201
|
Fooks K, Galicia-Vazquez G, Gife V, Schcolnik-Cabrera A, Nouhi Z, Poon WWL, Luo V, Rys RN, Aloyz R, Orthwein A, Johnson NA, Hulea L, Mercier FE. EIF4A inhibition targets bioenergetic homeostasis in AML MOLM-14 cells in vitro and in vivo and synergizes with cytarabine and venetoclax. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:340. [PMID: 36482393 PMCID: PMC9733142 DOI: 10.1186/s13046-022-02542-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is an aggressive hematological cancer resulting from uncontrolled proliferation of differentiation-blocked myeloid cells. Seventy percent of AML patients are currently not cured with available treatments, highlighting the need of novel therapeutic strategies. A promising target in AML is the mammalian target of rapamycin complex 1 (mTORC1). Clinical inhibition of mTORC1 is limited by its reactivation through compensatory and regulatory feedback loops. Here, we explored a strategy to curtail these drawbacks through inhibition of an important effector of the mTORC1signaling pathway, the eukaryotic initiation factor 4A (eIF4A). METHODS We tested the anti-leukemic effect of a potent and specific eIF4A inhibitor (eIF4Ai), CR-1-31-B, in combination with cytosine arabinoside (araC) or the BCL2 inhibitor venetoclax. We utilized the MOLM-14 human AML cell line to model chemoresistant disease both in vitro and in vivo. In eIF4Ai-treated cells, we assessed for changes in survival, apoptotic priming, de novo protein synthesis, targeted intracellular metabolite content, bioenergetic profile, mitochondrial reactive oxygen species (mtROS) and mitochondrial membrane potential (MMP). RESULTS eIF4Ai exhibits anti-leukemia activity in vivo while sparing non-malignant myeloid cells. In vitro, eIF4Ai synergizes with two therapeutic agents in AML, araC and venetoclax. EIF4Ai reduces mitochondrial membrane potential (MMP) and the rate of ATP synthesis from mitochondrial respiration and glycolysis. Furthermore, eIF4i enhanced apoptotic priming while reducing the expression levels of the antiapoptotic factors BCL2, BCL-XL and MCL1. Concomitantly, eIF4Ai decreases intracellular levels of specific metabolic intermediates of the tricarboxylic acid cycle (TCA cycle) and glucose metabolism, while enhancing mtROS. In vitro redox stress contributes to eIF4Ai cytotoxicity, as treatment with a ROS scavenger partially rescued the viability of eIF4A inhibition. CONCLUSIONS We discovered that chemoresistant MOLM-14 cells rely on eIF4A-dependent cap translation for survival in vitro and in vivo. EIF4A drives an intrinsic metabolic program sustaining bioenergetic and redox homeostasis and regulates the expression of anti-apoptotic proteins. Overall, our work suggests that eIF4A-dependent cap translation contributes to adaptive processes involved in resistance to relevant therapeutic agents in AML.
Collapse
Affiliation(s)
- Katie Fooks
- grid.414980.00000 0000 9401 2774Lady Davis Institute for Medical Research, Montreal, Canada ,grid.14709.3b0000 0004 1936 8649Department of Medicine, McGill University, Montreal, Canada
| | | | - Victor Gife
- grid.414216.40000 0001 0742 1666Maisonneuve-Rosemont Hospital Research Centre, Montreal, Canada ,grid.14848.310000 0001 2292 3357Present Address: Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montreal, Canada
| | | | - Zaynab Nouhi
- grid.414216.40000 0001 0742 1666Maisonneuve-Rosemont Hospital Research Centre, Montreal, Canada
| | - William W. L. Poon
- grid.414980.00000 0000 9401 2774Lady Davis Institute for Medical Research, Montreal, Canada ,grid.14709.3b0000 0004 1936 8649Department of Medicine, McGill University, Montreal, Canada
| | - Vincent Luo
- grid.414980.00000 0000 9401 2774Lady Davis Institute for Medical Research, Montreal, Canada ,grid.14709.3b0000 0004 1936 8649Department of Medicine, McGill University, Montreal, Canada
| | - Ryan N. Rys
- grid.414980.00000 0000 9401 2774Lady Davis Institute for Medical Research, Montreal, Canada ,grid.14709.3b0000 0004 1936 8649Department of Physiology, McGill University, Montreal, Canada
| | - Raquel Aloyz
- grid.414980.00000 0000 9401 2774Lady Davis Institute for Medical Research, Montreal, Canada ,grid.14709.3b0000 0004 1936 8649Department of Medicine, McGill University, Montreal, Canada ,grid.14709.3b0000 0004 1936 8649Gerald Bronfman Department of Oncology, McGill University, Montreal, Canada
| | - Alexandre Orthwein
- grid.414980.00000 0000 9401 2774Lady Davis Institute for Medical Research, Montreal, Canada ,grid.14709.3b0000 0004 1936 8649Department of Medicine, McGill University, Montreal, Canada ,grid.14709.3b0000 0004 1936 8649Gerald Bronfman Department of Oncology, McGill University, Montreal, Canada ,grid.189967.80000 0001 0941 6502Present Address: Department of Radiation Oncology, Emory School of Medicine, Atlanta, USA
| | - Nathalie A. Johnson
- grid.414980.00000 0000 9401 2774Lady Davis Institute for Medical Research, Montreal, Canada ,grid.14709.3b0000 0004 1936 8649Department of Medicine, McGill University, Montreal, Canada ,grid.14709.3b0000 0004 1936 8649Gerald Bronfman Department of Oncology, McGill University, Montreal, Canada
| | - Laura Hulea
- grid.414216.40000 0001 0742 1666Maisonneuve-Rosemont Hospital Research Centre, Montreal, Canada ,grid.14848.310000 0001 2292 3357Present Address: Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montreal, Canada ,grid.14848.310000 0001 2292 3357Département de Médecine, Université de Montréal, Montreal, Canada
| | - Francois E. Mercier
- grid.414980.00000 0000 9401 2774Lady Davis Institute for Medical Research, Montreal, Canada ,grid.14709.3b0000 0004 1936 8649Department of Medicine, McGill University, Montreal, Canada
| |
Collapse
|
202
|
Cyran L, Serfling J, Kirschner L, Raifer H, Lohoff M, Hermanns HM, Kerstan A, Bodem J, Lutz MB. Flt3L, LIF, and IL-10 combination promotes the selective in vitro development of ESAM low cDC2B from murine bone marrow. Eur J Immunol 2022; 52:1946-1960. [PMID: 35357005 DOI: 10.1002/eji.202149663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 02/12/2022] [Accepted: 03/29/2022] [Indexed: 01/05/2023]
Abstract
The development of two conventional dendritic cells (DC) subsets (cDC1 and cDC2) and the plasmacytoid DC (pDC) in vivo and in cultures of bone marrow (BM) cells is mediated by the growth factor Flt3L. However, little is known about the factors that direct the development of the individual DC subsets. Here, we describe the selective in vitro generation of murine ESAMlow CD103- XCR1- CD172a+ CD11b+ cDC2 from BM by treatment with a combination of Flt3L, LIF, and IL-10 (collectively named as FL10). FL10 promotes common dendritic cell progenitors (CDP) proliferation in the cultures, similar to Flt3L and CDP sorted and cultured in FL10 generate exclusively cDC2. These cDC2 express the transcription factors Irf4, Klf4, and Notch2, and their growth is reduced using BM from Irf4-/- mice, but the expression of Batf3 and Tcf4 is low. Functionally they respond to TLR3, TLR4, and TLR9 signals by upregulation of the surface maturation markers MHC II, CD80, CD86, and CD40, while they poorly secrete proinflammatory cytokines. Peptide presentation to TCR transgenic OT-II cells induced proliferation and IFN-γ production that was similar to GM-CSF-generated BM-DC and higher than Flt3L-generated DC. Together, our data support that FL10 culture of BM cells selectively promotes CDP-derived ESAMlow cDC2 (cDC2B) development and survival in vitro.
Collapse
Affiliation(s)
- Laura Cyran
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Julia Serfling
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Luisa Kirschner
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Hartmann Raifer
- Institute of Medical Microbiology and Hygiene, University of Marburg, Marburg, Germany
| | - Michael Lohoff
- Institute of Medical Microbiology and Hygiene, University of Marburg, Marburg, Germany
| | - Heike M Hermanns
- Department of Internal Medicine II, Hepatology Research Laboratory, University Hospital Würzburg, Würzburg, Germany
| | - Andreas Kerstan
- Department of Dermatology, Venereology, and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Jochen Bodem
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Manfred B Lutz
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
203
|
Haase P, Schäfer S, Gerlach RG, Winkler TH, Voehringer D. B cell fate mapping reveals their contribution to the memory immune response against helminths. Front Immunol 2022; 13:1016142. [PMID: 36505408 PMCID: PMC9730276 DOI: 10.3389/fimmu.2022.1016142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022] Open
Abstract
An estimated quarter of the human world population is infected with gastrointestinal helminths causing major socioeconomic problems in endemic countries. A better understanding of humoral immune responses against helminths is urgently needed to develop effective vaccination strategies. Here, we used a fate mapping (FM) approach to mark germinal center (GC) B cells and their developmental fates by induced expression of a fluorescent protein during infection of mice with the helminth Nippostrongylus brasiliensis. We could show that FM+ cells persist weeks after clearance of the primary infection mainly as CD80+CD73+PD-L2+ memory B cells. A secondary infection elicited expansion of helminth-specific memory B cells and plasma cells (PCs). Adoptive transfers and analysis of somatic mutations in immunoglobulin genes further revealed that FM+ B cells rapidly convert to PCs rather than participating again in a GC reaction. These results provide new insights in the population dynamics of the humoral immune response against helminths.
Collapse
Affiliation(s)
- Paul Haase
- Department of Infection Biology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Simon Schäfer
- Department of Genetics, Faculty of Sciences, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Roman G. Gerlach
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Thomas H. Winkler
- Department of Genetics, Faculty of Sciences, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - David Voehringer
- Department of Infection Biology, Universitätsklinikum Erlangen, Erlangen, Germany,Faculty of Medicine, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany,*Correspondence: David Voehringer,
| |
Collapse
|
204
|
Bonavia AS, Samuelsen A, Luthy J, Halstead ES. Integrated machine learning approaches for flow cytometric quantification of myeloid-derived suppressor cells in acute sepsis. Front Immunol 2022; 13:1007016. [PMID: 36466851 PMCID: PMC9714638 DOI: 10.3389/fimmu.2022.1007016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/04/2022] [Indexed: 09/05/2024] Open
Abstract
Highly heterogeneous cell populations require multiple flow cytometric markers for appropriate phenotypic characterization. This exponentially increases the complexity of 2D scatter plot analyses and exacerbates human errors due to variations in manual gating of flow data. We describe a semi-automated workflow, based entirely on the Flowjo Graphical User Interface (GUI), that involves the stepwise integration of several, newly available machine learning tools for the analysis of myeloid-derived suppressor cells (MDSCs) in septic and non-septic critical illness. Supervised clustering of flow cytometric data showed correlation with, but significantly different numbers of, MDSCs as compared with the cell numbers obtained by manual gating. Neither quantification method predicted 30-day clinical outcomes in a cohort of 16 critically ill and septic patients and 5 critically ill and non-septic patients. Machine learning identified a significant decrease in the proportion of PMN-MDSC in critically ill and septic patients as compared with healthy controls. There was no difference between the proportion of these MDSCs in septic and non-septic critical illness.
Collapse
Affiliation(s)
- Anthony S. Bonavia
- Division of Critical Care Medicine, Department of Anesthesiology and Perioperative Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
- Department of Anesthesiology and Perioperative Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Abigail Samuelsen
- Division of Critical Care Medicine, Department of Anesthesiology and Perioperative Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Joshua Luthy
- Product Innovation Division, BD Life Sciences - FlowJo, Ashland, OR, United States
| | - E. Scott Halstead
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| |
Collapse
|
205
|
Li Z, Xiang J, Zhang Q, Zhao M, Meng Y, Zhong J, Li T, Jia L, Li K, Lu X, Ao Z, Han D. An engineered hydrogel with low-dose antitumor drugs enhances tumor immunotherapy through tumor interstitial wrap. Front Bioeng Biotechnol 2022; 10:1072393. [PMID: 36452209 PMCID: PMC9701709 DOI: 10.3389/fbioe.2022.1072393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 11/15/2022] Open
Abstract
Stimulating immunogenic cell death (ICD) is the key to tumor immunotherapy. However, traditional chemoradiotherapy has limited effect on stimulating immunity and often requires repeated administration, which greatly reduces the tumor-killing effect. In this article, we created a sodium alginate hydrogel sustained-release system containing low-dose doxorubicin (Dox) and immune adjuvant R837, which were injected into the interstitial space to wrap around the tumor in situ, achieving a sustained release and long-lasting immune response. Cooperating with immune checkpoint blockade, Dox induced ICD, activated dendritic cells (DCs) and converted immunosuppressive M2-type tumor-associated macrophages (TAM) to tumor-killing M1-type TAMs. Simultaneously, it greatly promoted T cell proliferation and infiltration, and reduced tumor immunosuppressive factors, triggering a robust immune response to suppress tumors in vivo. In conclusion, this anti-tumor strategy based on interstitial injection can achieve continuous local immune stimulation by low-dose chemotherapy drugs, providing a potential approach for tumor immunotherapy.
Collapse
Affiliation(s)
- Zhongxian Li
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiawei Xiang
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qiang Zhang
- Hebei Key Lab of Nano-Biotechnology, Hebei Key Lab of Applied Chemistry, Yanshan University, Qinhuangdao, China
| | - Mingyuan Zhao
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Meng
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jie Zhong
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tingting Li
- College of Life Sciences, Bejing University of Chinese Medicine, Beijing, China
| | - Lanxin Jia
- University of Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Kai Li
- College of Life Sciences, Bejing University of Chinese Medicine, Beijing, China
| | - Xi Lu
- College of Life Sciences, Bejing University of Chinese Medicine, Beijing, China
| | - Zhuo Ao
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- *Correspondence: Zhuo Ao, ; Dong Han,
| | - Dong Han
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, Bejing University of Chinese Medicine, Beijing, China
- *Correspondence: Zhuo Ao, ; Dong Han,
| |
Collapse
|
206
|
Rückert T, Lareau CA, Mashreghi MF, Ludwig LS, Romagnani C. Clonal expansion and epigenetic inheritance of long-lasting NK cell memory. Nat Immunol 2022; 23:1551-1563. [PMID: 36289449 PMCID: PMC9663309 DOI: 10.1038/s41590-022-01327-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/09/2022] [Indexed: 11/08/2022]
Abstract
Clonal expansion of cells with somatically diversified receptors and their long-term maintenance as memory cells is a hallmark of adaptive immunity. Here, we studied pathogen-specific adaptation within the innate immune system, tracking natural killer (NK) cell memory to human cytomegalovirus (HCMV) infection. Leveraging single-cell multiomic maps of ex vivo NK cells and somatic mitochondrial DNA mutations as endogenous barcodes, we reveal substantial clonal expansion of adaptive NK cells in HCMV+ individuals. NK cell clonotypes were characterized by a convergent inflammatory memory signature enriched for AP1 motifs superimposed on a private set of clone-specific accessible chromatin regions. NK cell clones were stably maintained in specific epigenetic states over time, revealing that clonal inheritance of chromatin accessibility shapes the epigenetic memory repertoire. Together, we identify clonal expansion and persistence within the human innate immune system, suggesting that these mechanisms have evolved independent of antigen-receptor diversification.
Collapse
Affiliation(s)
- Timo Rückert
- Innate Immunity, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), ein Leibniz Institut, Berlin, Germany.
| | - Caleb A Lareau
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Mir-Farzin Mashreghi
- Therapeutic Gene Regulation, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), ein Leibniz Institut, Berlin, Germany
| | - Leif S Ludwig
- Berlin Institute of Health at Charité Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Chiara Romagnani
- Innate Immunity, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), ein Leibniz Institut, Berlin, Germany.
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Berlin, Germany.
- Leibniz-Science Campus Chronic Inflammation, Berlin, Germany.
| |
Collapse
|
207
|
Wilden A, Glaubitz J, Otto O, Biedenweg D, Nauck M, Mack M, Ribback S, Bröker BM, von Rheinbaben SF, Lerch MM, Aghdassi AA, Weiss FU, Sendler M. Mobilization of CD11b+/Ly6chi monocytes causes multi organ dysfunction syndrome in acute pancreatitis. Front Immunol 2022; 13:991295. [PMID: 36300116 PMCID: PMC9589437 DOI: 10.3389/fimmu.2022.991295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Acute pancreatitis (AP) is an inflammatory disorder, the severe form of which is burdened with multi-organ dysfunction and high mortality. The pathogenesis of life –threatening organ complications, such as respiratory and renal failure, is unknown. Design Organ dysfunction was investigated in a mouse model of AP. The influence of monocytes and neutrophils on multi organ dysfunction syndrome (MODS) was investigated in vivo by antibody depletion. Using real-time-fluorescence and deformability-cytometry (RT-DC) analysis we determined the mechanical properties of neutrophils and monocytes during AP. Furthermore, blood samples of pancreatitis patients were used to characterize severity-dependent chemokine profiles according to the revised Atlanta classification. Results Similar to AP in humans, severe disease in the mouse model associates with organ dysfunction mainly of lung and kidney, which is triggered by a mobilisation of Ly6g-/CD11b+/Ly6c hi monocytes, but not of Ly6g+/CD11b+ neutrophils. Monocyte depletion by anti-CCR2 antibody treatment ameliorated lung function (oxygen consumption) without interfering with the systemic immune response. RT-DC analysis of circulation monocytes showed a significant increase in cell size during SAP, but without a compensatory increase in elasticity. Patient chemokine profiles show a correlation of AP severity with monocyte attracting chemokines like MCP-1 or MIG and with leukocyte mobilisation. Conclusion In AP, the physical properties of mobilized monocytes, especially their large size, result in an obstruction of the fine capillary systems of the lung and of the kidney glomeruli. A selective depletion of monocytes may represent a treatment strategy for pancreatitis as well as for other inflammation-related disorders.
