1
|
Hickman HD, Moutsopoulos NM. Viral infection and antiviral immunity in the oral cavity. Nat Rev Immunol 2024:10.1038/s41577-024-01100-x. [PMID: 39533045 DOI: 10.1038/s41577-024-01100-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
Individual tissues have distinct antiviral properties garnered through various mechanisms, including physical characteristics, tissue-resident immune cells and commensal organisms. Although the oral mucosa has long been appreciated as a critical barrier tissue that is exposed to a continuous barrage of pathogens, many fundamental aspects of the antiviral immune response in this tissue remain unknown. Several viral pathogens, such as herpesviruses and human papillomaviruses, have been acknowledged both historically and at present for infections in the oral cavity that result in substantial clinical burden. However, recent viral outbreaks, including those with SARS-CoV-2 and mpox, featured oral symptoms even though these viruses are not generally considered oral pathogens. Ensuing studies have shown that the oral cavity is an important locale for viral infection and potential transmission of newly emergent or re-emergent pathogens, highlighting the need for an increased understanding of the mechanisms of antiviral immunity at this site. In this Review, we provide a broad overview of antiviral immune responses in the oral cavity and discuss common viral infections and their manifestations in the oral mucosa. In addition, we present current mouse models for the study of oral viral infections.
Collapse
Affiliation(s)
- Heather D Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Niki M Moutsopoulos
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Piersma SJ. Tissue-specific features of innate lymphoid cells in antiviral defense. Cell Mol Immunol 2024; 21:1036-1050. [PMID: 38684766 PMCID: PMC11364677 DOI: 10.1038/s41423-024-01161-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Innate lymphocytes (ILCs) rapidly respond to and protect against invading pathogens and cancer. ILCs include natural killer (NK) cells, ILC1s, ILC2s, ILC3s, and lymphoid tissue inducer (LTi) cells and include type I, type II, and type III immune cells. While NK cells have been well recognized for their role in antiviral immunity, other ILC subtypes are emerging as players in antiviral defense. Each ILC subset has specialized functions that uniquely impact the antiviral immunity and health of the host depending on the tissue microenvironment. This review focuses on the specialized functions of each ILC subtype and their roles in antiviral immune responses across tissues. Several viruses within infection-prone tissues will be highlighted to provide an overview of the extent of the ILC immunity within tissues and emphasize common versus virus-specific responses.
Collapse
Affiliation(s)
- Sytse J Piersma
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
3
|
Hu ST, Zhou G, Zhang J. Implications of innate lymphoid cells in oral diseases. Int Immunopharmacol 2024; 133:112122. [PMID: 38663313 DOI: 10.1016/j.intimp.2024.112122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024]
Abstract
Innate lymphoid cells (ILCs), as newly discovered antigen-independent innate immune cells, respond promptly to stimuli by secreting effector cytokines to exert effector functions similar to those of T cells. ILCs predominantly reside at mucosal sites and play critical roles in defending against infections, maintaining mucosal homeostasis, regulating inflammatory and immune responses, and participating in tumorigenesis. Recently, there has been a growing interest in the role of ILCs in oral diseases. This review outlines the classifications and the major characteristics of ILCs, and then comprehensively expatiates the research on ILCs in oral cancer, primary Sjogren's syndrome, periodontal diseases, oral lichen planus, oral candidiasis, Behcet's disease, and pemphigus vulgaris, aiming at summarising the implications of ILCs in oral diseases and providing new ideas for further research.
Collapse
Affiliation(s)
- Si-Ting Hu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China
| | - Gang Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China; Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, China
| | - Jing Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, China; Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, China.
| |
Collapse
|
4
|
Fol M, Karpik W, Zablotni A, Kulesza J, Kulesza E, Godkowicz M, Druszczynska M. Innate Lymphoid Cells and Their Role in the Immune Response to Infections. Cells 2024; 13:335. [PMID: 38391948 PMCID: PMC10886880 DOI: 10.3390/cells13040335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/07/2024] [Accepted: 02/10/2024] [Indexed: 02/24/2024] Open
Abstract
Over the past decade, a group of lymphocyte-like cells called innate lymphoid cells (ILCs) has gained considerable attention due to their crucial role in regulating immunity and tissue homeostasis. ILCs, lacking antigen-specific receptors, are a group of functionally differentiated effector cells that act as tissue-resident sentinels against infections. Numerous studies have elucidated the characteristics of ILC subgroups, but the mechanisms controlling protective or pathological responses to pathogens still need to be better understood. This review summarizes the functions of ILCs in the immunology of infections caused by different intracellular and extracellular pathogens and discusses their possible therapeutic potential.
Collapse
Affiliation(s)
- Marek Fol
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
| | - Wojciech Karpik
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
| | - Agnieszka Zablotni
- Department of Bacterial Biology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland;
| | - Jakub Kulesza
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, 91-347 Lodz, Poland;
| | - Ewelina Kulesza
- Department of Rheumatology and Internal Diseases, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Magdalena Godkowicz
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
- Lodz Institutes of the Polish Academy of Sciences, The Bio-Med-Chem Doctoral School, University of Lodz, 90-237 Lodz, Poland
| | - Magdalena Druszczynska
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
| |
Collapse
|
5
|
Pang XL, Li J, Wang J, Yan SS, Yang J. MiR-142-3p Regulates ILC1s by Targeting HMGB1 via the NF-κB Pathway in a Mouse Model of Early Pregnancy Loss. Curr Med Sci 2024; 44:195-211. [PMID: 38393528 DOI: 10.1007/s11596-024-2833-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/16/2023] [Indexed: 02/25/2024]
Abstract
OBJECTIVE Innate lymphoid cells (ILCs) are a class of newly discovered immunocytes. Group 1 ILCs (ILC1s) are identified in the decidua of humans and mice. High mobility group box 1 (HMGB1) is predicted to be one of the target genes of miR-142-3p, which is closely related to pregnancy-related diseases. Furthermore, miR-142-3p and HMGB1 are involved in regulating the NF-κB signaling pathway. This study aimed to examine the regulatory effect of miR-142-3p on ILC1s and the underlying mechanism involving HMGB1 and the NF-κB signaling pathway. METHODS Mouse models of normal pregnancy and abortion were constructed, and the alterations of ILC1s, miR-142-3p, ILC1 transcription factor (T-bet), and pro-inflammatory cytokines of ILC1s (TNF-α, IFN-γ and IL-2) were detected in mice from different groups. The targeting regulation of HMGB1 by miR-142-3p in ILC1s, and the expression of HMGB1 in normal pregnant mice and abortive mice were investigated. In addition, the regulatory effects of miR-142-3p and HMGB1 on ILC1s were detected in vitro by CCK-8, Annexin-V/PI, ELISA, and RT-PCR, respectively. Furthermore, changes of the NF-κB signaling pathway in ILC1s were examined in the different groups. For the in vivo studies, miR-142-3p-Agomir was injected in the uterus of abortive mice to evaluate the abortion rate and alterations of ILC1s at the maternal-fetal interface, and further detect the expression of HMGB1, pro-inflammatory cytokines, and the NF-κB signaling pathway. RESULTS The number of ILC1s was significantly increased, the level of HMGB1 was significantly upregulated, and that of miR-142-3p was considerably downregulated in the abortive mice as compared with the normal pregnant mice (all P<0.05). In addition, miR-142-3p was found to drastically inhibit the activation of the NF-κB signaling pathway (P<0.05). The number of ILC1s and the levels of pro-inflammatory cytokines were significantly downregulated and the activation of the NF-κB signaling pathway was inhibited in the miR-142-3p Agomir group (all P<0.05). CONCLUSION miR-142-3p can regulate ILC1s by targeting HMGB1 via the NF-κB signaling pathway, and attenuate the inflammation at the maternal-fetal interface in abortive mice.
