101
|
Ohno Y, Satoh K, Shitara A, Into T, Kashimata M. Arginase 1 is involved in lacrimal hyposecretion in male NOD mice, a model of Sjögren's syndrome, regardless of dacryoadenitis status. J Physiol 2020; 598:4907-4925. [PMID: 32780506 PMCID: PMC7693353 DOI: 10.1113/jp280090] [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: 05/13/2020] [Accepted: 08/01/2020] [Indexed: 01/14/2023] Open
Abstract
Key points Few reports have explored the possibility of involvement of non‐inflammatory factors in lacrimal hyposecretion in Sjögren's syndrome (SS). RNA‐sequencing analysis revealed that only four genes, including arginase 1, were downregulated in the lacrimal gland of SS model male mice (NOD mice) after onset of lacrimal hyposecretion and dacryoadenitis. Even in non‐dacryoadenitis‐type NOD mice, tear secretion and arginase 1 expression remained low. An arginase 1 inhibitor reduced tear secretion and partially reduced saliva secretion in BALB/c mice. The results indicate that a non‐inflammatory factor, arginase 1, is involved in lacrimal hyposecretion in male NOD mice, regardless of dacryoadenitis status.
Abstract Lacrimal fluid (tears) is important for preservation of the ocular surface, and thus lacrimal hyposecretion in Sjögren's syndrome (SS) leads to reduced quality of life. However, the cause(s) of lacrimal hyposecretion remains unknown, even though many studies have been conducted from the perspective of inflammation. Here, we hypothesized that a non‐inflammatory factor induces lacrimal hyposecretion in SS pathology, and to elucidate such a factor, we conducted transcriptome analysis of the lacrimal glands in male non‐obese diabetic (NOD) mice as an SS model. The NOD mice showed inflammatory cell infiltration and decreased pilocarpine‐induced tear secretion at and after 6 weeks of age compared to age‐matched BALB/c mice. RNA‐sequencing analysis revealed that only four genes, including arginase 1, were downregulated, whereas many genes relating to inflammation were upregulated, in the lacrimal glands of male NOD mice after onset of lacrimal hyposecretion and dacryoadenitis (lacrimal gland inflammation). Changes in the level of arginase 1 expression were confirmed by real‐time RT‐PCR and western blot analysis. Furthermore, non‐dacryoadenitis‐type NOD mice were used to investigate the relationships among arginase 1 expression, lacrimal hyposecretion and dacryoadenitis. Interestingly, these NOD mice retained the phenotype of dacryoadenitis with regard to tear secretion and arginase 1 expression level. An arginase 1 inhibitor reduced tear secretion and partially reduced saliva secretion in BALB/c mice. In conclusion, a non‐inflammatory factor, arginase 1, is involved in lacrimal hyposecretion in male NOD mice, regardless of dacryoadenitis status. These results shed light on the pathophysiological role of arginase 1 in SS (dry eye). Few reports have explored the possibility of involvement of non‐inflammatory factors in lacrimal hyposecretion in Sjögren's syndrome (SS). RNA‐sequencing analysis revealed that only four genes, including arginase 1, were downregulated in the lacrimal gland of SS model male mice (NOD mice) after onset of lacrimal hyposecretion and dacryoadenitis. Even in non‐dacryoadenitis‐type NOD mice, tear secretion and arginase 1 expression remained low. An arginase 1 inhibitor reduced tear secretion and partially reduced saliva secretion in BALB/c mice. The results indicate that a non‐inflammatory factor, arginase 1, is involved in lacrimal hyposecretion in male NOD mice, regardless of dacryoadenitis status.
Collapse
Affiliation(s)
- Yuta Ohno
- Department of Pharmacology, Asahi University School of Dentistry, 1851-1 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Keitaro Satoh
- Department of Pharmacology, Asahi University School of Dentistry, 1851-1 Hozumi, Mizuho, Gifu, 501-0296, Japan.,Department of Pharmacology, Meikai University School of Dentistry, 1-1 Keyakidai, Sakado, Saitama, 350-0283, Japan
| | - Akiko Shitara
- Department of Pharmacology, Asahi University School of Dentistry, 1851-1 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Takeshi Into
- Department of Oral Microbiology, Asahi University School of Dentistry, 1851-1 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Masanori Kashimata
- Department of Pharmacology, Asahi University School of Dentistry, 1851-1 Hozumi, Mizuho, Gifu, 501-0296, Japan
| |
Collapse
|
102
|
Cong J. Metabolism of Natural Killer Cells and Other Innate Lymphoid Cells. Front Immunol 2020; 11:1989. [PMID: 32983138 PMCID: PMC7484708 DOI: 10.3389/fimmu.2020.01989] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/23/2020] [Indexed: 12/15/2022] Open
Abstract
Natural killer (NK) cells are the host's first line of defense against tumors and viral infections without prior sensitization. It is increasingly accepted that NK cells belong to the innate lymphoid cell (ILC) family. Other ILCs, comprising ILC1s, ILC2s, ILC3s and lymphoid tissue inducer (LTi) cells, are largely non-cytotoxic, tissue-resident cells, which function to protect local microenvironments against tissue insults and maintain homeostasis. Recently, evidence has accumulated that metabolism supports many aspects of the biology of NK cells and other ILCs, and that metabolic reprogramming regulates their development and function. Here, we outline the current understanding of ILC metabolism, and describe how metabolic processes are affected, and how metabolic defects are coupled to dysfunction of ILCs, in disease settings. Furthermore, we summarize the current and potential directions for immunotherapy involving targeting of ILC metabolism. Finally, we discuss the open questions in the rapidly expanding field of ILC metabolism.
