1
|
Schulze TT, Neville AJ, Ehrhorn EG, Alsuleiman SA, Vennerstrom JL, Davis PH. Spleen and peripheral blood immunopathology in an outbred model of adult-stage murine schistosomiasis. Front Immunol 2025; 16:1527129. [PMID: 40375983 PMCID: PMC12078228 DOI: 10.3389/fimmu.2025.1527129] [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: 11/12/2024] [Accepted: 03/18/2025] [Indexed: 05/18/2025] Open
Abstract
Schistosomiasis, a parasitic disease caused by flatworms of genus Schistosoma, is a neglected tropical disease that causes significant morbidity in the developing world. Despite numerous efforts to eradicate the disease, the parasite remains a significant global burden, particularly within Sub-Saharan Africa. Schistosoma species possess an arsenal of potent mechanisms to defend against direct attack from host immune cells and a remarkable ability to modulate the host immune system through proximal and systemic modes that facilitate its stage-specific development, survival, and reproduction. Standardized animal models have been developed that serve as an important resource for dissecting parasite and host immunobiology and for drug and vaccine efficacy studies. However, a comprehensive understanding of the immune responses to Schistosoma mansoni in the standard outbred Swiss Webster mouse model is still lacking, particularly with the granulocyte composition of the spleen and the associated blood cytokine responses that occur during chronic infections. To continue characterization of this important secondary lymphoid tissue and the peripheral blood, we examined infected mouse spleens and additionally performed a detailed flow cytometric analysis of the splenic compartment from infected mice with specific attention to granulocytes and Th2-associated leukocytes. Peripheral blood from infected animals was used to evaluate a panel of Th1- and Th2-associated cytokines for comparison. Lastly, an analytical blood count and differential was also reported to provide a case study of late-stage chronic schistosomiasis. In mice infected with S. mansoni, we identified granulocytosis within the spleen including increased eosinophils, neutrophils, basophils, and mast cells. Additionally, ILC2s and dendritic cells were increased. The cytokine data suggests an IL33-independent mixed Th1/Th2 response with upregulation of granulocyte proliferative and recruitment factors. The late-stage chronic schistosomiasis case study identified blood neutrophilia and eosinophilia but with absent basophils. These data enhance our understanding of the complex immune response that occurs with schistosomiasis and may offer new insights to support therapeutic strategies against this important disease.
Collapse
Affiliation(s)
- Thomas T. Schulze
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, United States
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Andrew J. Neville
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, United States
| | - Evie G. Ehrhorn
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, United States
| | - Sarah A. Alsuleiman
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, United States
| | - Jonathan L. Vennerstrom
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Paul H. Davis
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, United States
| |
Collapse
|
2
|
Gosia M, Doshi G, Bagwe Parab S, Godad A. Innovative Approaches to Psoriasis: Small Molecules Targeting Key Signaling Pathways. Immunol Invest 2025:1-37. [PMID: 39819440 DOI: 10.1080/08820139.2025.2449960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
BACKGROUND Psoriasis (Pso) is a chronic, immune-mediated dermatological condition characterized by dysregulated inflammatory responses and the hyperproliferation of keratinocytes. Biologics, which target specific cytokines such as IL-17 and IL-23, have revolutionized the management by addressing key drivers of its pathophysiology. Despite their efficacy, biologics are not without limitations, including the need for intermittent administration and ongoing monitoring. In contrast, small molecules offer a promising alternative by selectively inhibiting key signaling pathways that modulate pro-inflammatory cytokines involved in the inflammatory cascade. METHODS AND RESULTS This review suggests a new therapeutic strategy for Pso treatment, emphasizing the intricate relationships between small molecules and important signaling pathways involved in the pathophysiology of skin conditions. Improving treatment outcomes and reducing the side effects associated with conventional medicines, this review aims to better understand how tailored small-molecule inhibitors might efficiently control these pathways. This creative approach promotes the creation of individualized treatment plans that can greatly enhance the quality of life of patients with Psoby utilizing the knowledge gathered from recent developments in signaling pathway research. CONCLUSION This review delves into the molecular mechanisms underlying Pso and explores how small molecules can be harnessed to enhance treatment outcomes, presenting a new paradigm for managing this chronic skin disorder.
Collapse
Affiliation(s)
- Meeral Gosia
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Siddhi Bagwe Parab
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Angel Godad
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| |
Collapse
|
3
|
Wang W, Li N, Guo X. The crosstalk between ILC3s and adaptive immunity in diseases. FEBS J 2024; 291:3965-3977. [PMID: 37994218 DOI: 10.1111/febs.17014] [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: 06/23/2023] [Revised: 09/26/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023]
Abstract
RORγt+ group 3 innate lymphoid cells (ILC3s), the innate counterpart of Th17 cells, are enriched in the mucosal area and lymphoid tissues. ILC3s interact with a variety of cells through their effector molecules and play an important role in the host defense against a spectrum of infections. Recent studies suggest that the extensive crosstalk between ILC3s and adaptive immune cells, especially T cells, is essential for maintaining tissue homeostasis. Here we discuss recent advances in the crosstalk between ILC3s and adaptive immune responses in multiple tissues and diseases. Understanding how ILC3s engage with adaptive immune cells will enhance our comprehension of diseases and facilitate the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Wenyan Wang
- Institute for Immunology, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Na Li
- Institute for Immunology, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Xiaohuan Guo
- Institute for Immunology, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| |
Collapse
|
4
|
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
|
5
|
Chen JH, Nieman LT, Spurrell M, Jorgji V, Elmelech L, Richieri P, Xu KH, Madhu R, Parikh M, Zamora I, Mehta A, Nabel CS, Freeman SS, Pirl JD, Lu C, Meador CB, Barth JL, Sakhi M, Tang AL, Sarkizova S, Price C, Fernandez NF, Emanuel G, He J, Van Raay K, Reeves JW, Yizhak K, Hofree M, Shih A, Sade-Feldman M, Boland GM, Pelka K, Aryee MJ, Mino-Kenudson M, Gainor JF, Korsunsky I, Hacohen N. Human lung cancer harbors spatially organized stem-immunity hubs associated with response to immunotherapy. Nat Immunol 2024; 25:644-658. [PMID: 38503922 DOI: 10.1038/s41590-024-01792-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 02/15/2024] [Indexed: 03/21/2024]
Abstract
The organization of immune cells in human tumors is not well understood. Immunogenic tumors harbor spatially localized multicellular 'immunity hubs' defined by expression of the T cell-attracting chemokines CXCL10/CXCL11 and abundant T cells. Here, we examined immunity hubs in human pre-immunotherapy lung cancer specimens and found an association with beneficial response to PD-1 blockade. Critically, we discovered the stem-immunity hub, a subtype of immunity hub strongly associated with favorable PD-1-blockade outcome. This hub is distinct from mature tertiary lymphoid structures and is enriched for stem-like TCF7+PD-1+CD8+ T cells, activated CCR7+LAMP3+ dendritic cells and CCL19+ fibroblasts as well as chemokines that organize these cells. Within the stem-immunity hub, we find preferential interactions between CXCL10+ macrophages and TCF7-CD8+ T cells as well as between mature regulatory dendritic cells and TCF7+CD4+ and regulatory T cells. These results provide a picture of the spatial organization of the human intratumoral immune response and its relevance to patient immunotherapy outcomes.
Collapse
Affiliation(s)
- Jonathan H Chen
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA.
- Department of Pathology, MGH, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Linda T Nieman
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Maxwell Spurrell
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA
- Department of Pathology, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Vjola Jorgji
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA
- Department of Pathology, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Liad Elmelech
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA
- Department of Pathology, MGH, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Peter Richieri
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA
| | - Katherine H Xu
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA
| | - Roopa Madhu
- Harvard Medical School, Boston, MA, USA
- Brigham and Women's Hospital, Division of Genetics, Boston, MA, USA
| | - Milan Parikh
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Izabella Zamora
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Arnav Mehta
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Christopher S Nabel
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Koch Institute for Integrative Cancer Research, Department of Biology, MIT, Cambridge, MA, USA
| | - Samuel S Freeman
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Joshua D Pirl
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Chenyue Lu
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA
| | - Catherine B Meador
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Division of Hematology/Oncology, MGH, HMS, Boston, MA, USA
| | | | | | - Alexander L Tang
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Siranush Sarkizova
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | | | | | | | | | | | | | - Keren Yizhak
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Matan Hofree
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Lautenberg Center for Immunology and Cancer Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Angela Shih
- Department of Pathology, MGH, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Moshe Sade-Feldman
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Genevieve M Boland
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Surgery, MGH, Boston, MA, USA
| | - Karin Pelka
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Gladstone-UCSF Institute of Genomic Immunology, Gladstone Institutes, San Francisco, CA, USA
| | - Martin J Aryee
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mari Mino-Kenudson
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA
- Department of Pathology, MGH, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Justin F Gainor
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Center for Thoracic Cancers, MGH, Boston, MA, USA.
| | - Ilya Korsunsky
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Brigham and Women's Hospital, Division of Genetics, Boston, MA, USA.
| | - Nir Hacohen
- Massachusetts General Hospital (MGH) Cancer Center, Harvard Medical School (HMS), Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Samborska I, Maievskyi O, Podzihun L, Lavrynenko V. Features of immune reactivity of the spleen and mechanisms of organ damage under the influence of animal venom toxins including scorpions (review). WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:120-125. [PMID: 38431816 DOI: 10.36740/wlek202401115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
OBJECTIVE Aim: To establish features of immune reactivity of the spleen and mechanisms of organ damage under the influence of animal venom toxins including scorpions. PATIENTS AND METHODS Materials and Methods: A thorough literature analysis was conducted on the basis of PubMed, Google Scholar, Web of Science, and Scopus databases. When processing the search results, we chose the newest publications up to 5 years old or the most thorough publications that vividly described the essence of our topic. CONCLUSION Conclusions: Spleen plays a leading role in the implementation of the body's defense processes, the elimination of structural elements affected by toxins, and the restoration of immune homeostasis. Its participation in the formation of the immune response can be accompanied by qualitative and quantitative changes in histological organization. Morpho-functional changes in the spleen under the action of animal venom toxins currently require careful study, because from the information available in the literature today, it is not possible to clearly construct a complete picture of lesions of certain components of the organ at the microscopic or submicroscopic levels. Therefore, this direction of research in the medical field is currently relevant, taking into account the existence of a large number of poisonous animals, including scorpions.
