mouse NKT cells injured both kidney vascular endothelial cells and tubular epithelial cells in vitro, causing acute kidney injury (AKI) with hematuria in middle-aged mice. The perforin-mediated pathway was mainly involved in glomerular endothelial cell injury, whereas the TNF-α/Fas ligand pathway played an important role in the injury of tubular epithelial cells. Kidney injury in young mice was mild but could be significantly exacerbated if NKT cells were strongly activated by NK cell depletion alone or in combination with IL-12 pretreatment. When stimulated by a combination of IL-2 and IL-12, human CD56+ T cells, a functional counterpart of mouse NKT cells, also damaged both glomerular endothelial cells and tubular epithelial cells, with the former being affected in a perforin-dependent manner. These data suggest that both mouse NKT cells and human CD56+ T cells are integral to the processes that mediate AKI. Targeting CD56+ T cells may, therefore, be a promising approach to treat AKI.
Collapse
Affiliation(s)
- Takahiro Uchida
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama , Japan
| | - Hiroyuki Nakashima
- Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Saitama , Japan
| | - Seigo Ito
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama , Japan
| | - Takuya Ishikiriyama
- Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Saitama , Japan
| | - Masahiro Nakashima
- Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Saitama , Japan
| | - Shuhji Seki
- Department of Immunology and Microbiology, National Defense Medical College, Tokorozawa, Saitama , Japan
| | - Hiroo Kumagai
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama , Japan
| | - Naoki Oshima
- Department of Nephrology and Endocrinology, National Defense Medical College, Tokorozawa, Saitama , Japan
| |
Collapse
|
40
|
Bolognese AC, Yang WL, Hansen LW, Sharma A, Nicastro JM, Coppa GF, Wang P. Activation of Invariant Natural Killer T Cells Redirects the Inflammatory Response in Neonatal Sepsis. Front Immunol 2018; 9:833. [PMID: 29720984 PMCID: PMC5922987 DOI: 10.3389/fimmu.2018.00833] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/05/2018] [Indexed: 12/20/2022] Open
Abstract
Sepsis is the third leading cause of death in the neonatal population, due to susceptibility to infection conferred by immaturity of both the innate and adaptive components of the immune system. Invariant natural killer T (iNKT) cells are specialized adaptive immune cells that possess important innate-like characteristics and have not yet been well-studied in septic neonates. We hypothesized that iNKT cells would play an important role in mediating the neonatal immune response to sepsis. To study this, we subjected 5- to 7-day-old neonatal C57BL/6 mice to sepsis by intraperitoneal (i.p.) cecal slurry (CS) injection. Thirty hours prior to or immediately following sepsis induction, pups received i.p. injection of the iNKT stimulator KRN7000 (KRN, 0.2 µg/g) or vehicle. Ten hours after CS injection, blood and tissues were collected for various analyses. Thirty-hour pretreatment with KRN resulted in better outcomes in inflammation, lung injury, and survival, while immediate treatment with KRN resulted in worse outcomes compared to vehicle treatment. We further analyzed the activation status of neonatal iNKT cells for 30 h after KRN administration, and showed a peak in frequency of CD69 expression on iNKT cells and serum IFN-γ levels at 5 and 10 h, respectively. We then used CD1d knockout neonatal mice to demonstrate that KRN acts through the major histocompatibility complex-like molecule CD1d to improve outcomes in neonatal sepsis. Finally, we identified that KRN pretreatment exerts its protective effect by increasing systemic levels of TGF-β1. These findings support the importance of iNKT cells for prophylactic immunomodulation in neonates susceptible to sepsis.