Collapse
Affiliation(s)
- Anika Wilden
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Juliane Glaubitz
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Oliver Otto
- Center for Innovation Competence: Humoral Immune Reactions in Cardiovascular Disorders, University of Greifswald, Greifswald, Germany
| | - Doreen Biedenweg
- Center for Innovation Competence: Humoral Immune Reactions in Cardiovascular Disorders, University of Greifswald, Greifswald, Germany
- Department of Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Diagnostics, University Medicine Greifswald, Greifswald, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) (German Centre for Cardiovascular Research), Partner Site Greifswald, University Medicine, Greifswald, Germany
| | - Matthias Mack
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Silvia Ribback
- Institute of Pathology, Universitat Greifswald, Greifswald, Mecklenburg-Vorpommern, Germany
| | - Barbara M. Bröker
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | | | - Markus M. Lerch
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | | | - Frank Ulrich Weiss
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Matthias Sendler
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
- *Correspondence: Matthias Sendler,
| |
Collapse
|
208
|
Li J, Reinke S, Shen Y, Schollmeyer L, Liu YC, Wang Z, Hardt S, Hipfl C, Hoffmann U, Frischbutter S, Chang HD, Alexander T, Perka C, Radbruch H, Qin Z, Radbruch A, Dong J. A ubiquitous bone marrow reservoir of preexisting SARS-CoV-2-reactive memory CD4+ T lymphocytes in unexposed individuals. Front Immunol 2022; 13:1004656. [PMID: 36268016 PMCID: PMC9576920 DOI: 10.3389/fimmu.2022.1004656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022] Open
Abstract
Circulating, blood-borne SARS-CoV-2-reactive memory T cells in persons so far unexposed to SARS-CoV-2 or the vaccines have been described in 20-100% of the adult population. They are credited with determining the efficacy of the immune response in COVID-19. Here, we demonstrate the presence of preexisting memory CD4+ T cells reacting to peptides of the spike, membrane, or nucleocapsid proteins of SARS-CoV-2 in the bone marrow of all 17 persons investigated that had previously not been exposed to SARS-CoV-2 or one of the vaccines targeting it, with only 15 of these persons also having such cells detectable circulating in the blood. The preexisting SARS-CoV-2-reactive memory CD4+ T cells of the bone marrow are abundant and polyfunctional, with the phenotype of central memory T cells. They are tissue-resident, at least in those persons who do not have such cells in the blood, and about 30% of them express CD69. Bone marrow resident SARS-CoV-2-reactive memory CD4+ memory T cells are also abundant in vaccinated persons analyzed 10-168 days after 1°-4° vaccination. Apart from securing the bone marrow, preexisting cross-reactive memory CD4+ T cells may play an important role in shaping the systemic immune response to SARS-CoV-2 and the vaccines, and contribute essentially to the rapid establishment of long-lasting immunity provided by memory plasma cells, already upon primary infection.
Collapse
Affiliation(s)
- Jinchan Li
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Simon Reinke
- Berlin Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Yu Shen
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Lena Schollmeyer
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Yuk-Chien Liu
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Zixu Wang
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Sebastian Hardt
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Christian Hipfl
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ute Hoffmann
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
- Schwiete-Laboratory for Microbiota and Inflammation, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Stefan Frischbutter
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Allergology and Immunology, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Berlin, Germany
| | - Hyun-Dong Chang
- Schwiete-Laboratory for Microbiota and Inflammation, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Tobias Alexander
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carsten Perka
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Helena Radbruch
- Institute of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Andreas Radbruch
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
| | - Jun Dong
- Cell Biology, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Institute of the Leibniz Association, Berlin, Germany
- *Correspondence: Jun Dong,
| |
Collapse
|
209
|
Barros-Martins J, Bruni E, Fichtner AS, Cornberg M, Prinz I. OMIP-084: 28-color full spectrum flow cytometry panel for the comprehensive analysis of human γδ T cells. Cytometry A 2022; 101:856-861. [PMID: 35521651 DOI: 10.1002/cyto.a.24564] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 03/21/2022] [Accepted: 04/25/2022] [Indexed: 01/27/2023]
Abstract
Using full spectrum flow cytometry, we designed a 28-color panel for the analysis of markers known to be associated with the γδ T cell immune response. This panel allows the classification of γδ T cell subsets via specific V gene usage (Vγ9, Vδ1, Vδ2, and Vδ3) of their T cell receptor (TCR) and according to their functional differentiation. Phenotypical surface receptors to distinguish different stages of cell maturation included CD45RA, CD27, CD28, CD127, CD57, and CD16; chemokine receptors CXCR6, CCR5, CCR6, and CX3CR1; NK-associated markers NKG2A, NKG2D, CD56, and CD161, checkpoint-inhibitor PD-1, and activating receptors CD38 and CD25. T cell lineage markers for the analysis of αβ T cells (CD4 and CD8) and MAIT cells (Vα7.2) were also included. This optimized multicolor panel allows a comprehensive immune-profiling of all main human γδ T cell subsets and is suitable for longitudinal or exploratory analysis of γδ T cell development and γδ T cell dynamics in clinical cohorts.
Collapse
Affiliation(s)
| | - Elena Bruni
- Hannover Medical School, Institute of Immunology, Hannover, Germany.,Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany.,Hamburg Center for Translational Immunology (HCTI), Institute of Systems Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Markus Cornberg
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany.,Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany.,Centre for Individualised Infection Medicine (CiiM), Hannover, Germany
| | - Immo Prinz
- Hannover Medical School, Institute of Immunology, Hannover, Germany.,Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany.,Hamburg Center for Translational Immunology (HCTI), Institute of Systems Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Centre for Individualised Infection Medicine (CiiM), Hannover, Germany
| |
Collapse
|
210
|
Panda S, Seelan DM, Faisal S, Arora A, Luthra K, Palanichamy JK, Mohan A, Vikram NK, Gupta NK, Ramakrishnan L, Singh A. Chronic hyperglycemia drives alterations in macrophage effector function in pulmonary tuberculosis. Eur J Immunol 2022; 52:1595-1609. [PMID: 36066992 DOI: 10.1002/eji.202249839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 07/05/2022] [Accepted: 09/05/2022] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus (DM) alters immune responses and given the rising prevalence of DM in tuberculosis (TB) endemic countries; hyperglycemia can be a potential risk factor for active TB development. However, the impact of hyperglycemia on TB-specific innate immune response in terms of macrophage functions remains poorly addressed. We assessed macrophage effector functions in uncontrolled DM patients with or without TB infection (PTB+DM and DM), non-diabetic TB patients (PTB), and non-diabetic-uninfected controls. Phagocytic capacity against BCG and surface expression of different pattern recognition receptors (PRRs) (CD11b, CD14, CD206, MARCO, and TLR-2) were measured via flow cytometry. Effector molecules (ROS and NO) required for bacterial killing were assessed via DCFDA and Griess reaction respectively. A systematic dysregulation in phagocytic capacity with concurrent alterations in the expression pattern of key PRRs (CD11b, MARCO, and CD206) was observed in PTB+DM. These altered PRR expressions were associated with decreased phagocytic capacity of macrophages. Similarly, ROS was aberrantly higher while NO was lower in PTB+DM. These altered macrophage functions were positively correlated with increasing disease severity. Our results highlight several key patterns of immune dysregulation against TB infection under hyperglycemic conditions and highlight a negative impact of hyperglycemia with etiology and progression of TB.
Collapse
Affiliation(s)
- Sudhasini Panda
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Diravya M Seelan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Shah Faisal
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Alisha Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | | | - Anant Mohan
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Naval K Vikram
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Neeraj Kumar Gupta
- Department of Pulmonary Medicine, VMMC and Safdarjung Hospital, New Delhi, 110029, India
| | - Lakshmy Ramakrishnan
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Archana Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| |
Collapse
|
211
|
Marsman C, Verstegen NJM, Streutker M, Jorritsma T, Boon L, ten Brinke A, van Ham SM. Termination of CD40L co-stimulation promotes human B cell differentiation into antibody-secreting cells. Eur J Immunol 2022; 52:1662-1675. [PMID: 36073009 PMCID: PMC9825913 DOI: 10.1002/eji.202249972] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/01/2022] [Accepted: 09/05/2022] [Indexed: 01/11/2023]
Abstract
Human naïve B cells are notoriously difficult to differentiate into antibody-secreting cells (ASCs) in vitro while maintaining sufficient cell numbers to evaluate the differentiation process. B cells require T follicular helper (TFH ) cell-derived signals like CD40L and IL-21 during germinal center (GC) responses to undergo differentiation into ASCs. Cognate interactions between B and TFH cells are transient; after TFH contact, B cells cycle between GC light and dark zones where TFH contact is present and absent, respectively. Here, we elucidated that the efficacy of naïve B cells in ACS differentiation is dramatically enhanced by the release of CD40L stimulation. Multiparameter phospho-flow and transcription factor (TF)-flow cytometry revealed that termination of CD40L stimulation downmodulates NF-κB and STAT3 signaling. Furthermore, the termination of CD40 signaling downmodulates C-MYC, while promoting ASC TFs BLIMP1 and XBP-1s. Reduced levels of C-MYC in the differentiating B cells are later associated with crucial downmodulation of the B cell signature TF PAX5 specifically upon the termination of CD40 signaling, resulting in the differentiation of BLIMP1 high expressing cells into ASCs. The data presented here are the first steps to provide further insights how the transient nature of CD40 signaling is in fact needed for efficient human naïve B cell differentiation to ASCs.
Collapse
Affiliation(s)
- Casper Marsman
- Sanquin ResearchDepartment of ImmunopathologyUniversity of AmsterdamAmsterdamThe Netherlands,Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Niels JM Verstegen
- Sanquin ResearchDepartment of ImmunopathologyUniversity of AmsterdamAmsterdamThe Netherlands,Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Marij Streutker
- Sanquin ResearchDepartment of ImmunopathologyUniversity of AmsterdamAmsterdamThe Netherlands,Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Tineke Jorritsma
- Sanquin ResearchDepartment of ImmunopathologyUniversity of AmsterdamAmsterdamThe Netherlands,Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | | | - Anja ten Brinke
- Sanquin ResearchDepartment of ImmunopathologyUniversity of AmsterdamAmsterdamThe Netherlands,Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - S. Marieke van Ham
- Sanquin ResearchDepartment of ImmunopathologyUniversity of AmsterdamAmsterdamThe Netherlands,Swammerdam Institute for Life SciencesUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
212
|
Mohr E, Hinnenthal T, Gryzik S, Hoang Y, Lischke T, Retzlaff J, Mekonnen A, Paul F, Valleriani A, Radbruch A, Vera J, Baumgrass R. Bin-based visualization of cytokine-co-expression patterns of IL-10-producing CD4 T cell subsets. Eur J Immunol 2022; 52:1684-1687. [PMID: 36067024 DOI: 10.1002/eji.202249829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 12/14/2022]
Affiliation(s)
- Elodie Mohr
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany
| | - Timo Hinnenthal
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany
| | - Stefanie Gryzik
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany
| | - Yen Hoang
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany
| | - Timo Lischke
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany
| | - Jimmy Retzlaff
- Systems Tumour Immunology, Friedrich-Alexander-University of Erlangen-Nürnberg, Universitätsklinikum, Erlangen, Germany
| | - Ariana Mekonnen
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité, Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Angelo Valleriani
- Max Planck Institute of Colloids and Interfaces, Biomaterials Department, Potsdam, Germany
| | - Andreas Radbruch
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany.,Charité, Campus Berlin Mitte, Berlin, Germany
| | - Julio Vera
- Systems Tumour Immunology, Friedrich-Alexander-University of Erlangen-Nürnberg, Universitätsklinikum, Erlangen, Germany
| | - Ria Baumgrass
- German Rheumatism Research Center, A Leibniz Institute, Berlin, Germany.,University of Potsdam, Potsdam, Germany
| |
Collapse
|
213
|
Li C, Lin YD, Wang WB, Xu M, Zhang N, Xiong N. Differential regulation of CD8 + CD86 + Vγ1.1 + γδT cell responses in skin barrier tissue protection and homeostatic maintenance. Eur J Immunol 2022; 52:1498-1509. [PMID: 35581932 DOI: 10.1002/eji.202249793] [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: 01/10/2022] [Revised: 04/13/2022] [Accepted: 05/16/2022] [Indexed: 11/11/2022]
Abstract
Compared to αβT cells, γδT cells are more innate-like and preferentially function as the first line of defense in barrier tissues. Certain populations of γδT cells possess adaptive immune cell properties but their regulation is not well understood. We herein report that while innate-like γδT17 cells dominated in the skin of WT mice, Vγ1.1+ γδT cells with adaptive T cell-like properties predominantly expanded in the skin of TCRβ-/- and B2m-/- mice. Commensal bacteria drove expansion of Vγ1.1+ skin γδT cells, functional properties of which correlated with local immune requirements. That is, Vγ1.1+ skin γδT cells in TCRβ-/- mice were a heterogeneous population; while Vγ1.1+ skin γδT cells in B2m-/- mice were mostly CD8+ CD86+ cells that had a similar function of CD8+ CD86+ skin αβT cells in supporting local Treg cells. We also found that intrinsic TGF-β receptor 2-derived signals in skin CD8+ αβT and γδT cells are required for their expression of CD86, a molecule important in supporting skin Treg cells. Our findings reveal broad functional potentials of γδT cells that are coordinately regulated with αβT cells to help maintain local tissue homeostasis.
Collapse
Affiliation(s)
- Chao Li
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, P. R. China
| | - Yang-Ding Lin
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Wei-Bei Wang
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Ming Xu
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Nu Zhang
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Na Xiong
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
- Division of Dermatology and Cutaneous Surgery, Department of Medicine, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| |
Collapse
|
214
|
Yang J, Zou M, Chu X, Floess S, Li Y, Delacher M, Huehn J. Inflammatory perturbations in early life long-lastingly shape the transcriptome and TCR repertoire of the first wave of regulatory T cells. Front Immunol 2022; 13:991671. [PMID: 36119090 PMCID: PMC9471859 DOI: 10.3389/fimmu.2022.991671] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
The first wave of Foxp3+ regulatory T cells (Tregs) generated in neonates is critical for the life-long prevention of autoimmunity. Although it is widely accepted that neonates are highly susceptible to infections, the impact of neonatal infections on this first wave of Tregs is completely unknown. Here, we challenged newborn Treg fate-mapping mice (Foxp3eGFPCreERT2xROSA26STOP-eYFP) with the Toll-like receptor (TLR) agonists LPS and poly I:C to mimic inflammatory perturbations upon neonatal bacterial or viral infections, respectively, and subsequently administrated tamoxifen during the first 8 days of life to selectively label the first wave of Tregs. Neonatally-tagged Tregs preferentially accumulated in non-lymphoid tissues (NLTs) when compared to secondary lymphoid organs (SLOs) irrespective of the treatment. One week post challenge, no differences in the frequency and phenotypes of neonatally-tagged Tregs were observed between challenged mice and untreated controls. However, upon aging, a decreased frequency of neonatally-tagged Tregs in both NLTs and SLOs was detected in challenged mice when compared to untreated controls. This decrease became significant 12 weeks post challenge, with no signs of altered Foxp3 stability. Remarkably, this late decrease in the frequency of neonatally-tagged Tregs only occurred when newborns were challenged, as treating 8-days-old mice with TLR agonists did not result in long-lasting alterations of the first wave of Tregs. Combined single-cell T cell receptor (TCR)-seq and RNA-seq revealed that neonatal inflammatory perturbations drastically diminished TCR diversity and long-lastingly altered the transcriptome of neonatally-tagged Tregs, exemplified by lower expression of Tigit, Foxp3, and Il2ra. Together, our data demonstrate that a single, transient encounter with a pathogen in early life can have long-lasting consequences for the first wave of Tregs, which might affect immunological tolerance, prevention of autoimmunity, and other non-canonical functions of tissue-resident Tregs in adulthood.