Collapse
Affiliation(s)
- Inha Samborska
- NATIONAL PIROGOV MEMORIAL MEDICAL UNIVERSITY, VINNYTSIA, UKRAINE
| | - Oleksandr Maievskyi
- EDUCATIONAL AND SCIENTIFIC CENTER "INSTITUTE OF BIOLOGY AND MEDICINE" OF TARAS SHEVCHENKO NATIONAL UNIVERSITY OF KYIV, KYIV, UKRAINE
| | | | - Victoriia Lavrynenko
- EDUCATIONAL AND SCIENTIFIC CENTER "INSTITUTE OF BIOLOGY AND MEDICINE" OF TARAS SHEVCHENKO NATIONAL UNIVERSITY OF KYIV, KYIV, UKRAINE
| |
Collapse
|
7
|
Wang M, Rajkumar S, Lai Y, Liu X, He J, Ishikawa T, Nallapothula D, Singh RR. Tertiary lymphoid structures as local perpetuators of organ-specific immune injury: implication for lupus nephritis. Front Immunol 2023; 14:1204777. [PMID: 38022566 PMCID: PMC10644380 DOI: 10.3389/fimmu.2023.1204777] [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: 04/12/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
In response to inflammatory stimuli in conditions such as autoimmune disorders, infections and cancers, immune cells organize in nonlymphoid tissues, which resemble secondary lymphoid organs. Such immune cell clusters are called tertiary lymphoid structures (TLS). Here, we describe the potential role of TLS in the pathogenesis of autoimmune disease, focusing on lupus nephritis, a condition that incurs major morbidity and mortality. In the kidneys of patients and animals with lupus nephritis, the presence of immune cell aggregates with similar cell composition, structure, and gene signature as lymph nodes and of lymphoid tissue-inducer and -organizer cells, along with evidence of communication between stromal and immune cells are indicative of the formation of TLS. TLS formation in kidneys affected by lupus may be instigated by local increases in lymphorganogenic chemokines such as CXCL13, and in molecules associated with leukocyte migration and vascularization. Importantly, the presence of TLS in kidneys is associated with severe tubulointerstitial inflammation, higher disease activity and chronicity indices, and poor response to treatment in patients with lupus nephritis. TLS may contribute to the pathogenesis of lupus nephritis by increasing local IFN-I production, facilitating the recruitment and supporting survival of autoreactive B cells, maintaining local production of systemic autoantibodies such as anti-dsDNA and anti-Sm/RNP autoantibodies, and initiating epitope spreading to local autoantigens. Resolution of TLS, along with improvement in lupus, by treating animals with soluble BAFF receptor, docosahexaenoic acid, complement inhibitor C4BP(β-), S1P1 receptor modulator Cenerimod, dexamethasone, and anti-CXCL13 further emphasizes a role of TLS in the pathogenesis of lupus. However, the mechanisms underlying TLS formation and their roles in the pathogenesis of lupus nephritis are not fully comprehended. Furthermore, the lack of non-invasive methods to visualize/quantify TLS in kidneys is also a major hurdle; however, recent success in visualizing TLS in lupus-prone mice by photon emission computed tomography provides hope for early detection and manipulation of TLS.
Collapse
Affiliation(s)
- Meiying Wang
- Department of Rheumatology and Immunology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Peking University Shenzhen Hosiptal, Shenzhen, China
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Snehin Rajkumar
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Yupeng Lai
- Department of Rheumatology and Immunology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xingjiao Liu
- Department of Rheumatology and Immunology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jing He
- Department of Rheumatology and Immunology, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Tatsuya Ishikawa
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Dhiraj Nallapothula
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
| | - Ram Raj Singh
- Autoimmunity and Tolerance Laboratory, Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
- Molecular Toxicology Interdepartmental Program, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| |
Collapse
|
8
|
Guilz NC, Ahn YO, Seo S, Mace EM. Unwinding the Role of the CMG Helicase in Inborn Errors of Immunity. J Clin Immunol 2023; 43:847-861. [PMID: 36809597 PMCID: PMC10789183 DOI: 10.1007/s10875-023-01437-3] [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: 10/31/2022] [Accepted: 01/20/2023] [Indexed: 02/23/2023]
Abstract
Inborn errors of immunity (IEI) are a collection of diseases resulting from genetic causes that impact the immune system through multiple mechanisms. Natural killer cell deficiency (NKD) is one such IEI where natural killer (NK) cells are the main immune lineage affected. Though rare, the deficiency of several genes has been described as underlying causes of NKD, including MCM4, GINS1, MCM10 , and GINS4 , all of which are involved in the eukaryotic CMG helicase. The CMG helicase is made up of C DC45 – M CM – G INS and accessory proteins including MCM10. The CMG helicase plays a critical role in DNA replication by unwinding the double helix and enabling access of polymerases to single-stranded DNA, and thus helicase proteins are active in any proliferating cell. Replication stress, DNA damage, and cell cycle arrest are among the cellular phenotypes attributed to loss of function variants in CMG helicase proteins. Despite the ubiquitous function of the CMG helicase, NK cells have an apparent susceptibility to the deficiency of helicase proteins. This review will examine the role of the CMG helicase in inborn errors of immunity through the lens of NKD and further discuss why natural killer cells can be so strongly affected by helicase deficiency.
Collapse
Affiliation(s)
- Nicole C Guilz
- Vagelos College of Physicians and Surgeons, Department of Pediatrics, Columbia University Irving Medical Center, 630 W 168th St., New York, NY, 10032, USA
| | - Yong-Oon Ahn
- Vagelos College of Physicians and Surgeons, Department of Pediatrics, Columbia University Irving Medical Center, 630 W 168th St., New York, NY, 10032, USA
| | - Seungmae Seo
- Vagelos College of Physicians and Surgeons, Department of Pediatrics, Columbia University Irving Medical Center, 630 W 168th St., New York, NY, 10032, USA
| | - Emily M Mace
- Vagelos College of Physicians and Surgeons, Department of Pediatrics, Columbia University Irving Medical Center, 630 W 168th St., New York, NY, 10032, USA.
| |
Collapse
|
9
|
Zoghi S, Masoumi F, Rezaei N. The immune system. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00005-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
10
|
Salles ÉL, Emami Naeini S, Bhandari B, Khodadadi H, Threlkeld E, Rezaee S, Meeks W, Meeks A, Awe A, El-Marakby A, Yu JC, Wang LP, Baban B. Sexual Dimorphism in the Polarization of Cardiac ILCs through Elabela. Curr Issues Mol Biol 2022; 45:223-232. [PMID: 36661503 PMCID: PMC9856941 DOI: 10.3390/cimb45010017] [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: 09/30/2022] [Revised: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023] Open
Abstract
Elabela is a component of the apelinergic system and may exert a cardioprotective role by regulating the innate immune responses. Innate lymphoid cells (ILCs) have a significant role in initiating and progressing immune-inflammatory responses. While ILCs have been intensively investigated during the last decade, little is known about their relationship with the apelinergic system and their cardiac diversity in a gender-based paradigm. In this study, we investigated the polarization of cardiac ILCs by Elabela in males versus females in a mouse model. Using flow cytometry and immunohistochemistry analyses, we showed a potential interplay between Elabela and cardiac ILCs and whether such interactions depend on sexual dimorphism. Our findings showed, for the first time, that Elabela is expressed by cardiac ILCs, and its expression is higher in females' ILC class 3 (ILC3s) compared to males. Females had higher frequencies of ILC1s, and Elabela was able to suppress T-cell activation and the expression of co-stimulatory CD28 in a mixed lymphocyte reaction assay (MLR). In conclusion, our results suggest, for the first time, a protective role for Elabela through its interplay with ILCs and that it can be used as an immunotherapeutic target in the treatment of cardiovascular disorders in a gender-based fashion.
Collapse
Affiliation(s)
- Évila Lopes Salles
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
- Correspondence: ; Tel.: +1-(706)-721-3181
| | - Sahar Emami Naeini
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Bidhan Bhandari
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Hesam Khodadadi
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Edie Threlkeld
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Sholeh Rezaee
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - William Meeks
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Avery Meeks
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Aderemi Awe
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Ahmed El-Marakby
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Jack C. Yu
- Department of Plastic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Lei P. Wang
- Medicinal Cannabis of Georgia LLC, Augusta, GA 30912, USA
| | - Babak Baban
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
11
|
Nanoadjuvants Actively targeting lymph node conduits and blocking tumor invasion in lymphatic vessels. J Control Release 2022; 352:497-506. [PMID: 36341931 DOI: 10.1016/j.jconrel.2022.10.053] [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: 07/26/2022] [Revised: 10/13/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
Great efforts have been made to manipulate nanoparticles (NPs) with a diameter of 10-100 nm to passively target lymph nodes (LNs) to magnitude anti-tumor activity of T cells. However, no attention has been paid to increasing the retention of NPs with active affinity in order to induce a prolonged release of antigens or molecular adjuvants in the LNs mattering the immune response. Here, we formulated two NPs encapsulated with imiquimod (IMQ), a TLR7/8 agonist, and paclitaxel (PTX) and further modified them with tannic acid (TA), respectively, to generate IMQ NP and PTX NP with a final diameter of approximately 40 nm. Attributing a strong affinity of TA molecules to the elastin of LN conduits, the TA modified IMQ NPs can bypass the gaps in the layer of lymphatic endothelial cells and enter the paracortex through the lymph node capsule-associated (LNC) conduits. Similarly, the TA modified PTX NPs increased delivery of PTX to the metastatic tumor site in LNs, where the tumor-associated antigens were released and presented by conduits-lining dendritic cells to activate T cells. Thus, the NPs with deposition to LN conduits showed excellent performance in preventing lymphovascular invasion of triple-negative breast cancer cells and lung metastasis thereafter. On the contrary, the NPs without TA flowed through the subcutaneous sinus existing LNs directly by efferent lymphatic vessels showing relatively poor therapeutic outcomes. This study reveals that TA may mediate the long retention of antigens and molecular adjuvants to be delivered to deep LNs for developing potent vaccination technology.
Collapse
|
12
|
Schulze TT, Neville AJ, Chapman RC, Davis PH. Mouse splenocyte enrichment strategies via negative selection for broadened single-cell transcriptomics. STAR Protoc 2022; 3:101402. [PMID: 35600930 PMCID: PMC9120244 DOI: 10.1016/j.xpro.2022.101402] [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: 01/18/2023] Open
Abstract
Mammalian splenic tissue is rich in functional immune cells, primarily lymphocytes which can mask low-abundance populations in downstream analyses. This protocol enriches minority immune cell populations from mouse spleen via immunomagnetic negative depletion to generate an untouched enriched cell fraction. Enriched cells are then spiked with untouched splenocytes in a controlled repopulation, validated by flow cytometry and results in a single-cell transcriptomic clustering analysis with a broadened cellular landscape.