Collapse
Affiliation(s)
- Jingjing Cong
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, School of Basic Medical Sciences, University of Science and Technology of China, Hefei, China
- Institue of Immunology, University of Science and Technology of China, Hefei, China
| |
Collapse
|
103
|
Basophils prime group 2 innate lymphoid cells for neuropeptide-mediated inhibition. Nat Immunol 2020; 21:1181-1193. [PMID: 32807943 PMCID: PMC9357342 DOI: 10.1038/s41590-020-0753-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/29/2020] [Indexed: 12/16/2022]
Abstract
Type 2 cytokine responses promote parasitic immunity and initiate tissue repair but, can also result in immunopathologies when not properly restricted. Basophilia is recognized as a common feature of type 2 inflammation, however, the roles basophils play in regulating these responses remain unknown. Here, we demonstrate that helminth-induced ILC2 responses are exaggerated in the absence of basophils, resulting in increased inflammation and diminished lung function. Additionally, we show that ILC2s from basophil-depleted mice express reduced amounts of the receptor for the neuropeptide, neuromedin B (NMB). Critically, NMB stimulation inhibited ILC2 responses from control but not basophil-depleted mice, and basophils were sufficient to directly enhance NMB receptor (NMBR) expression on ILC2s. These studies suggest that basophils prime ILC2s to respond to neuron-derived signals necessary to maintain tissue integrity. Further, these data provide mechanistic insight into the functions of basophils and identify NMB as a potent inhibitor of type 2 inflammation.
Collapse
|
104
|
Helou DG, Shafiei-Jahani P, Lo R, Howard E, Hurrell BP, Galle-Treger L, Painter JD, Lewis G, Soroosh P, Sharpe AH, Akbari O. PD-1 pathway regulates ILC2 metabolism and PD-1 agonist treatment ameliorates airway hyperreactivity. Nat Commun 2020; 11:3998. [PMID: 32778730 PMCID: PMC7417739 DOI: 10.1038/s41467-020-17813-1] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023] Open
Abstract
Allergic asthma is a leading chronic disease associated with airway hyperreactivity (AHR). Type-2 innate lymphoid cells (ILC2s) are a potent source of T-helper 2 (Th2) cytokines that promote AHR and lung inflammation. As the programmed cell death protein-1 (PD-1) inhibitory axis regulates a variety of immune responses, here we investigate PD-1 function in pulmonary ILC2s during IL-33-induced airway inflammation. PD-1 limits the viability of ILC2s and downregulates their effector functions. Additionally, PD-1 deficiency shifts ILC2 metabolism toward glycolysis, glutaminolysis and methionine catabolism. PD-1 thus acts as a metabolic checkpoint in ILC2s, affecting cellular activation and proliferation. As the blockade of PD-1 exacerbates AHR, we also develop a human PD-1 agonist and show that it can ameliorate AHR and suppresses lung inflammation in a humanized mouse model. Together, these results highlight the importance of PD-1 agonistic treatment in allergic asthma and underscore its therapeutic potential.
Collapse
Affiliation(s)
- Doumet Georges Helou
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Pedram Shafiei-Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Richard Lo
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Emily Howard
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Benjamin P Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lauriane Galle-Treger
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jacob D Painter
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Gavin Lewis
- Janssen Research and Development, San Diego, CA, USA
| | | | - Arlene H Sharpe
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
105
|
Miller MM, Reinhardt RL. The Heterogeneity, Origins, and Impact of Migratory iILC2 Cells in Anti-helminth Immunity. Front Immunol 2020; 11:1594. [PMID: 32793230 PMCID: PMC7390839 DOI: 10.3389/fimmu.2020.01594] [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: 04/22/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Soil-transmitted helminths represent a major global health burden with infections and infection-related comorbidities causing significant reductions in the quality of life for individuals living in endemic areas. Repeated infections and chronic colonization by these large extracellular worms in mammals led to the evolution of type-2 immunity characterized by the production of the type-2 cytokines interleukin (IL)-4, IL-5, and IL-13. Although a number of adaptive and innate immune cells produce type-2 cytokines, a key cellular source in the context of helminth infection is group 2 innate lymphoid cells (ILC2s). ILC2s promote mucosal barrier homeostasis, integrity, and repair by rapidly responding to epithelial cues in mucosal tissues. Though tissue-resident ILC2s (nILC2s) have been studied in detail over the last decade, considerably less is known with regard to a subset of inflammatory ILC2s (iILC2s) that migrate to the lungs of mice early after Nippostrongylus brasiliensis infection and are potent early producers of type-2 cytokines. This review will discuss the relationship and differences between nILC2s and iILC2s that establish their unique roles in anti-helminth immunity. We have placed particular emphasis on studies investigating iILC2 origin, function, and their potential long-term contribution to tissue-resident ILC2 reservoirs in settings of helminth infection.
