1
|
Mou YJ, Li FM, Zhang R, Sheng R, Han R, Zhang ZL, Hu LF, Zhao YZ, Wu JC, Qin ZH. The P2X7 receptor mediates NADPH transport across the plasma membrane. Biochem Biophys Res Commun 2024; 737:150500. [PMID: 39142135 DOI: 10.1016/j.bbrc.2024.150500] [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: 06/24/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
Nicotinamide Adenine Dinucleotide Phosphate (NADPH) plays a vital role in regulating redox homeostasis and reductive biosynthesis. However, if exogenous NADPH can be transported across the plasma membrane has remained elusive. In this study, we present evidence supporting that NADPH can traverse the plasma membranes of cells through a mechanism mediated by the P2X7 receptor (P2X7R). Notably, we observed an augmentation of intracellular NADPH levels in cultured microglia upon exogenous NADPH supplementation in the presence of ATP. The P2X7R-mediated transmembrane transportation of NADPH was validated with P2X7R antagonists, including OX-ATP, BBG, and A-438079, or through P2X7 knockdown, which impeded NADPH transportation into cells. Conversely, overexpression of P2X7 resulted in an enhanced capacity for NADPH transport. Furthermore, transfection of hP2X7 demonstrated the ability to complement NADPH uptake in native HEK293 cells. Our findings provide evidence for the first time that NADPH is transported across the plasma membrane via a P2X7R-mediated pathway. Additionally, we propose an innovative avenue for modulating intracellular NADPH levels. This discovery holds promise for advancing our understanding of the role of NADPH in redox homeostasis and neuroinflammation.
Collapse
Affiliation(s)
- Yu-Jie Mou
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Feng-Min Li
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Rong Zhang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Rong Han
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Zhong-Ling Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, 23 Post Street, Nangang District, Harbin, HeiLongjiang 150081, China.
| | - Li-Fang Hu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China.
| | - Yu-Zheng Zhao
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, 130 Mei Long Road, Shanghai 200237, China.
| | - Jun-Chao Wu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; Institute of Health Science and Technology, Suzhou Gaobo Vocational College, Qingshan Road, Suzhou Science and Technology Tower, Hi-Tech Area, Suzhou, Jiangsu 215163, China.
| |
Collapse
|
2
|
Junge M, Liaukouskaya N, Schwarz N, Pinto-Espinoza C, Schaffrath AZ, Rissiek B, Krebs CF, Rattay G, Mittrücker HW, Tomas NM, Nicke A, Haag F, Huber TB, Meyer-Schwesinger C, Koch-Nolte F, Wanner N. ATP-Gated P2X7-Ion Channel on Kidney-Resident Natural Killer T Cells and Memory T Cells in Intrarenal Inflammation. J Am Soc Nephrol 2024:00001751-990000000-00489. [PMID: 39675762 DOI: 10.1681/asn.0000000564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
Key Points
Parenchymal T cells in the kidney expressed much higher levels of P2X7 than vascular T cells.P2X7-blocking nanobodies uncover a large fraction of kidney-resident natural killer T and tissue-resident memory T cells.These cells were lost during cell preparation because of activation of P2X7 by NAD+ released from damaged cells, unless blocked by nanobodies.
Background
The P2X7 ion channel, a key sensor of sterile inflammation, has been implicated as a therapeutic target in GN, and P2X7-antagonistic nanobodies can attenuate experimental GN. However, little is known about the expression of P2X7 on renal immune cells.
Methods
We used conventional immunofluorescence of kidney sections and intraperitoneal injection of nanobodies in mice followed by flow cytometry analysis of parenchymal T cells and RNA sequencing to elucidate the expression and function of P2X7 on parenchymal and vascular immune cells in the mouse kidney.
Results
Our study showed that parenchymal T cells, including a large subset of natural killer T cells and CD69+ tissue-resident memory T cells, display much higher cell surface levels of P2X7 than vascular T cells. After a single intraperitoneal injection of P2X7-blocking nanobodies, P2X7 on parenchymal T cells was fully occupied by the injected nanobodies within 30 minutes. This resulted in an effective protection of these cells from nicotinamide adenine dinucleotide–induced cell death during cell preparation. Conversely, systemic injection of nicotinamide adenine dinucleotide that mimics sterile inflammation results in the selective depletion of P2X7hiCD69hi T cells from the kidney parenchyma.
Conclusions
Our study uncovered a novel purinergic regulatory mechanism affecting kidney-resident T-cell populations.
Collapse
Affiliation(s)
- Marten Junge
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nastassia Liaukouskaya
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicole Schwarz
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carolina Pinto-Espinoza
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alessa Z Schaffrath
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Rissiek
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian F Krebs
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Rattay
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Willi Mittrücker
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola M Tomas
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Annette Nicke
- Walther Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Catherine Meyer-Schwesinger
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Wanner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
3
|
Grassi F. An unconventional purine connection. J Exp Med 2024; 221:e20241527. [PMID: 39560664 PMCID: PMC11577275 DOI: 10.1084/jem.20241527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024] Open
Abstract
Xu et al. (https://doi.org/10.1084/jem.20240354) define NAD-induced cell death via purinergic P2RX7 receptor in type 1 unconventional T cells, particularly intrahepatic MAIT cells that are pivotal in liver homeostasis. Therefore, P2RX7 is a potential target to modulate unconventional T cells in immunopathological conditions and cancer.
Collapse
Affiliation(s)
- Fabio Grassi
- Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
4
|
Xu C, Obers A, Qin M, Brandli A, Wong J, Huang X, Clatch A, Fayed A, Starkey G, D’Costa R, Gordon CL, Mak JY, Fairlie DP, Beattie L, Mackay LK, Godfrey DI, Koay HF. Selective regulation of IFN-γ and IL-4 co-producing unconventional T cells by purinergic signaling. J Exp Med 2024; 221:e20240354. [PMID: 39560665 PMCID: PMC11577439 DOI: 10.1084/jem.20240354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 08/01/2024] [Accepted: 10/08/2024] [Indexed: 11/20/2024] Open
Abstract
Unconventional T cells, including mucosal-associated invariant T (MAIT), natural killer T (NKT), and gamma-delta T (γδT) cells, comprise distinct T-bet+, IFN-γ+ and RORγt+, IL-17+ subsets which play differential roles in health and disease. NKT1 cells are susceptible to ARTC2-mediated P2X7 receptor (P2RX7) activation, but the effects on other unconventional T-cell types are unknown. Here, we show that MAIT, γδT, and NKT cells express P2RX7 and are sensitive to P2RX7-mediated cell death. Mouse peripheral T-bet+ MAIT1, γδT1, and NKT1 cells, especially in liver, co-express ARTC2 and P2RX7. These markers could be further upregulated upon exposure to retinoic acid. Blocking ARTC2 or inhibiting P2RX7 protected MAIT1, γδT1, and NKT1 cells from cell death, enhanced their survival in vivo, and increased the number of IFN-γ-secreting cells without affecting IL-17 production. Importantly, this revealed the existence of IFN-γ and IL-4 co-producing unconventional T-cell populations normally lost upon isolation due to ARTC2/P2RX7-induced death. Administering extracellular NAD in vivo activated this pathway, depleting P2RX7-sensitive unconventional T cells. Our study reveals ARTC2/P2RX7 as a common regulatory axis modulating the unconventional T-cell compartment, affecting the viability of IFN-γ- and IL-4-producing T cells, offering important insights to facilitate future studies into how these cells can be regulated in health and disease.
Collapse
Affiliation(s)
- Calvin Xu
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Andreas Obers
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Minyi Qin
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Alice Brandli
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Australia
| | - Joelyn Wong
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Xin Huang
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Allison Clatch
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Aly Fayed
- Liver and Intestinal Transplant Unit, Austin Health, Melbourne, Australia
- Department of Surgery, The University of Melbourne, Austin Health, Melbourne, Australia
| | - Graham Starkey
- Liver and Intestinal Transplant Unit, Austin Health, Melbourne, Australia
- Department of Surgery, The University of Melbourne, Austin Health, Melbourne, Australia
| | - Rohit D’Costa
- DonateLife Victoria, Carlton, Australia
- Department of Intensive Care Medicine, Melbourne Health, Melbourne, Australia
| | - Claire L. Gordon
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, Austin Health, Melbourne, Australia
- North Eastern Public Health Unit, Austin Health, Melbourne, Australia
| | - Jeffrey Y.W. Mak
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - David P. Fairlie
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Lynette Beattie
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Laura K. Mackay
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Hui-Fern Koay
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| |
Collapse
|
5
|
English K, Kwan R, Holz LE, McGuffog C, Krol JMM, Kempe D, Kaisho T, Heath WR, Lisowski L, Biro M, McCaughan GW, Bowen DG, Bertolino P. A hepatic network of dendritic cells mediates CD4 T cell help outside lymphoid organs. Nat Commun 2024; 15:1261. [PMID: 38341416 PMCID: PMC10858872 DOI: 10.1038/s41467-024-45612-5] [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: 02/06/2023] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
While CD4+ T cells are a prerequisite for CD8+ T cell-mediated protection against intracellular hepatotropic pathogens, the mechanisms facilitating the transfer of CD4-help to intrahepatic CD8+ T cells are unknown. Here, we developed an experimental system to investigate cognate CD4+ and CD8+ T cell responses to a model-antigen expressed de novo in hepatocytes and reveal that after initial priming, effector CD4+ and CD8+ T cells migrate into portal tracts and peri-central vein regions of the liver where they cluster with type-1 conventional dendritic cells. These dendritic cells are locally licensed by CD4+ T cells and expand the number of CD8+ T cells in situ, resulting in larger effector and memory CD8+ T cell pools. These findings reveal that CD4+ T cells promote intrahepatic immunity by amplifying the CD8+ T cell response via peripheral licensing of hepatic type-1 conventional dendritic cells and identify intrahepatic perivascular compartments specialized in facilitating effector T cell-dendritic cell interactions.