Collapse
Affiliation(s)
- Thomas T. Schulze
- Department of Biology, University of Nebraska at Omaha, Omaha, NE 68182, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68106, USA
| | - Andrew J. Neville
- Department of Biology, University of Nebraska at Omaha, Omaha, NE 68182, USA
| | - Ryan C. Chapman
- Department of Biology, University of Nebraska at Omaha, Omaha, NE 68182, USA
| | - Paul H. Davis
- Department of Biology, University of Nebraska at Omaha, Omaha, NE 68182, USA
| |
Collapse
|
13
|
Sadeghi Hassanabadi N, Broux B, Marinović S, Gotthardt D. Innate Lymphoid Cells - Neglected Players in Multiple Sclerosis. Front Immunol 2022; 13:909275. [PMID: 35784374 PMCID: PMC9247827 DOI: 10.3389/fimmu.2022.909275] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/20/2022] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is a highly debilitating autoimmune disease affecting millions of individuals worldwide. Although classically viewed as T-cell mediated disease, the role of innate lymphoid cells (ILC) such as natural killer (NK) cells and ILC 1-3s has become a focal point as several findings implicate them in the disease pathology. The role of ILCs in MS is still not completely understood as controversial findings have been reported assigning them either a protective or disease-accelerating role. Recent findings in experimental autoimmune encephalomyelitis (EAE) suggest that ILCs infiltrate the central nervous system (CNS), mediate inflammation, and have a disease exacerbating role by influencing the recruitment of autoreactive T-cells. Elucidating the detailed role of ILCs and altered signaling pathways in MS is essential for a more complete picture of the disease pathology and novel therapeutic targets. We here review the current knowledge about ILCs in the development and progression of MS and preclinical models of MS and discuss their potential for therapeutic applications.
Collapse
Affiliation(s)
| | - Bieke Broux
- University MSCenter; Campus Diepenbeek, Diepenbeek, Belgium
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, UHasselt, Diepenbeek, Belgium
| | - Sonja Marinović
- Division of Molecular Medicine, Laboratory of Personalized Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Dagmar Gotthardt
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
- *Correspondence: Dagmar Gotthardt,
| |
Collapse
|
14
|
Chung DC, Jacquelot N, Ghaedi M, Warner K, Ohashi PS. Innate Lymphoid Cells: Role in Immune Regulation and Cancer. Cancers (Basel) 2022; 14:2071. [PMID: 35565201 PMCID: PMC9102917 DOI: 10.3390/cancers14092071] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023] Open
Abstract
Immune regulation is composed of a complex network of cellular and molecular pathways that regulate the immune system and prevent tissue damage. It is increasingly clear that innate lymphoid cells (ILCs) are also armed with immunosuppressive capacities similar to well-known immune regulatory cells (i.e., regulatory T cells). In cancer, immunoregulatory ILCs have been shown to inhibit anti-tumour immune response through various mechanisms including: (a) direct suppression of anti-tumour T cells or NK cells, (b) inhibiting T-cell priming, and (c) promoting other immunoregulatory cells. To provide a framework of understanding the role of immunosuppressive ILCs in the context of cancer, we first outline a brief history and challenges related to defining immunosuppressive ILCs. Furthermore, we focus on the mechanisms of ILCs in suppressing anti-tumour immunity and consequentially promoting tumour progression.
Collapse
Affiliation(s)
- Douglas C. Chung
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| | - Nicolas Jacquelot
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| | - Maryam Ghaedi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| | - Kathrin Warner
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| | - Pamela S. Ohashi
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| |
Collapse
|
15
|
Zhang L, Lin Q, Jiang L, Wu M, Huang L, Quan W, Li X. Increased circulating innate lymphoid cell (ILC)1 and decreased circulating ILC3 are involved in the pathogenesis of Henoch-Schonlein purpura. BMC Pediatr 2022; 22:201. [PMID: 35413831 PMCID: PMC9003988 DOI: 10.1186/s12887-022-03262-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 04/01/2022] [Indexed: 11/30/2022] Open
Abstract
Background Innate lymphoid cell (ILC) dysfunction is involved in numerous immune diseases, but this has not been demonstrated in Henoch-Schonlein purpura (HSP). This study aimed to investigate whether ILC dysfunction or imbalance participate in the pathogenesis of HSP. Methods This was a prospective study in patients with HSP who were hospitalized at the Children’s Hospital of Soochow University from June to December 2019. Age- and sex-matched controls were also enrolled. ILC subsets and lymphocyte subpopulations were determined by flow cytometry. The transmission immune turbidimetric method also facilitated the exploration of correlations between ILC subset frequency and lymphocyte subpopulation, as well as serum IgA in HSP patients. Results Fifty-one patients with HSP and 22 control patients were included. There were no differences in age and sex between the two groups. Compared with controls, patients with HSP had higher ILCs in relation to lymphocytes (P = 0.036), higher ILCs in relation to PBMCs (P = 0.026), higher ILC1s (P < 0.001), lower ILC3s (P < 0.05), and higher ILC1/ILC3 ratio (P < 0.001). Sixteen patients underwent routine therapy combined with methylprednisolone for 7–10 days; ILC1s were significantly decreased (P < 0.001) and ILC3s were increased (P = 0.033), and ILC1/ILC3 was significantly decreased (P < 0.001). Compared with the controls, the ratios of ILCs/lymphocytes and ILCs/PBMC were higher in patients in the arthritis and mixed groups (all P < 0.05). ILC1 were elevated in the purpura, arthritis, abdominal, and mixed groups (P = 0.027, P = 0.007, P < 0.001, and P < 0.001, respectively). ILC1/ILCs were positively correlated with CD3 + CD8 + T lymphocytes (r = 0.3701, P = 0.0075). The level of IgA did not correlate with ILCs. Conclusions Higher circulating ILC1s and lower circulating ILC3s appear to be involved in the pathogenesis of HSP. Supplementary Information The online version contains supplementary material available at 10.1186/s12887-022-03262-w.
Collapse
Affiliation(s)
- Lili Zhang
- Department of Nephrology, Children's Hospital of Soochow University, No. 92, Zhong Nan Street, Industrial Park, Suzhou, 215003, Jiangsu, China.,Department of Pediatrics, Affiliated Hospital of Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Qiang Lin
- Department of Nephrology, Children's Hospital of Soochow University, No. 92, Zhong Nan Street, Industrial Park, Suzhou, 215003, Jiangsu, China
| | - Lijun Jiang
- Department of Neonatology, Affiliated Hospital of Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Mingfu Wu
- Department of Neonatology, Affiliated Hospital of Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Linlin Huang
- Department of Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Suzhou, 215003, Jiangsu, China
| | - Wei Quan
- Department of Nephrology, Children's Hospital of Soochow University, No. 92, Zhong Nan Street, Industrial Park, Suzhou, 215003, Jiangsu, China
| | - Xiaozhong Li
- Department of Nephrology, Children's Hospital of Soochow University, No. 92, Zhong Nan Street, Industrial Park, Suzhou, 215003, Jiangsu, China.
| |
Collapse
|
16
|
Overview: Themes in Innate Lymphoid Cell Biology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1365:1-6. [DOI: 10.1007/978-981-16-8387-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
17
|
Korchagina AA, Koroleva E, Tumanov AV. Innate Lymphoid Cells in Response to Intracellular Pathogens: Protection Versus Immunopathology. Front Cell Infect Microbiol 2021; 11:775554. [PMID: 34938670 PMCID: PMC8685334 DOI: 10.3389/fcimb.2021.775554] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/03/2021] [Indexed: 12/23/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a heterogeneous group of cytokine-producing lymphocytes which are predominantly located at mucosal barrier surfaces, such as skin, lungs, and gastrointestinal tract. ILCs contribute to tissue homeostasis, regulate microbiota-derived signals, and protect against mucosal pathogens. ILCs are classified into five major groups by their developmental origin and distinct cytokine production. A recently emerged intriguing feature of ILCs is their ability to alter their phenotype and function in response to changing local environmental cues such as pathogen invasion. Once the pathogen crosses host barriers, ILCs quickly activate cytokine production to limit the spread of the pathogen. However, the dysregulated ILC responses can lead to tissue inflammation and damage. Furthermore, the interplay between ILCs and other immune cell types shapes the outcome of the immune response. Recent studies highlighted the important role of ILCs for host defense against intracellular pathogens. Here, we review recent advances in understanding the mechanisms controlling protective and pathogenic ILC responses to intracellular pathogens. This knowledge can help develop new ILC-targeted strategies to control infectious diseases and immunopathology.
Collapse
Affiliation(s)
- Anna A Korchagina
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Ekaterina Koroleva
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Alexei V Tumanov
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
18
|
Lymph node formation and B cell homeostasis require IKK-α in distinct endothelial cell-derived compartments. Proc Natl Acad Sci U S A 2021; 118:2100195118. [PMID: 34810256 DOI: 10.1073/pnas.2100195118] [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] [Accepted: 10/14/2021] [Indexed: 11/18/2022] Open
Abstract
Global inactivation of IκB kinase (IKK)-α results in defective lymph node (LN) formation and B cell maturation, and loss of IKK-α-dependent noncanonical NF-κB signaling in stromal organizer and hematopoietic cells is thought to underlie these distinct defects. We previously demonstrated that this pathway is also activated in vascular endothelial cells (ECs). To determine the physiologic function of EC-intrinsic IKK-α, we crossed IkkαF/F mice with Tie2-cre or Cdh5-cre mice to ablate IKK-α in ECs. Notably, the compound defects of global IKK-α inactivation were recapitulated in IkkαTie2 and IkkαCdh5 mice, as both lacked all LNs and mature follicular and marginal zone B cell numbers were markedly reduced. However, as Tie2-cre and Cdh5-cre are expressed in all ECs, including blood forming hemogenic ECs, IKK-α was also absent in hematopoietic cells (HC). To determine if loss of HC-intrinsic IKK-α affected LN development, we generated IkkαVav mice lacking IKK-α in only the hematopoietic compartment. While mature B cell numbers were significantly reduced in IkkαVav mice, LN formation was intact. As lymphatic vessels also arise during development from blood ECs, we generated IkkαLyve1 mice lacking IKK-α in lymphatic ECs (LECs) to determine if IKK-α in lymphatic vessels impacts LN development. Strikingly, while mature B cell numbers were normal, LNs were completely absent in IkkαLyve1 mice. Thus, our findings reveal that IKK-α in distinct EC-derived compartments is uniquely required to promote B cell homeostasis and LN development, and we establish that LEC-intrinsic IKK-α is absolutely essential for LN formation.
Collapse
|
19
|
Yeung SSH, Ho YS, Chang RCC. The role of meningeal populations of type II innate lymphoid cells in modulating neuroinflammation in neurodegenerative diseases. Exp Mol Med 2021; 53:1251-1267. [PMID: 34489558 PMCID: PMC8492689 DOI: 10.1038/s12276-021-00660-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 02/08/2023] Open
Abstract
Recent research into meningeal lymphatics has revealed a never-before appreciated role of type II innate lymphoid cells (ILC2s) in modulating neuroinflammation in the central nervous system (CNS). To date, the role of ILC2-mediated inflammation in the periphery has been well studied. However, the exact distribution of ILC2s in the CNS and therefore their putative role in modulating neuroinflammation in neurodegenerative diseases such as Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease (PD), and major depressive disorder (MDD) remain highly elusive. Here, we review the current evidence of ILC2-mediated modulation of neuroinflammatory cues (i.e., IL-33, IL-25, IL-5, IL-13, IL-10, TNFα, and CXCL16-CXCR6) within the CNS, highlight the distribution of ILC2s in both the periphery and CNS, and discuss some challenges associated with cell type-specific targeting that are important for therapeutics. A comprehensive understanding of the roles of ILC2s in mediating and responding to inflammatory cues may provide valuable insight into potential therapeutic strategies for many dementia-related disorders.