Collapse
Affiliation(s)
- Xiang-Li Pang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jie Li
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Clinic Research Center for Assisted Reproductive Technology and Embryonic Development in Hubei Province, Wuhan, 430060, China
| | - Jing Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Si-Si Yan
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Clinic Research Center for Assisted Reproductive Technology and Embryonic Development in Hubei Province, Wuhan, 430060, China.
| |
Collapse
|
6
|
Tougaard P, Pérez MR, Steels W, Huysentruyt J, Verstraeten B, Vetters J, Divert T, Gonçalves A, Roelandt R, Takahashi N, Janssens S, Buus TB, Taghon T, Leclercq G, Vandenabeele P. Type 1 immunity enables neonatal thymic ILC1 production. SCIENCE ADVANCES 2024; 10:eadh5520. [PMID: 38232171 PMCID: PMC10793954 DOI: 10.1126/sciadv.adh5520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 12/19/2023] [Indexed: 01/19/2024]
Abstract
Acute thymic atrophy occurs following type 1 inflammatory conditions such as viral infection and sepsis, resulting in cell death and disruption of T cell development. However, the impact type 1 immunity has on thymic-resident innate lymphoid cells (ILCs) remains unclear. Single-cell RNA sequencing revealed neonatal thymic-resident type 1 ILCs (ILC1s) as a unique and immature subset compared to ILC1s in other primary lymphoid organs. Culturing murine neonatal thymic lobes with the type 1 cytokines interleukin-12 (IL-12) and IL-18 resulted in a rapid expansion and thymic egress of KLRG1+CXCR6+ cytotoxic ILC1s. Live imaging showed the subcapsular thymic localization and exit of ILC1s following IL-12 + IL-18 stimulation. Similarly, murine cytomegalovirus infection in neonates resulted in thymic atrophy and subcapsular localization of thymic-resident ILC1s. Neonatal thymic grafting revealed that type 1 inflammation enhances the homing of cytokine-producing thymus-derived ILC1s to the liver and peritoneal cavity. Together, we show that type 1 immunity promotes the expansion and peripheral homing of thymic-derived ILC1s.
Collapse
Affiliation(s)
- Peter Tougaard
- Cell death and Inflammation Unit, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Mario R. Pérez
- Cell death and Inflammation Unit, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Wolf Steels
- Cell death and Inflammation Unit, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jelle Huysentruyt
- Cell death and Inflammation Unit, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Bruno Verstraeten
- Cell death and Inflammation Unit, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jessica Vetters
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Tatyana Divert
- Cell death and Inflammation Unit, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Amanda Gonçalves
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- VIB BioImaging Core, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent 9052, Belgium
| | - Ria Roelandt
- Cell death and Inflammation Unit, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Nozomi Takahashi
- Cell death and Inflammation Unit, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Sophie Janssens
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Terkild B. Buus
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Tom Taghon
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Georges Leclercq
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Peter Vandenabeele
- Cell death and Inflammation Unit, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
7
|
Pan L, Feng M, Chen J, Deng S, Han X, Wang Y, Tang G. Group 1 and 3 innate lymphoid cells are increased in oral lichen planus and oral lichenoid lesions. Oral Dis 2023; 29:3372-3380. [PMID: 36151914 DOI: 10.1111/odi.14384] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/25/2022] [Accepted: 09/14/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Innate lymphoid cells (ILCs) are vital innate immune cells cooperating with T cells. While their phenotypes and functions in oral mucosa kept unclear yet. In the present study, the relative proportions and distribution of different ILC subsets in oral mucosa of oral lichen planus (OLP), oral lichenoid lesions (OLL), and controls were compared. SUBJECTS AND METHODS Oral mucosal samples were collected from control (n = 29), OLP (n = 20), and OLL (n = 22) donors. ILCs subsets were characterized in single-cell suspensions by flow cytometry. Immunohistochemistry was performed to locate the CD127+ cells in situ. RESULTS ILCs were present in healthy and increased infiltration in OLP/OLL (p = 0.0092, p = 0.0216). Infiltration of ILC1 increased in OLP/OLL mucosa (p = 0.0225, p = 0.0399), as did the infiltration of ILC3 increase in OLL mucosa (p = 0.0128). The ILC2/ILCs ratio was significantly reduced in OLP and OLL (p = 0.0124, p = 0.0346). CD127+ cells were mainly located closely at the basement membrane. CONCLUSIONS The results of increased ILC1, decreased ILC2, and increased ILC3 suggested that changes of ILC distributions in oral mucosa may be relevant to persistent inflammation in local tissues, by promoting immune factors and weakening repair capacity.
Collapse
Affiliation(s)
- Lei Pan
- Department of 2nd Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Minghua Feng
- Shanghai Key Laboratory of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junjun Chen
- Shanghai Key Laboratory of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu Deng
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Harvard School of Dental Medicine Affiliate, Cambridge, Massachusetts, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| | - Xiaozhe Han
- Department of Oral Science and Translational Research, NSU College of Dental Medicine, Shanghai, China
| | - Yufeng Wang
- Shanghai Key Laboratory of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoyao Tang
- Shanghai Key Laboratory of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Stomatology, Shanghai Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Sharif K, Omar M, Lahat A, Patt YS, Amital H, Zoabi G, Bragazzi NL, Watad A. Big data- and machine learning-based analysis of a global pharmacovigilance database enables the discovery of sex-specific differences in the safety profile of dual IL4/IL13 blockade. Front Pharmacol 2023; 14:1271309. [PMID: 37954855 PMCID: PMC10637473 DOI: 10.3389/fphar.2023.1271309] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/04/2023] [Indexed: 11/14/2023] Open
Abstract
Background: Due to its apparent efficacy and safety, dupilumab, a monoclonal antibody that blocks Interleukin 4 (IL-4) and Interleukin 13 (IL-13), has been approved for treating T-helper 2 (Th2) disorders. However, adverse effects like local injection site reactions, conjunctivitis, headaches, and nasopharyngitis have been reported. Sex differences are known to influence both adaptive and innate immune responses and, thus, may have a bearing on the occurrence of these adverse effects. Nevertheless, the literature lacks a comprehensive exploration of this influence, a gap this study aims to bridge. Materials and Methods: A comprehensive data mining of VigiBase, the World Health Organization (WHO) global pharmacovigilance database which contains case safety reports of adverse drug reactions (ADRs) was performed to test for sex -specific safety response to dual IL4/IL13 blockade by dupilumab. The information component (IC), a measure of the disproportionality of ADR occurrence, was evaluated and compared between males and females to identify potential sexual dimorphism. Results: Of the 94,065 ADRs recorded in the WHO global pharmacovigilance database, 2,001 (57.4%) were reported among female dupilumab users, and 1,768 (50.7%) were among males. Immune/autoimmune T-helper 1 (Th1)-, innate- and T-helper 17 (Th17)-driven diseases and degenerative ones were consistently reported with a stronger association with Dupilumab in males than females. Some adverse events were more robustly associated with Dupilumab in females. Conclusion: Dupilumab has an excellent safety profile, even though some ADRs may occur. The risk is higher among male patients, further studies, including ad hoc studies, are needed to establish causality.