Collapse
Affiliation(s)
- Kieran English
- Centenary Institute and The University of Sydney, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Rain Kwan
- Centenary Institute and The University of Sydney, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Lauren E Holz
- Department of Microbiology and Immunology at The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Claire McGuffog
- Centenary Institute and The University of Sydney, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Jelte M M Krol
- Centenary Institute and The University of Sydney, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia
- Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Daryan Kempe
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - William R Heath
- Department of Microbiology and Immunology at The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Leszek Lisowski
- Children's Medical Research Institute, Translational Vectorology Research Unit, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Maté Biro
- EMBL Australia, Single Molecule Science node, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Geoffrey W McCaughan
- Centenary Institute and The University of Sydney, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - David G Bowen
- Centenary Institute and The University of Sydney, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
| | - Patrick Bertolino
- Centenary Institute and The University of Sydney, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
| |
Collapse
|
6
|
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
|
7
|
Xu C, Li S, Fulford TS, Christo SN, Mackay LK, Gray DH, Uldrich AP, Pellicci DG, I Godfrey D, Koay HF. Expansion of MAIT cells in the combined absence of NKT and γδ-T cells. Mucosal Immunol 2023; 16:446-461. [PMID: 37182737 DOI: 10.1016/j.mucimm.2023.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/11/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
Mucosal-associated invariant T (MAIT) cells, natural killer T (NKT) cells, and γδT cells are collectively referred to as 'unconventional T cells' due to their recognition of non-peptide antigens and restriction to MHC-I-like molecules. However, the factors controlling their widely variable frequencies between individuals and organs are poorly understood. We demonstrated that MAIT cells are increased in NKT or γδT cell-deficient mice and highly expand in mice lacking both cell types. TCRα repertoire analysis of γδT cell-deficient thymocytes revealed altered Trav segment usage relative to wild-type thymocytes, highlighting retention of the Tcra-Tcrd locus from the 129 mouse strain used to generate Tcrd-/- mice. This resulted in a moderate increase in distal Trav segment usage, including Trav1, potentially contributing to increased generation of Trav1-Traj33+ MAIT cells in the Tcrd-/- thymus. Importantly, adoptively transferred MAIT cells underwent increased homeostatic proliferation within NKT/gdT cell-deficient tissues, with MAIT cell subsets exhibiting tissue-specific homing patterns. Our data reveal a shared niche for unconventional T cells, where competition for common factors may be exploited to collectively modulate these cells in the immune response. Lastly, our findings emphasise careful assessment of studies using NKT or γδT cell-deficient mice when investigating the role of unconventional T cells in disease.
Collapse
Affiliation(s)
- Calvin Xu
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Shihan Li
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Thomas S Fulford
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Susan N Christo
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Laura K Mackay
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Daniel Hd Gray
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Adam P Uldrich
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Daniel G Pellicci
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia; Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia.
| | - Dale I Godfrey
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia.
| | - Hui-Fern Koay
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
8
|
Sluyter R, Adriouch S, Fuller SJ, Nicke A, Sophocleous RA, Watson D. Animal Models for the Investigation of P2X7 Receptors. Int J Mol Sci 2023; 24:ijms24098225. [PMID: 37175933 PMCID: PMC10179175 DOI: 10.3390/ijms24098225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
The P2X7 receptor is a trimeric ligand-gated cation channel activated by extracellular adenosine 5'-triphosphate. The study of animals has greatly advanced the investigation of P2X7 and helped to establish the numerous physiological and pathophysiological roles of this receptor in human health and disease. Following a short overview of the P2X7 distribution, roles and functional properties, this article discusses how animal models have contributed to the generation of P2X7-specific antibodies and nanobodies (including biologics), recombinant receptors and radioligands to study P2X7 as well as to the pharmacokinetic testing of P2X7 antagonists. This article then outlines how mouse and rat models have been used to study P2X7. These sections include discussions on preclinical disease models, polymorphic P2X7 variants, P2X7 knockout mice (including bone marrow chimeras and conditional knockouts), P2X7 reporter mice, humanized P2X7 mice and P2X7 knockout rats. Finally, this article reviews the limited number of studies involving guinea pigs, rabbits, monkeys (rhesus macaques), dogs, cats, zebrafish, and other fish species (seabream, ayu sweetfish, rainbow trout and Japanese flounder) to study P2X7.
Collapse
Affiliation(s)
- Ronald Sluyter
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Sahil Adriouch
- UniRouen, INSERM, U1234, Pathophysiology, Autoimmunity, and Immunotherapy, (PANTHER), Univ Rouen Normandie, University of Rouen, F-76000 Rouen, France
| | - Stephen J Fuller
- Sydney Medical School Nepean, Faculty of Medicine and Health, The University of Sydney, Nepean Hospital, Kingswood, NSW 2750, Australia
| | - Annette Nicke
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, 80336 Munich, Germany
| | - Reece A Sophocleous
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Debbie Watson
- Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| |
Collapse
|
9
|
Depew CE, Rixon JA, McSorley SJ. Optimal generation of hepatic tissue-resident memory CD4 T cells requires IL-1 and IL-2. Proc Natl Acad Sci U S A 2023; 120:e2214699120. [PMID: 37040404 PMCID: PMC10120061 DOI: 10.1073/pnas.2214699120] [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: 08/30/2022] [Accepted: 02/23/2023] [Indexed: 04/12/2023] Open
Abstract
Hepatic CD4 tissue-resident memory T cells (TRM) are required for robust protection against Salmonella infection; however, the generation of this T cell population is poorly understood. To interrogate the contribution of inflammation, we developed a simple Salmonella-specific T cell transfer system that allowed direct visualization of hepatic TRM formation. Salmonella-specific (SM1) T cell receptor (TCR) transgenic CD4 T cells were activated in vitro and adoptively transferred into C57BL/6 mice while hepatic inflammation was induced by acetaminophen overdose or L. monocytogenes infection. In both model systems, hepatic CD4 TRM formation was accentuated by local tissue responses. Liver inflammation also enhanced the suboptimal protection provided by a subunit Salmonella vaccine which typically induces circulating memory CD4 T cells. To further elucidate the mechanism of CD4 TRM formation in response to liver inflammation, various cytokines were examined by RNAseq, bone marrow chimeras, and in vivo neutralization. Surprisingly, IL-2 and IL-1 were found to enhance CD4 TRM formation. Thus, local inflammatory mediators enhance CD4 TRM populations and can boost the protective immunity provided by a suboptimal vaccine. This knowledge will be foundational for the development of a more effective vaccine against invasive nontyphoidal salmonellosis (iNTS).
Collapse
Affiliation(s)
- Claire E. Depew
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA95616
| | - Jordan A. Rixon
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA95616
| | - Stephen J. McSorley
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA95616
| |
Collapse
|
10
|
Complexing CpG adjuvants with cationic liposomes enhances vaccine-induced formation of liver T RM cells. Vaccine 2023; 41:1094-1107. [PMID: 36609029 DOI: 10.1016/j.vaccine.2022.12.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 12/05/2022] [Accepted: 12/19/2022] [Indexed: 01/06/2023]
Abstract
Tissue resident memory T cells (TRM cells) can provide effective tissue surveillance and can respond rapidly to infection. Vaccination strategies aimed at generating TRM cells have shown promise against a range of pathogens. We have previously shown that the choice of adjuvant critically influences CD8+ TRM cell formation in the liver. However, the range of adjuvants tested was limited. Here, we assessed the ability of a broad range of adjuvants stimulating membrane (TLR4), endosomal (TLR3, TLR7 and TLR9) and cytosolic (cGAS, RIG-I) pathogen recognition receptors for their capacity to induce CD8+ TRM formation in a subunit vaccination model. We show that CpG oligodeoxynucleotides (ODN) remain the most efficient inducers of liver TRM cells among all adjuvants tested. Moreover, their combination with the cationic liposome DOTAP further enhances the potency, particularly of the class B ODN CpG 1668 and the human TLR9 ligand CpG 2006 (CpG 7909). This study informs the design of efficient liver TRM-based vaccines for their potential translation.
Collapse
|
11
|
Real-Time Flow Cytometry as a Tool to Monitor Cellular Consequences of P2X7 Activation in Multiple Cell Populations Mixed in a Single FACS Tube. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2510:291-302. [PMID: 35776332 DOI: 10.1007/978-1-0716-2384-8_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The P2X7 receptor is an ATP-gated ion channel expressed by cells of the immune system. In murine T cells, P2X7 activation by high concentrations of ATP or by covalent ADP-ribosylation are potent triggers of cell death. In innate immune cells, such as macrophages or brain microglia, P2X7 is a key regulator of inflammasome activation and the release of mature interleukin 1 beta. ATP-mediated P2X7 activation is accompanied by several direct downstream events, including the influx of calcium, pore formation at the plasma membrane, ectodomain shedding, and cell shrinkage. With this chapter we provide a protocol to monitor all these immediate consequences of P2X7 activation in a time dependent fashion using real-time flow cytometry. We illustrate, for example, how to simultaneously monitor calcium influx and shedding of CD27 in four T cell subpopulations and how to simultaneously analyze calcium influx, pore formation and cell shrinkage in mouse primary microglia. We further provide an extended protocol to compare consequences of P2X7 activation among identical cell populations from two or more different donor mice mixed in a single FACS tube. Taken together, the here presented real-time flow cytometry protocol for measuring P2X7 activation is flexible, scalable and can easily be transferred to other experimental settings.