Collapse
Affiliation(s)
- Sherry Sin-Hang Yeung
- grid.194645.b0000000121742757Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Yuen-Shan Ho
- grid.16890.360000 0004 1764 6123School of Nursing, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR China
| | - Raymond Chuen-Chung Chang
- grid.194645.b0000000121742757Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China ,grid.194645.b0000000121742757State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| |
Collapse
|
20
|
Alternative pathways for the development of lymphoid structures in humans. Proc Natl Acad Sci U S A 2021; 118:2108082118. [PMID: 34261794 DOI: 10.1073/pnas.2108082118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Lymphoid tissue inducer (LTi) cells are critical for inducing the differentiation of most secondary lymphoid organs (SLOs) in mice. In humans, JAK3 and γc deficiencies result in severe combined immunodeficiency (SCIDs) characterized by an absence of T cells, natural killer cells, innate lymphoid cells (ILCs), and presumably LTi cells. Some of these patients have undergone allogeneic stem cell transplantation (HSCT) in the absence of myeloablation, which leads to donor T cell engraftment, while other leukocyte subsets are of host origin. By using MRI to look for SLOs in nine of these patients 16 to 44 y after HSCT, we discovered that SLOs were exclusively found in the three areas of the abdomen that drain the intestinal tract. A postmortem examination of a child with γc-SCID who had died 3.5 mo after HSCT showed corticomedullary differentiation in the thymus, T cell zones in the spleen, and the appendix, but in neither lymph nodes nor Peyer patches. Tertiary lymphoid organs were observed in the lung. No RAR-related orphan receptor-positive LTi cells could be detected in the existing lymphoid structures. These results suggest that while LTi cells are required for the genesis of most SLOs in humans, SLO in the appendix and in gut-draining areas, as well as tertiary lymphoid organs, can be generated likely by LTi cell-independent mechanisms.
Collapse
|
21
|
Kumar V. Innate Lymphoid Cells and Adaptive Immune Cells Cross-Talk: A Secret Talk Revealed in Immune Homeostasis and Different Inflammatory Conditions. Int Rev Immunol 2021; 40:217-251. [PMID: 33733998 DOI: 10.1080/08830185.2021.1895145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The inflammatory immune response has evolved to protect the host from different pathogens, allergens, and endogenous death or damage-associated molecular patterns. Both innate and adaptive immune components are crucial in inducing an inflammatory immune response depending on the stimulus type and its duration of exposure or the activation of the primary innate immune response. As the source of inflammation is removed, the aggravated immune response comes to its homeostatic level. However, the failure of the inflammatory immune response to subside to its normal level generates chronic inflammatory conditions, including autoimmune diseases and cancer. Innate lymphoid cells (ILCs) are newly discovered innate immune cells, which are present in abundance at mucosal surfaces, including lungs, gastrointestinal tract, and reproductive tract. Also, they are present in peripheral blood circulation, skin, and lymph nodes. They play a crucial role in generating the pro-inflammatory immune response during diverse conditions. On the other hand, adaptive immune cells, including different types of T and B cells are major players in the pathogenesis of autoimmune diseases (type 1 diabetes mellitus, rheumatoid arthritis, psoriasis, and systemic lupus erythematosus, etc.) and cancers. Thus the article is designed to discuss the immunological role of different ILCs and their interaction with adaptive immune cells in maintaining the immune homeostasis, and during inflammatory autoimmune diseases along with other inflammatory conditions (excluding pathogen-induced inflammation), including cancer, graft-versus-host diseases, and human pregnancy.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, St Lucia, Brisbane, Queensland, Australia.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, St Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
22
|
Humanization of Immunodeficient Animals for the Modeling of Transplantation, Graft Versus Host Disease, and Regenerative Medicine. Transplantation 2021; 104:2290-2306. [PMID: 32068660 PMCID: PMC7590965 DOI: 10.1097/tp.0000000000003177] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The humanization of animals is a powerful tool for the exploration of human disease pathogenesis in biomedical research, as well as for the development of therapeutic interventions with enhanced translational potential. Humanized models enable us to overcome biologic differences that exist between humans and other species, while giving us a platform to study human processes in vivo. To become humanized, an immune-deficient recipient is engrafted with cells, tissues, or organoids. The mouse is the most well studied of these hosts, with a variety of immunodeficient strains available for various specific uses. More recently, efforts have turned to the humanization of other animal species such as the rat, which offers some technical and immunologic advantages over mice. These advances, together with ongoing developments in the incorporation of human transgenes and additional mutations in humanized mouse models, have expanded our opportunities to replicate aspects of human allotransplantation and to assist in the development of immunotherapies. In this review, the immune and tissue humanization of various species is presented with an emphasis on their potential for use as models for allotransplantation, graft versus host disease, and regenerative medicine.
Collapse
|
23
|
Azeem M, Kader H, Kerstan A, Hetta HF, Serfling E, Goebeler M, Muhammad K. Intricate Relationship Between Adaptive and Innate Immune System in Allergic Contact Dermatitis. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2020; 93:699-709. [PMID: 33380932 PMCID: PMC7757059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Allergic contact dermatitis (ACD) is a complex immunological allergic disease characterized by the interplay between the innate and adaptive immune system. Initially, the role of the innate immune system was believed to be confined to the initial sensitization phase, while adaptive immune reactions were linked with the advanced elicitation phase. However, recent data predicted a comparatively mixed and interdependent role of both immune systems throughout the disease progression. Therefore, the actual mechanisms of disease progression are more complex and interlinked. The aim of this review is to combine such findings that enhanced our understanding of the pathomechanisms of ACD. Here, we focused on the main cell types from both immune domains, which are involved in ACD, such as CD4+ and CD8+ T cells, B cells, neutrophils, and innate lymphoid cells (ILCs). Such insights can be useful for devising future therapeutic interventions for ACD.
Collapse
Affiliation(s)
- Muhammad Azeem
- Department of Molecular Pathology, Institute of
Pathology, University of Würzburg, Würzburg, Germany
| | - Hidaya Kader
- Department of Biology, College of Science, United Arab
Emirates University, Al Ain, United Arab Emirates
| | - Andreas Kerstan
- Department of Dermatology, Venereology and Allergology,
University Hospital Würzburg, Würzburg, Germany
| | - Helal F. Hetta
- Department of Medical Microbiology and Immunology,
Faculty of Medicine, Assiut University, Assiut, Egypt
- Department of Internal Medicine, University of
Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Edgar Serfling
- Department of Molecular Pathology, Institute of
Pathology, University of Würzburg, Würzburg, Germany
| | - Matthias Goebeler
- Department of Dermatology, Venereology and Allergology,
University Hospital Würzburg, Würzburg, Germany
| | - Khalid Muhammad
- Department of Biology, College of Science, United Arab
Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
24
|
Krabbendam L, Bernink JH, Spits H. Innate lymphoid cells: from helper to killer. Curr Opin Immunol 2020; 68:28-33. [PMID: 32971468 DOI: 10.1016/j.coi.2020.08.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/20/2020] [Accepted: 08/23/2020] [Indexed: 12/28/2022]
Abstract
Five subsets of ILCs are extensively described, Lymphoid Tissue inducer (LTi) cells, cytotoxic NK cells and non-cytotoxic helper ILC1s, ILC2s and ILC3s. So far, the main focus has been on the potent cytokine production by helper ILCs and their plastic nature that allows them to switch function and phenotype upon environmental changes. Recent advances in the field indicate the presence of cytotoxic helper ILCs that are distinct from conventional NK cells. In humans, these cytotoxic ILCs can develop from conventional helper ILCs whereas in mice this remains to be elucidated. In this review we discuss the identification, development and function of cytotoxic helper ILCs subsets in humans and mice.
Collapse
Affiliation(s)
- Lisette Krabbendam
- Amsterdam UMC, University of Amsterdam, Department of Experimental Immunology, Amsterdam Infection & Immunity Institute (AI&II), Cancer Center Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Jochem H Bernink
- Amsterdam UMC, University of Amsterdam, Department of Experimental Immunology, Amsterdam Infection & Immunity Institute (AI&II), Cancer Center Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Hergen Spits
- Amsterdam UMC, University of Amsterdam, Department of Experimental Immunology, Amsterdam Infection & Immunity Institute (AI&II), Cancer Center Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.
| |
Collapse
|
25
|
Singh A, Kazer SW, Roider J, Krista KC, Millar J, Asowata OE, Ngoepe A, Ramsuran D, Fardoos R, Ardain A, Muenchhoff M, Kuhn W, Karim F, Ndung'u T, Shalek AK, Goulder P, Leslie A, Kløverpris HN. Innate Lymphoid Cell Activation and Sustained Depletion in Blood and Tissue of Children Infected with HIV from Birth Despite Antiretroviral Therapy. Cell Rep 2020; 32:108153. [PMID: 32937142 PMCID: PMC7495043 DOI: 10.1016/j.celrep.2020.108153] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 03/14/2020] [Accepted: 08/25/2020] [Indexed: 12/04/2022] Open
Abstract
Innate lymphoid cells (ILCs) are important for response to infection and for immune development in early life. HIV infection in adults depletes circulating ILCs, but the impact on children infected from birth remains unknown. We study vertically HIV-infected children from birth to adulthood and find severe and persistent depletion of all circulating ILCs that, unlike CD4+ T cells, are not restored by long-term antiretroviral therapy unless initiated at birth. Remaining ILCs upregulate genes associated with cellular activation and metabolic perturbation. Unlike HIV-infected adults, ILCs are also profoundly depleted in tonsils of vertically infected children. Transcriptional profiling of remaining ILCs reveals ongoing cell-type-specific activity despite antiretroviral therapy. Collectively, these data suggest an important and ongoing role for ILCs in lymphoid tissue of HIV-infected children from birth, where persistent depletion and sustained transcriptional activity are likely to have long-term immune consequences that merit further investigation.