Collapse
Affiliation(s)
- Kassem Sharif
- Department of Gastroenterology, Sheba Medical Centre, Ramat Gan, Israel
- Department of Medicine B, Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Mahmud Omar
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Adi Lahat
- Department of Gastroenterology, Sheba Medical Centre, Ramat Gan, Israel
- Department of Medicine B, Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Yonatan Shneor Patt
- Department of Medicine B, Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Howard Amital
- Department of Medicine B, Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Ghanem Zoabi
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Nicola Luigi Bragazzi
- Laboratory for Industrial and Applied Mathematics (LIAM), Department of Mathematics and Statistics, York University, Toronto, ON, Canada
| | - Abdulla Watad
- Department of Medicine B, Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
- Section of Musculoskeletal Disease, NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Institute of Molecular Medicine, Chapel Allerton Hospital, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
9
|
Affiliation(s)
- Irina Tsymala
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| | - Karl Kuchler
- Department of Medical Biochemistry, Max Perutz Labs Vienna, Medical University of Vienna, Campus Vienna Biocenter, Vienna, Austria
| |
Collapse
|
10
|
Lujan RA, Pei L, Shannon JP, Dábilla N, Dolan PT, Hickman HD. Widespread and dynamic expression of granzyme C by skin-resident antiviral T cells. Front Immunol 2023; 14:1236595. [PMID: 37809077 PMCID: PMC10552530 DOI: 10.3389/fimmu.2023.1236595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023] Open
Abstract
After recognition of cognate antigen (Ag), effector CD8+ T cells secrete serine proteases called granzymes in conjunction with perforin, allowing granzymes to enter and kill target cells. While the roles for some granzymes during antiviral immune responses are well characterized, the function of others, such as granzyme C and its human ortholog granzyme H, is still unclear. Granzyme C is constitutively expressed by mature, cytolytic innate lymphoid 1 cells (ILC1s). Whether other antiviral effector cells also produce granzyme C and whether it is continually expressed or responsive to the environment is unknown. To explore this, we analyzed granzyme C expression in different murine skin-resident antiviral lymphocytes. At steady-state, dendritic epidermal T cells (DETCs) expressed granzyme C while dermal γδ T cells did not. CD8+ tissue-resident memory T cells (TRM) generated in response to cutaneous viral infection with the poxvirus vaccinia virus (VACV) also expressed granzyme C. Both DETCs and virus-specific CD8+ TRM upregulated granzyme C upon local VACV infection. Continual Ag exposure was not required for maintained TRM expression of granzyme C, although re-encounter with cognate Ag boosted expression. Additionally, IL-15 treatment increased granzyme C expression in both DETCs and TRM. Together, our data demonstrate that granzyme C is widely expressed by antiviral T cells in the skin and that expression is responsive to both environmental stimuli and TCR engagement. These data suggest that granzyme C may have functions other than killing in tissue-resident lymphocytes.
Collapse
Affiliation(s)
- Ramon A. Lujan
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
- School of Nursing, Duke University, Durham, NC, United States
| | - Luxin Pei
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - John P. Shannon
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Nathânia Dábilla
- Quantitative Virology and Evolution Unit, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD, United States
| | - Patrick T. Dolan
- Quantitative Virology and Evolution Unit, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD, United States
| | - Heather D. Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
11
|
Jaber Y, Netanely Y, Naamneh R, Saar O, Zubeidat K, Saba Y, Georgiev O, Kles P, Barel O, Horev Y, Yosef O, Eli-Berchoer L, Nadler C, Betser-Cohen G, Shapiro H, Elinav E, Wilensky A, Hovav AH. Langerhans cells shape postnatal oral homeostasis in a mechanical-force-dependent but microbiota and IL17-independent manner. Nat Commun 2023; 14:5628. [PMID: 37699897 PMCID: PMC10497507 DOI: 10.1038/s41467-023-41409-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
The postnatal interaction between microbiota and the immune system establishes lifelong homeostasis at mucosal epithelial barriers, however, the barrier-specific physiological activities that drive the equilibrium are hardly known. During weaning, the oral epithelium, which is monitored by Langerhans cells (LC), is challenged by the development of a microbial plaque and the initiation of masticatory forces capable of damaging the epithelium. Here we show that microbial colonization following birth facilitates the differentiation of oral LCs, setting the stage for the weaning period, in which adaptive immunity develops. Despite the presence of the challenging microbial plaque, LCs mainly respond to masticatory mechanical forces, inducing adaptive immunity, to maintain epithelial integrity that is also associated with naturally occurring alveolar bone loss. Mechanistically, masticatory forces induce the migration of LCs to the lymph nodes, and in return, LCs support the development of immunity to maintain epithelial integrity in a microbiota-independent manner. Unlike in adult life, this bone loss is IL-17-independent, suggesting that the establishment of oral mucosal homeostasis after birth and its maintenance in adult life involve distinct mechanisms.
Collapse
Affiliation(s)
- Yasmin Jaber
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Yasmine Netanely
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Reem Naamneh
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Or Saar
- Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel; Department of Periodontology, Hadassah Medical Center, Jerusalem, Israel
| | - Khaled Zubeidat
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Yasmin Saba
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Olga Georgiev
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Paz Kles
- Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel; Department of Periodontology, Hadassah Medical Center, Jerusalem, Israel
| | - Or Barel
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Yael Horev
- Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel; Department of Periodontology, Hadassah Medical Center, Jerusalem, Israel
| | - Omri Yosef
- The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Luba Eli-Berchoer
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Chen Nadler
- Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
- Department of Oral Medicine, Sedation & Maxillofacial Imaging, Hadassah Medical Center, Jerusalem, Israel
| | - Gili Betser-Cohen
- Division of Identification and Forensic Science, Police National HQ, Jerusalem, Israel
| | - Hagit Shapiro
- System Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- System Immunology Department, Weizmann Institute of Science, Rehovot, Israel
- Microbe & Cancer Division, DKFZ, Heidelberg, Germany
| | - Asaf Wilensky
- Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel; Department of Periodontology, Hadassah Medical Center, Jerusalem, Israel
| | - Avi-Hai Hovav
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel.
| |
Collapse
|
12
|
Hu L, Han M, Deng Y, Gong J, Hou Z, Zeng Y, Zhang Y, He J, Zhong C. Genetic distinction between functional tissue-resident and conventional natural killer cells. iScience 2023; 26:107187. [PMID: 37404378 PMCID: PMC10316664 DOI: 10.1016/j.isci.2023.107187] [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: 04/20/2023] [Revised: 06/01/2023] [Accepted: 06/16/2023] [Indexed: 07/06/2023] Open
Abstract
Tissue-residential natural killer (trNK) cells act as pioneering responders during infectious challenges. However, their discrimination with conventional NK (cNK) cells is still an issue. Through an integrative transcriptome comparison of the two NK subgroups from different tissues, we have defined two genesets capable of efficiently distinguishing them. Based on the two genesets, a fundamental difference between the activation of trNK and cNK is identified and further confirmed. Mechanistically, we have discovered a particular role of chromatin landscape in regulating the trNK activation. In addition, IL-21R and IL-18R are respectively highly expressed by trNK and cNK, indicating a role of cytokine milieu in determining their differential activation. Indeed, IL-21 is particularly critical in accessorily promoting trNK activation using a bunch of bifunctional transcription factors. Together, this study sheds light on the bona fide difference between trNK and cNK, which will further expand our knowledge about their distinct functionalities during immune responses.