Collapse
Affiliation(s)
- Mindy M Miller
- Department of Biomedical Research, National Jewish Health, Denver, CO, United States
| | - R Lee Reinhardt
- Department of Biomedical Research, National Jewish Health, Denver, CO, United States.,Department of Immunology and Microbiology, University of Colorado-Anschutz Medical, Aurora, CO, United States
| |
Collapse
|
106
|
Wieczfinska J, Sitarek P, Kowalczyk T, Pawliczak R. Leonurus sibiricus root extracts decrease airway remodeling markers expression in fibroblasts. Clin Exp Immunol 2020; 202:28-46. [PMID: 32562256 DOI: 10.1111/cei.13481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/27/2020] [Accepted: 06/12/2020] [Indexed: 12/15/2022] Open
Abstract
Bronchial asthma is believed to be provoked by the interaction between airway inflammation and remodeling. Airway remodeling is a complex and poorly understood process, and controlling it appears key for halting the progression of asthma and other obstructive lung diseases. Plants synthesize a number of valuable compounds as constitutive products and as secondary metabolites, many of which have curative properties. The aim of this study was to evaluate the anti-remodeling properties of extracts from transformed and transgenic Leonurus sibiricus roots with transformed L. sibiricus roots extract with transcriptional factor AtPAP1 overexpression (AtPAP1). Two fibroblast cell lines, Wistar Institute-38 (WI-38) and human fetal lung fibroblast (HFL1), were incubated with extracts from transformed L. sibiricus roots (TR) and roots with transcriptional factor AtPAP1 over-expression (AtPAP1 TR). Additionally, remodeling conditions were induced in the cultures with rhinovirus 16 (HRV16). The expressions of metalloproteinase 9 (MMP)-9, tissue inhibitor of metalloproteinases 1 (TIMP-1), arginase I and transforming growth factor (TGF)-β were determined by quantitative polymerase chain reaction (qPCR) and immunoblotting methods. AtPAP1 TR decreased arginase I and MMP-9 expression with no effect on TIMP-1 or TGF-β mRNA expression. This extract also inhibited HRV16-induced expression of arginase I, MMP-9 and TGF-β in both cell lines (P < 0·05) Our study shows for the first time to our knowledge, that transformed AtPAP1 TR extract from L. sibiricus root may affect the remodeling process. Its effect can be attributed an increased amount of phenolic acids such as: chlorogenic acid, caffeic acid or ferulic acid and demonstrates the value of biotechnology in medicinal research.
Collapse
Affiliation(s)
- J Wieczfinska
- Department of Immunopathology, Medical University of Lodz, Lodz, Poland
| | - P Sitarek
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Lodz, Poland
| | - T Kowalczyk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Lodz, Poland
| | - R Pawliczak
- Department of Immunopathology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
107
|
Wu K, Wang X, Keeler SP, Gerovac BJ, Agapov EV, Byers DE, Gilfillan S, Colonna M, Zhang Y, Holtzman MJ. Group 2 Innate Lymphoid Cells Must Partner with the Myeloid-Macrophage Lineage for Long-Term Postviral Lung Disease. THE JOURNAL OF IMMUNOLOGY 2020; 205:1084-1101. [PMID: 32641386 DOI: 10.4049/jimmunol.2000181] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/05/2020] [Indexed: 12/16/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) are implicated in host defense and inflammatory disease, but these potential functional roles need more precise definition, particularly using advanced technologies to better target ILC2s and engaging experimental models that better manifest both acute infection and chronic, even lifelong, disease. In this study, we use a mouse model that applies an improved genetic definition of ILC2s via IL-7r-conditional Rora gene targeting and takes advantage of a distinct progression from acute illness to chronic disease, based on a persistent type 2 immune response to respiratory infection with a natural pathogen (Sendai virus). We first show that ILC2s are activated but are not required to handle acute illness after respiratory viral infection. In contrast, we find that this type of infection also activates ILC2s chronically for IL-13 production and consequent asthma-like disease traits that peak and last long after active viral infection is cleared. However, to manifest this type of disease, the Csf1-dependent myeloid-macrophage lineage is also active at two levels: first, at a downstream level, this lineage provides lung tissue macrophages (interstitial macrophages and tissue monocytes) that represent a major site of Il13 gene expression in the diseased lung; and second, at an upstream level, this same lineage is required for Il33 gene induction that is necessary to activate ILC2s for participation in disease at all, including IL-13 production. Together, these findings provide a revised scheme for understanding and controlling the innate immune response leading to long-term postviral lung diseases with features of asthma and related progressive conditions.
Collapse
Affiliation(s)
- Kangyun Wu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Xinyu Wang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Shamus P Keeler
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Benjamin J Gerovac
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Eugene V Agapov
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Derek E Byers
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Susan Gilfillan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; and
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110; and
| | - Yong Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J Holtzman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110; .,Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
108
|
Mitochondrial function in immune cells in health and disease. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165845. [PMID: 32473386 DOI: 10.1016/j.bbadis.2020.165845] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023]
Abstract
One of the main functions of mitochondria is production of ATP for cellular energy needs, however, it becomes more recognized that mitochondria are involved in differentiation and activation processes of immune cells. Upon activation, immune cells have a high need for energy. Immune cells have different strategies to generate this energy. In pro-inflammatory cells, such as activated monocytes and activated T and B cells, the energy is generated by increasing glycolysis, while in regulatory cells, such as regulatory T cells or M2 macrophages, energy is generated by increasing mitochondrial function and beta-oxidation. Except for being important for energy supply during activation, mitochondria also induce immune responses. During an infection, they release mitochondrial danger associated molecules (DAMPs) that resemble structures of bacterial derived pathogen associated molecular patterns (PAMPs). Such mitochondrial DAMPS are for instance mitochondrial DNA with hypomethylated CpG motifs or a specific lipid that is only present in prokaryotic bacteria and mitochondria, i.e. cardiolipin. Via release of such DAMPs, mitochondria guide the immune response towards an inflammatory response against pathogens. This is an important mechanism in early detection of an infection and in stimulating and sustaining immune responses to fight infections. However, mitochondrial DAMPs may also have a negative impact. If mitochondrial DAMPs are released by damaged cells, without the presence of an infection, such as after a trauma, mitochondrial DAMPs may induce an undesired inflammatory response, resulting in tissue damage and organ dysfunction. Thus, immune cells have developed mechanisms to prevent such undesired immune activation by mitochondrial components. In the present narrative review, we will describe the current view of mitochondria in regulation of immune responses. We will also discuss the current knowledge on disturbed mitochondrial function in immune cells in various immunological diseases.