Collapse
Affiliation(s)
- Alveera Singh
- Africa Health Research Institute (AHRI), Durban 4001, South Africa
| | - Samuel W Kazer
- Institute for Medical Engineering and Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139
| | - Julia Roider
- Africa Health Research Institute (AHRI), Durban 4001, South Africa; Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Rd, Oxford OX1 3SY, UK; HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban 4001, South Africa; Medizinische Klinik IV, Department of Infectious Diseases, Ludwig-Maximilians-University, Munich 80802, Germany
| | - Kami C Krista
- Institute for Medical Engineering and Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139
| | - Jane Millar
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Rd, Oxford OX1 3SY, UK; HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban 4001, South Africa
| | | | - Abigail Ngoepe
- Africa Health Research Institute (AHRI), Durban 4001, South Africa
| | - Duran Ramsuran
- Africa Health Research Institute (AHRI), Durban 4001, South Africa
| | - Rabiah Fardoos
- Africa Health Research Institute (AHRI), Durban 4001, South Africa; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen 2200N, Denmark
| | - Amanda Ardain
- Africa Health Research Institute (AHRI), Durban 4001, South Africa; School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Maximilian Muenchhoff
- Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Rd, Oxford OX1 3SY, UK; Max von Pettenkofer Institute, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Munich 81377, Germany; German Center for Infection Research (DZIF), partner site Munich 80333, Germany
| | - Warren Kuhn
- ENT Department General Justice Gizenga Mpanza Regional Hospital (Stanger Hospital), University of KwaZulu-Natal, Durban 4001, South Africa
| | - Farina Karim
- Africa Health Research Institute (AHRI), Durban 4001, South Africa
| | - Thumbi Ndung'u
- Africa Health Research Institute (AHRI), Durban 4001, South Africa; HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban 4001, South Africa; University College London, Division of Infection and Immunity, London WC1E 6AE, UK; Max Planck Institute for Infection Biology, Berlin 10117, Germany
| | - Alex K Shalek
- Institute for Medical Engineering and Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139
| | - Philip Goulder
- Africa Health Research Institute (AHRI), Durban 4001, South Africa; Department of Paediatrics, Peter Medawar Building for Pathogen Research, South Parks Rd, Oxford OX1 3SY, UK; HIV Pathogenesis Programme, The Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Alasdair Leslie
- Africa Health Research Institute (AHRI), Durban 4001, South Africa; University College London, Division of Infection and Immunity, London WC1E 6AE, UK; School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Henrik N Kløverpris
- Africa Health Research Institute (AHRI), Durban 4001, South Africa; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen 2200N, Denmark; University College London, Division of Infection and Immunity, London WC1E 6AE, UK; School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4001, South Africa.
| |
Collapse
|
26
|
Kumar V. Innate lymphoid cell and adaptive immune cell cross-talk: A talk meant not to forget. J Leukoc Biol 2020; 108:397-417. [PMID: 32557732 DOI: 10.1002/jlb.4mir0420-500rrr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 04/28/2020] [Accepted: 05/04/2020] [Indexed: 12/17/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a relatively new class of innate immune cells with phenotypical characters of lymphocytes but genotypically or functionally behave as typical innate immune cells. They have been classically divided into 3 groups (group 1 ILCs or ILC1s, group 2 ILCs or ILC2s, and group 3 ILCs or ILC3s). They serve as the first line of defense against invading pathogens and allergens at mucosal surfaces. The adaptive immune response works effectively in association with innate immunity as innate immune cells serve as APCs to directly stimulate the adaptive immune cells (various sets of T and B cells). Additionally, innate immune cells also secrete various effector molecules, including cytokines or chemokines impacting the function, differentiation, proliferation, and reprogramming among adaptive immune cells to maintain immune homeostasis. Only superantigens do not require their processing by innate immune cells as they are recognized directly by T cells and B cells. Thus, a major emphasis of the current article is to describe the cross-talk between different ILCs and adaptive immune cells during different conditions varying from normal physiological situations to different infectious diseases to allergic asthma.
Collapse
Affiliation(s)
- V Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, Brisbane, Queensland, Australia.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
27
|
Fu K, March K, Alexaki A, Fabozzi G, Moysi E, Petrovas C. Immunogenicity of Protein Therapeutics: A Lymph Node Perspective. Front Immunol 2020; 11:791. [PMID: 32477334 PMCID: PMC7240201 DOI: 10.3389/fimmu.2020.00791] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/07/2020] [Indexed: 12/31/2022] Open
Abstract
The continuous development of molecular biology and protein engineering technologies enables the expansion of the breadth and complexity of protein therapeutics for in vivo administration. However, the immunogenicity and associated in vivo development of antibodies against therapeutics are a major restriction factor for their usage. The B cell follicular and particularly germinal center areas in secondary lymphoid organs are the anatomical sites where the development of antibody responses against pathogens and immunogens takes place. A growing body of data has revealed the importance of the orchestrated function of highly differentiated adaptive immunity cells, including follicular helper CD4 T cells and germinal center B cells, for the optimal generation of these antibody responses. Understanding the cellular and molecular mechanisms mediating the antibody responses against therapeutics could lead to novel strategies to reduce their immunogenicity and increase their efficacy.
Collapse
Affiliation(s)
- Kristy Fu
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Kylie March
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Aikaterini Alexaki
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Giulia Fabozzi
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Eirini Moysi
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Constantinos Petrovas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
28
|
de Melo CVB, Hermida MDR, Mesquita BR, Fontes JLM, Koning JJ, Solcà MDS, Benevides BB, Mota GBS, Freitas LAR, Mebius RE, Dos-Santos WLC. Phenotypical Characterization of Spleen Remodeling in Murine Experimental Visceral Leishmaniasis. Front Immunol 2020; 11:653. [PMID: 32351510 PMCID: PMC7174685 DOI: 10.3389/fimmu.2020.00653] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/23/2020] [Indexed: 01/01/2023] Open
Abstract
Background: Visceral leishmaniasis (VL) is caused by Leishmania infantum or L. donovani infection. One of the main problems related to this disease is the emergence of severe clinical forms with a lethality of 5–20%, even while under specific treatment. In humans and other species susceptible to fatal VL, such as dogs and hamsters, the disruption of splenic white pulp (WP) is accompanied by disease progression. Control of VL progression is seen in BALB/c mice, as evidenced by a mild clinical presentation and controlled parasite replication in the liver and spleen. In this study, we investigated the features involved in the morphological remodeling of splenic compartments associated with the control of VL progression to death. Methods: We evaluated cohorts of BALB/c mice after 30, 60, and 90 days of infection by L. infantum. Spleen morphology, cell population subsets and cytokine production were studied in the spleen using flow- and histo-cytometry. Results: Intraperitoneal infection with 108 promastigotes of L. infantum led to progressive increases in spleen size at 60 and 90 days after infection. Splenomegaly was the only clinical sign of disease observed. At 30 days after infection, hyperplasia in the WP and decreased numbers of plasmacytoid dendritic cells were observed. The WP hyperplasia subsided at 60 days post-infection. However, the splenomegaly remained in association with increased numbers of macrophages, B and T lymphocytes and plasma cells. An increased number of lymphoid tissue inducer (LTi) cells was observed; these were distributed around the periarteriolar lymphoid sheath in control mice and scattered throughout the red pulp in the Leishmania-infected mice. After 90 days of infection, increased IL-6 and IFN-γ production was seen in the spleen, as well as higher frequencies of follicular and plasmacytoid dendritic cells. Conclusion: The data presented herein emphasizes the potential role of spleen remodeling in the control of severe forms of VL and highlights features potentially involved in this process.
Collapse
Affiliation(s)
- Caroline Vilas Boas de Melo
- Laboratório de Patologia Estrutural e Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Micely D'El-Rei Hermida
- Laboratório de Patologia Estrutural e Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Bianca R Mesquita
- Laboratório de Patologia Estrutural e Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Jonathan L M Fontes
- Laboratório de Patologia Estrutural e Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Jasper J Koning
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, VU University Medical Center, Amsterdam, Netherlands
| | - Manuela da Silva Solcà
- Laboratório de Patologia Estrutural e Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Bruno B Benevides
- Laboratório de Patologia Estrutural e Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Girlândia B S Mota
- Laboratório de Patologia Estrutural e Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Luiz A R Freitas
- Laboratório de Patologia Estrutural e Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, VU University Medical Center, Amsterdam, Netherlands
| | - Washington L C Dos-Santos
- Laboratório de Patologia Estrutural e Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| |
Collapse
|
29
|
Lewis SM, Williams A, Eisenbarth SC. Structure and function of the immune system in the spleen. Sci Immunol 2020; 4:4/33/eaau6085. [PMID: 30824527 DOI: 10.1126/sciimmunol.aau6085] [Citation(s) in RCA: 663] [Impact Index Per Article: 132.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/31/2019] [Indexed: 12/11/2022]
Abstract
The spleen is the largest secondary lymphoid organ in the body and, as such, hosts a wide range of immunologic functions alongside its roles in hematopoiesis and red blood cell clearance. The physical organization of the spleen allows it to filter blood of pathogens and abnormal cells and facilitate low-probability interactions between antigen-presenting cells (APCs) and cognate lymphocytes. APCs specific to the spleen regulate the T and B cell response to these antigenic targets in the blood. This review will focus on cell types, cell organization, and immunologic functions specific to the spleen and how these affect initiation of adaptive immunity to systemic blood-borne antigens. Potential differences in structure and function between mouse and human spleen will also be discussed.
Collapse
Affiliation(s)
- Steven M Lewis
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Adam Williams
- Jackson Laboratory for Genomic Medicine, University of Connecticut Health Center, Farmington, CT 06032, USA.,Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA. .,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
30
|
Molecular characteristics and possible functions of innate lymphoid cells in the uterus and gut. Cytokine Growth Factor Rev 2020; 52:15-24. [DOI: 10.1016/j.cytogfr.2019.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 12/23/2022]
|
31
|
Rivellese F, Pontarini E, Pitzalis C. Tertiary Lymphoid Organs in Rheumatoid Arthritis. Curr Top Microbiol Immunol 2020; 426:119-141. [PMID: 32483659 DOI: 10.1007/82_2020_216] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Rheumatoid Arthritis (RA) is a chronic systemic autoimmune disease. RA mainly affects the joints, with inflammation of the synovial membrane, characterized by hyperplasia, neo-angiogenesis, and immune cell infiltration that drives local inflammation and, if untreated, can lead to joint destruction and disability. In parallel to the well-known clinical heterogeneity, the underlying synovitis can also be significantly heterogeneous. In particular, in about 40% of patients with RA, synovitis is characterized by a dense lymphocytic infiltrate that can acquire the features of fully functional tertiary lymphoid organs (TLO). These structures amplify autoimmunity and inflammation locally associated with worse prognosis and potential implications for treatment response. Here, we will review the current knowledge on TLO in RA, with a focus on their pathogenetic and clinical relevance.