Collapse
Affiliation(s)
- Luni Hu
- Institute of Systems Biomedicine, Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Mengwei Han
- Institute of Systems Biomedicine, Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yichen Deng
- Institute of Systems Biomedicine, Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jingjing Gong
- Institute of Systems Biomedicine, Peking University Health Science Center, Beijing 100191, China
| | - Zhiyuan Hou
- Institute of Systems Biomedicine, Peking University Health Science Center, Beijing 100191, China
| | - Yanyu Zeng
- Institute of Systems Biomedicine, Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yime Zhang
- Institute of Systems Biomedicine, Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jing He
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Chao Zhong
- Institute of Systems Biomedicine, Department of Immunology, NHC Key Laboratory of Medical Immunology (Peking University), Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
13
|
Seo H, Verma A, Kinzel M, Huang Q, Mahoney DJ, Jacquelot N. Targeting Potential of Innate Lymphoid Cells in Melanoma and Other Cancers. Pharmaceutics 2023; 15:2001. [PMID: 37514187 PMCID: PMC10384206 DOI: 10.3390/pharmaceutics15072001] [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: 06/13/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Reinvigorating the killing function of tumor-infiltrating immune cells through the targeting of regulatory molecules expressed on lymphocytes has markedly improved the prognosis of cancer patients, particularly in melanoma. While initially thought to solely strengthen adaptive T lymphocyte anti-tumor activity, recent investigations suggest that other immune cell subsets, particularly tissue-resident innate lymphoid cells (ILCs), may benefit from immunotherapy treatment. Here, we describe the recent findings showing immune checkpoint expression on tissue-resident and tumor-infiltrating ILCs and how their effector function is modulated by checkpoint blockade-based therapies in cancer. We discuss the therapeutic potential of ILCs beyond the classical PD-1 and CTLA-4 regulatory molecules, exploring other possibilities to manipulate ILC effector function to further impede tumor growth and quench disease progression.
Collapse
Affiliation(s)
- Hobin Seo
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| | - Amisha Verma
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Megan Kinzel
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| | - Qiutong Huang
- The University of Queensland Frazer Institute, University of Queensland, Woolloongabba, QLD 4102, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Douglas J Mahoney
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| | - Nicolas Jacquelot
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Arnie Charbonneau Cancer Research Institute, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
14
|
Stolley JM, Scott MC, Joag V, Dale AJ, Johnston TS, Saavedra F, Gavil NV, Lotfi-Emran S, Soerens AG, Weyu E, Pierson MJ, Herzberg MC, Zhang N, Vezys V, Masopust D. Depleting CD103+ resident memory T cells in vivo reveals immunostimulatory functions in oral mucosa. J Exp Med 2023; 220:e20221853. [PMID: 37097449 PMCID: PMC10130744 DOI: 10.1084/jem.20221853] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/15/2023] [Accepted: 03/31/2023] [Indexed: 04/26/2023] Open
Abstract
The oral mucosa is a frontline for microbial exposure and juxtaposes several unique tissues and mechanical structures. Based on parabiotic surgery of mice receiving systemic viral infections or co-housing with microbially diverse pet shop mice, we report that the oral mucosa harbors CD8+ CD103+ resident memory T cells (TRM), which locally survey tissues without recirculating. Oral antigen re-encounter during the effector phase of immune responses potentiated TRM establishment within tongue, gums, palate, and cheek. Upon reactivation, oral TRM triggered changes in somatosensory and innate immune gene expression. We developed in vivo methods for depleting CD103+ TRM while sparing CD103neg TRM and recirculating cells. This revealed that CD103+ TRM were responsible for inducing local gene expression changes. Oral TRM putatively protected against local viral infection. This study provides methods for generating, assessing, and in vivo depleting oral TRM, documents their distribution throughout the oral mucosa, and provides evidence that TRM confer protection and trigger responses in oral physiology and innate immunity.
Collapse
Affiliation(s)
- J. Michael Stolley
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Milcah C. Scott
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Vineet Joag
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Alexander J. Dale
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Timothy S. Johnston
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Flavia Saavedra
- School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Noah V. Gavil
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Sahar Lotfi-Emran
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Andrew G. Soerens
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Eyob Weyu
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Mark J. Pierson
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Mark C. Herzberg
- School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Vaiva Vezys
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - David Masopust
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
15
|
Ma Z, Wang J, Hu L, Wang S. Function of Innate Lymphoid Cells in Periodontal Tissue Homeostasis: A Narrative Review. Int J Mol Sci 2023; 24:ijms24076099. [PMID: 37047071 PMCID: PMC10093809 DOI: 10.3390/ijms24076099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/16/2023] [Accepted: 03/18/2023] [Indexed: 04/14/2023] Open
Abstract
Periodontitis is an irreversible inflammatory response that occurs in periodontal tissues. Given the size and diversity of natural flora in the oral mucosa, host immunity must strike a balance between pathogen identification and a complicated system of tolerance. The innate immune system, which includes innate lymphoid cells (ILCs), certainly plays a crucial role in regulating this homeostasis because pathogens are quickly recognized and responded to. ILCs are a recently discovered category of tissue-resident lymphocytes that lack adaptive antigen receptors. ILCs are found in both lymphoid and non-lymphoid organs and are particularly prevalent at mucosal barrier surfaces, where they control inflammatory response and homeostasis. Recent studies have shown that ILCs are important players in periodontitis; however, the mechanisms that govern the innate immune response in periodontitis still require further investigation. This review focuses on the intricate crosstalk between ILCs and the microenvironment in periodontal tissue homeostasis, with the purpose of regulating or improving immune responses in periodontitis prevention and therapy.
Collapse
Affiliation(s)
- Zhiyu Ma
- Beijing Laboratory of Oral Health, School of Basic Medicine, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Jinsong Wang
- Beijing Laboratory of Oral Health, School of Basic Medicine, School of Stomatology, Capital Medical University, Beijing 100050, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Capital Medical University Beijing 100070, China
| | - Lei Hu
- Beijing Laboratory of Oral Health, School of Basic Medicine, School of Stomatology, Capital Medical University, Beijing 100050, China
- Department of Prosthodontics, School of Stomatology, Capital Medical University, Beijing 100050, China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing 100070, China
| | - Songlin Wang
- Beijing Laboratory of Oral Health, School of Basic Medicine, School of Stomatology, Capital Medical University, Beijing 100050, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Capital Medical University Beijing 100070, China
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing 100070, China
- Laboratory for Oral and General Health Integration and Translation, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing 100700, China
| |
Collapse
|
16
|
Laufer Britva R, Keren A, Bertolini M, Ullmann Y, Paus R, Gilhar A. Involvement of ILC1-like innate lymphocytes in human autoimmunity, lessons from alopecia areata. eLife 2023; 12:80768. [PMID: 36930216 PMCID: PMC10023162 DOI: 10.7554/elife.80768] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Here, we have explored the involvement of innate lymphoid cells-type 1 (ILC1) in the pathogenesis of alopecia areata (AA), because we found them to be significantly increased around lesional and non-lesional HFs of AA patients. To further explore these unexpected findings, we first co-cultured autologous circulating ILC1-like cells (ILC1lc) with healthy, but stressed, organ-cultured human scalp hair follicles (HFs). ILClc induced all hallmarks of AA ex vivo: they significantly promoted premature, apoptosis-driven HF regression (catagen), HF cytotoxicity/dystrophy, and most important for AA pathogenesis, the collapse of the HFs physiological immune privilege. NKG2D-blocking or IFNγ-neutralizing antibodies antagonized this. In vivo, intradermal injection of autologous activated, NKG2D+/IFNγ-secreting ILC1lc into healthy human scalp skin xenotransplanted onto SCID/beige mice sufficed to rapidly induce characteristic AA lesions. This provides the first evidence that ILC1lc, which are positive for the ILC1 phenotype and negative for the classical NK markers, suffice to induce AA in previously healthy human HFs ex vivo and in vivo, and further questions the conventional wisdom that AA is always an autoantigen-dependent, CD8 +T cell-driven autoimmune disease.