Collapse
|
12
|
Chen Z, Wang N, Yao Y, Yu D. Context-dependent regulation of follicular helper T cell survival. Trends Immunol 2022; 43:309-321. [DOI: 10.1016/j.it.2022.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 02/07/2023]
|
13
|
Leutert M, Duan Y, Winzer R, Menzel S, Tolosa E, Magnus T, Hottiger MO, Koch-Nolte F, Rissiek B. Identification of the Mouse T Cell ADP-Ribosylome Uncovers ARTC2.2 Mediated Regulation of CD73 by ADP-Ribosylation. Front Immunol 2021; 12:703719. [PMID: 34504490 PMCID: PMC8421852 DOI: 10.3389/fimmu.2021.703719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Mouse T cells express the ecto-ADP-ribosyltransferase ARTC2.2, which can transfer the ADP-ribose group of extracellular nicotinamide adenine dinucleotide (NAD+) to arginine residues of various cell surface proteins thereby influencing their function. Several targets of ARTC2.2, such as P2X7, CD8a and CD25 have been identified, however a comprehensive mouse T cell surface ADP-ribosylome analysis is currently missing. Using the Af1521 macrodomain-based enrichment of ADP-ribosylated peptides and mass spectrometry, we identified 93 ADP-ribsoylated peptides corresponding to 67 distinct T cell proteins, including known targets such as CD8a and CD25 but also previously unknown targets such as CD73. We evaluated the impact of ADP-ribosylation on the capability of CD73 to generate adenosine from adenosine monophosphate. Our results show that extracellular NAD+ reduces the enzymatic activity of CD73 HEK cells co-transfected with CD73/ARTC2.2. Importantly, NAD+ significantly reduced CD73 activity on WT CD8 T cells compared to ARTC2ko CD8 T cells or WT CD8 T cells treated with an ARTC2.2-blocking nanobody. Our study provides a comprehensive list of T cell membrane proteins that serve as targets for ADP-ribosylation by ARTC2.2 and whose function may be therefore affected by ADP-ribosylation.
Collapse
Affiliation(s)
- Mario Leutert
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland.,Department of Genome Sciences, University of Washington, Seattle, WA, United States
| | - Yinghui Duan
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Riekje Winzer
- Institute of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Menzel
- Institute of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.,Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eva Tolosa
- Institute of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Michael O Hottiger
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Rissiek
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
14
|
Gondé H, Demeules M, Hardet R, Scarpitta A, Junge M, Pinto-Espinoza C, Varin R, Koch-Nolte F, Boyer O, Adriouch S. A Methodological Approach Using rAAV Vectors Encoding Nanobody-Based Biologics to Evaluate ARTC2.2 and P2X7 In Vivo. Front Immunol 2021; 12:704408. [PMID: 34489954 PMCID: PMC8417108 DOI: 10.3389/fimmu.2021.704408] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 08/03/2021] [Indexed: 01/03/2023] Open
Abstract
On murine T cells, mono-ADP ribosyltransferase ARTC2.2 catalyzes ADP-ribosylation of various surface proteins when nicotinamide adenine dinucleotide (NAD+) is released into the extracellular compartment. Covalent ADP-ribosylation of the P2X7 receptor by ARTC2.2 thereby represents an additional mechanism of activation, complementary to its triggering by extracellular ATP. P2X7 is a multifaceted receptor that may represents a potential target in inflammatory, and neurodegenerative diseases, as well as in cancer. We present herein an experimental approach using intramuscular injection of recombinant AAV vectors (rAAV) encoding nanobody-based biologics targeting ARTC2.2 or P2X7. We demonstrate the ability of these in vivo generated biologics to potently and durably block P2X7 or ARTC2.2 activities in vivo, or in contrast, to potentiate NAD+- or ATP-induced activation of P2X7. We additionally demonstrate the ability of rAAV-encoded functional heavy chain antibodies to elicit long-term depletion of T cells expressing high levels of ARTC2.2 or P2X7. Our approach of using rAAV to generate functional nanobody-based biologics in vivo appears promising to evaluate the role of ARTC2.2 and P2X7 in murine acute as well as chronic disease models.
Collapse
Affiliation(s)
- Henri Gondé
- Normandie University, UNIROUEN, INSERM U1234, Pathophysiology, Autoimmunity, Neuromuscular Diseases and Regenerative THERapies, Rouen, France.,Rouen University Hospital, Department of Pharmacy, Rouen, France
| | - Mélanie Demeules
- Normandie University, UNIROUEN, INSERM U1234, Pathophysiology, Autoimmunity, Neuromuscular Diseases and Regenerative THERapies, Rouen, France
| | - Romain Hardet
- Normandie University, UNIROUEN, INSERM U1234, Pathophysiology, Autoimmunity, Neuromuscular Diseases and Regenerative THERapies, Rouen, France
| | - Allan Scarpitta
- Normandie University, UNIROUEN, INSERM U1234, Pathophysiology, Autoimmunity, Neuromuscular Diseases and Regenerative THERapies, Rouen, France
| | - Marten Junge
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Rémi Varin
- Normandie University, UNIROUEN, INSERM U1234, Pathophysiology, Autoimmunity, Neuromuscular Diseases and Regenerative THERapies, Rouen, France.,Rouen University Hospital, Department of Pharmacy, Rouen, France
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Olivier Boyer
- Normandie University, UNIROUEN, INSERM U1234, Pathophysiology, Autoimmunity, Neuromuscular Diseases and Regenerative THERapies, Rouen, France.,Rouen University Hospital, Department of Immunology and Biotherapy, Rouen, France
| | - Sahil Adriouch
- Normandie University, UNIROUEN, INSERM U1234, Pathophysiology, Autoimmunity, Neuromuscular Diseases and Regenerative THERapies, Rouen, France
| |
Collapse
|
15
|
Parga-Vidal L, Behr FM, Kragten NAM, Nota B, Wesselink TH, Kavazović I, Covill LE, Schuller MBP, Bryceson YT, Wensveen FM, van Lier RAW, van Dam TJP, Stark R, van Gisbergen KPJM. Hobit identifies tissue-resident memory T cell precursors that are regulated by Eomes. Sci Immunol 2021; 6:6/62/eabg3533. [PMID: 34417257 DOI: 10.1126/sciimmunol.abg3533] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 07/21/2021] [Indexed: 12/13/2022]
Abstract
Tissue-resident memory CD8+ T cells (TRM) constitute a noncirculating memory T cell subset that provides early protection against reinfection. However, how TRM arise from antigen-triggered T cells has remained unclear. Exploiting the TRM-restricted expression of Hobit, we used TRM reporter/deleter mice to study TRM differentiation. We found that Hobit was up-regulated in a subset of LCMV-specific CD8+ T cells located within peripheral tissues during the effector phase of the immune response. These Hobit+ effector T cells were identified as TRM precursors, given that their depletion substantially decreased TRM development but not the formation of circulating memory T cells. Adoptive transfer experiments of Hobit+ effector T cells corroborated their biased contribution to the TRM lineage. Transcriptional profiling of Hobit+ effector T cells underlined the early establishment of TRM properties including down-regulation of tissue exit receptors and up-regulation of TRM-associated molecules. We identified Eomes as a key factor instructing the early bifurcation of circulating and resident lineages. These findings establish that commitment of TRM occurs early in antigen-driven T cell differentiation and reveal the molecular mechanisms underlying this differentiation pathway.
Collapse
Affiliation(s)
- Loreto Parga-Vidal
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.
| | - Felix M Behr
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Natasja A M Kragten
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Benjamin Nota
- Department of Molecular and Cellular Hemostasis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Thomas H Wesselink
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Inga Kavazović
- Department of Histology and Embryology, University of Rijeka, Rijeka, Croatia
| | - Laura E Covill
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Margo B P Schuller
- Department of Molecular and Cellular Hemostasis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Yenan T Bryceson
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden.,Brogelmann Research Laboratory, Department of Clinical Sciences, University of Bergen, Bergen, Norway
| | - Felix M Wensveen
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Histology and Embryology, University of Rijeka, Rijeka, Croatia
| | - Rene A W van Lier
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Teunis J P van Dam
- Department of Molecular and Cellular Hemostasis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Regina Stark
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,BIH Center for Regenerative Therapies, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands. .,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
16
|
Fernandez-Ruiz D, de Menezes MN, Holz LE, Ghilas S, Heath WR, Beattie L. Harnessing liver-resident memory T cells for protection against malaria. Expert Rev Vaccines 2021; 20:127-141. [PMID: 33501877 DOI: 10.1080/14760584.2021.1881485] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Tissue-resident memory T cells (TRM cells) are powerful mediators of protracted adaptive immunity to infection in peripheral organs. Harnessing TRM cells through vaccination hence promises unprecedented potential for protection against infection. A paramount example of this is malaria, a major infectious disease for which immunity through traditional vaccination strategies remains challenging. Liver TRM cells appear to be highly protective against malaria, and recent developments in our knowledge of the biology of these cells have defined promising, novel strategies for their induction. AREAS COVERED Here, we describe the path that led to the discovery of TRM cells and discuss the importance of liver TRM cells in immunity against Plasmodium spp. infection; we summarize current knowledge on TRM cell biology and discuss the current state and potential of TRM-based vaccination against malaria. EXPERT OPINION TRM based vaccination has emerged as a promising means to achieve efficient protection against malaria. Recent advances provide a solid basis for continuing the development of this area of research. Deeper understanding of the mechanisms that mediate TRM formation and maintenance and identification of immunogenic and protective target epitopes suitable for human vaccination remain the main challenges for translation of these discoveries.