Collapse
|
109
|
Seo GY, Giles DA, Kronenberg M. The role of innate lymphoid cells in response to microbes at mucosal surfaces. Mucosal Immunol 2020; 13:399-412. [PMID: 32047273 PMCID: PMC7186215 DOI: 10.1038/s41385-020-0265-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 01/24/2020] [Accepted: 01/27/2020] [Indexed: 02/04/2023]
Abstract
Innate lymphoid cells (ILCs) are a lymphocyte population that is mostly resident at mucosal surfaces. They help to induce an appropriate immune response to the microbiome at homeostasis. In healthy people, the mucosal immune system works symbiotically with organisms that make up the microbiota. ILCs play a critical role in orchestrating this balance, as they can both influence and in turn be influenced by the microbiome. ILCs also are important regulators of the early response to infections by diverse types of pathogenic microbes at mucosal barriers. Their rapid responses initiate inflammatory programs, production of antimicrobial products and repair processes. This review will focus on the role of ILCs in response to the microbiota and to microbial infections of the lung and intestine.
Collapse
Affiliation(s)
- Goo-Young Seo
- Division of Developmental Immunology, La Jolla Institute for Immunology, 9420 Athena Circle La Jolla, CA, 92037, USA
| | - Daniel A Giles
- Division of Developmental Immunology, La Jolla Institute for Immunology, 9420 Athena Circle La Jolla, CA, 92037, USA
| | - Mitchell Kronenberg
- Division of Developmental Immunology, La Jolla Institute for Immunology, 9420 Athena Circle La Jolla, CA, 92037, USA,Division of Biology, University of California San Diego, La Jolla, CA 92037, USA,Correspondence:
| |
Collapse
|
110
|
Karagiannis F, Masouleh SK, Wunderling K, Surendar J, Schmitt V, Kazakov A, Michla M, Hölzel M, Thiele C, Wilhelm C. Lipid-Droplet Formation Drives Pathogenic Group 2 Innate Lymphoid Cells in Airway Inflammation. Immunity 2020; 52:620-634.e6. [PMID: 32268121 DOI: 10.1016/j.immuni.2020.03.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/31/2020] [Accepted: 03/12/2020] [Indexed: 12/26/2022]
Abstract
Innate lymphoid cells (ILCs) play an important role in the control and maintenance of barrier immunity. However, chronic activation of ILCs results in immune-mediated pathology. Here, we show that tissue-resident type 2 ILCs (ILC2s) display a distinct metabolic signature upon chronic activation. In the context of allergen-driven airway inflammation, ILC2s increase their uptake of both external lipids and glucose. Externally acquired fatty acids are transiently stored in lipid droplets and converted into phospholipids to promote the proliferation of ILC2s. This metabolic program is imprinted by interleukin-33 (IL-33) and regulated by the genes Pparg and Dgat1, which are both controlled by glucose availability and mTOR signaling. Restricting dietary glucose by feeding mice a ketogenic diet largely ablated ILC2-mediated airway inflammation by impairing fatty acid metabolism and the formation of lipid droplets. Together, these results reveal that pathogenic ILC2 responses require lipid metabolism and identify ketogenic diet as a potent intervention strategy to treat airway inflammation.
Collapse
Affiliation(s)
- Fotios Karagiannis
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Schekufe Kharabi Masouleh
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Klaus Wunderling
- Life & Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Jayagopi Surendar
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Vanessa Schmitt
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Alexander Kazakov
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Marcel Michla
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Michael Hölzel
- Institute of Experimental Oncology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Christoph Thiele
- Life & Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Christoph Wilhelm
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany.