Collapse
Affiliation(s)
- Juhao Yang
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, China Innovation Center of Roche, Shanghai, China
| | - Mangge Zou
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Xiaojing Chu
- Department Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), Helmholtz Centre for Infection Research and Hannover Medical School, Hannover, Germany
| | - Stefan Floess
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Yang Li
- Department Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), Helmholtz Centre for Infection Research and Hannover Medical School, Hannover, Germany
| | - Michael Delacher
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Centre for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- *Correspondence: Jochen Huehn,
| |
Collapse
|
215
|
Dongye Z, Wu X, Wen Y, Ding X, Wang C, Zhao T, Li J, Wu Y. Icaritin and intratumoral injection of CpG treatment synergistically promote T cell infiltration and antitumor immune response in mice. Int Immunopharmacol 2022; 111:109093. [PMID: 35930912 DOI: 10.1016/j.intimp.2022.109093] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/18/2022] [Accepted: 07/23/2022] [Indexed: 11/16/2022]
Abstract
The development of combination therapy that can modulate the tumor immunosuppressive microenvironment is highly desirable for cancer immunotherapy. Icaritin (ICT), a hydrolytic product of icariin from genus Epimedium, has been used as an anti-cancer immunoregulatory agent for many types of cancers. Herein, we design a novel therapeutic strategy for mice melanoma that combines systemic administration of icaritin with intratumoral injection of unmethylated cytosine-guanine oligodeoxynucleotide (CpG). Icaritin induces tumor cell apoptosis and increases tumor immunogenicity. The combination of icaritin with CpG synergistically suppresses tumor growth and significantly prolonged survival time of B16F10 melanoma bearing mice. importantly, the anti-tumor effects of this combination strategy are associated with the reversing of immunosuppressive microenvironment through increased recruitment of functional DCs and tumor-associated macrophages (TAM) in tumors, leading to the infiltration of cytotoxic CD8+ T cells expressing elevated levels of IFN-γ and TNF-α. Furthermore, the combination of icaritin with CpG augments the anti-tumor immune response to anti-PD-1/CTLA-4 immune checkpoint blockade treatment. These results support the combination of icaritin with CpG as a novel strategy to elicit effective T cell-mediated antitumor immune response.
Collapse
Affiliation(s)
- Zhangchi Dongye
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong 266071, PR China; Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaoping Wu
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuxiang Wen
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xuelei Ding
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chuanjie Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Tingting Zhao
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jian Li
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Yuzhang Wu
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
216
|
Thrun MC, Hoffmann J, Röhnert M, von Bonin M, Oelschlägel U, Brendel C, Ultsch A. Flow cytometry datasets consisting of peripheral blood and bone marrow samples for the evaluation of explainable artificial intelligence methods. Data Brief 2022; 43:108382. [PMID: 35799850 PMCID: PMC9253476 DOI: 10.1016/j.dib.2022.108382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 11/25/2022] Open
Abstract
Three different Flow Cytometry datasets consisting of diagnostic samples of either peripheral blood (pB) or bone marrow (BM) from patients without any sign of bone marrow disease at two different health care centers are provided. In Flow Cytometry, each cell rapidly passes through a laser beam one by one, and two light scatter, and eight surface parameters of more than 100.000 cells are measured per sample of each patient. The technology swiftly characterizes cells of the immune system at the single-cell level based on antigens presented on the cell surface that are targeted by a set of fluorochrome-conjugated antibodies. The first dataset consists of N=14 sample files measured in Marburg and the second dataset of N=44 data files measured in Dresden, of which half are BM samples and half are pB samples. The third dataset contains N=25 healthy bone marrow samples and N=25 leukemia bone marrow samples measured in Marburg. The data has been scaled to log between zero and six and used to identify cell populations that are simultaneously meaningful to the clinician and relevant to the distinction of pB vs BM, and BM vs leukemia. Explainable artificial intelligence methods should distinguish these samples and provide meaningful explanations for the classification without taking more than several hours to compute their results. The data described in this article are available in Mendeley Data [1].
Collapse
Affiliation(s)
- Michael C. Thrun
- Databionics, Mathematics and Computer Science, Philipps-Universität Marburg, Hans-Meerwein-Straße 6 D-35032 Marburg, Germany
- J Department of Hematology, Oncology and Immunology, Philipps-University, Baldinger Str., D-35032 Marburg, Germany
| | - Jörg Hoffmann
- J Department of Hematology, Oncology and Immunology, Philipps-University, Baldinger Str., D-35032 Marburg, Germany
| | - Maximilian Röhnert
- Medizinische Klinik und Poliklinik I Bereich Innere Medizin / Hämatologie und Onkologie, Universitätsklinikum Carl Gustav Carus Dresden, Germany
| | - Malte von Bonin
- Medizinische Klinik und Poliklinik I Bereich Innere Medizin / Hämatologie und Onkologie, Universitätsklinikum Carl Gustav Carus Dresden, Germany
| | - Uta Oelschlägel
- Medizinische Klinik und Poliklinik I Bereich Innere Medizin / Hämatologie und Onkologie, Universitätsklinikum Carl Gustav Carus Dresden, Germany
| | - Cornelia Brendel
- J Department of Hematology, Oncology and Immunology, Philipps-University, Baldinger Str., D-35032 Marburg, Germany
| | - Alfred Ultsch
- Databionics, Mathematics and Computer Science, Philipps-Universität Marburg, Hans-Meerwein-Straße 6 D-35032 Marburg, Germany
| |
Collapse
|
217
|
Bain CC, Louwe PA, Steers NJ, Bravo‐Blas A, Hegarty LM, Pridans C, Milling SW, MacDonald AS, Rückerl D, Jenkins SJ. CD11c identifies microbiota and EGR2-dependent MHCII + serous cavity macrophages with sexually dimorphic fate in mice. Eur J Immunol 2022; 52:1243-1257. [PMID: 35568024 PMCID: PMC7613339 DOI: 10.1002/eji.202149756] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/11/2022] [Accepted: 05/09/2022] [Indexed: 11/27/2022]
Abstract
The murine serous cavities contain a rare and enigmatic population of short-lived F4/80lo MHCII+ macrophages but what regulates their development, survival, and fate is unclear. Here, we show that mature F4/80lo MHCII+ peritoneal macrophages arise after birth, but that this occurs largely independently of colonization by microbiota. Rather, microbiota specifically regulate development of a subpopulation of CD11c+ cells that express the immunoregulatory cytokine RELM-α, are reliant on the transcription factor EGR2, and develop independently of the growth factor CSF1. Furthermore, we demonstrate that intrinsic expression of RELM-α, a signature marker shared by CD11c+ and CD11c- F4/80lo MHCII+ cavity macrophages, regulates survival and differentiation of these cells in the peritoneal cavity in a sex-specific manner. Thus, we identify a previously unappreciated diversity in serous cavity F4/80lo MHCII+ macrophages that is regulated by microbiota, and describe a novel sex and site-specific function for RELM-α in regulating macrophage endurance that reveals the unique survival challenge presented to monocyte-derived macrophages by the female peritoneal environment.
Collapse
Affiliation(s)
- Calum C. Bain
- Queens Medical Research InstituteUniversity of Edinburgh Centre for Inflammation ResearchEdinburghUK
| | - Pieter A. Louwe
- Queens Medical Research InstituteUniversity of Edinburgh Centre for Inflammation ResearchEdinburghUK
| | | | - Alberto Bravo‐Blas
- Institute of Infection, Immunity, and InflammationUniversity of GlasgowGlasgowUK
| | - Lizi M. Hegarty
- Queens Medical Research InstituteUniversity of Edinburgh Centre for Inflammation ResearchEdinburghUK
| | - Clare Pridans
- Queens Medical Research InstituteUniversity of Edinburgh Centre for Inflammation ResearchEdinburghUK
- Simons Initiative for the Developing Brain, Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Simon W.F. Milling
- Institute of Infection, Immunity, and InflammationUniversity of GlasgowGlasgowUK
| | - Andrew S. MacDonald
- Lydia Becker Institute for Immunology and Infection, School of Biological Sciences, Faculty of Biology, Medicine & HealthUniversity of ManchesterManchesterUK
| | - Dominik Rückerl
- Lydia Becker Institute for Immunology and Infection, School of Biological Sciences, Faculty of Biology, Medicine & HealthUniversity of ManchesterManchesterUK
| | - Stephen J. Jenkins
- Queens Medical Research InstituteUniversity of Edinburgh Centre for Inflammation ResearchEdinburghUK
| |
Collapse
|
218
|
Dong X, Liang Z, Zhang J, Zhang Q, Xu Y, Zhang Z, Zhang L, Zhang B, Zhao Y. Trappc1 deficiency impairs thymic epithelial cell development by breaking endoplasmic reticulum homeostasis. Eur J Immunol 2022; 52:1789-1804. [PMID: 35908180 DOI: 10.1002/eji.202249915] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/28/2022] [Accepted: 07/26/2022] [Indexed: 11/05/2022]
Abstract
Thymic epithelial cells (TECs) are important for T cell development and immune tolerance establishment. Although comprehensive molecular regulation of TEC development has been studied, the role of transport protein particle complexes (Trappcs) in TECs is not clear. Using TEC-specific homozygous or heterozygous Trappc1 deleted mice model, we found that Trappc1 deficiency caused severe thymus atrophy with decreased cell number and blocked maturation of TECs. Mice with a TEC-specific Trappc1 deletion show poor thymic T cell output and have a greater percentage of activated/memory T cells, suffered from spontaneous autoimmune disorders. Our RNA-seq and molecular studies indicated that the decreased endoplasmic reticulum (ER) and Golgi apparatus, enhanced unfolded protein response (UPR) and subsequent Atf4-CHOP-mediated apoptosis, and reactive oxygen species (ROS)-mediated ferroptosis coordinately contributed to the reduction of Trappc1-deleted TECs. Additionally, reduced Aire+ mTECs accompanied by the decreased expression of Irf4, Irf8, and Tbx21 in Trappc1 deficiency mTECs, may further coordinately block the tissue-restricted antigen expression. In this study, we reveal that Trappc1 plays an indispensable role in TEC development and maturation and provide evidence for the importance of inter-organelle traffic and ER homeostasis in TEC development. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Xue Dong
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences.,University of Chinese Academy of Sciences
| | - Zhanfeng Liang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences.,University of Chinese Academy of Sciences.,Beijing Institute for Stem Cell and Regeneration
| | - Jiayu Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences.,University of Chinese Academy of Sciences
| | - Qian Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences.,University of Chinese Academy of Sciences
| | - Yanan Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences.,University of Chinese Academy of Sciences
| | - Zhaoqi Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences.,University of Chinese Academy of Sciences
| | - Lianfeng Zhang
- Key Laboratory of Human Diseases and Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences.,University of Chinese Academy of Sciences.,Beijing Institute for Stem Cell and Regeneration
| |
Collapse
|
219
|
Lo Tartaro D, Camiro-Zúñiga A, Nasi M, De Biasi S, Najera-Avila MA, Jaramillo-Jante MDR, Gibellini L, Pinti M, Neroni A, Mussini C, Soto-Ramírez LE, Calva JJ, Belaunzarán-Zamudio F, Crabtree-Ramirez B, Hernández-Leon C, Mosqueda-Gómez JL, Navarro-Álvarez S, Perez-Patrigeon S, Cossarizza A. Effective Treatment of Patients Experiencing Primary, Acute HIV Infection Decreases Exhausted/Activated CD4+ T Cells and CD8+ T Memory Stem Cells. Cells 2022; 11:cells11152307. [PMID: 35954153 PMCID: PMC9367582 DOI: 10.3390/cells11152307] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Several studies have identified main changes in T- and B-lymphocyte subsets during chronic HIV infection, but few data exist on how these subsets behave during the initial phase of HIV infection. We enrolled 22 HIV-infected patients during the acute stage of infection before the initiation of antiretroviral therapy (ART). Patients had blood samples drawn previous to ART initiation (T0), and at 2 (T1) and 12 (T2) months after ART initiation. We quantified cellular HIV-DNA content in sorted naïve and effector memory CD4 T cells and identified the main subsets of T- and B-lymphocytes using an 18-parameter flow cytometry panel. We identified correlations between the patients’ clinical and immunological data using PCA. Effective HIV treatment reduces integrated HIV DNA in effector memory T cells after 12 months (T2) of ART. The main changes in CD4+ T cells occurred at T2, with a reduction of activated memory, cytolytic and activated/exhausted stem cell memory T (TSCM) cells. Changes were present among CD8+ T cells since T1, with a reduction of several activated subsets, including activated/exhausted TSCM. At T2 a reduction of plasmablasts and exhausted B cells was also observed. A negative correlation was found between the total CD4+ T-cell count and IgM-negative plasmablasts. In patients initiating ART immediately following acute/early HIV infection, the fine analysis of T- and B-cell subsets has allowed us to identify and follow main modifications due to effective treatment, and to identify significant changes in CD4+ and CD8+ T memory stem cells.
Collapse
Affiliation(s)
- Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (D.L.T.); (S.D.B.); (L.G.); (A.N.)
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Antonio Camiro-Zúñiga
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Infectious Diseases, Mexico City 14080, Mexico; (A.C.-Z.); (M.A.N.-A.); (M.D.R.J.-J.); (L.E.S.-R.); (J.J.C.); (F.B.-Z.); (B.C.-R.); (S.P.-P.)
| | - Milena Nasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy
- Correspondence: (M.N.); (A.C.); Tel.: +39-059-205-5415 (M.N.); +39-059-205-5422 (A.C.)
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (D.L.T.); (S.D.B.); (L.G.); (A.N.)
| | - Marco A. Najera-Avila
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Infectious Diseases, Mexico City 14080, Mexico; (A.C.-Z.); (M.A.N.-A.); (M.D.R.J.-J.); (L.E.S.-R.); (J.J.C.); (F.B.-Z.); (B.C.-R.); (S.P.-P.)
| | - Maria Del Rocio Jaramillo-Jante
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Infectious Diseases, Mexico City 14080, Mexico; (A.C.-Z.); (M.A.N.-A.); (M.D.R.J.-J.); (L.E.S.-R.); (J.J.C.); (F.B.-Z.); (B.C.-R.); (S.P.-P.)
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (D.L.T.); (S.D.B.); (L.G.); (A.N.)
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (D.L.T.); (S.D.B.); (L.G.); (A.N.)
| | - Cristina Mussini
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria Policlinico di Modena, 41124 Modena, Italy;
| | - Luis E. Soto-Ramírez
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Infectious Diseases, Mexico City 14080, Mexico; (A.C.-Z.); (M.A.N.-A.); (M.D.R.J.-J.); (L.E.S.-R.); (J.J.C.); (F.B.-Z.); (B.C.-R.); (S.P.-P.)
| | - Juan J. Calva
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Infectious Diseases, Mexico City 14080, Mexico; (A.C.-Z.); (M.A.N.-A.); (M.D.R.J.-J.); (L.E.S.-R.); (J.J.C.); (F.B.-Z.); (B.C.-R.); (S.P.-P.)
| | - Francisco Belaunzarán-Zamudio
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Infectious Diseases, Mexico City 14080, Mexico; (A.C.-Z.); (M.A.N.-A.); (M.D.R.J.-J.); (L.E.S.-R.); (J.J.C.); (F.B.-Z.); (B.C.-R.); (S.P.-P.)
| | - Brenda Crabtree-Ramirez
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Infectious Diseases, Mexico City 14080, Mexico; (A.C.-Z.); (M.A.N.-A.); (M.D.R.J.-J.); (L.E.S.-R.); (J.J.C.); (F.B.-Z.); (B.C.-R.); (S.P.-P.)
| | - Christian Hernández-Leon
- Centro Ambulatorio para la Prevención y Atención del Sida e Infecciones de Transmisión Sexual (CAPASITS), Puebla 72410, Mexico;
| | - Juan L. Mosqueda-Gómez
- Centro Ambulatorio para la Prevención y Atención del Sida e Infecciones de Transmisión Sexual (CAPASITS), Leon 37320, Mexico;
| | | | - Santiago Perez-Patrigeon
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Infectious Diseases, Mexico City 14080, Mexico; (A.C.-Z.); (M.A.N.-A.); (M.D.R.J.-J.); (L.E.S.-R.); (J.J.C.); (F.B.-Z.); (B.C.-R.); (S.P.-P.)
- Division of Infectious Diseases, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (D.L.T.); (S.D.B.); (L.G.); (A.N.)
- National Institute for Cardiovascular Research—INRC, 40126 Bologna, Italy
- Correspondence: (M.N.); (A.C.); Tel.: +39-059-205-5415 (M.N.); +39-059-205-5422 (A.C.)
| |
Collapse
|
220
|
Jacobsen H, Cobos Jiménez V, Sitaras I, Bar-Zeev N, Čičin-Šain L, Higdon MM, Deloria-Knoll M. Post-vaccination T cell immunity to omicron. Front Immunol 2022; 13:944713. [PMID: 35990661 PMCID: PMC9386871 DOI: 10.3389/fimmu.2022.944713] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
In late 2021, the omicron variant of SARS Coronavirus 2 (SARS-CoV-2) emerged and replaced the previously dominant delta strain. Effectiveness of COVID-19 vaccines against omicron has been challenging to estimate in clinical studies or is not available for all vaccines or populations of interest. T cell function can be predictive of vaccine longevity and effectiveness against disease, likely in a more robust way than antibody neutralization. In this mini review, we summarize the evidence on T cell immunity against omicron including effects of boosters, homologous versus heterologous regimens, hybrid immunity, memory responses and vaccine product. Overall, T cell reactivity in post-vaccine specimens is largely preserved against omicron, indicating that vaccines utilizing the parental antigen continue to be protective against disease caused by the omicron variant.