Collapse
Affiliation(s)
- Daniel Fernandez-Ruiz
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| | - Maria N de Menezes
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia
| | - Lauren E Holz
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| | - Sonia Ghilas
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| | - William R Heath
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| | - Lynette Beattie
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| |
Collapse
|
17
|
Er-Lukowiak M, Duan Y, Rassendren F, Ulmann L, Nicke A, Ufer F, Friese MA, Koch-Nolte F, Magnus T, Rissiek B. A P2rx7 Passenger Mutation Affects the Vitality and Function of T cells in Congenic Mice. iScience 2020; 23:101870. [PMID: 33336163 PMCID: PMC7733020 DOI: 10.1016/j.isci.2020.101870] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/26/2020] [Accepted: 11/23/2020] [Indexed: 12/20/2022] Open
Abstract
Among laboratory mouse strains many genes are differentially expressed in the same cell population. As consequence, gene targeting in 129-derived embryonic stem cells (ESCs) and backcrossing the modified mice onto the C57BL/6 background can introduce passenger mutations in the close proximity of the targeted gene. Here, we demonstrate that several transgenic mice carry a P2rx7 passenger mutation that affects the function of T cells. By the example of P2rx4tm1Rass we demonstrate that P2X4ko T cells express higher levels of P2X7 and are more sensitive toward the P2X7 activators ATP and NAD+, rendering these cells more vulnerable toward NAD-induced cell death (NICD) compared with wild type (WT). The enhanced NICD sensitivity confounded functional assays e.g. cytokine production and cell migration. Our results need to be considered when working with P2rx4tm1Rass mice or other 129-based transgenic strains that target P2rx7 neighboring genes. T cells from 129 mice express higher level of P2X7 compared with T cells from B6 mice P2rx4tm1Rass T cells express high level of P2X7 due to a P2rx7 passenger mutation P2rx4tm1Rass T cells are highly susceptible to NAD-induced cell death (NICD) NICD susceptibility of P2rx4tm1Rass T cells confounds the outcome of functional assays
Collapse
Affiliation(s)
- Marco Er-Lukowiak
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg-Eppendorf, Germany
| | - Yinghui Duan
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg-Eppendorf, Germany
| | - Francois Rassendren
- IGF, Univ. Montpellier, CNRS, INSERM, 34094 Montpellier, France.,LabEx ICST, 34094 Montpellier, France
| | - Lauriane Ulmann
- IGF, Univ. Montpellier, CNRS, INSERM, 34094 Montpellier, France.,LabEx ICST, 34094 Montpellier, France
| | - Annette Nicke
- Walther Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| | - Friederike Ufer
- Institute of Neuroimmunology und Multiple Sclerosis (INIMS), University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology und Multiple Sclerosis (INIMS), University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg-Eppendorf, Germany
| | - Björn Rissiek
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg-Eppendorf, Germany
| |
Collapse
|
18
|
Li L, Zeng Z. Live Imaging of Innate and Adaptive Immune Responses in the Liver. Front Immunol 2020; 11:564768. [PMID: 33042143 PMCID: PMC7527534 DOI: 10.3389/fimmu.2020.564768] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/13/2020] [Indexed: 12/21/2022] Open
Abstract
Immune response in the liver is determined by the spatial organization and cellular dynamics of hepatic immune cells. The liver vasculature accommodates abundant tissue-resident innate immune cells, such as Kupffer cells, natural killer cells, and natural killer T cells, to ensure efficient intravascular immunosurveillance. The fenestrated sinusoids also allow direct contact between circulating T cells and non-canonical antigen-presenting cells, such as hepatocytes, to instruct adaptive immune responses. Distinct cellular behaviors are exploited by liver immune cells to exert proper functions. Intravital imaging enables real-time visualization of individual immune cell in living animals, representing a powerful tool in dissecting the spatiotemporal features of intrahepatic immune cells during steady state and liver diseases. This review summarizes current advances in liver immunology prompted by in vivo imaging, with a particular focus on liver-resident innate immune cells and hepatic T cells.
Collapse
Affiliation(s)
- Lu Li
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhutian Zeng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
19
|
Grassi F. The P2X7 Receptor as Regulator of T Cell Development and Function. Front Immunol 2020; 11:1179. [PMID: 32587592 PMCID: PMC7297980 DOI: 10.3389/fimmu.2020.01179] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/13/2020] [Indexed: 12/24/2022] Open
Abstract
Unique structural features characterize the P2X7 receptor with respect to other P2X family members. Dual gating by eATP and regulated expression of P2X7 can imprint distinct outcomes to the T cell depending on the metabolic fitness and/or developmental stage. In the thymus, signaling by P2X7 contributes to γδ T cell lineage choice. In secondary lymphoid organs, P2X7 stimulation promotes Th1/Th17 polarization of CD4+ naïve cells, Tregs conversion to Th17 cells and cell death of Tfh cells that are not stimulated by cognate antigen. Moreover, P2X7 stimulation in eATP rich microenvironments, such as damaged and/or inflamed tissues as well as tumors, induces cell death of various T cell effector subsets.
Collapse
Affiliation(s)
- Fabio Grassi
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| |
Collapse
|
20
|
Bovens AA, Wesselink TH, Behr FM, Kragten NAM, van Lier RAW, van Gisbergen KPJM, Stark R. Murine iNKT cells are depleted by liver damage via activation of P2RX7. Eur J Immunol 2020; 50:1515-1524. [PMID: 32390174 DOI: 10.1002/eji.201948509] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/21/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
Abstract
Invariant natural killer T cells (iNKT) constitute up to 50% of liver lymphocytes and contribute to immunosurveillance as well as pathogenesis of the liver. Systemic activation of iNKT cells induces acute immune-mediated liver injury. However, how tissue damage events regulate iNKT cell function and homeostasis remains unclear. We found that specifically tissue-resident iNKT cells in liver and spleen express the tissue-damage receptor P2RX7 and the P2RX7-activating ectoenzyme ARTC2. P2RX7 expression restricted formation of iNKT cells in the liver suggesting that liver iNKT cells are actively restrained under homeostatic conditions. Deliberate activation of P2RX7 in vivo by exogenous NAD resulted in a nearly complete iNKT cell ablation in liver and spleen in a P2RX7-dependent manner. Tissue damage generated by acetaminophen-induced liver injury reduced the number of iNKT cells in the liver. The tissue-damage-induced iNKT cell depletion was driven by P2RX7 and localized to the site of injury, as iNKT cells in the spleen remained intact. The depleted liver iNKT cells reconstituted only slowly compared to other lymphocytes such as regulatory T cells. These findings suggest that tissue-damage-mediated depletion of iNKT cells acts as a feedback mechanism to limit iNKT cell-induced pathology resulting in the establishment of a tolerogenic environment.
Collapse
Affiliation(s)
- Astrid A Bovens
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Thomas H Wesselink
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Felix M Behr
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Natasja A M Kragten
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - René A W van Lier
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Regina Stark
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, BIH Center for Regenerative Therapies, Berlin, Germany
| |
Collapse
|
21
|
Schwarz N, Junge M, Haag F, Koch-Nolte F. Flow Cytometry of Membrane Purinoreceptors. Methods Mol Biol 2020; 2041:117-136. [PMID: 31646484 DOI: 10.1007/978-1-4939-9717-6_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Mammalian purinoreceptors respond to extracellular nucleotides and their metabolites, for example, following the release of ATP or NAD+ from cells and their hydrolysis by ectonucleotidases. Membrane purinoreceptors are expressed as ionotropic ligand-gated ion channels designated P2X receptors, or as metabotropic G-protein coupled receptors designated P1 or P2Y receptors, on the cell surface of different cell types. In this chapter, we provide protocols to monitor the expression and activity of purinoreceptors on the cell membrane of living cells by flow cytometry.
Collapse
Affiliation(s)
- Nicole Schwarz
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marten Junge
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
22
|
Stark R, Wesselink TH, Behr FM, Kragten NAM, Arens R, Koch-Nolte F, van Gisbergen KPJM, van Lier RAW. T RM maintenance is regulated by tissue damage via P2RX7. Sci Immunol 2019; 3:3/30/eaau1022. [PMID: 30552101 DOI: 10.1126/sciimmunol.aau1022] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 11/13/2018] [Indexed: 12/16/2022]
Abstract
Tissue-resident memory T cells (TRM) are noncirculating immune cells that contribute to the first line of local defense against reinfections. Their location at hotspots of pathogen encounter frequently exposes TRM to tissue damage. This history of danger-signal exposure is an important aspect of TRM-mediated immunity that has been overlooked so far. RNA profiling revealed that TRM from liver and small intestine express P2RX7, a damage/danger-associated molecular pattern (DAMP) receptor that is triggered by extracellular nucleotides (ATP, NAD+). We confirmed that P2RX7 protein was expressed in CD8+ TRM but not in circulating T cells (TCIRC) across different infection models. Tissue damage induced during routine isolation of liver lymphocytes led to P2RX7 activation and resulted in selective cell death of TRM P2RX7 activation in vivo by exogenous NAD+ led to a specific depletion of TRM while retaining TCIRC The effect was absent in P2RX7-deficient mice and after P2RX7 blockade. TCR triggering down-regulated P2RX7 expression and made TRM resistant to NAD-induced cell death. Physiological triggering of P2RX7 by sterile tissue damage during acetaminophen-induced liver injury led to a loss of previously acquired pathogen-specific local TRM in wild-type but not in P2RX7 KO T cells. Our results highlight P2RX7-mediated signaling as a critical pathway for the regulation of TRM maintenance. Extracellular nucleotides released during infection and tissue damage could deplete TRM locally and free niches for new and infection-relevant specificities. This suggests that the recognition of tissue damage promotes persistence of antigen-specific over bystander TRM in the tissue niche.