Collapse
Affiliation(s)
- Alexandra C Bolognese
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States.,Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Weng-Lang Yang
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States.,Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States.,Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Laura W Hansen
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Archna Sharma
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Jeffrey M Nicastro
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Gene F Coppa
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Ping Wang
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States.,Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States.,Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| |
Collapse
|
41
|
Kling JC, Jordan MA, Pitt LA, Meiners J, Thanh-Tran T, Tran LS, Nguyen TTK, Mittal D, Villani R, Steptoe RJ, Khosrotehrani K, Berzins SP, Baxter AG, Godfrey DI, Blumenthal A. Temporal Regulation of Natural Killer T Cell Interferon Gamma Responses by β-Catenin-Dependent and -Independent Wnt Signaling. Front Immunol 2018; 9:483. [PMID: 29616022 PMCID: PMC5864864 DOI: 10.3389/fimmu.2018.00483] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/23/2018] [Indexed: 12/13/2022] Open
Abstract
Natural killer T (NKT) cells are prominent innate-like lymphocytes in the liver with critical roles in immune responses during infection, cancer, and autoimmunity. Interferon gamma (IFN-γ) and IL-4 are key cytokines rapidly produced by NKT cells upon recognition of glycolipid antigens presented by antigen-presenting cells (APCs). It has previously been reported that the transcriptional coactivator β-catenin regulates NKT cell differentiation and functionally biases NKT cell responses toward IL-4, at the expense of IFN-γ production. β-Catenin is not only a central effector of Wnt signaling but also contributes to other signaling networks. It is currently unknown whether Wnt ligands regulate NKT cell functions. We thus investigated how Wnt ligands and β-catenin activity shape liver NKT cell functions in vivo in response to the glycolipid antigen, α-galactosylceramide (α-GalCer) using a mouse model. Pharmacologic targeting of β-catenin activity with ICG001, as well as myeloid-specific genetic ablation of Wntless (Wls), to specifically target Wnt protein release by APCs, enhanced early IFN-γ responses. By contrast, within several hours of α-GalCer challenge, myeloid-specific Wls deficiency, as well as pharmacologic targeting of Wnt release using the small molecule inhibitor IWP-2 impaired α-GalCer-induced IFN-γ responses, independent of β-catenin activity. These data suggest that myeloid cell-derived Wnt ligands drive early Wnt/β-catenin signaling that curbs IFN-γ responses, but that, subsequently, Wnt ligands sustain IFN-γ expression independent of β-catenin activity. Our analyses in ICG001-treated mice confirmed a role for β-catenin activity in driving early IL-4 responses by liver NKT cells. However, neither pharmacologic nor genetic perturbation of Wnt production affected the IL-4 response, suggesting that IL-4 production by NKT cells in response to α-GalCer is not driven by released Wnt ligands. Collectively, these data reveal complex temporal roles of Wnt ligands and β-catenin signaling in the regulation of liver NKT cell activation, and highlight Wnt-dependent and -independent contributions of β-catenin to NKT cell functions.
Collapse
Affiliation(s)
- Jessica C Kling
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Margaret A Jordan
- Comparative Genomics Centre, James Cook University, Townsville, QLD, Australia
| | - Lauren A Pitt
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Jana Meiners
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Thao Thanh-Tran
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Le Son Tran
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Tam T K Nguyen
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Deepak Mittal
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Rehan Villani
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Raymond J Steptoe
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Kiarash Khosrotehrani
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Stuart P Berzins
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia.,Fiona Elsey Cancer Research Institute; and Federation University, Ballarat, VIC, Australia
| | - Alan G Baxter
- Comparative Genomics Centre, James Cook University, Townsville, QLD, Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, VIC, Australia
| | - Antje Blumenthal
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
42
|
Van Kaer L, Wu L. Therapeutic Potential of Invariant Natural Killer T Cells in Autoimmunity. Front Immunol 2018; 9:519. [PMID: 29593743 PMCID: PMC5859017 DOI: 10.3389/fimmu.2018.00519] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 02/28/2018] [Indexed: 11/13/2022] Open
Abstract
Tolerance against self-antigens is regulated by a variety of cell types with immunoregulatory properties, such as CD1d-restricted invariant natural killer T (iNKT) cells. In many experimental models of autoimmunity, iNKT cells promote self-tolerance and protect against autoimmunity. These findings are supported by studies with patients suffering from autoimmune diseases. Based on these studies, the therapeutic potential of iNKT cells in autoimmunity has been explored. Many of these studies have been performed with the potent iNKT cell agonist KRN7000 or its structural variants. These findings have generated promising results in several autoimmune diseases, although mechanisms by which iNKT cells modulate autoimmunity remain incompletely understood. Here, we will review these preclinical studies and discuss the prospects for translating their findings to patients suffering from autoimmune diseases.