Collapse
Affiliation(s)
- Henning Jacobsen
- Department of Viral Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Viviana Cobos Jiménez
- Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Ioannis Sitaras
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Naor Bar-Zeev
- International Vaccine Access Center, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Luka Čičin-Šain
- Department of Viral Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany
- Centre for Individualised Infection Medicine (CIIM), a joint venture of HZI and MHH, Hannover, Germany
- German Centre for Infection Research (DZIF), Hannover-Braunschweig site, Germany
| | - Melissa M. Higdon
- International Vaccine Access Center, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Maria Deloria-Knoll
- International Vaccine Access Center, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
221
|
Paolini A, Borella R, Neroni A, Lo Tartaro D, Mattioli M, Fidanza L, Di Nella A, Santacroce E, Gozzi L, Busani S, Trenti T, Meschiari M, Guaraldi G, Girardis M, Mussini C, Gibellini L, De Biasi S, Cossarizza A. Patients Recovering from Severe COVID-19 Develop a Polyfunctional Antigen-Specific CD4+ T Cell Response. Int J Mol Sci 2022; 23:8004. [PMID: 35887351 PMCID: PMC9323836 DOI: 10.3390/ijms23148004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 12/10/2022] Open
Abstract
Specific T cells are crucial to control SARS-CoV-2 infection, avoid reinfection and confer protection after vaccination. We have studied patients with severe or moderate COVID-19 pneumonia, compared to patients who recovered from a severe or moderate infection that had occurred about 4 months before the analyses. In all these subjects, we assessed the polyfunctionality of virus-specific CD4+ and CD8+ T cells by quantifying cytokine production after in vitro stimulation with different SARS-CoV-2 peptide pools covering different proteins (M, N and S). In particular, we quantified the percentage of CD4+ and CD8+ T cells simultaneously producing interferon-γ, tumor necrosis factor, interleukin (IL)-2, IL-17, granzyme B, and expressing CD107a. Recovered patients who experienced a severe disease display high proportions of antigen-specific CD4+ T cells producing Th1 and Th17 cytokines and are characterized by polyfunctional SARS-CoV-2-specific CD4+ T cells. A similar profile was found in patients experiencing a moderate form of COVID-19 pneumonia. No main differences in polyfunctionality were observed among the CD8+ T cell compartments, even if the proportion of responding cells was higher during the infection. The identification of those functional cell subsets that might influence protection can thus help in better understanding the complexity of immune response to SARS-CoV-2.
Collapse
Affiliation(s)
- Annamaria Paolini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Marco Mattioli
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Lucia Fidanza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Alessia Di Nella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Elena Santacroce
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Licia Gozzi
- Infectious Diseases Clinics, AOU Policlinico di Modena, Via del Pozzo 71, 41124 Modena, Italy; (L.G.); (M.M.); (G.G.); (C.M.)
| | - Stefano Busani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (S.B.); (M.G.)
- Department of Anesthesia and Intensive Care, AOU Policlinico and University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124 Modena, Italy;
| | - Marianna Meschiari
- Infectious Diseases Clinics, AOU Policlinico di Modena, Via del Pozzo 71, 41124 Modena, Italy; (L.G.); (M.M.); (G.G.); (C.M.)
| | - Giovanni Guaraldi
- Infectious Diseases Clinics, AOU Policlinico di Modena, Via del Pozzo 71, 41124 Modena, Italy; (L.G.); (M.M.); (G.G.); (C.M.)
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (S.B.); (M.G.)
| | - Massimo Girardis
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (S.B.); (M.G.)
- Department of Anesthesia and Intensive Care, AOU Policlinico and University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy
| | - Cristina Mussini
- Infectious Diseases Clinics, AOU Policlinico di Modena, Via del Pozzo 71, 41124 Modena, Italy; (L.G.); (M.M.); (G.G.); (C.M.)
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (S.B.); (M.G.)
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125 Modena, Italy; (A.P.); (R.B.); (A.N.); (D.L.T.); (M.M.); (L.F.); (A.D.N.); (E.S.); (L.G.); (A.C.)
- National Institute for Cardiovascular Research, Via Irnerio 48, 40126 Bologna, Italy
| |
Collapse
|
222
|
Lamacchia G, Mazzoni A, Spinicci M, Vanni A, Salvati L, Peruzzi B, Bencini S, Capone M, Carnasciali A, Farahvachi P, Rocca A, Kiros ST, Graziani L, Zammarchi L, Mencarini J, Colao MG, Caporale R, Liotta F, Cosmi L, Rossolini GM, Bartoloni A, Maggi L, Annunziato F. Clinical and Immunological Features of SARS-CoV-2 Breakthrough Infections in Vaccinated Individuals Requiring Hospitalization. J Clin Immunol 2022; 42:1379-1391. [PMID: 35809212 PMCID: PMC9674730 DOI: 10.1007/s10875-022-01325-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/30/2022] [Indexed: 12/29/2022]
Abstract
Background and Purpose Waning immunity and the surge of SARS-CoV-2 variants are responsible for breakthrough infections, i.e., infections in fully vaccinated individuals. Although the majority of vaccinated infected subjects report mild or no symptoms, some others require hospitalization. The clinical and immunological features of vaccinated hospitalized COVID-19 patients are currently unknown. Methods Twenty-nine unvaccinated and 36 vaccinated hospitalized COVID-19 patients were prospectively enrolled and clinical and laboratory data were gathered. Immunophenotyping of leukocytes’ subsets, T and B cell SARS-CoV-2-specific responses were evaluated via flow cytometry. Anti-IFN-α autoantibodies were measured via ELISA. Results Despite vaccinated patients were older and with more comorbidities, unvaccinated subjects showed higher levels of pro-inflammatory markers, more severe disease, and increased mortality rate. Accordingly, they presented significant alterations in the circulating leukocyte composition, typical of severe COVID-19. Vaccinated patients displayed higher levels of anti-Spike IgGs and Spike-specific B cells. Of all participants, survivors showed higher levels of anti-Spike IgGs and Spike-specific CD4+ T cells than non-survivors. At hospital admission, 6 out of 65 patients (9.2%) displayed high serum concentrations of autoantibodies targeting IFN-α. Remarkably, 3 were unvaccinated and eventually died, while the other 3 were vaccinated and survived. Conclusion Despite more severe pre-existing clinical conditions, vaccinated patients have good outcome. A rapid activation of anti-SARS-CoV-2-specific immunity is fundamental for the resolution of the infection. Therefore, prior immunization through vaccination provides a significant contribution to prevention of disease worsening and can even overcome the presence of high-risk factors (i.e., older age, comorbidities, anti-IFN-α autoantibodies). Supplementary Information The online version contains supplementary material available at 10.1007/s10875-022-01325-2.
Collapse
Affiliation(s)
- Giulia Lamacchia
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Michele Spinicci
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy.,Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Anna Vanni
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Lorenzo Salvati
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Benedetta Peruzzi
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| | - Sara Bencini
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Alberto Carnasciali
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Parham Farahvachi
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Arianna Rocca
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Seble Tekle Kiros
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Lucia Graziani
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy
| | - Lorenzo Zammarchi
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy.,Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Jessica Mencarini
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Maria Grazia Colao
- Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Roberto Caporale
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy.,Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy.,Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy.,Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy.,Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Alessandro Bartoloni
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy.,Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy.
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Viale Pieraccini, 6, 50134, Florence, Italy. .,Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
223
|
Milardi G, Di Lorenzo B, Gerosa J, Barzaghi F, Di Matteo G, Omrani M, Jofra T, Merelli I, Barcella M, Filippini M, Conti A, Ferrua F, Pozzo Giuffrida F, Dionisio F, Rovere‐Querini P, Marktel S, Assanelli A, Piemontese S, Brigida I, Zoccolillo M, Cirillo E, Giardino G, Danieli MG, Specchia F, Pacillo L, Di Cesare S, Giancotta C, Romano F, Matarese A, Chetta AA, Trimarchi M, Laurenzi A, De Pellegrin M, Darin S, Montin D, Marinoni M, Dellepiane RM, Sordi V, Lougaris V, Vacca A, Melzi R, Nano R, Azzari C, Bongiovanni L, Pignata C, Cancrini C, Plebani A, Piemonti L, Petrovas C, Di Micco R, Ponzoni M, Aiuti A, Cicalese MP, Fousteri G. Follicular helper T cell signature of replicative exhaustion, apoptosis, and senescence in common variable immunodeficiency. Eur J Immunol 2022; 52:1171-1189. [PMID: 35562849 PMCID: PMC9542315 DOI: 10.1002/eji.202149480] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 02/08/2022] [Accepted: 05/09/2022] [Indexed: 11/06/2022]
Abstract
Common variable immunodeficiency (CVID) is the most frequent primary antibody deficiency whereby follicular helper T (Tfh) cells fail to establish productive responses with B cells in germinal centers. Here, we analyzed the frequency, phenotype, transcriptome, and function of circulating Tfh (cTfh) cells in CVID patients displaying autoimmunity as an additional phenotype. A group of patients showed a high frequency of cTfh1 cells and a prominent expression of PD-1 and ICOS as well as a cTfh mRNA signature consistent with highly activated, but exhausted, senescent, and apoptotic cells. Plasmatic CXCL13 levels were elevated in this group and positively correlated with cTfh1 cell frequency and PD-1 levels. Monoallelic variants in RTEL1, a telomere length- and DNA repair-related gene, were identified in four patients belonging to this group. Their blood lymphocytes showed shortened telomeres, while their cTfh were more prone to apoptosis. These data point toward a novel pathogenetic mechanism in CVID, whereby alterations in DNA repair and telomere elongation might predispose to antibody deficiency. A Th1, highly activated but exhausted and apoptotic cTfh phenotype was associated with this form of CVID.
Collapse
Affiliation(s)
- Giulia Milardi
- Division of Immunology, Transplantation, and Infectious DiseasesDiabetes Research InstituteIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Biagio Di Lorenzo
- Division of Immunology, Transplantation, and Infectious DiseasesDiabetes Research InstituteIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Jolanda Gerosa
- Division of Immunology, Transplantation, and Infectious DiseasesDiabetes Research InstituteIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Federica Barzaghi
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
- Pathogenesis and therapy of primary immunodeficiencies UnitSan Raffaele Telethon Institute for Gene TherapySr‐TIGETIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Gigliola Di Matteo
- Department of Systems Medicine, University of Rome Tor VergataVia Cracovia 50Rome00133Italy
- Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Academic Department of PediatricsBambino Gesù Children's HospitalIRCCSPiazza di Sant'Onofrio 4Rome00165Italy
| | - Maryam Omrani
- Pathogenesis and therapy of primary immunodeficiencies UnitSan Raffaele Telethon Institute for Gene TherapySr‐TIGETIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
- Department of Computer Science, Systems and Communication, University of Milano‐BicoccaPiazza dell'Ateneo Nuovo 1Milan20126Italy
| | - Tatiana Jofra
- Division of Immunology, Transplantation, and Infectious DiseasesDiabetes Research InstituteIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Ivan Merelli
- Pathogenesis and therapy of primary immunodeficiencies UnitSan Raffaele Telethon Institute for Gene TherapySr‐TIGETIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
- Department of BioinformaticsInstitute for Biomedical TechnologiesNational Research CouncilVia Fratelli Cervi 93Segrate20090Italy
| | - Matteo Barcella
- Pathogenesis and therapy of primary immunodeficiencies UnitSan Raffaele Telethon Institute for Gene TherapySr‐TIGETIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Matteo Filippini
- Division of Immunology, Transplantation, and Infectious DiseasesDiabetes Research InstituteIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Anastasia Conti
- Pathogenesis and therapy of primary immunodeficiencies UnitSan Raffaele Telethon Institute for Gene TherapySr‐TIGETIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Francesca Ferrua
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
- Pathogenesis and therapy of primary immunodeficiencies UnitSan Raffaele Telethon Institute for Gene TherapySr‐TIGETIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Francesco Pozzo Giuffrida
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
- Pathogenesis and therapy of primary immunodeficiencies UnitSan Raffaele Telethon Institute for Gene TherapySr‐TIGETIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Francesca Dionisio
- Pathogenesis and therapy of primary immunodeficiencies UnitSan Raffaele Telethon Institute for Gene TherapySr‐TIGETIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Patrizia Rovere‐Querini
- Department of ImmunologyTransplantation and Infectious DiseasesIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Sarah Marktel
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Andrea Assanelli
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Simona Piemontese
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Immacolata Brigida
- Pathogenesis and therapy of primary immunodeficiencies UnitSan Raffaele Telethon Institute for Gene TherapySr‐TIGETIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Matteo Zoccolillo
- Pathogenesis and therapy of primary immunodeficiencies UnitSan Raffaele Telethon Institute for Gene TherapySr‐TIGETIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Emilia Cirillo
- Department of Translational Medical SciencesSection of PediatricsFederico II University of NaplesCorso Umberto I, 40, 80138Italy
| | - Giuliana Giardino
- Department of Translational Medical SciencesSection of PediatricsFederico II University of NaplesCorso Umberto I, 40, 80138Italy
| | - Maria Giovanna Danieli
- Department of Clinical and Molecular SciencesMarche Polytechnic University of AnconaClinica MedicaVia Tronto 10/aAncona60126Italy
| | - Fernando Specchia
- Department of PediatricsS. Orsola‐Malpighi HospitalUniversity of BolognaVia Giuseppe Massarenti 9Bologna40138Italy
| | - Lucia Pacillo
- Department of Systems Medicine, University of Rome Tor VergataVia Cracovia 50Rome00133Italy
- Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Academic Department of PediatricsBambino Gesù Children's HospitalIRCCSPiazza di Sant'Onofrio 4Rome00165Italy
| | - Silvia Di Cesare
- Department of Systems Medicine, University of Rome Tor VergataVia Cracovia 50Rome00133Italy
- Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Academic Department of PediatricsBambino Gesù Children's HospitalIRCCSPiazza di Sant'Onofrio 4Rome00165Italy
| | - Carmela Giancotta
- Department of Systems Medicine, University of Rome Tor VergataVia Cracovia 50Rome00133Italy
- Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Academic Department of PediatricsBambino Gesù Children's HospitalIRCCSPiazza di Sant'Onofrio 4Rome00165Italy
| | - Francesca Romano
- Pediatric Immunology DivisionDepartment of PediatricsAnna Meyer Children's University HospitalViale Gaetano Pieraccini 24Florence50139Italy
| | - Alessandro Matarese
- Department of Respiratory MedicineSanti AntonioBiagio and Cesare Arrigo HospitalVia Venezia 16Alessandria15121Italy
| | - Alfredo Antonio Chetta
- Department of Medicine and SurgeryRespiratory Disease and Lung Function UnitUniversity of ParmaStr. dell'Università 12Parma43121Italy
| | - Matteo Trimarchi
- Otorhinolaryngology Unit, Head and Neck Department, IRCCS San Raffaele Scientific InstituteVia Olgettina 60Milan20132Italy
- Pathology UnitIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Andrea Laurenzi
- Division of Immunology, Transplantation, and Infectious DiseasesDiabetes Research InstituteIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Maurizio De Pellegrin
- Unit of Orthopaedics, IRCCS San Raffaele Scientific InstituteVia Olgettina 60Milan20132Italy
| | - Silvia Darin
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Davide Montin
- Department of Pediatrics and Public HealthRegina Margherita HospitalPiazza Polonia 94Turin10126Italy
| | - Maddalena Marinoni
- Pediatric UnitOspedale “F. Del Ponte”Via Filippo del Ponte 19Varese21100Italy
| | - Rosa Maria Dellepiane
- Department of PediatricsFondazione IRCCS Cà Granda Ospedale Maggiore PoliclinicoUniversity of MilanVia Francesco Sforza 35Milan20122Italy
| | - Valeria Sordi
- Division of Immunology, Transplantation, and Infectious DiseasesDiabetes Research InstituteIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Vassilios Lougaris
- Department of Clinical and Experimental SciencesPediatrics Clinic and Institute for Molecular Medicine A. NocivelliUniversity of BresciaPiazza del Mercato 15Brescia25121Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human OncologyUniversity of Bari Medical SchoolPiazza Umberto I, 1Bari70121Italy
| | - Raffaella Melzi
- Division of Immunology, Transplantation, and Infectious DiseasesDiabetes Research InstituteIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Rita Nano
- Division of Immunology, Transplantation, and Infectious DiseasesDiabetes Research InstituteIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Chiara Azzari
- Pediatric Immunology DivisionDepartment of PediatricsAnna Meyer Children's University HospitalViale Gaetano Pieraccini 24Florence50139Italy
| | - Lucia Bongiovanni
- Pathology UnitIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Claudio Pignata
- Department of Translational Medical SciencesSection of PediatricsFederico II University of NaplesCorso Umberto I, 40, 80138Italy
| | - Caterina Cancrini
- Department of Systems Medicine, University of Rome Tor VergataVia Cracovia 50Rome00133Italy
- Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Academic Department of PediatricsBambino Gesù Children's HospitalIRCCSPiazza di Sant'Onofrio 4Rome00165Italy
| | - Alessandro Plebani
- Department of Clinical and Experimental SciencesPediatrics Clinic and Institute for Molecular Medicine A. NocivelliUniversity of BresciaPiazza del Mercato 15Brescia25121Italy
| | - Lorenzo Piemonti
- Division of Immunology, Transplantation, and Infectious DiseasesDiabetes Research InstituteIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
- Faculty of MedicineUniversity Vita‐Salute San RaffaeleVia Olgettina 60Milan20132Italy
| | - Constantinos Petrovas
- Tissue Analysis Core, Immunology LaboratoryVaccine Research CenterNational Institute of Allergy and Infectious DiseasesNational Institutes of Health9000 Rockville PikeBethesdaMD20892USA
| | - Raffaella Di Micco
- Pathogenesis and therapy of primary immunodeficiencies UnitSan Raffaele Telethon Institute for Gene TherapySr‐TIGETIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Maurilio Ponzoni
- Pathology UnitIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
- Faculty of MedicineUniversity Vita‐Salute San RaffaeleVia Olgettina 60Milan20132Italy
| | - Alessandro Aiuti
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
- Pathogenesis and therapy of primary immunodeficiencies UnitSan Raffaele Telethon Institute for Gene TherapySr‐TIGETIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
- Faculty of MedicineUniversity Vita‐Salute San RaffaeleVia Olgettina 60Milan20132Italy
| | - Maria Pia Cicalese
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
- Pathogenesis and therapy of primary immunodeficiencies UnitSan Raffaele Telethon Institute for Gene TherapySr‐TIGETIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| | - Georgia Fousteri
- Division of Immunology, Transplantation, and Infectious DiseasesDiabetes Research InstituteIRCCS San Raffaele HospitalVia Olgettina 60Milan20132Italy
| |
Collapse
|
224
|
Wiech M, Chroscicki P, Swatler J, Stepnik D, De Biasi S, Hampel M, Brewinska-Olchowik M, Maliszewska A, Sklinda K, Durlik M, Wierzba W, Cossarizza A, Piwocka K. Remodeling of T Cell Dynamics During Long COVID Is Dependent on Severity of SARS-CoV-2 Infection. Front Immunol 2022; 13:886431. [PMID: 35757700 PMCID: PMC9226563 DOI: 10.3389/fimmu.2022.886431] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/22/2022] [Indexed: 12/14/2022] Open
Abstract
Several COVID-19 convalescents suffer from the post-acute COVID-syndrome (PACS)/long COVID, with symptoms that include fatigue, dyspnea, pulmonary fibrosis, cognitive dysfunctions or even stroke. Given the scale of the worldwide infections, the long-term recovery and the integrative health-care in the nearest future, it is critical to understand the cellular and molecular mechanisms as well as possible predictors of the longitudinal post-COVID-19 responses in convalescent individuals. The immune system and T cell alterations are proposed as drivers of post-acute COVID syndrome. However, despite the number of studies on COVID-19, many of them addressed only the severe convalescents or the short-term responses. Here, we performed longitudinal studies of mild, moderate and severe COVID-19-convalescent patients, at two time points (3 and 6 months from the infection), to assess the dynamics of T cells immune landscape, integrated with patients-reported symptoms. We show that alterations among T cell subsets exhibit different, severity- and time-dependent dynamics, that in severe convalescents result in a polarization towards an exhausted/senescent state of CD4+ and CD8+ T cells and perturbances in CD4+ Tregs. In particular, CD8+ T cells exhibit a high proportion of CD57+ terminal effector cells, together with significant decrease of naïve cell population, augmented granzyme B and IFN-γ production and unresolved inflammation 6 months after infection. Mild convalescents showed increased naïve, and decreased central memory and effector memory CD4+ Treg subsets. Patients from all severity groups can be predisposed to the long COVID symptoms, and fatigue and cognitive dysfunctions are not necessarily related to exhausted/senescent state and T cell dysfunctions, as well as unresolved inflammation that was found only in severe convalescents. In conclusion, the post-COVID-19 functional remodeling of T cells could be seen as a two-step process, leading to distinct convalescent immune states at 6 months after infection. Our data imply that attenuation of the functional polarization together with blocking granzyme B and IFN-γ in CD8+ cells might influence post-COVID alterations in severe convalescents. However, either the search for long COVID predictors or any treatment to prevent PACS and further complications is mandatory in all patients with SARS-CoV-2 infection, and not only in those suffering from severe COVID-19.