Collapse
Affiliation(s)
- Felice Rivellese
- Barts and the London School of Medicine & Dentistry, Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, John Vane Science Centre, London, UK
| | - Elena Pontarini
- Barts and the London School of Medicine & Dentistry, Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, John Vane Science Centre, London, UK
| | - Costantino Pitzalis
- Barts and the London School of Medicine & Dentistry, Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, John Vane Science Centre, London, UK.
| |
Collapse
|
32
|
Chen L, Strohmeier V, He Z, Deshpande M, Catalan-Dibene J, Durum SK, Moran TM, Kraus T, Xiong H, Faith JJ, Sodhi CP, Hackam DJ, Lira SA, Furtado GC. Interleukin 22 disrupts pancreatic function in newborn mice expressing IL-23. Nat Commun 2019; 10:4517. [PMID: 31586069 PMCID: PMC6778080 DOI: 10.1038/s41467-019-12540-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 09/17/2019] [Indexed: 12/19/2022] Open
Abstract
Neonatal inflammatory diseases are associated with severe morbidity, but the inflammatory factors underlying them and their potential effector mechanisms are poorly defined. Here we show that necrotizing enterocolitis in neonate mice is accompanied by elevation of IL-23 and IL-22 and decreased production of pancreatic enzymes. These phenotypes are mirrored in neonate mice overexpressing IL-23 in CX3CR1+ myeloid cells or in keratinocytes. The mice fail to grow and die prematurely, displaying systemic inflammation, nutrient malabsorption and decreased expression of intestinal and pancreatic genes mediating digestion and absorption of carbohydrates, proteins, and lipids. Germ-free environment improves, and genetic ablation of IL-22 restores normal growth in mice overexpressing IL-23. Mechanistically, IL-22 acts directly at the level of pancreatic acinar cells to decrease expression of the pancreas associated transcription factor 1a (PTF1a). These results show that augmented production of IL-23 and IL-22 in early life has a negative impact on pancreatic enzyme secretion and food absorption.
Collapse
Affiliation(s)
- Lili Chen
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Valentina Strohmeier
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, 79104, Freiburg, Germany
| | - Zhengxiang He
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Madhura Deshpande
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jovani Catalan-Dibene
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Scott K Durum
- Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Thomas M Moran
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Therapeutic Antibody Development, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Thomas Kraus
- Center for Therapeutic Antibody Development, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Huabao Xiong
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jeremiah J Faith
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Chhinder P Sodhi
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD, 21287, USA
| | - David J Hackam
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD, 21287, USA
| | - Sergio A Lira
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Glaucia C Furtado
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
33
|
Maoz A, Dennis M, Greenson JK. The Crohn's-Like Lymphoid Reaction to Colorectal Cancer-Tertiary Lymphoid Structures With Immunologic and Potentially Therapeutic Relevance in Colorectal Cancer. Front Immunol 2019; 10:1884. [PMID: 31507584 PMCID: PMC6714555 DOI: 10.3389/fimmu.2019.01884] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 07/25/2019] [Indexed: 12/17/2022] Open
Abstract
The Crohn's-like lymphoid reaction (CLR) to colorectal cancer (CRC), a CRC-specific ectopic lymphoid reaction, is thought to play an important role in the host response to CRC. CLR is characterized by peritumoral lymphocytic aggregates that are found at the advancing edge of the tumor. Spatial and molecular characterization of CLR within the tumor microenvironment (TME) have uncovered a spectrum of peritumoral lymphoid aggregates with varying levels of organization and maturation. In early stages of CLR development, CD4+ T-cells cluster predominantly with mature antigen presenting dendritic cells. As CLR matures, increasing numbers of B-cells, as well as follicular dendritic cells are recruited to create lymphoid follicles. When highly organized, CLR resembles functional tertiary lymphoid structures (TLS), allowing for lymphocyte recruitment to the TME and promoting a tumor-specific adaptive immune response. CLR has been consistently associated with favorable prognostic factors and improved survival among CRC patients, often providing more prognostic information than current clinical staging systems. However, consensus is lacking regarding CLR scoring and it is not clinically assessed or reported. Differences between CLR and other cancer-associated lymphoid structures exist both in primary and metastatic disease, underscoring the need to characterize organ-specific TLS. Further research is needed to explore the role of CLR in predicting response to immunotherapy and to leverage CLR to promote immunotherapeutic strategies in CRC.
Collapse
Affiliation(s)
- Asaf Maoz
- Boston University School of Medicine and Boston Medical Center, Boston, MA, United States
| | - Michael Dennis
- Boston University School of Medicine and Boston Medical Center, Boston, MA, United States
| | - Joel K Greenson
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
34
|
Almeida FF, Jacquelot N, Belz GT. Deconstructing deployment of the innate immune lymphocyte army for barrier homeostasis and protection. Immunol Rev 2019; 286:6-22. [PMID: 30294966 PMCID: PMC6446816 DOI: 10.1111/imr.12709] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/16/2018] [Indexed: 12/30/2022]
Abstract
The study of the immune system has shifted from a purely dichotomous separation between the innate and adaptive arms to one that is now highly complex and reshaping our ideas of how steady‐state health is assured. It is now clear that immune cells do not neatly fit into these two streams and immune homeostasis depends on continual dialogue between multiple lineages of the innate (including dendritic cells, innate lymphoid cells, and unconventional lymphocytes) and adaptive (T and B lymphocytes) arms together with a finely tuned synergy between the host and microbes which is essential to ensure immune homeostasis. Innate lymphoid cells are critical players in this new landscape. Here, we discuss recent studies that have elucidated in detail the development of ILCs from their earliest progenitors and examine factors that influence their identification and ability to drive immune homeostasis and long‐term immune protection.
Collapse
Affiliation(s)
- Francisca F Almeida
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Nicolas Jacquelot
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Gabrielle T Belz
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
35
|
Shultz LD, Keck J, Burzenski L, Jangalwe S, Vaidya S, Greiner DL, Brehm MA. Humanized mouse models of immunological diseases and precision medicine. Mamm Genome 2019; 30:123-142. [PMID: 30847553 PMCID: PMC6610695 DOI: 10.1007/s00335-019-09796-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/02/2019] [Indexed: 12/25/2022]
Abstract
With the increase in knowledge resulting from the sequencing of the human genome, the genetic basis for the underlying differences in individuals, their diseases, and how they respond to therapies is starting to be understood. This has formed the foundation for the era of precision medicine in many human diseases that is beginning to be implemented in the clinic, particularly in cancer. However, preclinical testing of therapeutic approaches based on individual biology will need to be validated in animal models prior to translation into patients. Although animal models, particularly murine models, have provided significant information on the basic biology underlying immune responses in various diseases and the response to therapy, murine and human immune systems differ markedly. These fundamental differences may be the underlying reason why many of the positive therapeutic responses observed in mice have not translated directly into the clinic. There is a critical need for preclinical animal models in which human immune responses can be investigated. For this, many investigators are using humanized mice, i.e., immunodeficient mice engrafted with functional human cells, tissues, and immune systems. We will briefly review the history of humanized mice, the remaining limitations, approaches to overcome them and how humanized mouse models are being used as a preclinical bridge in precision medicine for evaluation of human therapies prior to their implementation in the clinic.
Collapse
Affiliation(s)
- Leonard D Shultz
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA.
| | - James Keck
- The Jackson Laboratory, 1650 Santa Ana Avenue, Sacramento, CA, 95838, USA
| | - Lisa Burzenski
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Sonal Jangalwe
- Diabetes Center of Excellence, The University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| | - Shantashri Vaidya
- Diabetes Center of Excellence, The University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| | - Dale L Greiner
- Diabetes Center of Excellence, The University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| | - Michael A Brehm
- Diabetes Center of Excellence, The University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| |
Collapse
|
36
|
Komori S, Saito Y, Respatika D, Nishimura T, Kotani T, Murata Y, Matozaki T. SIRPα + dendritic cells promote the development of fibroblastic reticular cells in murine peripheral lymph nodes. Eur J Immunol 2019; 49:1364-1371. [PMID: 31099900 DOI: 10.1002/eji.201948103] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 04/01/2019] [Accepted: 05/15/2019] [Indexed: 11/06/2022]
Abstract
Nonhematopoietic stromal cells contribute to the organization and homeostasis of secondary lymphoid organs by producing cytokines and chemokines. The development and maintenance of these stromal cells are thought to be regulated by innate immune cells. Indeed, we recently showed that signal regulatory protein α (SIRPα)-positive dendritic cells (DCs) are essential for the proliferation and survival of podoplanin (Pdpn)-positive fibroblastic reticular cells (FRCs) in mouse spleen. We have now established an in vitro culture system for lymph node stromal cells (LNSCs) isolated from mouse peripheral LNs. Activated DCs and TNF-α each promoted the proliferation of cultured LNSCs, most of which were found to be Pdpn+ FRCs. Furthermore, ablation of SIRPα in CD11c+ cells attenuated this effect of DCs on LNSC proliferation. Transplantation of activated DCs together with cultured LNSCs into the renal subcapsular space markedly increased the number of ER-TR7+ stromal cells as well as induced the accumulation of T cells and increased the expression of Ccl19 in the transplants. Ablation of SIRPα in CD11c+ cells greatly impaired the development of LN-like structure in the transplants. Our findings thus suggest that SIRPα+ DCs are important for the proliferation and differentiation of Pdpn+ FRCs in peripheral LNs.
Collapse
Affiliation(s)
- Satomi Komori
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yasuyuki Saito
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Datu Respatika
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan.,Division of Reconstruction, Oculoplasty, and Oncology, Faculty of Medicine, Department of Ophthalmology, Public Health, and Nursing, Gadjah Mada University, Yogyakarta, Indonesia
| | - Taichi Nishimura
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takenori Kotani
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoji Murata
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takashi Matozaki
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
37
|
Zeng B, Shi S, Ashworth G, Dong C, Liu J, Xing F. ILC3 function as a double-edged sword in inflammatory bowel diseases. Cell Death Dis 2019; 10:315. [PMID: 30962426 PMCID: PMC6453898 DOI: 10.1038/s41419-019-1540-2] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/13/2019] [Accepted: 03/19/2019] [Indexed: 12/19/2022]
Abstract
Inflammatory bowel diseases (IBD), composed mainly of Crohn's disease (CD) and ulcerative colitis (UC), are strongly implicated in the development of intestinal inflammation lesions. Its exact etiology and pathogenesis are still undetermined. Recently accumulating evidence supports that group 3 innate lymphoid cells (ILC3) are responsible for gastrointestinal mucosal homeostasis through moderate generation of IL-22, IL-17, and GM-CSF in the physiological state. ILC3 contribute to the progression and aggravation of IBD while both IL-22 and IL-17, along with IFN-γ, are overexpressed by the dysregulation of NCR- ILC3 or NCR+ ILC3 function and the bias of NCR+ ILC3 towards ILC1 as well as regulatory ILC dysfunction in the pathological state. Herein, we feature the group 3 innate lymphoid cells' development, biological function, maintenance of gut homeostasis, mediation of IBD occurrence, and potential application to IBD therapy.
Collapse
Affiliation(s)
- Boning Zeng
- Institute of Tissue Transplantation and Immunology, Department of Immunobiology, Jinan University, Guangzhou, China
- Key Laboratory of Functional Protein Research of Guangdong, Higher Education Institutes, Jinan University, Guangzhou, China
| | - Shengnan Shi
- Institute of Tissue Transplantation and Immunology, Department of Immunobiology, Jinan University, Guangzhou, China
| | | | | | - Jing Liu
- BioMedical Research Centre, University of East Anglia, NR4 7TJ, Norwich, UK.
| | - Feiyue Xing
- Institute of Tissue Transplantation and Immunology, Department of Immunobiology, Jinan University, Guangzhou, China.