Collapse
Affiliation(s)
- Regina Stark
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands. .,Department of Experimental Immunology, Amsterdam UMC, Amsterdam, Netherlands
| | - Thomas H Wesselink
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Felix M Behr
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Immunology, Amsterdam UMC, Amsterdam, Netherlands
| | - Natasja A M Kragten
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Immunology, Amsterdam UMC, Amsterdam, Netherlands
| | - René A W van Lier
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Immunology, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
23
|
Liu Q, Kim CH. Control of Tissue-Resident Invariant NKT Cells by Vitamin A Metabolites and P2X7-Mediated Cell Death. THE JOURNAL OF IMMUNOLOGY 2019; 203:1189-1197. [PMID: 31308092 DOI: 10.4049/jimmunol.1900398] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 06/21/2019] [Indexed: 12/11/2022]
Abstract
Invariant NKT (iNKT) cells provide rapid innate T cell responses to glycolipid Ags from host cells and microbes. The numbers of CD1d-restricted iNKT cells are tightly controlled in mucosal tissues, but the mechanisms have been largely unclear. We found that vitamin A is a dominant factor that controls the population size of mucosal iNKT cells in mice. This negative regulation is mediated by the induction of the purinergic receptor P2X7 on iNKT cells. The expression of P2X7 is particularly high on intestinal iNKT cells, making iNKT cells highly susceptible to P2X7-mediated cell death. In vitamin A deficiency, iNKT cells fail to express P2X7 and are, therefore, resistant to P2X7-mediated cell death, leading to iNKT cell overpopulation. This phenomenon is most prominent in the intestine. We found that iNKT cells are divided into CD69+ sphingosine-1-phosphate receptor 1 (S1P1)- tissue resident and CD69- S1P1+ nonresident iNKT cells. The CD69+ S1P1- tissue-resident iNKT cells highly express P2X7 and are effectively controlled by the P2X7 pathway. The regulation of iNKT cells by vitamin A by the P2X7 pathway is important to prevent aberrant expansion of effector cytokine-producing iNKT cells. Our findings identify a novel role of vitamin A in regulating iNKT cell homeostasis in many tissues throughout the body.
Collapse
Affiliation(s)
- Qingyang Liu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109; Mary H. Weiser Food Allergy Center, University of Michigan School of Medicine, Ann Arbor, MI 48109; and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI 48109
| | - Chang H Kim
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109; Mary H. Weiser Food Allergy Center, University of Michigan School of Medicine, Ann Arbor, MI 48109; and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI 48109
| |
Collapse
|
24
|
Lalfer M, Chappert P, Carpentier M, Urbain D, Davoust JM, Gross DA. Foxp3 + Regulatory and Conventional CD4 + T Cells Display Similarly High Frequencies of Alloantigen-Reactive Cells. Front Immunol 2019; 10:521. [PMID: 30941146 PMCID: PMC6434998 DOI: 10.3389/fimmu.2019.00521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 02/26/2019] [Indexed: 01/22/2023] Open
Abstract
Foxp3+ regulatory T cells (Tregs) play a major role in acquired immune tolerance to allogenic transplants. Their suppressive activity is thought to require T cell receptor (TCR)-driven antigen recognition; little, however, is known about the fraction of Tregs able to recognize alloantigens within this T cell subset primarily educated against self-antigens. Performing transfer experiments of Tregs or conventional T cells (Tconv) into both lymphoreplete and lymphopenic mice, we observed a similarly high proportion of cells signaling through their TCR and proliferating in allogenic hosts. Furthermore, using an in vivo proliferation assay with limited T cell numbers infused into lymphopenic mice, we found that the overall frequency of alloreactive Tregs was similar if not higher to that of alloreactive Tconv. Overall our study highlights a noticeably high level of alloreactive Foxp3+ regulatory T cells accounting for their predominant role in transplantation tolerance.
Collapse
Affiliation(s)
- Mélanie Lalfer
- Institut National de la Santé et de la Recherche Médicale U1151 - Centre National de la Recherche Scientifique UMR 8253, Institut Necker-Enfants Malades, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Pascal Chappert
- Institut National de la Santé et de la Recherche Médicale U1151 - Centre National de la Recherche Scientifique UMR 8253, Institut Necker-Enfants Malades, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maxime Carpentier
- Institut National de la Santé et de la Recherche Médicale U1151 - Centre National de la Recherche Scientifique UMR 8253, Institut Necker-Enfants Malades, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Dominique Urbain
- Institut National de la Santé et de la Recherche Médicale U1151 - Centre National de la Recherche Scientifique UMR 8253, Institut Necker-Enfants Malades, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jean M Davoust
- Institut National de la Santé et de la Recherche Médicale U1151 - Centre National de la Recherche Scientifique UMR 8253, Institut Necker-Enfants Malades, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - David-Alexandre Gross
- Institut National de la Santé et de la Recherche Médicale U1151 - Centre National de la Recherche Scientifique UMR 8253, Institut Necker-Enfants Malades, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
25
|
Monitoring the Sensitivity of T Cell Populations Towards NAD + Released During Cell Preparation. Methods Mol Biol 2019; 1813:317-326. [PMID: 30097878 DOI: 10.1007/978-1-4939-8588-3_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Mouse T cells express the toxin-related ecto-ADP-ribosyltransferase ARTC2 that catalyzes the posttranslational ADP-ribosylation of cell surface proteins by transferring the ADP-ribose group of its substrate nicotinamide adenine dinucleotide (NAD+) to arginine residues of its target proteins. One well known target of ARTC2 is the ATP-gated P2X7 ion channel. ADP-ribosylation of P2X7 induces gating of the channel, calcium influx, ecto-domain shedding, phosphatidylserine externalization, and finally cell death. Previous studies have shown that the ARTC2 substrate NAD+ is released during T cell preparation. Since P2X7 is differentially expressed among T cell subpopulations, preparation-related ADP-ribosylation has a strong impact on the vitality of T cells that express high levels of P2X7. With this chapter we provide a protocol to monitor the consequences of preparation-related P2X7 ADP-ribosylation on T cells using regulatory T cells as generic T cell subpopulation known to express high levels of P2X7. However, this protocol could be easily adapted to other T cell populations.
Collapse
|
26
|
Borges da Silva H, Wang H, Qian LJ, Hogquist KA, Jameson SC. ARTC2.2/P2RX7 Signaling during Cell Isolation Distorts Function and Quantification of Tissue-Resident CD8 + T Cell and Invariant NKT Subsets. THE JOURNAL OF IMMUNOLOGY 2019; 202:2153-2163. [PMID: 30777922 DOI: 10.4049/jimmunol.1801613] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 01/22/2019] [Indexed: 12/19/2022]
Abstract
Peripheral invariant NKT cells (iNKT) and CD8+ tissue-resident memory T cells (TRM) express high levels of the extracellular ATP receptor P2RX7 in mice. High extracellular ATP concentrations or NAD-mediated P2RX7 ribosylation by the enzyme ARTC2.2 can induce P2RX7 pore formation and cell death. Because both ATP and NAD are released during tissue preparation for analysis, cell death through these pathways may compromise the analysis of iNKT and CD8+ TRM Indeed, ARTC2.2 blockade enhanced recovery of viable liver iNKT and TRM The expression of ARTC2.2 and P2RX7 on distinct iNKT subsets and TRM is unclear, however, as is the impact of recovery from other nonlymphoid sites. In this study, we performed a comprehensive analysis of ARTC2.2 and P2RX7 expression in iNKT and CD8+ T cells in diverse tissues, at steady-state and after viral infection. NKT1 cells and CD8+ TRM express high levels of both ARTC2.2 and P2RX7 compared with NKT2, NKT17, and CD8+ circulating memory subsets. Using nanobody-mediated ARTC2.2 antagonism, we showed that ARTC2.2 blockade enhanced NKT1 and TRM recovery from nonlymphoid tissues during cell preparation. Moreover, blockade of this pathway was essential to preserve functionality, viability, and proliferation of both populations. We also showed that short-term direct P2RX7 blockade enhanced recovery of TRM, although to a lesser degree. In summary, our data show that short-term in vivo blockade of the ARTC2.2/P2RX7 axis permits much improved flow cytometry-based phenotyping and enumeration of murine iNKT and TRM from nonlymphoid tissues, and it represents a crucial step for functional studies of these populations.
Collapse
Affiliation(s)
- Henrique Borges da Silva
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Haiguang Wang
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Lily J Qian
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Kristin A Hogquist
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Stephen C Jameson
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
27
|
Linden J, Koch-Nolte F, Dahl G. Purine Release, Metabolism, and Signaling in the Inflammatory Response. Annu Rev Immunol 2019; 37:325-347. [PMID: 30676821 DOI: 10.1146/annurev-immunol-051116-052406] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
ATP, NAD+, and nucleic acids are abundant purines that, in addition to having critical intracellular functions, have evolved extracellular roles as danger signals released in response to cell lysis, apoptosis, degranulation, or membrane pore formation. In general ATP and NAD+ have excitatory and adenosine has anti-inflammatory effects on immune cells. This review focuses on recent advances in our understanding of purine release mechanisms, ectoenzymes that metabolize purines (CD38, CD39, CD73, ENPP1, and ENPP2/autotaxin), and signaling by key P2 purinergic receptors (P2X7, P2Y2, and P2Y12). In addition to metabolizing ATP or NAD+, some purinergic ectoenzymes metabolize other inflammatory modulators, notably lysophosphatidic acid and cyclic GMP-AMP (cGAMP). Also discussed are extracellular signaling effects of NAD+ mediated by ADP-ribosylation, and epigenetic effects of intracellular adenosine mediated by modification of S-adenosylmethionine-dependent DNA methylation.
Collapse
Affiliation(s)
- Joel Linden
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, California 92037, USA; .,Department of Pharmacology, University of California, San Diego, La Jolla, California 92093, USA
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany;
| | - Gerhard Dahl
- Department of Physiology and Biophysics, University of Miami School of Medicine, Miami, Florida 33136, USA;
| |
Collapse
|
28
|
Janks L, Sprague RS, Egan TM. ATP-Gated P2X7 Receptors Require Chloride Channels To Promote Inflammation in Human Macrophages. THE JOURNAL OF IMMUNOLOGY 2018; 202:883-898. [PMID: 30598517 DOI: 10.4049/jimmunol.1801101] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/25/2018] [Indexed: 12/28/2022]
Abstract
Immune cells of myeloid origin show robust expression of ATP-gated P2X7 receptors, two-transmembrane ion channels permeable to Na+, K+, and Ca2+ Receptor activation promotes inflammasome activation and release of the proinflammatory cytokines IL-1β and IL-18. In this study, we show that ATP generates facilitating cationic currents in monocyte-derived human macrophages and permeabilizes the plasma membrane to polyatomic cationic dyes. We find that antagonists of PLA2 and Cl- channels abolish P2X7 receptor-mediated current facilitation, membrane permeabilization, blebbing, phospholipid scrambling, inflammasome activation, and IL-1β release. Our data demonstrate significant differences in the actions of ATP in murine and human macrophages and suggest that PLA2 and Cl- channels mediate innate immunity downstream of P2X7 receptors in human macrophages.