Collapse
Affiliation(s)
- Milena Wiech
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Piotr Chroscicki
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Julian Swatler
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Dawid Stepnik
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Michal Hampel
- Department of Gastroenterological Surgery and Transplantology, Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland
| | - Marta Brewinska-Olchowik
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Maliszewska
- Department of Gastroenterological Surgery and Transplantology, Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland
| | - Katarzyna Sklinda
- Department of Radiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Marek Durlik
- Department of Gastroenterological Surgery and Transplantology, Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland.,Departament of Gastroenterological Surgery and Transplantology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Waldemar Wierzba
- Central Clinical Hospital of the Ministry of Interior, Warsaw, Poland.,University of Humanities and Economics, Lodz, Poland
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy.,National Institute for Cardiovascular Research, Bologna, Italy
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
225
|
Lam AJ, Haque M, Ward-Hartstonge KA, Uday P, Wardell CM, Gillies JK, Speck M, Mojibian M, Klein Geltink RI, Levings MK. PTEN is required for human Treg suppression of costimulation in vitro. Eur J Immunol 2022; 52:1482-1497. [PMID: 35746855 DOI: 10.1002/eji.202249888] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/18/2022] [Accepted: 06/10/2022] [Indexed: 11/07/2022]
Abstract
Regulatory T cell (Treg) therapy is under clinical investigation for the treatment of transplant rejection, autoimmune disease, and graft-versus-host disease. With the advent of genome editing, attention has turned to reinforcing Treg function for therapeutic benefit. A hallmark of Tregs is dampened activation of PI3K-AKT signalling, of which PTEN is a major negative regulator. Loss-of-function studies of PTEN, however, have not conclusively shown a requirement for PTEN in upholding Treg function and stability. Using CRISPR-based genome editing in human Tregs, we show that PTEN ablation does not cause a global defect in Treg function and stability; rather, it selectively blocks their ability to suppress antigen-presenting cells. PTEN-KO Tregs exhibit elevated glycolytic activity, upregulate FOXP3, maintain a Treg phenotype, and have no discernable defects in lineage stability. Functionally, PTEN is dispensable for human Treg-mediated inhibition of T cell activity in vitro and in vivo, but is required for suppression of costimulatory molecule expression by antigen-presenting cells. These data are the first to define a role for a signalling pathway in controlling a subset of human Treg activity. Moreover, they point to the functional necessity of PTEN-regulated PI3K-AKT activity for optimal human Treg function. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Avery J Lam
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Manjurul Haque
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Kirsten A Ward-Hartstonge
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Prakruti Uday
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Christine M Wardell
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Jana K Gillies
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Madeleine Speck
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Majid Mojibian
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Ramon I Klein Geltink
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada.,Department of Molecular Oncology, BC Cancer Research, Vancouver, BC, V5Z 1L3, Canada
| | - Megan K Levings
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, V6T 1Z3, Canada
| |
Collapse
|
226
|
Moschetti G, Vasco C, Clemente F, Galeota E, Carbonara M, Pluderi M, Locatelli M, Stocchetti N, Abrignani S, Zanier ER, Ortolano F, Zoerle T, Geginat J. Deep Phenotyping of T-Cells Derived From the Aneurysm Wall in a Pediatric Case of Subarachnoid Hemorrhage. Front Immunol 2022; 13:866558. [PMID: 35711453 PMCID: PMC9197186 DOI: 10.3389/fimmu.2022.866558] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Intracranial aneurysms (IAs) are very rare in children, and the characteristics of the T-cells in the IA wall are largely unknown. A comatose 7-years-old child was admitted to our center because of a subarachnoid hemorrhage due to a ruptured giant aneurysm of the right middle cerebral artery. Two days after the aneurysm clipping the patient was fully awake with left hemiparesis. T-cells from the IA wall and from peripheral blood of this patient were analyzed by multi-dimensional flow cytometry. Unbiased analysis, based on the use of FlowSOM clustering and dimensionality reduction technique UMAP, indicated that there was virtually no overlap between circulating and tissue-infiltrating T-cells. Thus, naïve T-cells and canonical memory T-cells were largely restricted to peripheral blood, while CD4-CD8-T-cells were strongly enriched in the IA wall. The unique CD4+, CD8+ and CD4-CD8-T-cell clusters from the IA wall expressed high levels of CCR5, Granzyme B and CD69, displaying thus characteristics of cytotoxic and tissue-resident effector cells. Low Ki67 expression indicated that they were nevertheless in a resting state. Among regulatory T-cell subsets, Eomes+Tr1-like cells were strongly enriched in the IA wall. Finally, analysis of cytokine producing capacities unveiled that the IA wall contained poly-functional T-cells, which expressed predominantly IFN-γ, TNF and IL-2. CD4+T-cells co-expressed also CD40L, and produced some IL-17, GM-CSF and IL-10. This report provides to our knowledge the first detailed characterization of the human T-cell compartment in the IA wall.
Collapse
Affiliation(s)
| | - Chiara Vasco
- National Institute for Molecular Genetics (INGM), Milan, Italy
| | | | - Eugenia Galeota
- National Institute for Molecular Genetics (INGM), Milan, Italy
| | - Marco Carbonara
- Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mauro Pluderi
- Department of Neurosurgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Locatelli
- Department of Neurosurgery, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Nino Stocchetti
- Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Sergio Abrignani
- National Institute for Molecular Genetics (INGM), Milan, Italy.,Department of Clinical Sciences and Community Health University Milan, Milan, Italy
| | - Elisa R Zanier
- Laboratory of Acute Brain Injury and Therapeutic Strategies, Department of Neuroscience, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| | - Fabrizio Ortolano
- Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Tommaso Zoerle
- Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Jens Geginat
- National Institute for Molecular Genetics (INGM), Milan, Italy.,Department of Clinical Sciences and Community Health University Milan, Milan, Italy
| |
Collapse
|
227
|
Kucykowicz S, Amin OE, Burton AR, Swadling L, Schmidt NM, Zakeri N, Davies J, Aidoo-Micah G, Stegmann KA, Easom NJ, Jeffery-Smith A, Maini MK, Pallett LJ. Isolation of human intrahepatic leukocytes for phenotypic and functional characterization by flow cytometry. STAR Protoc 2022; 3:101356. [PMID: 35516846 PMCID: PMC9065431 DOI: 10.1016/j.xpro.2022.101356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
With the growing appreciation of tissue-resident immunity, studying tissue-specific immune cells contributing to both homeostasis and disease is imperative. Here, we provide a protocol for the isolation of human intrahepatic leukocytes (IHL) maximizing viability, purity, and yield. Our protocol is scalable by tissue weight, allowing for reproducible and efficient IHL liberation suitable for functional characterization, cell isolation, and profiling by flow (or mass) cytometry. Furthermore, we provide a "guide" to determine an expected IHL yield per gram of tissue processed. For complete details on the use and execution of this protocol, please refer to Stegmann et al. (2016), Pallett et al. (2017), Easom et al. (2018), Swadling et al. (2020), Pallett et al. (2020), and Zakeri et al. (2022).
Collapse
Affiliation(s)
- Stephanie Kucykowicz
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, London, UK
| | - Oliver E. Amin
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, London, UK
- Corresponding author
| | - Alice R. Burton
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, London, UK
| | - Leo Swadling
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, London, UK
| | - Nathalie M. Schmidt
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, London, UK
| | - Nekisa Zakeri
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, London, UK
| | - Jessica Davies
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, London, UK
| | - Gloryanne Aidoo-Micah
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, London, UK
| | - Kerstin A. Stegmann
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, London, UK
| | - Nicholas J. Easom
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, London, UK
| | - Anna Jeffery-Smith
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, London, UK
| | - Mala K. Maini
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, London, UK
| | - Laura J. Pallett
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, London, UK
- Corresponding author
| |
Collapse
|
228
|
Lo Tartaro D, Neroni A, Paolini A, Borella R, Mattioli M, Fidanza L, Quong A, Petes C, Awong G, Douglas S, Lin D, Nieto J, Gozzi L, Franceschini E, Busani S, Nasi M, Mattioli AV, Trenti T, Meschiari M, Guaraldi G, Girardis M, Mussini C, Gibellini L, Cossarizza A, De Biasi S. Molecular and cellular immune features of aged patients with severe COVID-19 pneumonia. Commun Biol 2022; 5:590. [PMID: 35710943 PMCID: PMC9203559 DOI: 10.1038/s42003-022-03537-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/26/2022] [Indexed: 12/12/2022] Open
Abstract
Aging is a major risk factor for developing severe COVID-19, but few detailed data are available concerning immunological changes after infection in aged individuals. Here we describe main immune characteristics in 31 patients with severe SARS-CoV-2 infection who were >70 years old, compared to 33 subjects <60 years of age. Differences in plasma levels of 62 cytokines, landscape of peripheral blood mononuclear cells, T cell repertoire, transcriptome of central memory CD4+ T cells, specific antibodies are reported along with features of lung macrophages. Elderly subjects have higher levels of pro-inflammatory cytokines, more circulating plasmablasts, reduced plasmatic level of anti-S and anti-RBD IgG3 antibodies, lower proportions of central memory CD4+ T cells, more immature monocytes and CD56+ pro-inflammatory monocytes, lower percentages of circulating follicular helper T cells (cTfh), antigen-specific cTfh cells with a less activated transcriptomic profile, lung resident activated macrophages that promote collagen deposition and fibrosis. Our study underlines the importance of inflammation in the response to SARS-CoV-2 and suggests that inflammaging, coupled with the inability to mount a proper anti-viral response, could exacerbate disease severity and the worst clinical outcome in old patients. Patients over the age of 70 show inflammaging and a weaker anti-viral response to SARS-CoV-2, pointing at the immunological changes associated with COVID-19 severity and outcome for aged patients.
Collapse
Affiliation(s)
- Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy
| | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy
| | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy
| | - Marco Mattioli
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy
| | - Lucia Fidanza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy
| | - Andrew Quong
- Fluidigm Corporation, 2 Tower Place, Suite 2000, South San Francisco, 94080, CA, USA
| | - Carlene Petes
- Fluidigm Corporation, 2 Tower Place, Suite 2000, South San Francisco, 94080, CA, USA
| | - Geneve Awong
- Fluidigm Corporation, 2 Tower Place, Suite 2000, South San Francisco, 94080, CA, USA
| | - Samuel Douglas
- Fluidigm Corporation, 2 Tower Place, Suite 2000, South San Francisco, 94080, CA, USA
| | - Dongxia Lin
- Fluidigm Corporation, 2 Tower Place, Suite 2000, South San Francisco, 94080, CA, USA
| | - Jordan Nieto
- Fluidigm Corporation, 2 Tower Place, Suite 2000, South San Francisco, 94080, CA, USA
| | - Licia Gozzi
- Infectious Diseases Clinics, AOU Policlinico di Modena, via del Pozzo 71, 41124, Modena, Italy
| | - Erica Franceschini
- Infectious Diseases Clinics, AOU Policlinico di Modena, via del Pozzo 71, 41124, Modena, Italy
| | - Stefano Busani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy.,Department of Anesthesia and Intensive Care, AOU Policlinico and University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy
| | - Milena Nasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy
| | - Anna Vittoria Mattioli
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy.,National Institute for Cardiovascular Research, via Irnerio 48, 40126, Bologna, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, AUSL/AOU Policlinico, 41124, Modena, Italy
| | - Marianna Meschiari
- Infectious Diseases Clinics, AOU Policlinico di Modena, via del Pozzo 71, 41124, Modena, Italy
| | - Giovanni Guaraldi
- Infectious Diseases Clinics, AOU Policlinico di Modena, via del Pozzo 71, 41124, Modena, Italy.,Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy
| | - Massimo Girardis
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy.,Department of Anesthesia and Intensive Care, AOU Policlinico and University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy
| | - Cristina Mussini
- Infectious Diseases Clinics, AOU Policlinico di Modena, via del Pozzo 71, 41124, Modena, Italy.,Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, via del Pozzo 71, 41124, Modena, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy. .,National Institute for Cardiovascular Research, via Irnerio 48, 40126, Bologna, Italy.