| |
Collapse
|
111
|
Do AN, Watson CT, Cohain AT, Griffin RS, Grishin A, Wood RA, Wesley Burks A, Jones SM, Scurlock A, Leung DYM, Sampson HA, Sicherer SH, Sharp AJ, Schadt EE, Bunyavanich S. Dual transcriptomic and epigenomic study of reaction severity in peanut-allergic children. J Allergy Clin Immunol 2020; 145:1219-1230. [PMID: 31838046 PMCID: PMC7192362 DOI: 10.1016/j.jaci.2019.10.040] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/27/2019] [Accepted: 10/29/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Unexpected allergic reactions to peanut are the most common cause of fatal food-related anaphylaxis. Mechanisms underlying the variable severity of peanut-allergic reactions remain unclear. OBJECTIVES We sought to expand mechanistic understanding of reaction severity in peanut allergy. METHODS We performed an integrated transcriptomic and epigenomic study of peanut-allergic children as they reacted in vivo during double-blind, placebo-controlled peanut challenges. We integrated whole-blood transcriptome and CD4+ T-cell epigenome profiles to identify molecular signatures of reaction severity (ie, how severely a peanut-allergic child reacts when exposed to peanut). A threshold-weighted reaction severity score was calculated for each subject based on symptoms experienced during peanut challenge and the eliciting dose. Through linear mixed effects modeling, network construction, and causal mediation analysis, we identified genes, CpGs, and their interactions that mediate reaction severity. Findings were replicated in an independent cohort. RESULTS We identified 318 genes with changes in expression during the course of reaction associated with reaction severity, and 203 CpG sites with differential DNA methylation associated with reaction severity. After replicating these findings in an independent cohort, we constructed interaction networks with the identified peanut severity genes and CpGs. These analyses and leukocyte deconvolution highlighted neutrophil-mediated immunity. We identified NFKBIA and ARG1 as hubs in the networks and 3 groups of interacting key node CpGs and peanut severity genes encompassing immune response, chemotaxis, and regulation of macroautophagy. In addition, we found that gene expression of PHACTR1 and ZNF121 causally mediates the association between methylation at corresponding CpGs and reaction severity, suggesting that methylation may serve as an anchor upon which gene expression modulates reaction severity. CONCLUSIONS Our findings enhance current mechanistic understanding of the genetic and epigenetic architecture of reaction severity in peanut allergy.
Collapse
Affiliation(s)
- Anh N Do
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Corey T Watson
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Ky
| | - Ariella T Cohain
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Robert S Griffin
- Department of Anesthesia, Hospital for Special Surgery, New York, NY
| | - Alexander Grishin
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Robert A Wood
- Department of Pediatrics, Johns Hopkins University, Baltimore, Md
| | - A Wesley Burks
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC
| | - Stacie M Jones
- Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock, Ark
| | - Amy Scurlock
- Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock, Ark
| | | | - Hugh A Sampson
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Scott H Sicherer
- Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Andrew J Sharp
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Supinda Bunyavanich
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY; Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
112
|
Abstract
PURPOSE OF REVIEW Allergic diseases represent a growing global health concern, especially among pediatric populations. Current strategies for the treatment of allergies and asthma focus on limiting the severity of the symptoms; however, additional research investigating the mechanisms promoting inflammation in the context of allergic reactions may lead to the development of more effective therapeutic strategies. RECENT FINDINGS Novel studies have highlighted the contributions of innate lymphocytes to the induction of inflammatory responses to allergens. Remarkably, neuron-derived signals, hormones, and even vitamins have been suggested to modulate the activity of innate lymphocytes, opening new windows of opportunity for the treatment of allergic inflammation. SUMMARY These studies highlight the complex interactions of the nervous, endocrine, and immune system that promote pathology in the context of allergic inflammation. Further studies are required to understand these interactions in order to aid in the development of novel and much-needed therapies to treat allergic conditions.
Collapse
|
113
|
Ghaedi M, Shen ZY, Orangi M, Martinez-Gonzalez I, Wei L, Lu X, Das A, Heravi-Moussavi A, Marra MA, Bhandoola A, Takei F. Single-cell analysis of RORα tracer mouse lung reveals ILC progenitors and effector ILC2 subsets. J Exp Med 2020; 217:e20182293. [PMID: 31816636 PMCID: PMC7062532 DOI: 10.1084/jem.20182293] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 07/12/2019] [Accepted: 10/30/2019] [Indexed: 12/16/2022] Open
Abstract
Lung group 2 innate lymphoid cells (ILC2s) drive allergic inflammation and promote tissue repair. ILC2 development is dependent on the transcription factor retinoic acid receptor-related orphan receptor (RORα), which is also expressed in common ILC progenitors. To elucidate the developmental pathways of lung ILC2s, we generated RORα lineage tracer mice and performed single-cell RNA sequencing, flow cytometry, and functional analyses. In adult mouse lungs, we found an IL-18Rα+ST2- population different from conventional IL-18Rα-ST2+ ILC2s. The former was GATA-3intTcf7EGFP+Kit+, produced few cytokines, and differentiated into multiple ILC lineages in vivo and in vitro. In neonatal mouse lungs, three ILC populations were identified, namely an ILC progenitor population similar to that in adult lungs and two distinct effector ILC2 subsets that differentially produced type 2 cytokines and amphiregulin. Lung ILC progenitors might actively contribute to ILC-poiesis in neonatal and inflamed adult lungs. In addition, neonatal lung ILC2s include distinct proinflammatory and tissue-repairing subsets.