- Key Laboratory of Functional Protein Research of Guangdong, Higher Education Institutes, Jinan University, Guangzhou, China.
| |
Collapse
|
38
|
Pulmonary group 2 innate lymphoid cells: surprises and challenges. Mucosal Immunol 2019; 12:299-311. [PMID: 30664706 PMCID: PMC6436699 DOI: 10.1038/s41385-018-0130-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 02/04/2023]
Abstract
Group 2 innate lymphoid cells (ILC2s) are a recently described subset of innate lymphocytes with important immune and homeostatic functions at multiple tissue sites, especially the lung. These cells expand locally after birth and during postnatal lung maturation and are present in the lung and other peripheral organs. They are modified by a variety of processes and mediate inflammatory responses to respiratory pathogens, inhaled allergens and noxious particles. Here, we review the emerging roles of ILC2s in pulmonary homeostasis and discuss recent and surprising advances in our understanding of how hormones, age, neurotransmitters, environmental challenges, and infection influence ILC2s. We also review how these responses may underpin the development, progression and severity of pulmonary inflammation and chronic lung diseases and highlight some of the remaining challenges for ILC2 biology.
Collapse
|
39
|
Colonna M. Innate Lymphoid Cells: Diversity, Plasticity, and Unique Functions in Immunity. Immunity 2019; 48:1104-1117. [PMID: 29924976 DOI: 10.1016/j.immuni.2018.05.013] [Citation(s) in RCA: 280] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 05/13/2018] [Accepted: 05/30/2018] [Indexed: 01/12/2023]
Abstract
Type 1, 2, and 3 innate lymphoid cells (ILCs) have emerged as tissue-resident innate correlates of T helper 1 (Th1), Th2, and Th17 cells. Recent studies suggest that ILCs are more diverse than originally proposed; this might reflect truly distinct lineages or adaptation of ILCs to disparate tissue microenvironments, known as plasticity. Given that ILCs strikingly resemble T cells, are they redundant? While the regulation, timing, and magnitude of ILC and primary T cell responses differ, tissue-resident memory T cells may render ILCs redundant during secondary responses. The unique impact of ILCs in immunity is probably embodied in the extensive array of surface and intracellular receptors that endow these cells with the ability to distinguish between normal and pathogenic components, interact with other cells, and calibrate their cytokine secretion accordingly. Here I review recent advances in elucidating the diversity of ILCs and discuss their unique and redundant functions.
Collapse
Affiliation(s)
- Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
40
|
Abstract
Dendritic cells (DCs) can be viewed as translators between innate and adaptive immunity. They integrate signals derived from tissue infection or damage and present processed antigen from these sites to naive T cells in secondary lymphoid organs while also providing multiple soluble and surface-bound signals that help to guide T cell differentiation. DC-mediated tailoring of the appropriate T cell programme ensures a proper cascade of immune responses that adequately targets the insult. Recent advances in our understanding of the different types of DC subsets along with the cellular organization and orchestration of DC and lymphocyte positioning in secondary lymphoid organs over time has led to a clearer understanding of how the nature of the T cell response is shaped. This Review discusses how geographical organization and ordered sequences of cellular interactions in lymph nodes and the spleen regulate immunity.
Collapse
Affiliation(s)
- S C Eisenbarth
- Department of Laboratory Medicine, Immunobiology, Section of Allergy & Immunology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
41
|
Cooper MA, Fehniger TA, Colonna M. Is There Natural Killer Cell Memory and Can It Be Harnessed by Vaccination? Vaccination Strategies Based on NK Cell and ILC Memory. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a029512. [PMID: 29254976 DOI: 10.1101/cshperspect.a029512] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Studies over the last decade have decisively shown that innate immune natural killer (NK) cells exhibit enhanced long-lasting functional responses following a single activation event. With the increased recognition of memory and memory-like properties of NK cells, questions have arisen with regard to their ability to effectively mediate vaccination responses in humans. Moreover, recently discovered innate lymphoid cells (ILCs) could also potentially exhibit memory-like functions. Here, we review different forms of NK cell memory, and speculate about the ability of these cells and ILCs to meaningfully contribute to vaccination responses.
Collapse
Affiliation(s)
- Megan A Cooper
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, St. Louis, Missouri 63110.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Todd A Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
42
|
Kindstedt E, Koskinen Holm C, Palmqvist P, Sjöström M, Lejon K, Lundberg P. Innate lymphoid cells are present in gingivitis and periodontitis. J Periodontol 2018; 90:200-207. [PMID: 30070705 DOI: 10.1002/jper.17-0750] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 04/02/2018] [Accepted: 04/02/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Innate lymphoid cells (ILCs) are the most recently identified leukocytes of the immune system and these cells are increasingly acknowledged to play important roles in host defence and tissue repair. ILCs are also contributors of inflammatory diseases such as asthma and colitis. We analyzed the presence and relative proportions of the different ILC subsets (ILC1, ILC2 and ILC3) in gingivitis and periodontitis. Further, we investigated if ILCs express receptor activator of nuclear factor kappa-B ligand (RANKL), a cytokine crucial for osteoclast differentiation and bone resorption. METHODS We collected gingivitis and periodontitis soft tissue and characterized ILC subsets including RANKL expression in single-cell suspensions using flow cytometry. RESULTS ILCs were detected both in gingivitis and periodontitis. The majority of ILCs, in both conditions, were ILC1s. Furthermore, RANKL expression was detected on a fraction of the ILC1s. CONCLUSIONS Our discovery of the presence of ILCs both in gingivitis and periodontitis and concomitant expression of RANKL on a fraction of the ILC1 population suggest that these cells may be of importance in periodontal disease. In addition, our findings provide a new insight into the field of oral immunology.
Collapse
Affiliation(s)
- Elin Kindstedt
- Department of Odontology, Division of Molecular Periodontology, Umeå University, SE-901 85 Umeå, Sweden
| | - Cecilia Koskinen Holm
- Department of Odontology, Division of Molecular Periodontology, Umeå University, SE-901 85 Umeå, Sweden
| | - Py Palmqvist
- Department of Odontology, Division of Molecular Periodontology, Umeå University, SE-901 85 Umeå, Sweden
| | - Mats Sjöström
- Department of Odontology, Division of Oral and Maxillofacial Surgery, Umeå University, SE-901 85 Umeå, Sweden
| | - Kristina Lejon
- Department of Clinical Microbiology, Division of Immunology, Umeå University, SE-901 85 Umeå, Sweden
| | - Pernilla Lundberg
- Department of Odontology, Division of Molecular Periodontology, Umeå University, SE-901 85 Umeå, Sweden
| |
Collapse
|
43
|
Perez-Shibayama C, Gil-Cruz C, Cheng HW, Onder L, Printz A, Mörbe U, Novkovic M, Li C, Lopez-Macias C, Buechler MB, Turley SJ, Mack M, Soneson C, Robinson MD, Scandella E, Gommerman J, Ludewig B. Fibroblastic reticular cells initiate immune responses in visceral adipose tissues and secure peritoneal immunity. Sci Immunol 2018; 3:3/26/eaar4539. [DOI: 10.1126/sciimmunol.aar4539] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 04/18/2018] [Accepted: 06/07/2018] [Indexed: 12/19/2022]
|
44
|
Bar-Ephraim YE, Cornelissen F, Papazian N, Konijn T, Hoogenboezem RM, Sanders MA, Westerman BA, Gönültas M, Kwekkeboom J, Den Haan JMM, Reijmers RM, Mebius RE, Cupedo T. Cross-Tissue Transcriptomic Analysis of Human Secondary Lymphoid Organ-Residing ILC3s Reveals a Quiescent State in the Absence of Inflammation. Cell Rep 2018; 21:823-833. [PMID: 29045847 DOI: 10.1016/j.celrep.2017.09.070] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/21/2017] [Accepted: 09/21/2017] [Indexed: 12/31/2022] Open
Abstract
A substantial number of human and mouse group 3 innate lymphoid cells (ILC3s) reside in secondary lymphoid organs, yet the phenotype and function of these ILC3s is incompletely understood. Here, we employed an unbiased cross-tissue transcriptomic approach to compare human ILC3s from non-inflamed lymph nodes and spleen to their phenotypic counterparts in inflamed tonsils and from circulation. These analyses revealed that, in the absence of inflammation, lymphoid organ-residing ILC3s lack transcription of cytokines associated with classical ILC3 functions. This was independent of expression of the natural cytotoxicity receptor NKp44. However, and in contrast to ILC3s from peripheral blood, lymphoid organ-residing ILC3s express activating cytokine receptors and have acquired the ability to be recruited into immune responses by inflammatory cytokines. This comprehensive cross-tissue dataset will allow for identification of functional changes in human lymphoid organ ILC3s associated with human disease.
Collapse
Affiliation(s)
- Yotam E Bar-Ephraim
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1081 HZ Amsterdam, the Netherlands
| | - Ferry Cornelissen
- Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Natalie Papazian
- Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Tanja Konijn
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1081 HZ Amsterdam, the Netherlands
| | - Remco M Hoogenboezem
- Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Mathijs A Sanders
- Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Bart A Westerman
- Department of Neurosurgery, VU University Medical Center, Cancer Center Amsterdam, 1081 HV Amsterdam, the Netherlands
| | - Mehmet Gönültas
- Department of Otolaryngology, Slotervaart Hospital, 1066 EC Amsterdam, the Netherlands
| | - Jaap Kwekkeboom
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Joke M M Den Haan
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1081 HZ Amsterdam, the Netherlands
| | - Rogier M Reijmers
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1081 HZ Amsterdam, the Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1081 HZ Amsterdam, the Netherlands.
| | - Tom Cupedo
- Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands.
| |
Collapse
|
45
|
Bonne-Année S, Nutman TB. Human innate lymphoid cells (ILCs) in filarial infections. Parasite Immunol 2018; 40:10.1111/pim.12442. [PMID: 28504838 PMCID: PMC5685925 DOI: 10.1111/pim.12442] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/11/2017] [Indexed: 12/22/2022]
Abstract
Filarial infections are characteristically chronic and can cause debilitating diseases governed by parasite-induced innate and adaptive immune responses. Filarial parasites traverse or establish niches in the skin (migrating infective larvae), in nonmucosal tissues (adult parasite niche) and in the blood or skin (circulating microfilariae) where they intersect with the host immune response. While several studies have demonstrated that filarial parasites and their antigens can modulate myeloid cells (monocyte, macrophage and dendritic cell subsets), T- and B-lymphocytes and skin resident cell populations, the role of innate lymphoid cells during filarial infections has only recently emerged. Despite the identification and characterization of innate lymphoid cells (ILCs) in murine helminth infections, little is actually known about the role of human ILCs during parasitic infections. The focus of this review will be to highlight the composition of ILCs in the skin, lymphatics and blood; where the host-parasite interaction is well-defined and to examine the role of ILCs during filarial infections.