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Via Campi 287, 41125, Modena, Italy.
| |
Collapse
|
229
|
Schulz SR, Hoffmann M, Roth E, Pracht K, Burnett DL, Mazigi O, Schuh W, Manger B, Mielenz D, Goodnow CC, Christ D, Pöhlmann S, Jäck H. Augmented neutralization of SARS-CoV-2 Omicron variant by boost vaccination and monoclonal antibodies. Eur J Immunol 2022; 52:970-977. [PMID: 35253229 PMCID: PMC9087419 DOI: 10.1002/eji.202249841] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/18/2022] [Accepted: 02/25/2022] [Indexed: 11/08/2022]
Abstract
Effective vaccines and monoclonal antibodies have been developed against coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the appearance of virus variants with higher transmissibility and pathogenicity is a major concern because of their potential to escape vaccines and clinically approved SARS-CoV-2- antibodies. Here, we use flow cytometry-based binding and pseudotyped SARS-CoV-2 neutralization assays to determine the efficacy of boost immunization and therapeutic antibodies to neutralize the dominant Omicron variant. We provide compelling evidence that the third vaccination with BNT162b2 increases the amount of neutralizing serum antibodies against Delta and Omicron variants, albeit to a lower degree when compared to the parental Wuhan strain. Therefore, a third vaccination is warranted to increase titers of protective serum antibodies, especially in the case of the Omicron variant. We also found that most clinically approved and otherwise potent therapeutic antibodies against the Delta variant failed to recognize and neutralize the Omicron variant. In contrast, some antibodies under preclinical development potentially neutralized the Omicron variant. Our studies also support using a flow cytometry-based antibody binding assay to rapidly monitor therapeutic candidates and serum titers against emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Sebastian R. Schulz
- Division of Molecular ImmunologyDepartment of Internal Medicine 3Nikolaus‐Fiebiger‐ZentrumFriedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| | - Markus Hoffmann
- Infection Biology UnitGerman Primate Center‐Leibniz Institute for Primate ResearchGöttingenGermany
- Faculty of Biology and PsychologyUniversity of GöttingenGöttingenGermany
| | - Edith Roth
- Division of Molecular ImmunologyDepartment of Internal Medicine 3Nikolaus‐Fiebiger‐ZentrumFriedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| | - Katharina Pracht
- Division of Molecular ImmunologyDepartment of Internal Medicine 3Nikolaus‐Fiebiger‐ZentrumFriedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| | - Deborah L. Burnett
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia
- Faculty of Medicine, UNSWSt. Vincent's Clinical SchoolSydneyNew South WalesAustralia
| | - Ohan Mazigi
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia
- Faculty of Medicine, UNSWSt. Vincent's Clinical SchoolSydneyNew South WalesAustralia
| | - Wolfgang Schuh
- Division of Molecular ImmunologyDepartment of Internal Medicine 3Nikolaus‐Fiebiger‐ZentrumFriedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| | - Bernhard Manger
- Department of Internal Medicine 3University Hospital ErlangenErlangenBavariaGermany
| | - Dirk Mielenz
- Division of Molecular ImmunologyDepartment of Internal Medicine 3Nikolaus‐Fiebiger‐ZentrumFriedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| | - Christopher C. Goodnow
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia
- UNSWCellular Genomics Futures InstituteSydneyNew South WalesAustralia
| | - Daniel Christ
- Garvan Institute of Medical ResearchSydneyNew South WalesAustralia
- Faculty of Medicine, UNSWSt. Vincent's Clinical SchoolSydneyNew South WalesAustralia
| | - Stefan Pöhlmann
- Infection Biology UnitGerman Primate Center‐Leibniz Institute for Primate ResearchGöttingenGermany
- Faculty of Biology and PsychologyUniversity of GöttingenGöttingenGermany
| | - Hans‐Martin Jäck
- Division of Molecular ImmunologyDepartment of Internal Medicine 3Nikolaus‐Fiebiger‐ZentrumFriedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| |
Collapse
|
230
|
Pohar J, O'Connor R, Manfroi B, Behi ME, Jouneau L, Boudinot P, Bunse M, Uckert W, Luka M, Ménager M, Liblau R, Anderton SM, Fillatreau S. Antigen receptor-engineered Tregs inhibit CNS autoimmunity in cell therapy using non-redundant immune mechanisms in mice. Eur J Immunol 2022; 52:1335-1349. [PMID: 35579560 PMCID: PMC9542066 DOI: 10.1002/eji.202249845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/28/2022] [Accepted: 05/16/2022] [Indexed: 11/21/2022]
Abstract
CD4+FOXP3+ Tregs are currently explored to develop cell therapies against immune‐mediated disorders, with an increasing focus on antigen receptor‐engineered Tregs. Deciphering their mode of action is necessary to identify the strengths and limits of this approach. Here, we addressed this issue in an autoimmune disease of the CNS, EAE. Following disease induction, autoreactive Tregs upregulated LAG‐3 and CTLA‐4 in LNs, while IL‐10 and amphiregulin (AREG) were increased in CNS Tregs. Using genetic approaches, we demonstrated that IL‐10, CTLA‐4, and LAG‐3 were nonredundantly required for the protective function of antigen receptor‐engineered Tregs against EAE in cell therapy whereas AREG was dispensable. Treg‐derived IL‐10 and CTLA‐4 were both required to suppress acute autoreactive CD4+ T‐cell activation, which correlated with disease control. These molecules also affected the accumulation in the recipients of engineered Tregs themselves, underlying complex roles for these molecules. Noteworthy, despite the persistence of the transferred Tregs and their protective effect, autoreactive T cells eventually accumulated in the spleen of treated mice. In conclusion, this study highlights the remarkable power of antigen receptor‐engineered Tregs to appropriately provide multiple suppressive factors nonredundantly necessary to prevent autoimmune attacks.
Collapse
Affiliation(s)
- Jelka Pohar
- Institut Necker Enfants Malades, Institut National de la Santé et de la Recherche Médicale INSERM U1151 - Centre National de la Recherche Scientifique CNRS UMR 8253, 156-160, rue de Vaugirard, Paris, 75015, France
| | | | - Benoît Manfroi
- Institut Necker Enfants Malades, Institut National de la Santé et de la Recherche Médicale INSERM U1151 - Centre National de la Recherche Scientifique CNRS UMR 8253, 156-160, rue de Vaugirard, Paris, 75015, France
| | - Mohamed El Behi
- Institut Necker Enfants Malades, Institut National de la Santé et de la Recherche Médicale INSERM U1151 - Centre National de la Recherche Scientifique CNRS UMR 8253, 156-160, rue de Vaugirard, Paris, 75015, France
| | - Luc Jouneau
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, 78350, France
| | - Pierre Boudinot
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, 78350, France
| | - Mario Bunse
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Wolfgang Uckert
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Marine Luka
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, Paris, F-75015, France.,Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, Paris, F-75015, France
| | - Mickael Ménager
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, Paris, F-75015, France.,Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, Paris, F-75015, France
| | - Roland Liblau
- Infinity - Institut Toulousain des Maladies Infectieuses et Inflammatoires, NSERM UMR1291 - CNRS UMR5051 - Université Toulouse III, Toulouse, France
| | | | - Simon Fillatreau
- Institut Necker Enfants Malades, Institut National de la Santé et de la Recherche Médicale INSERM U1151 - Centre National de la Recherche Scientifique CNRS UMR 8253, 156-160, rue de Vaugirard, Paris, 75015, France.,Université de Paris, Faculté de Médecine, Paris, France.,AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| |
Collapse
|
231
|
Wiechers C, Pezoldt J, Beckstette M, Berner J, Schraml BU, Huehn J. Lymph node stromal cells support the maturation of pre‐DCs into cDC‐like cells via colony‐stimulating factor 1. Immunology 2022; 166:475-491. [DOI: 10.1111/imm.13497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/18/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Carolin Wiechers
- Department Experimental Immunology Helmholtz Centre for Infection Research Braunschweig Germany
| | - Joern Pezoldt
- Department Experimental Immunology Helmholtz Centre for Infection Research Braunschweig Germany
- Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne Lausanne Switzerland
| | - Michael Beckstette
- Department Experimental Immunology Helmholtz Centre for Infection Research Braunschweig Germany
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine Helmholtz Centre for Infection Research and Hannover Medical School Hannover Germany
| | - Johanna Berner
- Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, Faculty of Medicine, LMU Munich Planegg‐Martinsried Germany
- Walter‐Brendel‐Centre of Experimental Medicine University Hospital, LMU Munich Planegg‐Martinsried Germany
| | - Barbara U. Schraml
- Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, Faculty of Medicine, LMU Munich Planegg‐Martinsried Germany
- Walter‐Brendel‐Centre of Experimental Medicine University Hospital, LMU Munich Planegg‐Martinsried Germany
| | - Jochen Huehn
- Department Experimental Immunology Helmholtz Centre for Infection Research Braunschweig Germany
| |
Collapse
|
232
|
Schmitz J, Brauns N, Hüsing AM, Flechsig M, Glomb T, Bräsen JH, Haller H, von Vietinghoff S. Renal medullary osmolytes NaCl and urea differentially modulate human tubular cell cytokine expression and monocyte recruitment. Eur J Immunol 2022; 52:1258-1272. [DOI: 10.1002/eji.202149723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/20/2022] [Accepted: 05/06/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Jessica Schmitz
- Nephropathology Unit Institute for Pathology University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
| | - Nicolas Brauns
- Department of Internal Medicine Division of Nephrology and Hypertension University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
| | - Anne M. Hüsing
- Department of Internal Medicine Division of Nephrology and Hypertension University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
| | - Martina Flechsig
- Department of Internal Medicine Division of Nephrology and Hypertension University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
| | - Thorsten Glomb
- Core Facility Transcriptomics Hannover Medical School Hannover Germany
| | - Jan Hinrich Bräsen
- Nephropathology Unit Institute for Pathology University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
| | - Hermann Haller
- Department of Internal Medicine Division of Nephrology and Hypertension University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
| | - Sibylle von Vietinghoff
- Department of Internal Medicine Division of Nephrology and Hypertension University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
- Nephrology Section First Medical Clinic University Clinic and Rheinische Friedrich‐Wilhelms Universität Bonn Bonn Germany
| |
Collapse
|
233
|
De Biasi S, Guida A, Lo Tartaro D, Fanelli M, Depenni R, Dominici M, Finak G, Porta C, Paolini A, Borella R, Bertoldi C, Cossarizza A, Sabbatini R, Gibellini L. Redistribution of CD8+ T cell subsets in metastatic renal cell carcinoma patients treated with anti-PD-1 therapy. Cytometry A 2022; 101:597-605. [PMID: 35507402 PMCID: PMC9542732 DOI: 10.1002/cyto.a.24562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 11/20/2022]
Abstract
Renal‐cell carcinoma (RCC) is responsible for the majority of tumors arising from the kidney parenchyma. Although a progressive improvement in median overall survival has been observed after the introduction of anti‐PD‐1 therapy, many patients do not benefit from this treatment. Therefore, we have investigated T cell dynamics to find immune modification induced by anti‐PD‐1 therapy. Here, we show that, after therapy, RCC patients (5 responders and 14 nonresponders) are characterized by a redistribution of different subsets across the memory T cell compartment.
Collapse
Affiliation(s)
- Sara De Biasi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Emilia-Romagna
| | - Annalisa Guida
- Azienda Ospedaliera Santa Maria, Terni, Italy.,Department of Oncology, University of Modena & Reggio Emilia, Modena, Italy
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Emilia-Romagna.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Martina Fanelli
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Emilia-Romagna.,Department of Oncology, University of Modena & Reggio Emilia, Modena, Italy
| | - Roberta Depenni
- Department of Oncology, University of Modena & Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Emilia-Romagna.,Department of Oncology, University of Modena & Reggio Emilia, Modena, Italy
| | - Greg Finak
- Fred Hutchinson Cancer Research Center, Seattle, USA
| | | | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Emilia-Romagna.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Emilia-Romagna.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Carlo Bertoldi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Emilia-Romagna.,National Institute for Cardiovascular Research, Bologna
| | - Roberto Sabbatini
- Department of Oncology, University of Modena & Reggio Emilia, Modena, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Emilia-Romagna
| |
Collapse
|
234
|
Haschka D, Petzer V, Burkert FR, Fritsche G, Wildner S, Bellmann-Weiler R, Tymoszuk P, Weiss G. Alterations of blood monocyte subset distribution and surface phenotype are linked to infection severity in COVID-19 inpatients. Eur J Immunol 2022; 52:1285-1296. [PMID: 35491910 PMCID: PMC9348104 DOI: 10.1002/eji.202149680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 12/13/2022]
Abstract
Severe coronavirus disease 19 (COVID‐19) manifests with systemic immediate proinflammatory innate immune activation and altered iron turnover. Iron homeostasis, differentiation, and function of myeloid leukocytes are interconnected. Therefore, we characterized the cellularity, surface marker expression, and iron transporter phenotype of neutrophils and monocyte subsets in COVID‐19 patients within 72 h from hospital admission, and analyzed how these parameters relate to infection severity. Between March and November 2020, blood leukocyte samples from hospitalized COVID‐19 patients (n = 48) and healthy individuals (n = 7) were analyzed by flow cytometry enabling comparative analysis of 40 features. Inflammation‐driven neutrophil expansion, depletion of CD16+ nonclassical monocytes, and changes in surface expression of neutrophil and monocyte CD64 and CD86 were associated with COVID‐19 severity. By unsupervised self‐organizing map clustering, four patterns of innate myeloid response were identified and linked to varying levels of systemic inflammation, altered cellular iron trafficking and the severity of disease. These alterations of the myeloid leukocyte compartment during acute COVID‐19 may be hallmarks of inefficient viral control and immune hyperactivation and may help at risk prediction and treatment optimization.
Collapse
Affiliation(s)
- David Haschka
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Verena Petzer
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Gernot Fritsche
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Sophie Wildner
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Rosa Bellmann-Weiler
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria.,Data Analytics As a Service Tirol, Innsbruck, Austria
| | - Guenter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
235
|
Hennings V, Thörn K, Albinsson S, Lingblom C, Andersson K, Andersson C, Järbur K, Pullerits R, Idorn M, Paludan SR, Eriksson K, Wennerås C. The presence of serum anti-SARS-CoV-2 IgA appears to protect primary health care workers from COVID-19. Eur J Immunol 2022; 52:800-809. [PMID: 35128644 PMCID: PMC9087394 DOI: 10.1002/eji.202149655] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 12/14/2021] [Accepted: 01/21/2022] [Indexed: 11/15/2022]
Abstract
The patterns of humoral and cellular responses to SARS-CoV-2 were studied in Swedish primary health care workers (n = 156) for 6 months during the Covid-19 pandemic. Serum IgA and IgG to SARS-CoV-2, T-cell proliferation and cytokine secretion, demographic and clinical data, PCR-verified infection, and self-reported symptoms were monitored. The multivariate method OPLS-DA was used to identify immune response patterns coupled to protection from Covid-19. Contracting Covid-19 was associated with SARS-CoV-2-specific neutralizing serum IgG, T cell, IFN-γ, and granzyme B responses to SARS-CoV-2, self-reported typical Covid-19 symptoms, male sex, higher BMI, and hypertension. Not contracting Covid-19 was associated with female sex, IgA-dominated, or no antibody responses to SARS-CoV-2, airborne allergy, and smoking. The IgG-responders had SARS-CoV-2-specific T-cell responses including a cytotoxic CD4+ T-cell population expressing CD25, CD38, CD69, CD194, CD279, CTLA-4, and granzyme B. IgA-responders with no IgG response to SARS-CoV-2 constituted 10% of the study population. The IgA responses were partially neutralizing and only seen in individuals who did not succumb to Covid-19. To conclude, serum IgG-dominated responses correlated with T-cell responses to SARS-CoV-2 and PCR-confirmed Covid-19, whereas IgA-dominated responses correlated with not contracting the infection.
Collapse
Affiliation(s)
- Viktoria Hennings
- Department of Clinical MicrobiologyRegion Västra GötalandSahlgrenska University HospitalGöteborgSweden
| | - Karolina Thörn
- Department of Rheumatology and Inflammation ResearchSahlgrenska AcademyInstitute of MedicineUniversity of GothenburgGöteborgSweden
| | - Sofie Albinsson
- Department of Infectious DiseasesInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGöteborgSweden
| | - Christine Lingblom
- Department of Clinical MicrobiologyRegion Västra GötalandSahlgrenska University HospitalGöteborgSweden
| | - Kerstin Andersson
- Department of Clinical MicrobiologyRegion Västra GötalandSahlgrenska University HospitalGöteborgSweden
| | | | - Katarina Järbur
- Nötkärnan Primary Health Care Center ConsortiumGöteborgSweden
| | - Rille Pullerits
- Department of Clinical MicrobiologyRegion Västra GötalandSahlgrenska University HospitalGöteborgSweden
- Department of Rheumatology and Inflammation ResearchSahlgrenska AcademyInstitute of MedicineUniversity of GothenburgGöteborgSweden
| | - Manja Idorn
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Søren R. Paludan
- Department of Rheumatology and Inflammation ResearchSahlgrenska AcademyInstitute of MedicineUniversity of GothenburgGöteborgSweden
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Kristina Eriksson
- Department of Clinical MicrobiologyRegion Västra GötalandSahlgrenska University HospitalGöteborgSweden
- Department of Rheumatology and Inflammation ResearchSahlgrenska AcademyInstitute of MedicineUniversity of GothenburgGöteborgSweden
| | - Christine Wennerås
- Department of Clinical MicrobiologyRegion Västra GötalandSahlgrenska University HospitalGöteborgSweden
- Department of Infectious DiseasesInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGöteborgSweden
| |
Collapse
|
236
|
Mair F, Erickson JR, Frutoso M, Konecny AJ, Greene E, Voillet V, Maurice NJ, Rongvaux A, Dixon D, Barber B, Gottardo R, Prlic M. Extricating human tumour immune alterations from tissue inflammation. Nature 2022; 605:728-735. [PMID: 35545675 PMCID: PMC9132772 DOI: 10.1038/s41586-022-04718-w] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/01/2022] [Indexed: 12/17/2022]
Abstract
Immunotherapies have achieved remarkable successes in the treatment of cancer, but major challenges remain1,2. An inherent weakness of current treatment approaches is that therapeutically targeted pathways are not restricted to tumours, but are also found in other tissue microenvironments, complicating treatment3,4. Despite great efforts to define inflammatory processes in the tumour microenvironment, the understanding of tumour-unique immune alterations is limited by a knowledge gap regarding the immune cell populations in inflamed human tissues. Here, in an effort to identify such tumour-enriched immune alterations, we used complementary single-cell analysis approaches to interrogate the immune infiltrate in human head and neck squamous cell carcinomas and site-matched non-malignant, inflamed tissues. Our analysis revealed a large overlap in the composition and phenotype of immune cells in tumour and inflamed tissues. Computational analysis identified tumour-enriched immune cell interactions, one of which yields a large population of regulatory T (Treg) cells that is highly enriched in the tumour and uniquely identified among all haematopoietically-derived cells in blood and tissue by co-expression of ICOS and IL-1 receptor type 1 (IL1R1). We provide evidence that these intratumoural IL1R1+ Treg cells had responded to antigen recently and demonstrate that they are clonally expanded with superior suppressive function compared with IL1R1- Treg cells. In addition to identifying extensive immunological congruence between inflamed tissues and tumours as well as tumour-specific changes with direct disease relevance, our work also provides a blueprint for extricating disease-specific changes from general inflammation-associated patterns.