Collapse
Affiliation(s)
- Maryam Ghaedi
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Terry Fox Laboratory, B.C. Cancer, Vancouver, British Columbia, Canada
| | - Zi Yi Shen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Terry Fox Laboratory, B.C. Cancer, Vancouver, British Columbia, Canada
| | - Mona Orangi
- Terry Fox Laboratory, B.C. Cancer, Vancouver, British Columbia, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, British Columbia, Canada
| | - Itziar Martinez-Gonzalez
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Terry Fox Laboratory, B.C. Cancer, Vancouver, British Columbia, Canada
| | - Lisa Wei
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Xiaoxiao Lu
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Arundhoti Das
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Alireza Heravi-Moussavi
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Marco A. Marra
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Avinash Bhandoola
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Fumio Takei
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Terry Fox Laboratory, B.C. Cancer, Vancouver, British Columbia, Canada
| |
Collapse
|
114
|
Agnello G, Alters SE, Rowlinson SW. Preclinical safety and antitumor activity of the arginine-degrading therapeutic enzyme pegzilarginase, a PEGylated, cobalt-substituted recombinant human arginase 1. Transl Res 2020; 217:11-22. [PMID: 31954097 DOI: 10.1016/j.trsl.2019.12.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 11/24/2022]
Abstract
Metabolic remodeling contributes to the development and progression of some cancers and exposes them to vulnerabilities, including specific nutrient dependencies that can be targeted therapeutically. Arginine is a semiessential amino acid, and several cancers are unable to endogenously synthesize sufficient levels of arginine for survival and proliferation, most commonly due to reduced expression of argininosuccinate synthase (ASS1). Such cancers are dependent on arginine and they can be targeted via enzyme-mediated depletion of systemic arginine. We report the preclinical safety, antitumor efficacy, and immune-potentiating effects of pegzilarginase, a highly potent human arginine-degrading enzyme. Toxicology studies showed that pegzilarginase-mediated arginine depletion is well tolerated at therapeutic levels that elicit an antitumor growth effect. To determine which tumor types are best suited for clinical development, we profiled clinical tumor samples for ASS1 expression, which correlated with pegzilarginase sensitivity in vivo in patient-derived xenograft (PDx) models. Among the histologies tested, malignant melanoma, small cell lung cancer and Merkel cell carcinoma had the highest prevalence of low ASS1 expression, the highest proportion of PDx models responding to pegzilarginase, and the strongest correlation between low or no ASS1 expression and sensitivity to pegzilarginase. In an immune-competent syngeneic mouse model, pegzilarginase slowed tumor growth and promoted the recruitment of CD8+ tumor infiltrating lymphocytes. This is consistent with the known autophagy-inducing effects of arginine depletion, and the link between autophagy and major histocompatibility complex antigen presentation to T cells. Our work supports the ongoing clinical investigations of pegzilarginase in solid tumors and clinical combination of pegzilarginase with immune checkpoint inhibitors.
Collapse
|
115
|
Lee JD, Kim HY, Kang K, Jeong HG, Song MK, Tae IH, Lee SH, Kim HR, Lee K, Chae S, Hwang D, Kim S, Kim HS, Kim KB, Lee BM. Integration of transcriptomics, proteomics and metabolomics identifies biomarkers for pulmonary injury by polyhexamethylene guanidine phosphate (PHMG-p), a humidifier disinfectant, in rats. Arch Toxicol 2020; 94:887-909. [PMID: 32080758 DOI: 10.1007/s00204-020-02657-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 02/03/2020] [Indexed: 12/16/2022]
|
116
|
ILC2s amplify PD-1 blockade by activating tissue-specific cancer immunity. Nature 2020; 579:130-135. [PMID: 32076273 PMCID: PMC7060130 DOI: 10.1038/s41586-020-2015-4] [Citation(s) in RCA: 237] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 12/31/2019] [Indexed: 12/14/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) regulate inflammation and immunity in mammalian tissues1,2. Although ILC2s are found in cancers of these tissues3, their roles in cancer immunity and immunotherapy are unclear. Here we show that ILC2s infiltrate pancreatic ductal adenocarcinomas (PDACs) to activate tissue-specific tumour immunity. Interleukin-33 (IL33) activates tumour ILC2s (TILC2s) and CD8+ T cells in orthotopic pancreatic tumours but not heterotopic skin tumours in mice to restrict pancreas-specific tumour growth. Resting and activated TILC2s express the inhibitory checkpoint receptor PD-1. Antibody-mediated PD-1 blockade relieves ILC2 cell-intrinsic PD-1 inhibition to expand TILC2s, augment anti-tumour immunity, and enhance tumour control, identifying activated TILC2s as targets of anti-PD-1 immunotherapy. Finally, both PD-1+ TILC2s and PD-1+ T cells are present in most human PDACs. Our results identify ILC2s as anti-cancer immune cells for PDAC immunotherapy. More broadly, ILC2s emerge as tissue-specific enhancers of cancer immunity that amplify the efficacy of anti-PD-1 immunotherapy. As ILC2s and T cells co-exist in human cancers and share stimulatory and inhibitory pathways, immunotherapeutic strategies to collectively target anti-cancer ILC2s and T cells may be broadly applicable.
Collapse
|
117
|
Living with Yourself: Innate Lymphoid Cell Immunometabolism. Cells 2020; 9:cells9020334. [PMID: 32024050 PMCID: PMC7072664 DOI: 10.3390/cells9020334] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 01/29/2020] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Innate lymphoid cells (ILCs) are tissue-resident sentinels of the immune system that function to protect local tissue microenvironments against pathogens and maintain homeostasis. However, because ILCs are sensitively tuned to perturbations within tissues, they can also contribute to host pathology when critical activating signals become dysregulated. Recent work has demonstrated that the crosstalk between ILCs and their environment has a significant impact on host metabolism in health and disease. In this review, we summarize studies that support evidence for the ability of ILCs to influence tissue and systemic metabolism, as well as how ILCs can be regulated by environmental changes in systemic host metabolism. We also highlight studies demonstrating how ILC- intrinsic metabolism influences their activation, proliferation, and homeostasis. Finally, this review discusses the challenges and open questions in the rapidly expanding field of ILCs and immunometabolism.