Collapse
Affiliation(s)
- Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Lan Wu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
43
|
Yang G, Richt JA, Driver JP. Harnessing Invariant NKT Cells to Improve Influenza Vaccines: A Pig Perspective. Int J Mol Sci 2017; 19:68. [PMID: 29280974 PMCID: PMC5796018 DOI: 10.3390/ijms19010068] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 12/21/2017] [Accepted: 12/25/2017] [Indexed: 12/20/2022] Open
Abstract
Invariant natural killer T (iNKT) cells are an "innate-like" T cell lineage that recognize glycolipid rather than peptide antigens by their semi-invariant T cell receptors. Because iNKT cells can stimulate an extensive array of immune responses, there is considerable interest in targeting these cells to enhance human vaccines against a wide range of microbial pathogens. However, long overlooked is the potential to harness iNKT cell antigens as vaccine adjuvants for domestic animal species that express the iNKT cell-CD1d system. In this review, we discuss the prospect of targeting porcine iNKT cells as a strategy to enhance the efficiency of swine influenza vaccines. In addition, we compare the phenotype and tissue distribution of porcine iNKT cells. Finally, we discuss the challenges that must be overcome before iNKT cell agonists can be contemplated for veterinary use in livestock.
Collapse
Affiliation(s)
- Guan Yang
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, USA.
| | - Jürgen A Richt
- College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
- Diagnostic Medicine/Pathobiology and Center of Excellence for Emerging and Zoonotic Animal Diseases (CEEZAD), Manhattan, KS 66502, USA.
| | - John P Driver
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
44
|
Teyton L. New Directions for Natural Killer T Cells in the Immunotherapy of Cancer. Front Immunol 2017; 8:1480. [PMID: 29209309 PMCID: PMC5701619 DOI: 10.3389/fimmu.2017.01480] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/23/2017] [Indexed: 01/07/2023] Open
Abstract
Natural killer T (NKT) cells have been placed at the interface between innate and adaptive immunity by a long series of experiments that convincingly showed that beyond cytokine secretion and NK cell recruitment, NKT cells were coordinating dendritic cell and B cell maturation through direct membrane contacts and initiate productive responses. As such, NKT cells are the cellular adjuvant of many immune reactions and have functions that go much beyond what their name encapsulates. In addition, the initial discovery of the ligands of NKT cells is deeply linked to cancer biology and therapy. However, for a host of reasons, animal models in which agonists of NKT cells were used did not translate well to human cancers. A systematic reassessment of NKT cells role in tumorigenesis, especially spontaneous one, is now accessible using single cell analysis technologies both in mouse and man, and should be taken advantage of. Similarly, the migration, localization, phenotype of NKT cells following induced expansion after injection of an agonist can be examined at the single cell level. This technological revolution will help evaluate where and how NKT cells can be used in cancer.
Collapse
Affiliation(s)
- Luc Teyton
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
45
|
Keller CW, Freigang S, Lünemann JD. Reciprocal Crosstalk between Dendritic Cells and Natural Killer T Cells: Mechanisms and Therapeutic Potential. Front Immunol 2017; 8:570. [PMID: 28596767 PMCID: PMC5442181 DOI: 10.3389/fimmu.2017.00570] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 04/28/2017] [Indexed: 12/23/2022] Open
Abstract
Natural killer T cells carrying a highly conserved, semi-invariant T cell receptor (TCR) [invariant natural killer T (iNKT) cells] are a subset of unconventional T lymphocytes that recognize glycolipids presented by CD1d molecules. Although CD1d is expressed on a variety of hematopoietic and non-hematopoietic cells, dendritic cells (DCs) are key presenters of glycolipid antigen in vivo. When stimulated through their TCR, iNKT cells rapidly secrete copious amounts of cytokines and induce maturation of DCs, thereby facilitating coordinated stimulation of innate and adaptive immune responses. The bidirectional crosstalk between DCs and iNKT cells determines the functional outcome of iNKT cell-targeted responses and iNKT cell agonists are used and currently being evaluated as adjuvants to enhance the efficacy of antitumor immunotherapy. This review illustrates mechanistic underpinnings of reciprocal DCs and iNKT cell interactions and discusses how those can be harnessed for cancer therapy.