Collapse
Affiliation(s)
- Rimma Laufer Britva
- Skin Research Laboratory, Rappaport Faculty of Medicine, Technion – Israel Institute of TechnologyHaifaIsrael
- Department of Dermatology, Rambam Health Care CampusHaifaIsrael
| | - Aviad Keren
- Skin Research Laboratory, Rappaport Faculty of Medicine, Technion – Israel Institute of TechnologyHaifaIsrael
| | | | - Yehuda Ullmann
- Department of Plastic Surgery, Rambam Medical CenterHaifaIsrael
| | - Ralf Paus
- Monasterium LaboratoryMünsterGermany
- Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, Miller School of Medicine, University of MiamiMiamiUnited States
- CUTANEONHamburgGermany
| | - Amos Gilhar
- Skin Research Laboratory, Rappaport Faculty of Medicine, Technion – Israel Institute of TechnologyHaifaIsrael
| |
Collapse
|
17
|
Taggenbrock RLRE, van Gisbergen KPJM. ILC1: Development, maturation, and transcriptional regulation. Eur J Immunol 2023; 53:e2149435. [PMID: 36408791 PMCID: PMC10099236 DOI: 10.1002/eji.202149435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/26/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022]
Abstract
Type 1 Innate Lymphoid cells (ILC1s) are tissue-resident cells that partake in the regulation of inflammation and homeostasis. A major feature of ILC1s is their ability to rapidly respond after infections. The effector repertoire of ILC1s includes the pro-inflammatory cytokines IFN-γ and TNF-α and cytotoxic mediators such as granzymes, which enable ILC1s to establish immune responses and to directly kill target cells. Recent advances in the characterization of ILC1s have considerably furthered our understanding of ILC1 development and maintenance in tissues. In particular, it has become clear how ILC1s operate independently from conventional natural killer cells, with which they share many characteristics. In this review, we discuss recent developments with regards to the differentiation, polarization, and effector maturation of ILC1s. These processes may underlie the observed heterogeneity in ILC1 populations within and between different tissues. Next, we highlight transcriptional programs that control each of the separate steps in the differentiation of ILC1s. These transcriptional programs are shared with other tissue-resident type-1 lymphocytes, such as tissue-resident memory T cells (TRM ) and invariant natural killer T cells (iNKT), highlighting that ILC1s utilize networks of transcriptional regulation that are conserved between lymphocyte lineages to respond effectively to tissue-invading pathogens.
Collapse
Affiliation(s)
- Renske L R E Taggenbrock
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Krämer B, Nalin AP, Ma F, Eickhoff S, Lutz P, Leonardelli S, Goeser F, Finnemann C, Hack G, Raabe J, ToVinh M, Ahmad S, Hoffmeister C, Kaiser KM, Manekeller S, Branchi V, Bald T, Hölzel M, Hüneburg R, Nischalke HD, Semaan A, Langhans B, Kaczmarek DJ, Benner B, Lordo MR, Kowalski J, Gerhardt A, Timm J, Toma M, Mohr R, Türler A, Charpentier A, van Bremen T, Feldmann G, Sattler A, Kotsch K, Abdallah AT, Strassburg CP, Spengler U, Carson WE, Mundy-Bosse BL, Pellegrini M, O'Sullivan TE, Freud AG, Nattermann J. Single-cell RNA sequencing identifies a population of human liver-type ILC1s. Cell Rep 2023; 42:111937. [PMID: 36640314 PMCID: PMC9950534 DOI: 10.1016/j.celrep.2022.111937] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/30/2022] [Accepted: 12/15/2022] [Indexed: 01/02/2023] Open
Abstract
Group 1 innate lymphoid cells (ILCs) comprise a heterogeneous family of cytotoxic natural killer (NK) cells and ILC1s. We identify a population of "liver-type" ILC1s with transcriptional, phenotypic, and functional features distinct from those of conventional and liver-resident NK cells as well as from other previously described human ILC1 subsets. LT-ILC1s are CD49a+CD94+CD200R1+, express the transcription factor T-BET, and do not express the activating receptor NKp80 or the transcription factor EOMES. Similar to NK cells, liver-type ILC1s produce IFN-γ, TNF-α, and GM-CSF; however, liver-type ILC1s also produce IL-2 and lack perforin and granzyme-B. Liver-type ILC1s are expanded in cirrhotic liver tissues, and they can be produced from blood-derived ILC precursors in vitro in the presence of TGF-β1 and liver sinusoidal endothelial cells. Cells with similar signature and function can also be found in tonsil and intestinal tissues. Collectively, our study identifies and classifies a population of human cross-tissue ILC1s.
Collapse
Affiliation(s)
- Benjamin Krämer
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), 53127 Bonn, Germany.
| | - Ansel P Nalin
- Medical Scientist Training Program, The Ohio State University, Columbus, OH 43210, USA; Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Feiyang Ma
- Molecular Cell and Developmental Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sarah Eickhoff
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Philipp Lutz
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), 53127 Bonn, Germany
| | - Sonia Leonardelli
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Felix Goeser
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), 53127 Bonn, Germany
| | - Claudia Finnemann
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), 53127 Bonn, Germany
| | - Gudrun Hack
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), 53127 Bonn, Germany
| | - Jan Raabe
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), 53127 Bonn, Germany
| | - Michael ToVinh
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), 53127 Bonn, Germany
| | - Sarah Ahmad
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), 53127 Bonn, Germany
| | - Christoph Hoffmeister
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), 53127 Bonn, Germany
| | - Kim M Kaiser
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), 53127 Bonn, Germany
| | | | | | - Tobias Bald
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Michael Hölzel
- Institute of Experimental Oncology (IEO), Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Robert Hüneburg
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany
| | | | | | - Bettina Langhans
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), 53127 Bonn, Germany
| | | | - Brooke Benner
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Matthew R Lordo
- Medical Scientist Training Program, The Ohio State University, Columbus, OH 43210, USA; Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | | | - Adam Gerhardt
- College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jörg Timm
- Institute of Virology, University of Duesseldorf, 40225 Düsseldorf, Germany
| | - Marieta Toma
- Department of Pathology, University of Bonn, 53127 Bonn, Germany
| | - Raphael Mohr
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany
| | - Andreas Türler
- General and Visceral Surgery, Johanniter Hospital, 53113 Bonn, Germany
| | - Arthur Charpentier
- Department of Otorhinolaryngology/Head and Neck Surgery, University of Bonn, 53127 Bonn, Germany; Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Tobias van Bremen
- Department of Otorhinolaryngology/Head and Neck Surgery, University of Bonn, 53127 Bonn, Germany
| | - Georg Feldmann
- Department of Internal Medicine III, University of Bonn, 53127 Bonn, Germany
| | - Arne Sattler
- Clinic for Surgery, Transplant Immunology Lab, Charité University Hospital Berlin, 10117 Berlin, Germany
| | - Katja Kotsch
- Clinic for Surgery, Transplant Immunology Lab, Charité University Hospital Berlin, 10117 Berlin, Germany
| | - Ali T Abdallah
- Interdisciplinary Center for Clinical Research, RWTH Aachen University, 52074 Aachen, Germany
| | | | - Ulrich Spengler
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), 53127 Bonn, Germany
| | - William E Carson
- Division of Surgical Oncology, Department of Surgery, Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - Bethany L Mundy-Bosse
- Division of Hematology, Department of Internal Medicine, Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Matteo Pellegrini
- Molecular Cell and Developmental Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 900953, USA
| | - Aharon G Freud
- Department of Pathology, Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA.