Collapse
Affiliation(s)
- Laura Janks
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Randy S Sprague
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Terrance M Egan
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104
| |
Collapse
|
29
|
Regulation of P2X7 receptor expression and function in the brain. Brain Res Bull 2018; 151:153-163. [PMID: 30593878 DOI: 10.1016/j.brainresbull.2018.12.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/19/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023]
Abstract
Because of its prominent role in driving inflammatory processes, the ATP-gated purinergic P2X7 receptor has attracted much attention over the past decade as a potential therapeutic target for numerous human conditions, particularly diseases of the central nervous system, including neurodegenerative diseases (e.g. Alzheimer's and Huntington's disease), psychiatric disorders (e.g. schizophrenia and depression) and the neurological disease, epilepsy. Evidence stems from studies using experimental models and patient tissue showing changes in P2X7 expression and function under pathological conditions and beneficial effects provided by P2X7 antagonism. Apart from promoting neuroinflammation, P2X7, however, also impacts on other pathological processes in the brain, including cell death, hyperexcitability, changes in neurotransmitter release and neurogenesis. Reports also suggest a role for P2X7 in the maintenance of blood-brain-barrier integrity. It therefore comes as no surprise that the regulation of P2X7 expression and function is complex, providing tight control on P2X7 activation. Much progress has been made in understanding how P2X7 is regulated during physiological and pathological conditions and what the consequences are of pathological P2X7 expression and function. Regulatory mechanisms altering P2X7 expression include transcriptional and post-translational regulation including nucleotide polymorphisms, promoter regulation via DNA methylation, transcription factors (e.g. Sp1 and HIF-1α), the generation of different splice variants and receptor phosphorylation, glycosylation and palmitoylation. Finally, more recently, reports have also shown P2X7-targeting by microRNAs, blocking P2X7 translation into functional proteins. The present review provides a broad overview of what is known to-date about the complex regulation of P2X7 expression with a particular emphasis on the brain and how each of these regulatory mechanisms impacts on receptor function and pathology.
Collapse
|
30
|
Optimal protection against Salmonella infection requires noncirculating memory. Proc Natl Acad Sci U S A 2018; 115:10416-10421. [PMID: 30254173 DOI: 10.1073/pnas.1808339115] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
While CD4 Th1 cells are required for resistance to intramacrophage infections, adoptive transfer of Th1 cells is insufficient to protect against Salmonella infection. Using an epitope-tagged vaccine strain of Salmonella, we found that effective protection correlated with expanded Salmonella-specific memory CD4 T cells in circulation and nonlymphoid tissues. However, naive mice that previously shared a blood supply with vaccinated partners lacked T cell memory with characteristics of tissue residence and did not acquire robust protective immunity. Using a YFP-IFN-γ reporter system, we identified Th1 cells in the liver of immunized mice that displayed markers of tissue residence, including P2X7, ARTC2, LFA-1, and CD101. Adoptive transfer of liver memory cells after ARTC2 blockade increased protection against highly virulent bacteria. Taken together, these data demonstrate that noncirculating memory Th1 cells are a vital component of immunity to Salmonella infection and should be the focus of vaccine strategies.
Collapse
|
31
|
Othman AS, Franke-Fayard BM, Imai T, van der Gracht ETI, Redeker A, Salman AM, Marin-Mogollon C, Ramesar J, Chevalley-Maurel S, Janse CJ, Arens R, Khan SM. OX40 Stimulation Enhances Protective Immune Responses Induced After Vaccination With Attenuated Malaria Parasites. Front Cell Infect Microbiol 2018; 8:247. [PMID: 30073152 PMCID: PMC6060232 DOI: 10.3389/fcimb.2018.00247] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/28/2018] [Indexed: 01/15/2023] Open
Abstract
Protection against a malaria infection can be achieved by immunization with live-attenuated Plasmodium sporozoites and while the precise mechanisms of protection remain unknown, T cell responses are thought to be critical in the elimination of infected liver cells. In cancer immunotherapies, agonistic antibodies that target T cell surface proteins, such as CD27, OX40 (CD134), and 4-1BB (CD137), have been used to enhance T cell function by increasing co-stimulation. In this study, we have analyzed the effect of agonistic OX40 monoclonal antibody treatment on protective immunity induced in mice immunized with genetically attenuated parasites (GAPs). OX40 stimulation enhanced protective immunity after vaccination as shown by an increase in the number of protected mice and delay to blood-stage infection after challenge with wild-type sporozoites. Consistent with the enhanced protective immunity enforced OX40 stimulation resulted in an increased expansion of antigen-experienced effector (CD11ahiCD44hi) CD8+ and CD4+ T cells in the liver and spleen and also increased IFN-γ and TNF producing CD4+ T cells in the liver and spleen. In addition, GAP immunization plus α-OX40 treatment significantly increased sporozoite-specific IgG responses. Thus, we demonstrate that targeting T cell costimulatory receptors can improve sporozoite-based vaccine efficacy.
Collapse
Affiliation(s)
- Ahmad Syibli Othman
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands.,Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Terengganu, Malaysia
| | - Blandine M Franke-Fayard
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Takashi Imai
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Esmé T I van der Gracht
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Anke Redeker
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Ahmed M Salman
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands.,The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Catherin Marin-Mogollon
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Jai Ramesar
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Chris J Janse
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Shahid M Khan
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
32
|
Rissiek B, Lukowiak M, Raczkowski F, Magnus T, Mittrücker HW, Koch-Nolte F. In Vivo Blockade of Murine ARTC2.2 During Cell Preparation Preserves the Vitality and Function of Liver Tissue-Resident Memory T Cells. Front Immunol 2018; 9:1580. [PMID: 30038627 PMCID: PMC6046629 DOI: 10.3389/fimmu.2018.01580] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/26/2018] [Indexed: 01/13/2023] Open
Abstract
On murine T cells, GPI-anchored ADP-ribosyltransferase 2.2 (ARTC2.2) ADP-ribosylates the P2X7 ion channel at arginine 125 in response to nicotinamide adenine dinucleotide (NAD+) released during cell preparation. We have previously shown that chronic gating of P2X7 by ADP-ribosylation reduces the vitality and function of regulatory T cells and natural killer T cells that co-express high levels of ARTC2.2 and P2X7. Here, we evaluated the expression of ARTC2.2 and P2X7 by effector and memory T cells in the liver of naïve mice and after infection with Listeria monocytogenes (Lm). We found that KLRG1−/CD69+ tissue-resident memory T cells (Trm) in the liver of naïve mice and 7 weeks after infection with Lm express high levels of ARTC2.2 and P2X7. Isolation of liver Trm and subsequent incubation at 37°C resulted in cell death of the majority of CD4+ and CD8+ Trm. Injection of the ARTC2.2-blocking nanobody s+16a 30 min prior to organ harvesting effectively prevented ADP-ribosylation of P2X7 during cell preparation and thereby prevented NAD-induced cell death of the isolated Trm upon subsequent incubation at 37°C. Consequently, preserving Trm vitality by s+16a injection enabled a highly sensitive in vitro cytokine expression profile analyses of FACS sorted liver Trm. We conclude that in vivo blockade of ARTC2.2 during cell preparation by nanobody s+16a injection represents a valuable strategy to study the role and function of liver Trm in mice.
Collapse
Affiliation(s)
- Björn Rissiek
- Department of Neurology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Marco Lukowiak
- Department of Neurology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Friederike Raczkowski
- Institute of Immunology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Willi Mittrücker
- Institute of Immunology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
33
|
Menzel S, Schwarz N, Haag F, Koch-Nolte F. Nanobody-Based Biologics for Modulating Purinergic Signaling in Inflammation and Immunity. Front Pharmacol 2018; 9:266. [PMID: 29636685 PMCID: PMC5880931 DOI: 10.3389/fphar.2018.00266] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 03/09/2018] [Indexed: 12/18/2022] Open
Abstract
Adenosine triphosphate (ATP) and nicotinamide adenine dinucleotide (NAD+) are released as danger signals from cells during infection and sterile inflammation. In the extracellular compartment ATP is converted by CD39, CD73, and other ecto-enzymes into metabolites that modulate the activity of T cells and macrophages. While ATP mediates pro-inflammatory signals via P2X7 and other P2 receptors, adenosine triggers anti-inflammatory signaling via the adenosine 2a receptor (Adora2a) and other P1 receptors. The latter also plays a role in maintaining an immunosuppressive tumor microenvironment. NAD+ is converted by CD38, CD203 and other ecto-enzymes to the Ca2+ mobilizing messengers cyclic ADP-ribose and ADP-ribose, and to adenosine. Recent findings on the roles of CD38, CD39, CD73, CD203, P2X7, and Adora2a in inflammation and immunity underscore the potential of these proteins as drug targets. However, available small molecule inhibitors often lack specificity and mediate unwanted off-target toxicity. Nanobodies – single domain antibodies derived from heavy chain antibodies that naturally occur in camelids – display a propensity to bind functional epitopes not accessible to conventional antibodies. Like conventional antibodies, nanobodies and nanobody-based biologics are highly specific and have well-understood, tunable in vivo pharmacodynamics with little if any toxicity. Nanobodies thus represent attractive alternatives to small molecule inhibitors for modulating purinergic signaling in inflammation and immunity. Here we review recent progress made in developing nanobodies against key targets of purinergic signaling.