Collapse
Affiliation(s)
- Christian W Keller
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland
| | - Stefan Freigang
- Institute of Pathology, Laboratory of Immunopathology, University of Bern, Bern, Switzerland
| | - Jan D Lünemann
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland.,Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
46
|
Keller CW, Loi M, Ewert S, Quast I, Theiler R, Gannagé M, Münz C, De Libero G, Freigang S, Lünemann JD. The autophagy machinery restrains iNKT cell activation through CD1D1 internalization. Autophagy 2017; 13:1025-1036. [PMID: 28296542 DOI: 10.1080/15548627.2017.1297907] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Invariant natural killer T (iNKT) cells are innate T cells with powerful immune regulatory functions that recognize glycolipid antigens presented by the CD1D protein. While iNKT cell-activating glycolipids are currently being explored for their efficacy to improve immunotherapy against infectious diseases and cancer, little is known about the mechanisms that control CD1D antigen presentation and iNKT cell activation in vivo. CD1D molecules survey endocytic pathways to bind lipid antigens in MHC class II-containing compartments (MIICs) before recycling to the plasma membrane. Autophagosomes intersect with MIICs and autophagy-related proteins are known to support antigen loading for increased CD4+ T cell immunity. Here, we report that mice with dendritic cell (DC)-specific deletion of the essential autophagy gene Atg5 showed better CD1D1-restricted glycolipid presentation in vivo. These effects led to enhanced iNKT cell cytokine production upon antigen recognition and lower bacterial loads during Sphingomonas paucimobilis infection. Enhanced iNKT cell activation was independent of receptor-mediated glycolipid uptake or costimulatory signals. Instead, loss of Atg5 in DCs impaired clathrin-dependent internalization of CD1D1 molecules via the adaptor protein complex 2 (AP2) and, thus, increased surface expression of stimulatory CD1D1-glycolipid complexes. These findings indicate that the autophagic machinery assists in the recruitment of AP2 to CD1D1 molecules resulting in attenuated iNKT cell activation, in contrast to the supporting role of macroautophagy in CD4+ T cell stimulation.
Collapse
Affiliation(s)
- Christian W Keller
- a Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich , Zurich , Switzerland
| | - Monica Loi
- b Institute of Experimental Immunology, Laboratory of Viral Immunobiology, University of Zurich , Zurich , Switzerland
| | - Svenja Ewert
- c Institute of Pathology, Laboratory of Immunopathology, University of Bern , Bern , Switzerland
| | - Isaak Quast
- a Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich , Zurich , Switzerland.,d Department of Immunology & Pathology , Central Clinical School, Monash University , Melbourne , Australia
| | - Romina Theiler
- c Institute of Pathology, Laboratory of Immunopathology, University of Bern , Bern , Switzerland
| | - Monique Gannagé
- e Department of Pathology and Immunology , School of Medicine - CMU, University of Geneva , Geneva , Switzerland.,f Division of Rheumatology , Department of Internal Medicine , University Hospital , Geneva , Geneva , Switzerland
| | - Christian Münz
- b Institute of Experimental Immunology, Laboratory of Viral Immunobiology, University of Zurich , Zurich , Switzerland
| | - Gennaro De Libero
- g Singapore Immunology Network , Agency for Science, Technology and Research (A*STAR) , Singapore.,h Department of Biomedicine, Laboratory of Experimental Immunology , University Hospital Basel, University of Basel , Basel , Switzerland
| | - Stefan Freigang
- c Institute of Pathology, Laboratory of Immunopathology, University of Bern , Bern , Switzerland
| | - Jan D Lünemann
- a Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich , Zurich , Switzerland
| |
Collapse
|
47
|