| | - Jacob Nattermann
- Department of Internal Medicine I, University of Bonn, 53127 Bonn, Germany; German Center for Infection Research (DZIF), 53127 Bonn, Germany
| |
Collapse
|
19
|
Differential Regulation of Innate Lymphoid Cells in Human and Murine Oral Squamous Cell Carcinoma. Int J Mol Sci 2023; 24:ijms24021627. [PMID: 36675138 PMCID: PMC9865302 DOI: 10.3390/ijms24021627] [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: 12/02/2022] [Revised: 01/09/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Oral squamous cell carcinomas (OSCC) remain a major healthcare burden in Asian countries. In Pakistan alone, it is the most common cancer in males and second only to breast cancer in females. Alarmingly, treatment options for OSCC remain limited. With this context, investigations made to explore the inflammatory milieu of OSCC become highly relevant, with the hope of practicing immunotherapeutic approaches to address this highly prevalent tumor. We investigated the newly identified innate lymphoid cells (ILCs) and associated cytokines in well-defined human oral squamous cell carcinoma (OSCC) as well as in a 7,12-dimethylbenz[a]anthracene (DMBA)-induced murine model of OSCC using flow cytometry and quantitative real-time polymerase chain reaction (qPCR). We further went on to explore molecular circuitry involved in OSCC by developing a murine model of OSCC and using an α-Thy1 antibody to inhibit ILCs. Amongst the ILCs that we found in human OSCC, ILC3 (23%) was the most abundant, followed by ILC2 (17%) and ILC1 (1%). Mice were divided into four groups: DMBA (n = 33), DMBA+antibody (Ab) (n = 30), acetone (n = 5), and control (n = 5). In murine OSCC tissues, ILC1 and ILC3 were down-infiltrated, while ILC2 remained unchanged compared to controls. Interestingly, compared to the controls (DMBA group), mice treated with the α-Thy1 antibody showed fewer numbers of large tumors, and a larger percentage of these mice were tumor-free at this study's end point. We present novel data on the differential expansion/downsizing of ILCs in OSCC, which provides a pivotal basis to dive deeper into molecular circuitry and the OSCC tumor niche to devise novel diagnostic, therapeutic, and prognostic strategies to prevent/treat oral cancers.
Collapse
|
20
|
Hu Q, Liu B, Fan Y, Zheng Y, Wen F, Yu U, Wang W. Multi-omics association analysis reveals interactions between the oropharyngeal microbiome and the metabolome in pediatric patients with influenza A virus pneumonia. Front Cell Infect Microbiol 2022; 12:1011254. [PMID: 36389138 PMCID: PMC9651038 DOI: 10.3389/fcimb.2022.1011254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/03/2022] [Indexed: 11/30/2022] Open
Abstract
Children are at high risk for influenza A virus (IAV) infections, which can develop into severe illnesses. However, little is known about interactions between the microbiome and respiratory tract metabolites and their impact on the development of IAV pneumonia in children. Using a combination of liquid chromatography tandem mass spectrometry (LC-MS/MS) and 16S rRNA gene sequencing, we analyzed the composition and metabolic profile of the oropharyngeal microbiota in 49 pediatric patients with IAV pneumonia and 42 age-matched healthy children. The results indicate that compared to healthy children, children with IAV pneumonia exhibited significant changes in the oropharyngeal macrobiotic structure (p = 0.001), and significantly lower microbial abundance and diversity (p < 0.05). These changes came with significant disturbances in the levels of oropharyngeal metabolites. Intergroup differences were observed in 204 metabolites mapped to 36 metabolic pathways. Significantly higher levels of sphingolipid (sphinganine and phytosphingosine) and propanoate (propionic acid and succinic acid) metabolism were observed in patients with IAV pneumonia than in healthy controls. Using Spearman’s rank-correlation analysis, correlations between IAV pneumonia-associated discriminatory microbial genera and metabolites were evaluated. The results indicate significant correlations and consistency in variation trends between Streptococcus and three sphingolipid metabolites (phytosphingosine, sphinganine, and sphingosine). Besides these three sphingolipid metabolites, the sphinganine-to-sphingosine ratio and the joint analysis of the three metabolites indicated remarkable diagnostic efficacy in children with IAV pneumonia. This study confirmed significant changes in the characteristics and metabolic profile of the oropharyngeal microbiome in pediatric patients with IAV pneumonia, with high synergy between the two factors. Oropharyngeal sphingolipid metabolites may serve as potential diagnostic biomarkers of IAV pneumonia in children.
Collapse
Affiliation(s)
- Qian Hu
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen, China
| | - Baiming Liu
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen, China
| | - Yanqun Fan
- Department of Trans-omics Research, Biotree Metabolomics Technology Research Center, Shanghai, China
| | - Yuejie Zheng
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen, China
| | - Feiqiu Wen
- Department of Hematology and Oncology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Uet Yu
- Department of Hematology and Oncology, Shenzhen Children’s Hospital, Shenzhen, China
- *Correspondence: Wenjian Wang, ; Uet Yu,
| | - Wenjian Wang
- Department of Respiratory Diseases, Shenzhen Children’s Hospital, Shenzhen, China
- *Correspondence: Wenjian Wang, ; Uet Yu,
| |
Collapse
|
21
|
Sparano C, Solís-Sayago D, Vijaykumar A, Rickenbach C, Vermeer M, Ingelfinger F, Litscher G, Fonseca A, Mussak C, Mayoux M, Friedrich C, Nombela-Arrieta C, Gasteiger G, Becher B, Tugues S. Embryonic and neonatal waves generate distinct populations of hepatic ILC1s. Sci Immunol 2022; 7:eabo6641. [PMID: 36054340 DOI: 10.1126/sciimmunol.abo6641] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Group 1 innate lymphoid cells (ILCs) comprising circulating natural killer (cNK) cells and tissue-resident ILC1s are critical for host defense against pathogens and tumors. Despite a growing understanding of their role in homeostasis and disease, the ontogeny of group 1 ILCs remains largely unknown. Here, we used fate mapping and single-cell transcriptomics to comprehensively investigate the origin and turnover of murine group 1 ILCs. Whereas cNK cells are continuously replaced throughout life, we uncovered tissue-dependent development and turnover of ILC1s. A first wave of ILC1s emerges during embryogenesis in the liver and transiently colonizes fetal tissues. After birth, a second wave quickly replaces ILC1s in most tissues apart from the liver, where they layer with embryonic ILC1s, persist until adulthood, and undergo a specific developmental program. Whereas embryonically derived ILC1s give rise to a cytotoxic subset, the neonatal wave establishes the full spectrum of ILC1s. Our findings uncover key ontogenic features of murine group 1 ILCs and their association with cellular identities and functions.