Collapse
Affiliation(s)
- Florian Mair
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA, USA
| | - Jami R Erickson
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Marie Frutoso
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA, USA
| | - Andrew J Konecny
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Evan Greene
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA, USA
| | - Valentin Voillet
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA, USA
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, NPC (HCRISA), Cape Town, South Africa
| | - Nicholas J Maurice
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA
| | - Anthony Rongvaux
- Department of Immunology, University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, USA
| | - Douglas Dixon
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA, USA
- Department of Periodontics, University of Tennessee Health Science Center, College of Dentistry, Memphis, TN, USA
| | - Brittany Barber
- Department of Otolaryngology-Head and Neck Surgery, University of Washington, Seattle, WA, USA
| | - Raphael Gottardo
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA, USA
- Department of Statistics, University of Washington, Seattle, WA, USA
- University of Lausanne and Lausanne University Hospital, Switzerland, Lausanne, Switzerland
| | - Martin Prlic
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
237
|
Hagemann K, Riecken K, Jung J, Hildebrandt H, Menzel S, Bunders M, Fehse B, Koch-Nolte F, Heinrich F, Peine S, Schulze Zur Wiesch J, Brehm TT, Addo MM, Lütgehetmann M, Altfeld M. Natural killer cell-mediated ADCC in SARS-CoV-2-infected individuals and vaccine recipients. Eur J Immunol 2022; 52:1297-1307. [PMID: 35416291 PMCID: PMC9087393 DOI: 10.1002/eji.202149470] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 03/11/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022]
Abstract
COVID‐19, caused by SARS‐CoV‐2, has emerged as a global pandemic. While immune responses of the adaptive immune system have been in the focus of research, the role of NK cells in COVID‐19 remains less well understood. Here, we characterized NK cell‐mediated SARS‐CoV‐2 antibody‐dependent cellular cytotoxicity (ADCC) against SARS‐CoV‐2 spike‐1 (S1) and nucleocapsid (NC) protein. Serum samples from SARS‐CoV‐2 resolvers induced significant CD107a‐expression by NK cells in response to S1 and NC, while serum samples from SARS‐CoV‐2‐negative individuals did not. Furthermore, serum samples from individuals that received the BNT162b2 vaccine induced strong CD107a expression by NK cells that increased with the second vaccination and was significantly higher than observed in infected individuals. As expected, vaccine‐induced responses were only directed against S1 and not against NC protein. S1‐specific CD107a responses by NK cells were significantly correlated to NK cell‐mediated killing of S1‐expressing cells. Interestingly, screening of serum samples collected prior to the COVID‐19 pandemic identified two individuals with cross‐reactive antibodies against SARS‐CoV‐2 S1, which also induced degranulation of NK cells. Taken together, these data demonstrate that antibodies induced by SARS‐CoV‐2 infection and anti‐SARS‐CoV‐2 vaccines can trigger significant NK cell‐mediated ADCC activity, and identify some cross‐reactive ADCC‐activity against SARS‐CoV‐2 by endemic coronavirus‐specific antibodies.
Collapse
Affiliation(s)
- Kerri Hagemann
- Leibniz Institute for Experimental Virology, Department of Virus Immunology, Hamburg, 20251, Germany
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Johannes Jung
- Leibniz Institute for Experimental Virology, Department of Virus Immunology, Hamburg, 20251, Germany
| | - Heike Hildebrandt
- Leibniz Institute for Experimental Virology, Department of Virus Immunology, Hamburg, 20251, Germany
| | - Stephan Menzel
- University Medical Center Hamburg-Eppendorf, Institute of Immunology, Hamburg, 20246, Germany
| | - Madeleine Bunders
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany.,German Center for Infection Disease (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Friedrich Koch-Nolte
- University Medical Center Hamburg-Eppendorf, Institute of Immunology, Hamburg, 20246, Germany
| | - Fabian Heinrich
- Center for Diagnostics, Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany.,Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Sven Peine
- Institute for Transfusion Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Julian Schulze Zur Wiesch
- German Center for Infection Disease (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany.,Division of Infectious Diseases, I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Thomas T Brehm
- German Center for Infection Disease (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany.,Division of Infectious Diseases, I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Marylyn M Addo
- German Center for Infection Disease (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany.,Division of Infectious Diseases, I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, 20359, Germany
| | - Marc Lütgehetmann
- Center for Diagnostics, Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Marcus Altfeld
- Leibniz Institute for Experimental Virology, Department of Virus Immunology, Hamburg, 20251, Germany.,German Center for Infection Disease (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
238
|
García-González P, Fernández D, Gutiérrez D, Parra-Cordero M, Osorio F. Human cDC1 display constitutive activation of the UPR Sensor IRE1. Eur J Immunol 2022; 52:1069-1076. [PMID: 35419836 PMCID: PMC9541385 DOI: 10.1002/eji.202149774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/08/2022] [Accepted: 04/08/2022] [Indexed: 12/04/2022]
Abstract
The intracellular mechanisms safeguarding DC function are of biomedical interest in several immune‐related diseases. Type 1 conventional DCs (cDC1s) are prominent targets of immunotherapy typified by constitutive activation of the unfolded protein response (UPR) sensor IRE1. Through its RNase domain, IRE1 regulates key processes in cDC1s including survival, ER architecture and function. However, most evidence linking IRE1 RNase with cDC1 biology emerges from mouse studies and it is currently unknown whether human cDC1s also activate the enzyme to preserve cellular homeostasis. In this work, we report that human cDC1s constitutively activate IRE1 RNase in steady state, which is evidenced by marked expression of IRE1, XBP1s, and target genes, and low levels of mRNA substrates of the IRE1 RNase domain. On a functional level, pharmacological inhibition of the IRE1 RNase domain curtailed IL‐12 and TNF production by cDC1s upon stimulation with TLR agonists. Altogether, this work demonstrates that activation of the IRE1/XBP1s axis is a conserved feature of cDC1s across species and suggests that the UPR sensor may also play a relevant role in the biology of the human lineage.
Collapse
Affiliation(s)
- Paulina García-González
- Laboratory of Immunology and Cellular Stress, Immunology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Dominique Fernández
- Laboratory of Immunology and Cellular Stress, Immunology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Diane Gutiérrez
- Fetal Medicine Unit, Clinical Hospital of University of Chile, Santiago, Chile
| | - Mauro Parra-Cordero
- Fetal Medicine Unit, Clinical Hospital of University of Chile, Santiago, Chile
| | - Fabiola Osorio
- Laboratory of Immunology and Cellular Stress, Immunology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
239
|
A Mutated Prostatic Acid Phosphatase (PAP) Peptide-Based Vaccine Induces PAP-Specific CD8 + T Cells with Ex Vivo Cytotoxic Capacities in HHDII/DR1 Transgenic Mice. Cancers (Basel) 2022; 14:cancers14081970. [PMID: 35454873 PMCID: PMC9032647 DOI: 10.3390/cancers14081970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Current treatments for castrate (hormone)-resistant prostate cancer (CRPC) remain limited and are not curative, with a median survival from diagnosis of 23 months. The PAP-specific Sipuleucel-T vaccine, which was approved by the FDA in 2010, increases the Overall Survival (OS) by 4 months, but is extremely expensive. We have previously shown that a 15 amino accid (AA) PAP sequence-derived peptide could induce strong immune responses and delay the growth of murine TRAMP-C1 prostate tumors. We have now substituted one amino acid and elongated the sequence to include epitopes predicted to bind to several additional HLA haplotypes. Herein, we present the immunological properties of this 42mer-mutated PAP-derived sequence (MutPAP42mer). METHODS The presence of PAP-135-143 epitope-specific CD8+ T cells in the blood of patients with prostate cancer (PCa) was assessed by flow cytometry using Dextramer™ technology. HHDII/DR1 transgenic mice were immunized with mutated and non-mutated PAP-derived 42mer peptides in the presence of CAF®09 or CpG ODN1826 (TLR-9 agonist) adjuvants. Vaccine-induced immune responses were measured by assessing the proportion and functionality of splenic PAP-specific T cells in vitro. RESULTS PAP-135-143 epitope-specific CD8+ T cells were detected in the blood of patients with PCa and stimulation of PBMCs from patients with PCa with mutPAP42mer enhanced their capacity to kill human LNCaP PCa target cells expressing PAP. The MutPAP42mer peptide was significantly more immunogenic in HHDII/DR1 mice than the wild type sequence, and immunogenicity was further enhanced when combined with the CAF®09 adjuvant. The vaccine induced secretory (IFNγ and TNFα) and cytotoxic CD8+ T cells and effector memory splenic T cells. CONCLUSIONS The periphery of patients with PCa exhibits immune responsiveness to the MutPAP42mer peptide and immunization of mice induces/expands T cell-driven, wild-type PAP immunity, and therefore, has the potential to drive protective anti-tumor immunity in patients with PCa.
Collapse
|
240
|
Peng YQ, Wu ZC, Xu ZB, Fang SB, Chen DH, Zhang HY, Liu XQ, He BX, Chen D, Akdis CA, Fu QL. Mesenchymal stromal cells-derived small extracellular vesicles modulate DC function to suppress Th2 responses via IL-10 in patients with allergic rhinitis. Eur J Immunol 2022; 52:1129-1140. [PMID: 35415925 PMCID: PMC9545324 DOI: 10.1002/eji.202149497] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 02/01/2022] [Accepted: 04/11/2022] [Indexed: 12/02/2022]
Abstract
Mesenchymal stromal cells (MSCs) are well known for their immunoregulatory roles on allergic inflammation particularly by acting on T cells, B cells, and dendritic cells (DCs). MSC‐derived small extracellular vesicles (MSC‐sEV) are increasingly considered as one of the main factors for the effects of MSCs on immune responses. However, the effects of MSC‐sEV on DCs in allergic diseases remain unclear.
Collapse
Affiliation(s)
- Ya-Qi Peng
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zi-Cong Wu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Bin Xu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shu-Bin Fang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - De-Hua Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hong-Yu Zhang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Qing Liu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bi-Xin He
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dong Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.,Christine Kühne - Center for Research and Education (CK-CARE), Davos, Switzerland
| | - Qing-Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
241
|
Gibellini L, De Biasi S, Meschiari M, Gozzi L, Paolini A, Borella R, Mattioli M, Lo Tartaro D, Fidanza L, Neroni A, Busani S, Girardis M, Guaraldi G, Mussini C, Cozzi-Lepri A, Cossarizza A. Plasma Cytokine Atlas Reveals the Importance of TH2 Polarization and Interferons in Predicting COVID-19 Severity and Survival. Front Immunol 2022; 13:842150. [PMID: 35386702 PMCID: PMC8979161 DOI: 10.3389/fimmu.2022.842150] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/21/2022] [Indexed: 12/11/2022] Open
Abstract
Although it is now widely accepted that host inflammatory response contributes to COVID-19 immunopathogenesis, the pathways and mechanisms driving disease severity and clinical outcome remain poorly understood. In the effort to identify key soluble mediators that characterize life-threatening COVID-19, we quantified 62 cytokines, chemokines and other factors involved in inflammation and immunity in plasma samples, collected at hospital admission, from 80 hospitalized patients with severe COVID-19 disease who were stratified on the basis of clinical outcome (mechanical ventilation or death by day 28). Our data confirm that age, as well as neutrophilia, lymphocytopenia, procalcitonin, D-dimer and lactate dehydrogenase are strongly associated with the risk of fatal COVID-19. In addition, we found that cytokines related to TH2 regulations (IL-4, IL-13, IL-33), cell metabolism (lep, lep-R) and interferons (IFNα, IFNβ, IFNγ) were also predictive of life-threatening COVID-19.
Collapse
Affiliation(s)
- Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Marianna Meschiari
- Infectious Diseases Clinics, Azienda Ospedaliera-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Licia Gozzi
- Infectious Diseases Clinics, Azienda Ospedaliera-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Marco Mattioli
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Lucia Fidanza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Stefano Busani
- Department of Anesthesia and Intensive Care, Azienda Ospedaliera-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Girardis
- Department of Anesthesia and Intensive Care, Azienda Ospedaliera-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanni Guaraldi
- Infectious Diseases Clinics, Azienda Ospedaliera-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Mussini
- Infectious Diseases Clinics, Azienda Ospedaliera-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro Cozzi-Lepri
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation, Institute for Global Health, London, United Kingdom
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy.,National Institute for Cardiovascular Research, Bologna, Italy
| |
Collapse
|
242
|
Kong Y, Jing Y, Allard D, Scavuzzo MA, Sprouse ML, Borowiak M, Bettini ML, Bettini M. A dormant T cell population with autoimmune potential exhibits low self-reactivity and infiltrates islets in type 1 diabetes. Eur J Immunol 2022; 52:1158-1170. [PMID: 35389516 DOI: 10.1002/eji.202149690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 11/09/2022]
Abstract
The contribution of low affinity T cells to autoimmunity in the context of polyclonal T cell responses is understudied due to the limitations in their capture by tetrameric reagents and low level of activation in response to antigenic stimulation. As a result, low affinity T cells are often disregarded as non-antigen specific cells irrelevant to the immune response. Our study aimed to assess how the level of self-antigen reactivity shapes T cell lineage and effector responses in the context of spontaneous tissue specific autoimmunity observed in NOD mice. Using multi-color flow cytometry in combination with Nur77GFP reporter of TCR signaling we identified a dormant population of T cells that infiltrated the pancreatic islets of pre-diabetic NOD mice, which exhibited reduced level of self-tissue reactivity based on expression of CD5 and Nur77GFP . We showed that these CD5low T cells had a unique TCR repertoire, exhibited low activation and minimal effector function; however, induced rapid diabetes upon transfer. The CD4+ CD5low T cell population displayed transcriptional signature of central memory T cells, consistent with the ability to acquire effector function post-transfer. Transcriptional profile of CD5low T cells was similar to T cells expressing a low affinity TCR, indicating TCR affinity to be the important factor in shaping CD5low T cell phenotype and function at the tissue site. Overall, our study suggests that autoimmune tissue can maintain a reservoir of undifferentiated central memory-like autoreactive T cells with pathogenic effector potential that might be an important source for effector T cells during long-term chronic autoimmunity. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yuelin Kong
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, 77030
| | - Yi Jing
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, 77030.,Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, 84112
| | - Denise Allard
- Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, 84112
| | - Marissa A Scavuzzo
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030
| | - Maran L Sprouse
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, 77030
| | - Malgorzata Borowiak
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030.,Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, 77030.,McNair Medical Institute, Houston, TX, 77030
| | - Matthew L Bettini
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, 77030.,Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, 84112.,McNair Medical Institute, Houston, TX, 77030
| | - Maria Bettini
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, 77030.,Department of Pathology, Microbiology and Immunology, University of Utah, Salt Lake City, UT, 84112.,McNair Medical Institute, Houston, TX, 77030
| |
Collapse
|
243
|
Parga-Vidal L, Taggenbrock RLRE, Beumer-Chuwonpad A, Aglmous H, Kragten NAM, Behr FM, Bovens AA, van Lier RAW, Stark R, van Gisbergen KPJM. Hobit and Blimp-1 regulate T RM abundance after LCMV infection by suppressing tissue exit pathways of T RM precursors. Eur J Immunol 2022; 52:1095-1111. [PMID: 35389518 PMCID: PMC9545210 DOI: 10.1002/eji.202149665] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/17/2022] [Accepted: 03/31/2022] [Indexed: 11/12/2022]
Abstract
Tissue‐resident memory T cells (Trm) are retained in peripheral tissues after infection for enhanced protection against secondary encounter with the same pathogen. We have previously shown that the transcription factor Hobit and its homolog Blimp‐1 drive Trm development after viral infection, but how and when these transcription factors mediate Trm formation remains poorly understood. In particular, the major impact of Blimp‐1 in regulating several aspects of effector T‐cell differentiation impairs study of its specific role in Trm development. Here, we used the restricted expression of Hobit in the Trm lineage to develop mice with a conditional deletion of Blimp‐1 in Trm, allowing us to specifically investigate the role of both transcription factors in Trm differentiation. We found that Hobit and Blimp‐1 were required for the upregulation of CD69 and suppression of CCR7 and S1PR1 on virus‐specific Trm precursors after LCMV infection, underlining a role in their retention within tissues. The early impact of Hobit and Blimp‐1 favored Trm formation and prevented the development of circulating memory T cells. Thus, our findings highlight a role of Hobit and Blimp‐1 at the branching point of circulating and resident memory lineages by suppressing tissue egress of Trm precursors early during infection.