Tsuchiya K, Ikeda T, Batmunkh B, Choijookhuu N, Ishizaki H, Hotokezaka M, Hishikawa Y, Nanashima A. Frequency of CD4+CD161+ T Cell and Interleukin-10 Expression in Inflammatory Bowel Diseases. Acta Histochem Cytochem 2017; 50:21-28. [PMID: 28386147 PMCID: PMC5374100 DOI: 10.1267/ahc.16035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/05/2016] [Indexed: 12/19/2022] Open
Abstract
Mucosal immune dysregulation associated with T cells plays a critical role in the development of inflammatory bowel diseases (IBD). However, the definite significances of these cells in IBD still remain unclear. Therefore, we investigated the population and expression of CD4+CD161+ T cells in the colonic lamina propria mononuclear cells (LPMCs) in patients with IBD by analyses using flow cytometry and immunohistochemistry. Interleukin-10 (IL-10) mRNA levels in both LPMCs and CD4+ T cells in lamina propria (LP-CD4+ T cells) were measured using a real-time quantitative reverse transcription-polymerase chain reaction. IL-10 production was investigated with immunohistochemistry. The results revealed that the population of CD4+CD161+ T cells was significantly decreased in active ulcerative colitis (UC) compared with inactive UC (P < 0.05). The CD4+CD161+ T cell population was inversely correlated with disease activity in patients with UC (r = −0.6326, P = 0.0055), but there was no significant correlation in those with Crohn’s disease. Over-expression of IL-10 mRNA in both LPMCs and LP-CD4+ T cells were detected in active UC. Immunohistochemistry revealed decreased frequency of CD161+ cells and increased IL-10 positive cells in active UC. The frequency of CD4+CD161+ T cells and IL-10 expression was supposed to be associated with the pathological status of mucosal immunoregulation in IBD.
Collapse
Affiliation(s)
- Kazuyo Tsuchiya
- Department of Surgery, Faculty of Medicine, University of Miyazaki
| | - Takuto Ikeda
- Department of Surgery, Faculty of Medicine, University of Miyazaki
| | - Baatarsuren Batmunkh
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki
| | - Narantsog Choijookhuu
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki
| | | | | | - Yoshitaka Hishikawa
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki
| | | |
Collapse
|
48
|
Wei Y, Zeng B, Chen J, Cui G, Lu C, Wu W, Yang J, Wei H, Xue R, Bai L, Chen Z, Li L, Iwabuchi K, Uede T, Van Kaer L, Diao H. Enterogenous bacterial glycolipids are required for the generation of natural killer T cells mediated liver injury. Sci Rep 2016; 6:36365. [PMID: 27821872 PMCID: PMC5099575 DOI: 10.1038/srep36365] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/13/2016] [Indexed: 02/08/2023] Open
Abstract
Glycolipids are potent activator of natural killer T (NKT) cells. The relationship between NKT cells and intestinal bacterial glycolipids in liver disorders remained unclear. We found that, in sharp contrast to specific pathogen-free (SPF) mice, germ-free (GF) mice are resistant to Concanavalin A (ConA)-induced liver injury. ConA treatment failed to trigger the activation of hepatic NKT cells in GF mice. These defects correlated with the sharply reduced levels of CD1d-presented glycolipid antigens in ConA-treated GF mice compared with SPF counterparts. Nevertheless, CD1d expression was similar between these two kinds of mice. The absence of intestinal bacteria did not affect the incidence of αGalCer-induced liver injury in GF mice. Importantly, we found the intestinal bacteria contain glycolipids which can be presented by CD1d and recognized by NKT cells. Furthermore, supplement of killed intestinal bacteria was able to restore ConA-mediated NKT cell activation and liver injury in GF mice. Our results suggest that glycolipid antigens derived from intestinal commensal bacteria are important hepatic NKT cell agonist and these antigens are required for the activation of NKT cells during ConA-induced liver injury. These finding provide a mechanistic explanation for the capacity of intestinal microflora to control liver inflammation.