Collapse
Affiliation(s)
- Colin Sparano
- Innate Lymphoid Cells and Cancer, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Darío Solís-Sayago
- Innate Lymphoid Cells and Cancer, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Anjali Vijaykumar
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Chiara Rickenbach
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Marijne Vermeer
- Innate Lymphoid Cells and Cancer, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Florian Ingelfinger
- Inflammation Research, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Gioana Litscher
- Innate Lymphoid Cells and Cancer, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - André Fonseca
- Innate Lymphoid Cells and Cancer, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Caroline Mussak
- Innate Lymphoid Cells and Cancer, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Maud Mayoux
- Innate Lymphoid Cells and Cancer, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Christin Friedrich
- Würzburg Institute of Systems Immunology, Max Planck Research Group, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Georg Gasteiger
- Würzburg Institute of Systems Immunology, Max Planck Research Group, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Burkhard Becher
- Inflammation Research, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sonia Tugues
- Innate Lymphoid Cells and Cancer, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
Dou X, Yu X, Du S, Han Y, Li L, Zhang H, Yao Y, Du Y, Wang X, Li J, Yang T, Zhang W, Yang C, Ma F, He S. Interferon‐mediated repression of
miR
‐324‐5p potentiates necroptosis to facilitate antiviral defense. EMBO Rep 2022; 23:e54438. [PMID: 35735238 PMCID: PMC9346494 DOI: 10.15252/embr.202154438] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/23/2022] [Accepted: 06/01/2022] [Indexed: 11/22/2022] Open
Abstract
Mixed lineage kinase domain‐like protein (MLKL) is the terminal effector of necroptosis, a form of regulated necrosis. Optimal activation of necroptosis, which eliminates infected cells, is critical for antiviral host defense. MicroRNAs (miRNAs) regulate the expression of genes involved in various biological and pathological processes. However, the roles of miRNAs in necroptosis‐associated host defense remain largely unknown. We screened a library of miRNAs and identified miR‐324‐5p as the most effective suppressor of necroptosis. MiR‐324‐5p downregulates human MLKL expression by specifically targeting the 3′UTR in a seed region‐independent manner. In response to interferons (IFNs), miR‐324‐5p is downregulated via the JAK/STAT signaling pathway, which removes the posttranscriptional suppression of MLKL mRNA and facilitates the activation of necroptosis. In influenza A virus (IAV)‐infected human primary macrophages, IFNs are induced, leading to the downregulation of miR‐324‐5p. MiR‐324‐5p overexpression attenuates IAV‐associated necroptosis and enhances viral replication, whereas deletion of miR‐324‐5p potentiates necroptosis and suppresses viral replication. Hence, miR‐324‐5p negatively regulates necroptosis by manipulating MLKL expression, and its downregulation by IFNs orchestrates optimal activation of necroptosis in host antiviral defense.
Collapse
Affiliation(s)
- Xiaoyan Dou
- Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology Soochow University Suzhou China
| | - Xiaoliang Yu
- CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Suzhou Institute of Systems Medicine Suzhou China
| | - Shujing Du
- CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Suzhou Institute of Systems Medicine Suzhou China
| | - Yu Han
- CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Suzhou Institute of Systems Medicine Suzhou China
| | - Liang Li
- CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Suzhou Institute of Systems Medicine Suzhou China
| | - Haoran Zhang
- Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology Soochow University Suzhou China
| | - Ying Yao
- Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology Soochow University Suzhou China
- CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Suzhou Institute of Systems Medicine Suzhou China
| | - Yayun Du
- CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Suzhou Institute of Systems Medicine Suzhou China
| | - Xinhui Wang
- CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Suzhou Institute of Systems Medicine Suzhou China
| | - Jingjing Li
- CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Suzhou Institute of Systems Medicine Suzhou China
| | - Tao Yang
- CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Suzhou Institute of Systems Medicine Suzhou China
| | - Wei Zhang
- CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Suzhou Institute of Systems Medicine Suzhou China
| | - Chengkui Yang
- CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Suzhou Institute of Systems Medicine Suzhou China
| | - Feng Ma
- CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Suzhou Institute of Systems Medicine Suzhou China
| | - Sudan He
- Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology Soochow University Suzhou China
- CAMS Key Laboratory of Synthetic Biology Regulatory Elements, Institute of Systems Medicine Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Suzhou Institute of Systems Medicine Suzhou China
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| |
Collapse
|
23
|
Li Z, Ma R, Ma S, Tian L, Lu T, Zhang J, Mundy-Bosse BL, Zhang B, Marcucci G, Caligiuri MA, Yu J. ILC1s control leukemia stem cell fate and limit development of AML. Nat Immunol 2022; 23:718-730. [PMID: 35487987 PMCID: PMC9106917 DOI: 10.1038/s41590-022-01198-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 03/23/2022] [Indexed: 12/13/2022]
Abstract
Type I innate lymphoid cells (ILC1s) are critical regulators of inflammation and immunity in mammalian tissues. However, their function in cancer is mostly undefined. Here, we show that a high density of ILC1s induces leukemia stem cell (LSC) apoptosis in mice. At a lower density, ILC1s prevent LSCs from differentiating into leukemia progenitors and promote their differentiation into non-leukemic cells, thus blocking the production of terminal myeloid blasts. All of these effects, which require ILC1s to produce interferon-γ after cell-cell contact with LSCs, converge to suppress leukemogenesis in vivo. Conversely, the antileukemia potential of ILC1s wanes when JAK-STAT or PI3K-AKT signaling is inhibited. The relevant antileukemic properties of ILC1s are also functional in healthy individuals and impaired in individuals with acute myeloid leukemia (AML). Collectively, these findings identify ILC1s as anticancer immune cells that might be suitable for AML immunotherapy and provide a potential strategy to treat AML and prevent relapse of the disease.
Collapse
Affiliation(s)
- Zhenlong Li
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Rui Ma
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Shoubao Ma
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Lei Tian
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Ting Lu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Jianying Zhang
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Bethany L Mundy-Bosse
- Division of Hematology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Bin Zhang
- Gehr Family Center for Leukemia Research, Department of Hematological Malignancies Translational Science, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Guido Marcucci
- Gehr Family Center for Leukemia Research, Department of Hematological Malignancies Translational Science, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Michael A Caligiuri
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA.
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA.
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, USA.
| | - Jianhua Yu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA.
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA.
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, USA.