Collapse
|
118
|
Monticelli LA, Diamond JM, Saenz SA, Tait Wojno ED, Porteous MK, Cantu E, Artis D, Christie JD. Lung Innate Lymphoid Cell Composition Is Altered in Primary Graft Dysfunction. Am J Respir Crit Care Med 2020; 201:63-72. [PMID: 31394048 PMCID: PMC6938146 DOI: 10.1164/rccm.201906-1113oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/07/2019] [Indexed: 01/08/2023] Open
Abstract
Rationale: Primary graft dysfunction (PGD) is the leading cause of early morbidity and mortality after lung transplantation, but the immunologic mechanisms are poorly understood. Innate lymphoid cells (ILC) are a heterogeneous family of immune cells regulating pathologic inflammation and beneficial tissue repair. However, whether changes in donor-derived lung ILC populations are associated with PGD development has never been examined.Objectives: To determine whether PGD in chronic obstructive pulmonary disease or interstitial lung disease transplant recipients is associated with alterations in ILC subset composition within the allograft.Methods: We performed a single-center cohort study of lung transplantation patients with surgical biopsies of donor tissue taken before, and immediately after, allograft reperfusion. Donor immune cells from 18 patients were characterized phenotypically by flow cytometry for single-cell resolution of distinct ILC subsets. Changes in the percentage of ILC subsets with reperfusion or PGD (grade 3 within 72 h) were assessed.Measurements and Main Results: Allograft reperfusion resulted in significantly decreased frequencies of natural killer cells and a trend toward reduced ILC populations, regardless of diagnosis (interstitial lung disease or chronic obstructive pulmonary disease). Seven patients developed PGD (38.9%), and PGD development was associated with selective reduction of the ILC2 subset after reperfusion. Conversely, patients without PGD exhibited significantly higher ILC1 frequencies before reperfusion, accompanied by elevated ILC2 frequencies after allograft reperfusion.Conclusions: The composition of donor ILC subsets is altered after allograft reperfusion and is associated with PGD development, suggesting that ILCs may be involved in regulating lung injury in lung transplant recipients.
Collapse
Affiliation(s)
- Laurel A. Monticelli
- Division of Pulmonary and Critical Care Medicine and
- Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, New York; and
| | | | - Steven A. Saenz
- Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, New York; and
| | - Elia D. Tait Wojno
- Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, New York; and
| | | | - Edward Cantu
- Division of Cardiovascular Surgery, Center for Translational Lung Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Artis
- Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, New York; and
| | | |
Collapse
|
119
|
Brys AK, Rodriguez-Homs LG, Suwanpradid J, Atwater AR, MacLeod AS. Shifting Paradigms in Allergic Contact Dermatitis: The Role of Innate Immunity. J Invest Dermatol 2020; 140:21-28. [PMID: 31101475 PMCID: PMC6854274 DOI: 10.1016/j.jid.2019.03.1133] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/07/2019] [Accepted: 03/07/2019] [Indexed: 12/30/2022]
Abstract
The role of the innate immune system in allergic contact dermatitis (ACD) has traditionally been confined to the initial antigen sensitization phase. However, more recent findings have shown the role of innate immunity in additional aspects of ACD, including the effector phase of the classic type IV hypersensitivity reaction. As a result, the precise immunologic mechanisms mediating ACD are more complex than previously believed. The aim of this review is to provide insight into recent advances in understanding the role of the innate immune system in the pathogenesis of ACD, including novel mechanistic roles for macrophages, innate lymphoid cells, natural killer cells, innate γδ T cells, and other signaling molecules. These insights provide new opportunities for therapeutic intervention in ACD.
Collapse
Affiliation(s)
- Adam K Brys
- Duke University Medical Center, Department of Dermatology, Duke University Medical Center, DUMC 3135, Durham, North Carolina, USA
| | - Larissa G Rodriguez-Homs
- Duke University Medical Center, Department of Dermatology, Duke University Medical Center, DUMC 3135, Durham, North Carolina, USA
| | - Jutamas Suwanpradid
- Duke University Medical Center, Department of Dermatology, Duke University Medical Center, DUMC 3135, Durham, North Carolina, USA
| | - Amber Reck Atwater
- Duke University Medical Center, Department of Dermatology, Duke University Medical Center, DUMC 3135, Durham, North Carolina, USA
| | - Amanda S MacLeod
- Duke University Medical Center, Department of Dermatology, Duke University Medical Center, DUMC 3135, Durham, North Carolina, USA.