Collapse
Affiliation(s)
- Yingfeng Wei
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Benhua Zeng
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China
| | - Jianing Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Guangying Cui
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Chong Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Wei Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Jiezuan Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China
| | - Rufeng Xue
- Institute of Immunology and Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Biology, Hefei National Laboratory for Physical Sciences at Microscale, Hefei 230027, China
| | - Li Bai
- Institute of Immunology and Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei 230027, China.,Innovation Center for Cell Biology, Hefei National Laboratory for Physical Sciences at Microscale, Hefei 230027, China
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - Kazuya Iwabuchi
- Department of Immunology, Kitasato University School of Medicine, Sagamihar, 108-8641, Japan
| | - Toshimitsu Uede
- Molecular Immunology, Institute for Genetic Medicine, Hokkaido University, Sapporo, 0600815, Japan
| | - Luc Van Kaer
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, 37232, USA
| | - Hongyan Diao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| |
Collapse
|
49
|
Abstract
Hepatitis B virus (HBV) infection is a worldwide health problem, with approximately one third of populations have been infected, among which 3-5% of adults and more than 90% of children developed to chronic HBV infection. Host immune factors play essential roles in the outcome of HBV infection. Thus, ineffective immune response against HBV may result in persistent virus replications and liver necroinflammations, then lead to chronic HBV infection, liver cirrhosis, and even hepatocellular carcinoma. Cytokine balance was shown to be an important immune characteristic in the development and progression of hepatitis B, as well as in an effective antiviral immunity. Large numbers of cytokines are not only involved in the initiation and regulation of immune responses but also contributing directly or indirectly to the inhibition of virus replication. Besides, cytokines initiate downstream signaling pathway activities by binding to specific receptors expressed on the target cells and play important roles in the responses against viral infections and, therefore, might affect susceptibility to HBV and/or the natural course of the infection. Since cytokines are the primary causes of inflammation and mediates liver injury after HBV infection, we have discussed recent advances on the roles of various cytokines [including T helper type 1 cells (Th1), Th2, Th17, regulatory T cells (Treg)-related cytokines] in different phases of HBV infection and cytokine-related mechanisms for impaired viral control and liver damage during HBV infection. We then focus on experimental therapeutic applications of cytokines to gain a better understanding of this newly emerging aspect of disease pathogenesis.
Collapse
|
50
|
Hägglöf T, Sedimbi SK, Yates JL, Parsa R, Salas BH, Harris RA, Leadbetter EA, Karlsson MCI. Neutrophils license iNKT cells to regulate self-reactive mouse B cell responses. Nat Immunol 2016; 17:1407-1414. [PMID: 27798616 DOI: 10.1038/ni.3583] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/13/2016] [Indexed: 12/13/2022]
Abstract
The innate responsiveness of the immune system is important not only for quick responses to pathogens but also for the initiation and shaping of the subsequent adaptive immune response. Activation via the cytokine IL-18, a product of inflammasomes, gives rise to a rapid response that includes the production of self-reactive antibodies. As increased concentrations of this cytokine are found in inflammatory diseases, we investigated the origin of the B cell response and its regulation. We identified an accumulation of B cell-helper neutrophils in the spleen that interacted with innate-type invariant natural killer T cells (iNKT cells) to regulate B cell responses. We found that neutrophil-dependent expression of the death-receptor ligand FasL by iNKT cells was needed to restrict autoantibody production. Neutrophils can thus license iNKT cells to regulate potentially harmful autoreactive B cell responses during inflammasome-driven inflammation.
Collapse
Affiliation(s)
- Thomas Hägglöf
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Saikiran K Sedimbi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | | | - Roham Parsa
- Department of Clinical Neuroscience, Karolinska Institutet, Centre for Molecular Medicine, Karolinska University Hospital at Solna, Solna, Sweden
| | - Briana Hauff Salas
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Robert A Harris
- Department of Clinical Neuroscience, Karolinska Institutet, Centre for Molecular Medicine, Karolinska University Hospital at Solna, Solna, Sweden
| | - Elizabeth A Leadbetter
- Trudeau Institute, Saranac Lake, New York, USA.,Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| |
Collapse
|