Collapse
Affiliation(s)
- Loreto Parga-Vidal
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Renske L R E Taggenbrock
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ammarina Beumer-Chuwonpad
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Hajar Aglmous
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, 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
| | - 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
| | - Astrid A Bovens
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Rene A W van Lier
- 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
| | - Klaas P J M van Gisbergen
- 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
| |
Collapse
|
244
|
Knop L, Spanier J, Larsen PK, Witte A, Bank U, Dunay IR, Kalinke U, Schüler T. IFNAR signaling in fibroblastic reticular cells can modulate CD8 + memory fate decision. Eur J Immunol 2022; 52:895-906. [PMID: 35365883 DOI: 10.1002/eji.202149760] [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: 12/08/2021] [Revised: 02/27/2022] [Accepted: 03/31/2022] [Indexed: 11/12/2022]
Abstract
CD8+ memory T cells (TM ) are crucial for the long-term protection from infections and cancer. Multiple cell types and cytokines are involved in the regulation of CD8+ T cell responses and subsequent TM formation. Besides their direct antiviral effects, type I interferons (IFN-α/β) modulate CD8+ T cell immunity via their action on several immune cell subsets. However, it is largely unclear how non-immune cells are involved in this multicellular network modulating CD8+ TM formation. Fibroblastic reticular cells (FRCs), form the three-dimensional scaffold of secondary lymphoid organs, express the IFN-α/β receptor (IFNAR) and modulate adaptive immune responses. However, it is unclear whether and how early IFNAR signals in lymph node (LN) FRCs affect CD8+ TM differentiation. Using peptide vaccination and viral infection, we studied CD8+ TM differentiation in mice with a FRC-specific IFNAR deletion (FRCΔIFNAR ). We show here that the differentiation of CD8+ TCR-transgenic T cells into central memory cells (TCM ) is enhanced in peptide-vaccinated FRCΔIFNAR mice. Conversely, vesicular stomatitis virus (VSV) infection of FRCΔIFNAR mice is associated with impaired TCM formation and the accumulation of VSV-specific double-positive (dp) CD127lo KLRG-1hi effector memory T cells. In summary, we provide evidence for a context-dependent contribution of FRC-specific IFNAR signaling to CD8+ TM differentiation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Laura Knop
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| | - Julia Spanier
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, 30625, Germany
| | - Pia-Katharina Larsen
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, 30625, Germany
| | - Amelie Witte
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| | - Ute Bank
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| | - Ildiko R Dunay
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, 30625, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, 39120, Germany
| |
Collapse
|
245
|
Lo JW, de Mucha MV, Henderson S, Roberts LB, Constable LE, Garrido‐Mesa N, Hertweck A, Stolarczyk E, Houlder EL, Jackson I, MacDonald AS, Powell N, Neves JF, Howard JK, Jenner RG, Lord GM. A population of naive-like CD4 + T cells stably polarized to the T H 1 lineage. Eur J Immunol 2022; 52:566-581. [PMID: 35092032 PMCID: PMC9304323 DOI: 10.1002/eji.202149228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 11/19/2021] [Accepted: 01/13/2022] [Indexed: 11/11/2022]
Abstract
T-bet is the lineage-specifying transcription factor for CD4+ TH 1 cells. T-bet has also been found in other CD4+ T cell subsets, including TH 17 cells and Treg, where it modulates their functional characteristics. However, we lack information on when and where T-bet is expressed during T cell differentiation and how this impacts T cell differentiation and function. To address this, we traced the ontogeny of T-bet-expressing cells using a fluorescent fate-mapping mouse line. We demonstrate that T-bet is expressed in a subset of CD4+ T cells that have naïve cell surface markers and transcriptional profile and that this novel cell population is phenotypically and functionally distinct from previously described populations of naïve and memory CD4+ T cells. Naïve-like T-bet-experienced cells are polarized to the TH 1 lineage, predisposed to produce IFN-γ upon cell activation, and resist repolarization to other lineages in vitro and in vivo. These results demonstrate that lineage-specifying factors can polarize T cells in the absence of canonical markers of T cell activation and that this has an impact on the subsequent T-helper response.
Collapse
Affiliation(s)
- Jonathan W. Lo
- School of Immunology and Microbial SciencesKing's College LondonLondonUK
- Division of Digestive DiseasesFaculty of MedicineImperial College LondonLondonUK
| | - Maria Vila de Mucha
- UCL Cancer Institute and CRUK UCL CentreUniversity College London (UCL)LondonUK
| | - Stephen Henderson
- UCL Cancer Institute and CRUK UCL CentreUniversity College London (UCL)LondonUK
| | - Luke B. Roberts
- School of Immunology and Microbial SciencesKing's College LondonLondonUK
| | - Laura E. Constable
- School of Immunology and Microbial SciencesKing's College LondonLondonUK
- Division of Digestive DiseasesFaculty of MedicineImperial College LondonLondonUK
| | - Natividad Garrido‐Mesa
- School of Immunology and Microbial SciencesKing's College LondonLondonUK
- School of Life Sciences, Pharmacy and ChemistryKingston UniversityLondonUK
| | - Arnulf Hertweck
- UCL Cancer Institute and CRUK UCL CentreUniversity College London (UCL)LondonUK
| | - Emilie Stolarczyk
- Abcam Plc.Cambridge Biomedical CampusCambridgeUK
- School of Cardiovascular Medicine and SciencesGuy's Campus, King's College LondonLondonUK
| | - Emma L. Houlder
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Ian Jackson
- School of Immunology and Microbial SciencesKing's College LondonLondonUK
| | - Andrew S. MacDonald
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| | - Nick Powell
- School of Immunology and Microbial SciencesKing's College LondonLondonUK
- Division of Digestive DiseasesFaculty of MedicineImperial College LondonLondonUK
| | - Joana F. Neves
- School of Immunology and Microbial SciencesKing's College LondonLondonUK
- Centre for Host‐Microbiome InteractionsKing's College LondonLondonUK
| | - Jane K. Howard
- School of Cardiovascular Medicine and SciencesGuy's Campus, King's College LondonLondonUK
| | - Richard G. Jenner
- UCL Cancer Institute and CRUK UCL CentreUniversity College London (UCL)LondonUK
| | - Graham M. Lord
- School of Immunology and Microbial SciencesKing's College LondonLondonUK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
- School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of ManchesterManchesterUK
| |
Collapse
|
246
|
Wu L, Shukla V, Yadavalli AD, Dinesh RK, Xu D, Rao A, Schatz DG. HMCES protects immunoglobulin genes specifically from deletions during somatic hypermutation. Genes Dev 2022; 36:433-450. [PMID: 35450882 PMCID: PMC9067407 DOI: 10.1101/gad.349438.122] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/29/2022] [Indexed: 01/07/2023]
Abstract
Somatic hypermutation (SHM) produces point mutations in immunoglobulin (Ig) genes in B cells when uracils created by the activation-induced deaminase are processed in a mutagenic manner by enzymes of the base excision repair (BER) and mismatch repair (MMR) pathways. Such uracil processing creates DNA strand breaks and is susceptible to the generation of deleterious deletions. Here, we demonstrate that the DNA repair factor HMCES strongly suppresses deletions without significantly affecting other parameters of SHM in mouse and human B cells, thereby facilitating the production of antigen-specific antibodies. The deletion-prone repair pathway suppressed by HMCES operates downstream from the uracil glycosylase UNG and is mediated by the combined action of BER factor APE2 and MMR factors MSH2, MSH6, and EXO1. HMCES's ability to shield against deletions during SHM requires its capacity to form covalent cross-links with abasic sites, in sharp contrast to its DNA end-joining role in class switch recombination but analogous to its genome-stabilizing role during DNA replication. Our findings lead to a novel model for the protection of Ig gene integrity during SHM in which abasic site cross-linking by HMCES intercedes at a critical juncture during processing of vulnerable gapped DNA intermediates by BER and MMR enzymes.
Collapse
Affiliation(s)
- Lizhen Wu
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut 06520, USA
| | - Vipul Shukla
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, California 92037, USA
| | | | - Ravi K Dinesh
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut 06520, USA
| | - Dijin Xu
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Anjana Rao
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, California 92037, USA
- Department of Pharmacology, Moores Cancer Center, University of California at San Diego, La Jolla, California 92093, USA
- Consortium for Regenerative Medicine, La Jolla, California 92037, USA
| | - David G Schatz
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut 06520, USA
| |
Collapse
|
247
|
Zhang H, Liu S, Li Y, Li J, Ni C, Yang M, Dong J, Wang Z, Qin Z. Dysfunction of S100A4 + effector memory CD8 + T cells aggravates asthma. Eur J Immunol 2022; 52:978-993. [PMID: 35340022 DOI: 10.1002/eji.202149572] [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: 08/11/2021] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/09/2022]
Abstract
Progressive loss of effector functions, especially IFN-γ secreting capability, in effector memory CD8+ T (CD8+ TEM ) cells plays a crucial role in asthma worsening. However, the mechanisms of CD8+ TEM cell dysfunction remain elusive. Here, we report that S100A4 drives CD8+ TEM cell dysfunction, impairing their protective memory response and promoting asthma worsening in an ovalbumin (OVA)-induced asthmatic murine model. We find that CD8+ TEM cells contain two subsets based on S100A4 expression. S100A4+ subsets exhibit dysfunctional effector phenotypes with increased proliferative capability, whereas S100A4- subsets retain effector function but are more inclined to apoptosis, giving rise a dysfunctional CD8+ TEM cell pool. Mechanistically, S100A4 upregulation of mitochondrial metabolism results in a decrease of acetyl-CoA levels, which impair the transcription of effector genes, especially ifn-γ, facilitating cell survival, tolerance and memory potential. Our findings thus reveal general insights into how S100A4 CD8+ TEM cells reprogram into dysfunctional and less protective phenotypes to aggravate asthma. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Huilei Zhang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuangqing Liu
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanan Li
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianru Li
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Chen Ni
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ming Yang
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW 2300, Australia
| | - Jun Dong
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, 10117, Germany
| | - Zhaoqing Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhihai Qin
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
248
|
La Manna MP, Orlando V, Badami GD, Tamburini B, Azgomi MS, Presti EL, Del Nonno F, Petrone L, Belmonte B, Falasca L, Carlo PD, Dieli F, Goletti D, Caccamo N. Platelets accumulate in lung lesions of tuberculosis patients and inhibit T-cell responses and Mycobacterium tuberculosis replication in macrophages. Eur J Immunol 2022; 52:784-799. [PMID: 35338775 PMCID: PMC9325462 DOI: 10.1002/eji.202149549] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/24/2021] [Accepted: 03/23/2022] [Indexed: 12/05/2022]
Abstract
Platelets regulate human inflammatory responses that lead to disease. However, the role of platelets in tuberculosis (TB) pathogenesis is still unclear. Here, we show that patients with active TB have a high number of platelets in peripheral blood and a low number of lymphocytes leading to a high platelets to lymphocytes ratio (PL ratio). Moreover, the serum concentration of different mediators promoting platelet differentiation or associated with platelet activation is increased in active TB. Immunohistochemistry analysis shows that platelets localise around the lung granuloma lesions in close contact with T lymphocytes and macrophages. Transcriptomic analysis of caseous tissue of human pulmonary TB granulomas, followed by Gene Ontology analysis, shows that 53 platelet activation‐associated genes are highly expressed compared to the normal lung tissue. In vitro activated platelets (or their supernatants) inhibit BCG‐induced T‐ lymphocyte proliferation and IFN‐γ production. Likewise, platelets inhibit the growth of intracellular macrophages of Mycobacterium (M.) tuberculosis. Soluble factors released by activated platelets mediate both immunological and M. tuberculosis replication activities. Furthermore, proteomic and neutralisation studies (by mAbs) identify TGF‐β and PF4 as the factors responsible for inhibiting T‐cell response and enhancing the mycobactericidal activity of macrophages, respectively. Altogether these results highlight the importance of platelets in TB pathogenesis.
Collapse
Affiliation(s)
- Marco P La Manna
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Valentina Orlando
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Giusto D Badami
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Bartolo Tamburini
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Mojtaba Shekarkar Azgomi
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Elena Lo Presti
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Franca Del Nonno
- Pathology Unit, National Institute for Infectious Diseases L. Spallanzani-IRCCS, Rome, Italy
| | - Linda Petrone
- Translational research Unit, National Institute for Infectious Diseases L. Spallanzani-IRCCS, Rome, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Health Science, Human Pathology Section, University of Palermo School of Medicine, Palermo, Italy
| | - Laura Falasca
- Pathology Unit, National Institute for Infectious Diseases L. Spallanzani-IRCCS, Rome, Italy
| | - Paola Di Carlo
- Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Delia Goletti
- Translational research Unit, National Institute for Infectious Diseases L. Spallanzani-IRCCS, Rome, Italy
| | - Nadia Caccamo
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| |
Collapse
|
249
|
Brdovčak MC, Materljan J, Šustić M, Ravlić S, Ružić T, Lisnić B, Miklić K, Brizić I, Matešić MP, Lisnić VJ, Halassy B, Rončević D, Knežević Z, Štefan L, Bertoglio F, Schubert M, Čičin-Šain L, Markotić A, Jonjić S, Krmpotić A. ChAdOx1-S adenoviral vector vaccine applied intranasally elicits superior mucosal immunity compared to the intramuscular route of vaccination. Eur J Immunol 2022; 52:936-945. [PMID: 35304741 PMCID: PMC9087383 DOI: 10.1002/eji.202249823] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 11/09/2022]
Abstract
COVID-19 vaccines prevent severe forms of the disease, but do not warrant complete protection against breakthrough infections. This could be due to suboptimal mucosal immunity at the site of virus entry, given that all currently approved vaccines are administered via the intramuscular route. In this study we assessed humoral and cellular immune responses in BALB/c mice after intranasal and intramuscular immunization with adenoviral vector ChAdOx1-S expressing full-length Spike protein of SARS-CoV-2. We showed that both routes of vaccination induced a potent IgG antibody response, as well as robust neutralizing capacity, but intranasal vaccination elicited a superior IgA antibody titer in the sera and in the respiratory mucosa. Bronchoalveolar lavage from intranasally immunized mice efficiently neutralized SARS-CoV-2, which has not been the case in intramuscularly immunized group. Moreover, substantially higher percentages of epitope-specific CD8 T cells exhibiting a tissue resident phenotype were found in the lungs of intranasally immunized animals. Finally, both intranasal and intramuscular vaccination with ChAdOx1-S efficiently protected the mice after the challenge with recombinant herpesvirus expressing the Spike protein. Our results demonstrate that intranasal application of adenoviral vector ChAdOx1-S induces superior mucosal immunity and therefore could be a promising strategy for putting the COVID-19 pandemic under control. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Maja Cokarić Brdovčak
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | - Jelena Materljan
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia.,Department of Histology and Embryology, University of Rijeka, Rijeka, 51000, Croatia
| | - Marko Šustić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | - Sanda Ravlić
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, 10000, Croatia
| | - Tina Ružić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | - Berislav Lisnić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | - Karmela Miklić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | - Ilija Brizić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | | | - Vanda Juranić Lisnić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | - Beata Halassy
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Zagreb, 10000, Croatia
| | | | | | - Leo Štefan
- JGL d.d. Jadran Galenski Laboratorij, Rijeka, 51 000, Croatia
| | - Federico Bertoglio
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, 38106, Germany
| | - Maren Schubert
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, 38106, Germany
| | - Luka Čičin-Šain
- Helmholtz Center for Infection Research, Department of Viral Immunology, Braunschweig, 38124, Germany
| | - Alemka Markotić
- University Hospital for Infectious Diseases "Fran Mihaljević", Zagreb, 10000, Croatia
| | - Stipan Jonjić
- Center for Proteomics, University of Rijeka, Faculty of Medicine, Rijeka, 51000, Croatia
| | - Astrid Krmpotić
- Department of Histology and Embryology, University of Rijeka, Rijeka, 51000, Croatia
| |
Collapse
|
250
|
Harelimana JDD, Ahor HS, Benner B, Hellmuth S, Adankwah E, Minadzi D, Aniagyei W, Lamptey M, Arthur J, Yeboah A, Abass MK, Debrah LB, Owusu DO, Mayatepek E, Seyfarth J, Phillips RO, Jacobsen M. Cytokine-induced transient monocyte IL-7Ra expression and the serum milieu in tuberculosis. Eur J Immunol 2022; 52:958-969. [PMID: 35279828 DOI: 10.1002/eji.202149661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 11/09/2022]
Abstract
Bacterial components and cytokines induce Interleukin-7 receptor (IL-7Rα) expression in monocytes. Aberrant low IL-7Rα expression of monocytes has been identified as a feature of tuberculosis immunopathology. Here, we investigated the mechanisms underlying IL-7Rα regulation of monocytes and tuberculosis serum effects IL-7Rα expression. Serum samples from tuberculosis patients and healthy controls, cytokine candidates, and mycobacterial components were analyzed for in vitro effects on IL-7Rα expression of primary monocytes, monocyte-derived macrophages (MDM), and monocyte cell lines. IL-7Rα regulation during culture and the role of FoxO1 was characterized. In vitro activation induced IL-7Rα expression in human monocytes and serum samples from tuberculosis patients boosted IL-7Rα expression. Although pathognomonic tuberculosis cytokines were not associated with serum effects, we identified cytokines (i.e., GM-CSF, IL-1β, TNFα, IFNγ) that induced IL-7Rα expression in monocytes and/or MDM comparable to mycobacterial components. Blocking of cytokine subsets (i.e., IL-1β/TNFα in monocytes, GM-CSF in MDM) largely diminished IL-7Rα expression induced by mycobacterial components. Finally, we showed that in vitro induced IL-7Rα expression was transient and dependent on constitutive FoxO1 expression in primary monocytes and monocyte cell lines. This study demonstrated the crucial roles of cytokines and constitutive FoxO1 expression for transient IL-7Rα expression in monocytes. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jean De Dieu Harelimana
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, 40225, Germany
| | - Hubert Senanu Ahor
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, 40225, Germany
| | - Bastian Benner
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, 40225, Germany
| | - Sabine Hellmuth
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, 40225, Germany
| | - Ernest Adankwah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
| | - Difery Minadzi
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
| | - Wilfred Aniagyei
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
| | - Millicent Lamptey
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
| | - Joseph Arthur
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
| | - Augustine Yeboah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
| | | | - Linda Batsa Debrah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
| | - Dorcas O Owusu
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, 40225, Germany
| | - Julia Seyfarth
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, 40225, Germany
| | - Richard O Phillips
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kumasi, Ghana.,School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Marc Jacobsen
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, 40225, Germany
| |
Collapse
|