Collapse
Affiliation(s)
- Stephan Menzel
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicole Schwarz
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
34
|
Georgiev H, Ravens I, Papadogianni G, Malissen B, Förster R, Bernhardt G. Blocking the ART2.2/P2X7-system is essential to avoid a detrimental bias in functional CD4 T cell studies. Eur J Immunol 2018; 48:1078-1081. [PMID: 29508376 DOI: 10.1002/eji.201747420] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/06/2018] [Accepted: 02/27/2018] [Indexed: 11/10/2022]
Abstract
Murine T cell subsets differ in their expression level of P2X7. Depending on several parameters like extracellular NAD+ , P2X7 can be ADP-ribosylated rapidly by adjacent ARTC2.2 resulting in susceptibilities to apoptosis to a varying extent. This detrimental effect can be prevented when drugs like KN-62 are present during cell preparations.
Collapse
Affiliation(s)
- Hristo Georgiev
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Inga Ravens
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | | | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Günter Bernhardt
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
35
|
Menzel S, Adriouch S, Bannas P, Haag F, Koch-Nolte F. Monitoring Expression and Enzyme Activity of Ecto-ARTCs. Methods Mol Biol 2018; 1813:167-186. [PMID: 30097867 DOI: 10.1007/978-1-4939-8588-3_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mammalian ARTCs are expressed as glycosylphosphatidylinositol (GPI)-anchored ectoenzymes (ARTC1-ARTC4) or secretory proteins (ARTC5) by different cell types. The ARTC2 enzymes catalyze mono-ADP-ribosylation of arginine residues in the extracellular domain of membrane proteins or secretory proteins. In this chapter we provide protocols to monitor the expression and activity of ARTCs on the cell membrane of living cells and in soluble form in biological fluids.
Collapse
Affiliation(s)
- Stephan Menzel
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sahil Adriouch
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Faculty of Medicine, University of Rouen, Rouen, France
| | - Peter Bannas
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
36
|
Danquah W, Meyer-Schwesinger C, Rissiek B, Pinto C, Serracant-Prat A, Amadi M, Iacenda D, Knop JH, Hammel A, Bergmann P, Schwarz N, Assunção J, Rotthier W, Haag F, Tolosa E, Bannas P, Boué-Grabot E, Magnus T, Laeremans T, Stortelers C, Koch-Nolte F. Nanobodies that block gating of the P2X7 ion channel ameliorate inflammation. Sci Transl Med 2017; 8:366ra162. [PMID: 27881823 DOI: 10.1126/scitranslmed.aaf8463] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 04/11/2016] [Accepted: 10/27/2016] [Indexed: 12/17/2022]
Abstract
Ion channels are desirable therapeutic targets, yet ion channel-directed drugs with high selectivity and few side effects are still needed. Unlike small-molecule inhibitors, antibodies are highly selective for target antigens but mostly fail to antagonize ion channel functions. Nanobodies-small, single-domain antibody fragments-may overcome these problems. P2X7 is a ligand-gated ion channel that, upon sensing adenosine 5'-triphosphate released by damaged cells, initiates a proinflammatory signaling cascade, including release of cytokines, such as interleukin-1β (IL-1β). To further explore its function, we generated and characterized nanobodies against mouse P2X7 that effectively blocked (13A7) or potentiated (14D5) gating of the channel. Systemic injection of nanobody 13A7 in mice blocked P2X7 on T cells and macrophages in vivo and ameliorated experimental glomerulonephritis and allergic contact dermatitis. We also generated nanobody Dano1, which specifically inhibited human P2X7. In endotoxin-treated human blood, Dano1 was 1000 times more potent in preventing IL-1β release than small-molecule P2X7 antagonists currently in clinical development. Our results show that nanobody technology can generate potent, specific therapeutics against ion channels, confirm P2X7 as a therapeutic target for inflammatory disorders, and characterize a potent new drug candidate that targets P2X7.
Collapse
Affiliation(s)
- Welbeck Danquah
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Catherine Meyer-Schwesinger
- Department of Nephrology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Björn Rissiek
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.,Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Carolina Pinto
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Arnau Serracant-Prat
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Miriam Amadi
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Domenica Iacenda
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.,Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Jan-Hendrik Knop
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.,Department of Nephrology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Anna Hammel
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.,Department of Nephrology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Philine Bergmann
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.,Université de Bordeaux, Institut des Maladies Neurodégénératives, CNRS UMR 5293, Bordeaux 33076, France
| | - Nicole Schwarz
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Joana Assunção
- Ablynx NV, Technologiepark 21, B-9052 Zwijnaarde, Belgium
| | - Wendy Rotthier
- Ablynx NV, Technologiepark 21, B-9052 Zwijnaarde, Belgium
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Eva Tolosa
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Peter Bannas
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.,Department of Radiology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Eric Boué-Grabot
- Université de Bordeaux, Institut des Maladies Neurodégénératives, CNRS UMR 5293, Bordeaux 33076, France
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Toon Laeremans
- Ablynx NV, Technologiepark 21, B-9052 Zwijnaarde, Belgium
| | | | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.
| |
Collapse
|
37
|
Sag D, Özkan M, Kronenberg M, Wingender G. Improved Detection of Cytokines Produced by Invariant NKT Cells. Sci Rep 2017; 7:16607. [PMID: 29192280 PMCID: PMC5709402 DOI: 10.1038/s41598-017-16832-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/16/2017] [Indexed: 12/01/2022] Open
Abstract
Invariant Natural killer T (iNKT) cells rapidly produce copious amounts of multiple cytokines after in vivo activation, allowing for the direct detection of a number of cytokines directly ex vivo. However, for some cytokines this approach is suboptimal. Here, we report technical variations that allow the improved detection of IL-4, IL-10, IL-13 and IL-17A ex vivo. Furthermore, we describe an alternative approach for stimulation of iNKT cells in vitro that allows a significantly improved detection of cytokines produced by iNKT cells. Together, these protocols allow the detection of iNKT cell cytokines ex vivo and in vitro with increased sensitivity.
Collapse
Affiliation(s)
- Duygu Sag
- Izmir Biomedicine and Genome Center (IBG), 35340, Balcova, Izmir, Turkey.,Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, 35340, Balcova, Izmir, Turkey
| | - Müge Özkan
- Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, 35340, Balcova, Izmir, Turkey
| | - Mitchell Kronenberg
- La Jolla Institute for Allergy and Immunology (LJI), 9420 Athena Circle, La Jolla, CA, 92037, USA.,Division of Biological Sciences, University of California San Diego, La Jolla, CA, 92037, USA
| | - Gerhard Wingender
- Izmir Biomedicine and Genome Center (IBG), 35340, Balcova, Izmir, Turkey. .,La Jolla Institute for Allergy and Immunology (LJI), 9420 Athena Circle, La Jolla, CA, 92037, USA.
| |
Collapse
|
38
|
Shissler SC, Lee MS, Webb TJ. Mixed Signals: Co-Stimulation in Invariant Natural Killer T Cell-Mediated Cancer Immunotherapy. Front Immunol 2017; 8:1447. [PMID: 29163518 PMCID: PMC5671952 DOI: 10.3389/fimmu.2017.01447] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 10/17/2017] [Indexed: 12/31/2022] Open
Abstract
Invariant natural killer T (iNKT) cells are an integral component of the immune system and play an important role in antitumor immunity. Upon activation, iNKT cells can directly kill malignant cells as well as rapidly produce cytokines that stimulate other immune cells, making them a front line defense against tumorigenesis. Unfortunately, iNKT cell number and activity are reduced in multiple cancer types. This anergy is often associated with upregulation of co-inhibitory markers such as programmed death-1. Similar to conventional T cells, iNKT cells are influenced by the conditions of their activation. Conventional T cells receive signals through the following three types of receptors: (1) T cell receptor (TCR), (2) co-stimulation molecules, and (3) cytokine receptors. Unlike conventional T cells, which recognize peptide antigen presented by MHC class I or II, the TCRs of iNKT cells recognize lipid antigen in the context of the antigen presentation molecule CD1d (Signal 1). Co-stimulatory molecules can positively and negatively influence iNKT cell activation and function and skew the immune response (Signal 2). This study will review the background of iNKT cells and their co-stimulatory requirements for general function and in antitumor immunity. We will explore the impact of monoclonal antibody administration for both blocking inhibitory pathways and engaging stimulatory pathways on iNKT cell-mediated antitumor immunity. This review will highlight the incorporation of co-stimulatory molecules in antitumor dendritic cell vaccine strategies. The use of co-stimulatory intracellular signaling domains in chimeric antigen receptor-iNKT therapy will be assessed. Finally, we will explore the influence of innate-like receptors and modification of immunosuppressive cytokines (Signal 3) on cancer immunotherapy.
Collapse
Affiliation(s)
- Susannah C Shissler
- Department of Microbiology and Immunology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Michael S Lee
- Department of Microbiology and Immunology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tonya J Webb
- Department of Microbiology and Immunology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
39
|
Di Virgilio F, Dal Ben D, Sarti AC, Giuliani AL, Falzoni S. The P2X7 Receptor in Infection and Inflammation. Immunity 2017; 47:15-31. [PMID: 28723547 DOI: 10.1016/j.immuni.2017.06.020] [Citation(s) in RCA: 837] [Impact Index Per Article: 104.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/14/2017] [Accepted: 06/27/2017] [Indexed: 12/11/2022]
Abstract
Adenosine triphosphate (ATP) accumulates at sites of tissue injury and inflammation. Effects of extracellular ATP are mediated by plasma membrane receptors named P2 receptors (P2Rs). The P2R most involved in inflammation and immunity is the P2X7 receptor (P2X7R), expressed by virtually all cells of innate and adaptive immunity. P2X7R mediates NLRP3 inflammasome activation, cytokine and chemokine release, T lymphocyte survival and differentiation, transcription factor activation, and cell death. Ten human P2RX7 gene splice variants and several SNPs that produce complex haplotypes are known. The P2X7R is a potent stimulant of inflammation and immunity and a promoter of cancer cell growth. This makes P2X7R an appealing target for anti-inflammatory and anti-cancer therapy. However, an in-depth knowledge of its structure and of the associated signal transduction mechanisms is needed for an effective therapeutic development.