Collapse
Affiliation(s)
- S Bonne-Année
- Laboratory of Parasitic Diseases, Helminth Immunology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - T B Nutman
- Laboratory of Parasitic Diseases, Helminth Immunology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
46
|
Jetten AM, Takeda Y, Slominski A, Kang HS. Retinoic acid-related Orphan Receptor γ (RORγ): connecting sterol metabolism to regulation of the immune system and autoimmune disease. CURRENT OPINION IN TOXICOLOGY 2018; 8:66-80. [PMID: 29568812 DOI: 10.1016/j.cotox.2018.01.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cholesterol and its metabolites are bioactive lipids that interact with and regulate the activity of various proteins and signaling pathways that are implicated in the control of a variety of physiological and pathological processes. Recent studies revealed that retinoic acid-related orphan receptors, RORα and γ, members of the ligand-dependent nuclear receptor superfamily, exhibit quite a wide binding specificity for a number of sterols. Several cholesterol intermediates and metabolites function as natural ligands of RORα and RORγ and act as agonists or inverse agonists. Changes in cholesterol homeostasis that alter the level or type of sterol metabolites in cells, can either enhance or inhibit ROR transcriptional activity that subsequently result in changes in the physiological processes regulated by RORs, including various immune responses and metabolic pathways. Consequently, this might negatively or positively impact pathologies, in which RORs are implicated, such as autoimmune disease, inflammation, metabolic syndrome, cancer, and several neurological disorders. Best studied are the links between cholesterol metabolism, RORγt activity, and their regulation of Th17 differentiation and autoimmune disease. The discovery that Th17-dependent inflammation is significantly attenuated in RORγ-deficient mice in several experimental autoimmune disease models, initiated a search for ROR modulators that led to the identification of a number of small molecular weight RORγ inverse agonists. The inverse agonists suppress Th17 differentiation and IL-17 production and protect against autoimmunity. Together, these studies suggest that RORγt may provide an attractive therapeutic target in the management of several (inflammatory) diseases.
Collapse
Affiliation(s)
- Anton M Jetten
- Cell Biology Section, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Yukimasa Takeda
- Cell Biology Section, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Andrzej Slominski
- Department of Dermatology, Comprehensive Cancer Center Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL 35249, USA
| | - Hong Soon Kang
- Cell Biology Section, Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| |
Collapse
|
47
|
Kurashima Y, Yamamoto D, Nelson S, Uematsu S, Ernst PB, Nakayama T, Kiyono H. Mucosal Mesenchymal Cells: Secondary Barrier and Peripheral Educator for the Gut Immune System. Front Immunol 2017; 8:1787. [PMID: 29321781 PMCID: PMC5733542 DOI: 10.3389/fimmu.2017.01787] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/29/2017] [Indexed: 01/08/2023] Open
Abstract
Stromal connective tissue contains mesenchymal cells, including fibroblasts and myofibroblasts, which line the tissue structure. However, it has been identified that the function of mesenchymal cells is not just structural-they also play critical roles in the creation and regulation of intestinal homeostasis. Thus, mucosal mesenchymal cells instruct intestinal immune cell education (or peripheral immune education) and epithelial cell differentiation thereby shaping the local environment of the mucosal immune system. Malfunction of the mesenchymal cell-mediated instruction system (e.g., fibrosis) leads to pathological conditions such as intestinal stricture.
Collapse
Affiliation(s)
- Yosuke Kurashima
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Division of Clinical Vaccinology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Institute for Global Prominent Research, Chiba University, Chiba, Japan.,Department of Mucosal Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.,Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines (CU-UCSD cMAV), San Diego, CA, Unites States
| | - Daiki Yamamoto
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sean Nelson
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoshi Uematsu
- Department of Mucosal Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines (CU-UCSD cMAV), San Diego, CA, Unites States.,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Peter B Ernst
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines (CU-UCSD cMAV), San Diego, CA, Unites States.,Center for Veterinary Sciences and Comparative Medicine, University of California, San Diego, CA, Unites States.,Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, CA, Unites States
| | - Toshinori Nakayama
- Institute for Global Prominent Research, Chiba University, Chiba, Japan.,Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines (CU-UCSD cMAV), San Diego, CA, Unites States.,Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Division of Clinical Vaccinology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines (CU-UCSD cMAV), San Diego, CA, Unites States.,Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
48
|
Elemam NM, Hannawi S, Maghazachi AA. Innate Lymphoid Cells (ILCs) as Mediators of Inflammation, Release of Cytokines and Lytic Molecules. Toxins (Basel) 2017; 9:toxins9120398. [PMID: 29232860 PMCID: PMC5744118 DOI: 10.3390/toxins9120398] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 12/31/2022] Open
Abstract
Innate lymphoid cells (ILCs) are an emerging group of immune cells that provide the first line of defense against various pathogens as well as contributing to tissue repair and inflammation. ILCs have been classically divided into three subgroups based on their cytokine secretion and transcription factor profiles. ILC nomenclature is analogous to that of T helper cells. Group 1 ILCs composed of natural killer (NK) cells as well as IFN-γ secreting ILC1s. ILC2s have the capability to produce TH2 cytokines while ILC3s and lymphoid tissue inducer (LTis) are subsets of cells that are able to secrete IL-17 and/or IL-22. A recent subset of ILC known as ILC4 was discovered, and the cells of this subset were designated as NK17/NK1 due to their release of IL-17 and IFN-γ. In this review, we sought to explain the subclasses of ILCs and their roles as mediators of lytic enzymes and inflammation.
Collapse
Affiliation(s)
- Noha Mousaad Elemam
- Department of Clinical Sciences, College of Medicine, and Sharjah Institute for Medical Research (SIMR), University of Sharjah, Sharjah 27272, UAE.
| | - Suad Hannawi
- Medical Department, Ministry of Health and Prevention, Dubai 65522, UAE.
| | - Azzam A Maghazachi
- Department of Clinical Sciences, College of Medicine, and Sharjah Institute for Medical Research (SIMR), University of Sharjah, Sharjah 27272, UAE.
| |
Collapse
|
49
|
Miah SMS, Jayasuriya CT, Salter AI, Reilly EC, Fugere C, Yang W, Chen Q, Brossay L. Ptpn11 Deletion in CD4 + Cells Does Not Affect T Cell Development and Functions but Causes Cartilage Tumors in a T Cell-Independent Manner. Front Immunol 2017; 8:1326. [PMID: 29085371 PMCID: PMC5650614 DOI: 10.3389/fimmu.2017.01326] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 09/29/2017] [Indexed: 12/01/2022] Open
Abstract
The ubiquitously expressed tyrosine phosphatase Src homology region 2 domain-containing phosphatase-2 (SHP-2, encoded by Ptpn11) is required for constitutive cellular processes including proliferation, differentiation, and the regulation of immune responses. During development and maturation, subsets of T cells express a variety of inhibitory receptors known to associate with phosphatases, which in turn, dephosphorylate key players of activating receptor signaling pathways. We hypothesized that SHP-2 deletion would have major effects on T cell development by altering the thresholds for activation, as well as positive and negative selection. Surprisingly, using mice conditionally deficient for SHP-2 in the T cell lineage, we show that the development of these lymphocytes is globally intact. In addition, our data demonstrate that SHP-2 absence does not compromise T cell effector functions, suggesting that SHP-2 is dispensable in these cells. Unexpectedly, in aging mice, Ptpn11 gene deletion driven by CD4 Cre recombinase leads to cartilage tumors in wrist bones in a T cell-independent manner. These tumors resemble miniature cartilaginous growth plates and contain CD4-lineage positive chondrocyte-like cells. Altogether these results indicate that SHP-2 is a cartilage tumor suppressor during aging.
Collapse
Affiliation(s)
- S M Shahjahan Miah
- Department of Molecular Microbiology and Immunology, Graduate Program in Pathobiology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, United States
| | - Chathuraka T Jayasuriya
- Department of Orthopaedics, Rhode Island Hospital and Brown University Alpert Medical School, Providence, RI, United States
| | - Alexander I Salter
- Department of Molecular Microbiology and Immunology, Graduate Program in Pathobiology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, United States
| | - Emma C Reilly
- Department of Molecular Microbiology and Immunology, Graduate Program in Pathobiology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, United States
| | - Céline Fugere
- Department of Molecular Microbiology and Immunology, Graduate Program in Pathobiology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, United States
| | - Wentian Yang
- Department of Orthopaedics, Rhode Island Hospital and Brown University Alpert Medical School, Providence, RI, United States
| | - Qian Chen
- Department of Orthopaedics, Rhode Island Hospital and Brown University Alpert Medical School, Providence, RI, United States
| | - Laurent Brossay
- Department of Molecular Microbiology and Immunology, Graduate Program in Pathobiology, Division of Biology and Medicine, Brown University Alpert Medical School, Providence, RI, United States
| |
Collapse
|
50
|
Geremia A, Arancibia-Cárcamo CV. Innate Lymphoid Cells in Intestinal Inflammation. Front Immunol 2017; 8:1296. [PMID: 29081776 PMCID: PMC5645495 DOI: 10.3389/fimmu.2017.01296] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/27/2017] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the intestine that encompasses Crohn's disease (CD) and ulcerative colitis. The cause of IBD is unknown, but the evidence suggests that an aberrant immune response toward the commensal bacterial flora is responsible for disease in genetically susceptible individuals. Results from animal models of colitis and human studies indicate a role for innate lymphoid cells (ILC) in the pathogenesis of chronic intestinal inflammation in IBD. ILC are a population of lymphocytes that are enriched at mucosal sites, where they play a protective role against pathogens including extracellular bacteria, helminthes, and viruses. ILC lack an antigen-specific receptor, but can respond to environmental stress signals contributing to the rapid orchestration of an early immune response. Several subsets of ILC reflecting functional characteristics of T helper subsets have been described. ILC1 express the transcription factor T-bet and are characterized by secretion of IFNγ, ILC2 are GATA3+ and secrete IL5 and IL13 and ILC3 depend on expression of RORγt and secrete IL17 and IL22. However, ILC retain a degree of plasticity depending on exposure to cytokines and environmental factors. IL23 responsive ILC have been implicated in the pathogenesis of colitis in several innate murine models through the production of IL17, IFNγ, and GM-CSF. We have previously identified IL23 responsive ILC in the human intestine and found that they accumulate in the inflamed colon and small bowel of patients with CD. Other studies have confirmed accumulation of ILC in CD with increased frequencies of IFNγ-secreting ILC1 in both the intestinal lamina propria and the epithelium. Moreover, IL23 driven IL22 producing ILC have been shown to drive bacteria-induced colitis-associated cancer in mice. Interestingly, our data show increased ILC accumulation in patients with IBD and primary sclerosing cholangitis, who carry an increased risk of developing colorectal cancer. ILC may play an important amplifying role in IBD and IBD-associated cancer, through secretion of inflammatory cytokines and interaction with other immune and non-immune cells. Here, we will review the evidence indicating a role for ILC in the pathogenesis of chronic intestinal inflammation.
Collapse
Affiliation(s)
- Alessandra Geremia
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom
| | - Carolina V Arancibia-Cárcamo
- Translational Gastroenterology Unit, Nuffield Department of Experimental Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|