- Department of Immuno-Oncology, City of Hope, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Lai D, Chen W, Zhang K, Scott MJ, Li Y, Billiar TR, Wilson MA, Fan J. GRK2 regulates group 2 innate lymphoid cell mobilization in sepsis. Mol Med 2022; 28:32. [PMID: 35272622 PMCID: PMC8908620 DOI: 10.1186/s10020-022-00459-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 02/28/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Sepsis induces group 2 innate lymphoid cell (ILC2) expansion in the lung. However, the origin of these lung-recruited ILC2 and the mechanism of ILC2 expansion are unclear. This study aims to determine the origin of lung-recruited ILC2 and its underlying mechanism in sepsis. METHODS Sepsis was induced by cecal ligation and puncture (CLP) model in wild-type, IL-33-deficient and ST2-deficient mice. The frequency, cell number and C-X-C chemokine receptor 4 (CXCR4) expression of ILC2 in bone marrow (BM), blood and lung were measured by flow cytometry. In the in vitro studies, purified ILC2 progenitor (ILC2p) were challenged with IL-33 or G protein-coupled receptor kinase 2 (GRK2) inhibitor, the CXCR4 expression and GRK2 activity were detected by confocal microscopy or flow cytometry. RESULTS We show that IL-33 acts through its receptor, ST2, on BM ILC2p to induce GRK2 expression and subsequent downregulation of cell surface expression of CXCR4, which results in decreasing retention of ILC2p in the BM and promoting expansion of ILC2 in the lung. Importantly, we demonstrate that reduced IL-33 level in aging mice contributes to impaired ILC2 mobilization from BM and accumulation in the lung following sepsis. CONCLUSION This study identifies a novel pathway in regulating ILC2p mobilization and expansion during sepsis and indicates BM as the main source of ILC2 in the lung following sepsis.
Collapse
Affiliation(s)
- Dengming Lai
- grid.21925.3d0000 0004 1936 9000Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, 15213 USA ,grid.13402.340000 0004 1759 700XDepartment of Neonatal Surgery, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052 China
| | - Weiwei Chen
- grid.21925.3d0000 0004 1936 9000Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, 15213 USA
| | - Kai Zhang
- grid.21925.3d0000 0004 1936 9000Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, 15213 USA
| | - Melanie J. Scott
- grid.21925.3d0000 0004 1936 9000Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, 15213 USA
| | - Yuehua Li
- grid.21925.3d0000 0004 1936 9000Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, 15213 USA
| | - Timothy R. Billiar
- grid.21925.3d0000 0004 1936 9000Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, 15213 USA ,grid.21925.3d0000 0004 1936 9000McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219 USA
| | - Mark A. Wilson
- grid.21925.3d0000 0004 1936 9000Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, 15213 USA ,grid.413935.90000 0004 0420 3665Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240 USA
| | - Jie Fan
- grid.21925.3d0000 0004 1936 9000Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, 15213 USA ,grid.21925.3d0000 0004 1936 9000McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219 USA ,grid.413935.90000 0004 0420 3665Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240 USA ,grid.21925.3d0000 0004 1936 9000Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, 15213 USA
| |
Collapse
|
25
|
Vrba SM, Hickman HD. Imaging viral infection in vivo to gain unique perspectives on cellular antiviral immunity. Immunol Rev 2022; 306:200-217. [PMID: 34796538 PMCID: PMC9073719 DOI: 10.1111/imr.13037] [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: 10/14/2021] [Accepted: 10/17/2021] [Indexed: 11/29/2022]
Abstract
The past decade has seen near continual global public health crises caused by emerging viral infections. Extraordinary increases in our knowledge of the mechanisms underlying successful antiviral immune responses in animal models and during human infection have accompanied these viral outbreaks. Keeping pace with the rapidly advancing field of viral immunology, innovations in microscopy have afforded a previously unseen view of viral infection occurring in real-time in living animals. Here, we review the contribution of intravital imaging to our understanding of cell-mediated immune responses to viral infections, with a particular focus on studies that visualize the antiviral effector cells responding to infection as well as virus-infected cells. We discuss methods to visualize viral infection in vivo using intravital microscopy (IVM) and significant findings arising through the application of IVM to viral infection. Collectively, these works underscore the importance of developing a comprehensive spatial understanding of the relationships between immune effectors and virus-infected cells and how this has enabled unique discoveries about virus/host interactions and antiviral effector cell biology.
Collapse
Affiliation(s)
- Sophia M. Vrba
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Heather D. Hickman
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Correspondence to: HDH. . 10 Center Drive, Rm 11N244A. Bethesda, MD. 20892. 301-761-6330
| |
Collapse
|
26
|
Shannon JP, Cherry CR, Vrba SM, Hickman HD. Protocol for analyzing and visualizing antiviral immune responses after acute infection of the murine oral mucosa. STAR Protoc 2021; 2:100790. [PMID: 34622218 PMCID: PMC8479830 DOI: 10.1016/j.xpro.2021.100790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The oral mucosa is an important site for virus infection and transmission, yet few animal models exist to examine the virology, pathology, and immunology of acute oral mucosal viral infection. Here, we provide a protocol for infecting and imaging the inner lip (labial mucosa) of mice with the poxvirus vaccinia virus (VACV). Inoculation of the labial mucosa with a bifurcated needle results in viral replication and priming of an adaptive antiviral response that can be imaged using intravital microscopy. For complete details on the use and execution of this protocol, please refer to Shannon et al. (2021). The oral mucosa is a critical barrier tissue during viral infection Vaccinia virus infection provides an acute oral mucosal viral challenge model Antiviral immune responses can be analyzed using ex vivo and in vivo approaches The murine inner lip (labial mucosa) can be imaged using intravital microscopy
Collapse
Affiliation(s)
- John P Shannon
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christian R Cherry
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sophia M Vrba
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Heather D Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
27
|
Zubeidat K, Hovav AH. Shaped by the epithelium - postnatal immune mechanisms of oral homeostasis. Trends Immunol 2021; 42:622-634. [PMID: 34083119 DOI: 10.1016/j.it.2021.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023]
Abstract
The first encounter of mucosal barriers with the microbiota initiates host-microbiota feedback loops instructing the tailored development of both the immune system and microbiota at each mucosal site. Once established, balanced immunological interactions enable symbiotic relationships with the microbiota in adult life. This process has been extensively investigated in the mammalian monolayer epithelium-covered intestine and lung mucosae; however, the postnatal mechanisms engaged by the oral mucosa to establish homeostasis are currently being discovered. Here, we discuss the early life dialogue between the oral mucosa and the microbiota, with particular emphasis on the central role the multilayer epithelium plays to protect the oral mucosa. These intricate and unique postnatal immunological processes shape oral homeostasis, which can potentially affect buccal and systemic health in adult life.
Collapse
Affiliation(s)
- Khaled Zubeidat
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Avi-Hai Hovav
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel.
| |
Collapse
|
28
|
Abstract
Type 1 innate lymphoid cells (ILC1s) regulate inflammation in the tissues; however, their role in anti-viral immunity remains largely unknown. In this issue of Immunity, Shannon et al. report that ILC1s invoke an anti-viral effect by producing interferon (IFN)γ at homeostasis, thereby limiting viral replication in the oral mucosa.
Collapse
Affiliation(s)
- Tsukasa Nabekura
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan; Department of Immunology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan; R&D Center for Innovative Drug Discovery, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Akira Shibuya
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan; Department of Immunology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan; R&D Center for Innovative Drug Discovery, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|