Collapse
Affiliation(s)
- Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.
| | - Diego Dal Ben
- School of Pharmacy, University of Camerino, Camerino, Italy
| | - Alba Clara Sarti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Anna Lisa Giuliani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Simonetta Falzoni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
40
|
Pupovac A, Sluyter R. Roles of extracellular nucleotides and P2 receptors in ectodomain shedding. Cell Mol Life Sci 2016; 73:4159-4173. [PMID: 27180276 PMCID: PMC11108277 DOI: 10.1007/s00018-016-2274-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/10/2016] [Indexed: 02/03/2023]
Abstract
Ectodomain shedding of integral membrane receptors results in the release of soluble molecules and modification of the transmembrane portions to mediate or modulate extracellular and intracellular signalling. Ectodomain shedding is stimulated by a variety of mechanisms, including the activation of P2 receptors by extracellular nucleotides. This review describes in detail the roles of extracellular nucleotides and P2 receptors in the shedding of various cell surface molecules, including amyloid precursor protein, CD23, CD62L, and members of the epidermal growth factor, immunoglobulin and tumour necrosis factor families. This review discusses the mechanisms involved in P2 receptor-mediated shedding, demonstrating central roles for the P2 receptors, P2X7 and P2Y2, and the sheddases, ADAM10 and ADAM17, in this process in a number of cell types.
Collapse
Affiliation(s)
- Aleta Pupovac
- School of Biological Sciences, University of Wollongong, Wollongong, NSW, 2522, Australia
- Centre for Medical and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Ronald Sluyter
- School of Biological Sciences, University of Wollongong, Wollongong, NSW, 2522, Australia.
- Centre for Medical and Molecular Bioscience, University of Wollongong, Wollongong, NSW, 2522, Australia.
- Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
41
|
Liver-Resident Memory CD8 + T Cells Form a Front-Line Defense against Malaria Liver-Stage Infection. Immunity 2016; 45:889-902. [DOI: 10.1016/j.immuni.2016.08.011] [Citation(s) in RCA: 264] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 06/21/2016] [Accepted: 07/07/2016] [Indexed: 01/10/2023]
|
42
|
Steeland S, Vandenbroucke RE, Libert C. Nanobodies as therapeutics: big opportunities for small antibodies. Drug Discov Today 2016; 21:1076-113. [DOI: 10.1016/j.drudis.2016.04.003] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 02/26/2016] [Accepted: 04/04/2016] [Indexed: 12/28/2022]
|
43
|
Rissiek B, Haag F, Boyer O, Koch-Nolte F, Adriouch S. P2X7 on Mouse T Cells: One Channel, Many Functions. Front Immunol 2015; 6:204. [PMID: 26042119 PMCID: PMC4436801 DOI: 10.3389/fimmu.2015.00204] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 04/14/2015] [Indexed: 12/12/2022] Open
Abstract
The P2X7 receptor is an adenosine triphosphate (ATP)-gated cation channel that is expressed by several cells of the immune system. P2X7 is best known for its proinflammatory role in promoting inflammasome formation and release of mature interleukin (IL)-1β by innate immune cells. Mounting evidence indicates that P2X7 is also an important regulatory receptor of murine and human T cell functions. Murine T cells express a sensitive splice variant of P2X7 that can be activated either by non-covalent binding of ATP or, in the presence of nicotinamide adenine dinucleotide, by its covalent ADP-ribosylation catalyzed by the ecto-ADP-ribosyltransferase ARTC2.2. Prolonged activation of P2X7 by either one of these pathways triggers the induction of T cell death. Conversely, lower concentrations of ATP can activate P2X7 to enhance T cell proliferation and production of IL-2. In this review, we will highlight the molecular and cellular consequences of P2X7 activation on mouse T cells and its versatile role in T cell homeostasis and activation. Further, we will discuss important differences in the function of P2X7 on human and murine T cells.
Collapse
Affiliation(s)
- Björn Rissiek
- Insitute of Immunology, University Medical Center , Hamburg , Germany ; Department of Neurology, University Medical Center , Hamburg , Germany
| | - Friedrich Haag
- Insitute of Immunology, University Medical Center , Hamburg , Germany
| | - Olivier Boyer
- U905, INSERM , Rouen , France ; Institute for Research and Innovation in Biomedicine (IRIB), Normandy University , Rouen , France ; Department of Immunology, Rouen University Hospital , Rouen , France
| | | | - Sahil Adriouch
- U905, INSERM , Rouen , France ; Institute for Research and Innovation in Biomedicine (IRIB), Normandy University , Rouen , France
| |
Collapse
|
44
|
Teege S, Hann A, Miksiewicz M, MacMillan C, Rissiek B, Buck F, Menzel S, Nissen M, Bannas P, Haag F, Boyer O, Seman M, Adriouch S, Koch-Nolte F. Tuning IL-2 signaling by ADP-ribosylation of CD25. Sci Rep 2015; 5:8959. [PMID: 25753532 PMCID: PMC4354014 DOI: 10.1038/srep08959] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 02/12/2015] [Indexed: 01/18/2023] Open
Abstract
Control of immunologic tolerance and homeostasis rely on Foxp3+CD4+CD25+ regulatory T cells (Tregs) that constitutively express the high affinity receptor for Interleukin-2, CD25. Tregs proliferate in response to injections of IL-2/anti-IL-2 antibody complexes or low doses of IL-2. However, little is known about endogenous mechanisms that regulate the sensitivity of CD25 to signaling by IL-2. Here we demonstrate that CD25 is ADP-ribosylated at Arg35 in the IL-2 binding site by ecto-ADP-ribosyltransferase ARTC2.2, a toxin-related GPI-anchored ecto-enzyme. ADP-ribosylation inhibits binding of IL-2 by CD25, IL-2- induced phosphorylation of STAT5, and IL-2-dependent cell proliferation. Our study elucidates an as-yet-unrecognized mechanism to tune IL-2 signaling. This newly found mechanism might thwart Tregs at sites of inflammation and thereby permit a more potent response of activated effector T cells.
Collapse
Affiliation(s)
- Sophie Teege
- Institute of Immunology, University Medical Center, 20246 Hamburg, Germany
| | - Alexander Hann
- Institute of Immunology, University Medical Center, 20246 Hamburg, Germany
| | - Maria Miksiewicz
- Institute of Immunology, University Medical Center, 20246 Hamburg, Germany
| | - Cary MacMillan
- Institute of Immunology, University Medical Center, 20246 Hamburg, Germany
| | - Björn Rissiek
- Institute of Immunology, University Medical Center, 20246 Hamburg, Germany
| | - Friedrich Buck
- Department of Clinical Chemistry, University Medical Center, 20246 Hamburg, Germany
| | - Stephan Menzel
- Institute of Immunology, University Medical Center, 20246 Hamburg, Germany
| | - Marion Nissen
- Institute of Immunology, University Medical Center, 20246 Hamburg, Germany
| | - Peter Bannas
- 1] Institute of Immunology, University Medical Center, 20246 Hamburg, Germany [2] Department of Radiology, University Medical Center, 20246 Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center, 20246 Hamburg, Germany
| | - Olivier Boyer
- 1] Normandy University, Institute for Research and Innovation in Biomedicine, 76183 Rouen, France [2] Inserm, U905, 76000 Rouen, France [3] Rouen University Hospital, Department of Immunology, 76000 Rouen, France
| | - Michel Seman
- 1] Normandy University, Institute for Research and Innovation in Biomedicine, 76183 Rouen, France [2] Inserm, U905, 76000 Rouen, France
| | - Sahil Adriouch
- 1] Normandy University, Institute for Research and Innovation in Biomedicine, 76183 Rouen, France [2] Inserm, U905, 76000 Rouen, France
| | - Friedrich Koch-Nolte
- 1] Institute of Immunology, University Medical Center, 20246 Hamburg, Germany [2] Normandy University, Institute for Research and Innovation in Biomedicine, 76183 Rouen, France
| |
Collapse
|
45
|
Rissiek B, Koch-Nolte F, Magnus T. Nanobodies as modulators of inflammation: potential applications for acute brain injury. Front Cell Neurosci 2014; 8:344. [PMID: 25374510 PMCID: PMC4204521 DOI: 10.3389/fncel.2014.00344] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 10/06/2014] [Indexed: 12/22/2022] Open
Abstract
Nanobodies are single domain antibodies derived from llama heavy-chain only antibodies (HCAbs). They represent a new generation of biologicals with unique properties: nanobodies show excellent tissue distribution, high temperature and pH stability, are easy to produce recombinantly and can readily be converted into different formats such as Fc-fusion proteins or hetero-dimers. Moreover, nanobodies have the unique ability to bind molecular clefts, such as the active site of enzymes, thereby interfering with the function of the target protein. Over the last decade, numerous nanobodies have been developed against proteins involved in inflammation with the aim to modulate their immune functions. Here, we give an overview about recently developed nanobodies that target immunological pathways linked to neuroinflammation. Furthermore, we highlight strategies to modify nanobodies so that they can overcome the blood brain barrier and serve as highly specific therapeutics for acute inflammatory brain injury.
Collapse
Affiliation(s)
- Björn Rissiek
- Department of Neurology, University Medical Center Hamburg-Eppendorf Hamburg, Germany
| | - Friedrich Koch-Nolte
- Department of Immunology, University Medical Center Hamburg-Eppendorf Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf Hamburg, Germany
| |
Collapse
|