| |
Collapse
|
120
|
Lowe MM, Boothby I, Clancy S, Ahn RS, Liao W, Nguyen DN, Schumann K, Marson A, Mahuron KM, Kingsbury GA, Liu Z, Munoz Sandoval P, Rodriguez RS, Pauli ML, Taravati K, Arron ST, Neuhaus IM, Harris HW, Kim EA, Shin US, Krummel MF, Daud A, Scharschmidt TC, Rosenblum MD. Regulatory T cells use arginase 2 to enhance their metabolic fitness in tissues. JCI Insight 2019; 4:129756. [PMID: 31852848 DOI: 10.1172/jci.insight.129756] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 11/13/2019] [Indexed: 12/31/2022] Open
Abstract
Distinct subsets of Tregs reside in nonlymphoid tissues where they mediate unique functions. To interrogate the biology of tissue Tregs in human health and disease, we phenotypically and functionally compared healthy skin Tregs with those in peripheral blood, inflamed psoriatic skin, and metastatic melanoma. The mitochondrial enzyme, arginase 2 (ARG2), was preferentially expressed in Tregs in healthy skin, increased in Tregs in metastatic melanoma, and reduced in Tregs from psoriatic skin. ARG2 enhanced Treg suppressive capacity in vitro and conferred a selective advantage for accumulation in inflamed tissues in vivo. CRISPR-mediated deletion of this gene in primary human Tregs was sufficient to skew away from a tissue Treg transcriptional signature. Notably, the inhibition of ARG2 increased mTOR signaling, whereas the overexpression of this enzyme suppressed it. Taken together, our results suggest that Tregs express ARG2 in human tissues to both regulate inflammation and enhance their metabolic fitness.
Collapse
Affiliation(s)
| | - Ian Boothby
- Department of Dermatology.,Medical Scientist Training Program
| | | | | | | | | | | | | | | | | | - Zheng Liu
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | | | | | | | | | | | | | | | - Esther A Kim
- Department of Surgery, UCSF, San Francisco, California, USA
| | - Uk Sok Shin
- Department of Surgery, UCSF, San Francisco, California, USA
| | | | | | | | | |
Collapse
|
121
|
Contribution of ROS and metabolic status to neonatal and adult CD8+ T cell activation. PLoS One 2019; 14:e0226388. [PMID: 31841528 PMCID: PMC6913967 DOI: 10.1371/journal.pone.0226388] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022] Open
Abstract
In neonatal T cells, a low response to infection contributes to a high incidence of morbidity and mortality of neonates. Here we have evaluated the impact of the cytoplasmic and mitochondrial levels of Reactive Oxygen Species of adult and neonatal CD8+ T cells on their activation potential. We have also constructed a logical model connecting metabolism and ROS with T cell signaling. Our model indicates the interplay between antigen recognition, ROS and metabolic status in T cell responses. This model displays alternative stable states corresponding to different cell fates, i.e. quiescent, activated and anergic states, depending on ROS levels. Stochastic simulations with this model further indicate that differences in ROS status at the cell population level contribute to the lower activation rate of neonatal, compared to adult, CD8+ T cells upon TCR engagement. These results are relevant for neonatal health care. Our model can serve to analyze the impact of metabolic shift during cancer in which, similar to neonatal cells, a high glycolytic rate and low concentrations of glutamine and arginine promote tumor tolerance.
Collapse
|
122
|
Wang R, Geller DA, Wink DA, Cheng B, Billiar TR. NO and hepatocellular cancer. Br J Pharmacol 2019; 177:5459-5466. [PMID: 31423564 PMCID: PMC7707086 DOI: 10.1111/bph.14838] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/26/2019] [Accepted: 07/01/2019] [Indexed: 12/27/2022] Open
Abstract
NO has broad and sometimes dichotomous roles in cancer. The effects of NO in tumours depend on the type and localization of NOS isoforms, concentration and duration of NO exposure, and cellular sensitivity to NO. Hepatocellular carcinoma (HCC) is a common and lethal disease for which no effective therapy other than surgical resection exists. Over two decades of research has yielded evidence that NO generated by the inducible NOS (iNOS or NOS2) contributes to HCC progression in at least a subset of patients with HCC. The co-expression of iNOS with COX-2 may portend a particularly aggressive cancer phenotype in HCC and at the same time reveal an opportunity for pharmacological intervention. In this review, we focus on what is known about the influence of NO in HCC neoplastic transformation, proliferation and apoptosis, angiogenesis, invasion, and metastasis, cancer stem cells, and the host immune response against the tumour. We discuss the implications of recent findings for targeting the NO pathways in HCC.
Collapse
Affiliation(s)
- Ronghua Wang
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David A Geller
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David A Wink
- Cancer Inflammation Program, NCI/NIH, Frederick, MD, USA
| | - Bin Cheng
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
123
|
Ziegler JF, Böttcher C, Letizia M, Yerinde C, Wu H, Freise I, Rodriguez-Sillke Y, Stoyanova AK, Kreis ME, Asbach P, Kunkel D, Priller J, Anagnostopoulos I, Kühl AA, Miehle K, Stumvoll M, Tran F, Fredrich B, Forster M, Franke A, Bojarski C, Glauben R, Löscher BS, Siegmund B, Weidinger C. Leptin induces TNFα-dependent inflammation in acquired generalized lipodystrophy and combined Crohn's disease. Nat Commun 2019; 10:5629. [PMID: 31822667 PMCID: PMC6904732 DOI: 10.1038/s41467-019-13559-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/14/2019] [Indexed: 12/27/2022] Open
Abstract
Leptin has been shown to modulate intestinal inflammation in mice. However, clinical evidence regarding its immune-stimulatory potential in human Crohn’s disease remains sparse. We here describe a patient with the unique combination of acquired generalized lipodystrophy and Crohn’s disease (AGLCD) featuring a lack of adipose tissue, leptin deficiency and intestinal inflammation. Using mass and flow cytometry, immunohistochemistry and functional metabolic analyses, the AGLCD patient was compared to healthy individuals and Crohn’s disease patients regarding immune cell composition, function and metabolism and the effects of recombinant N-methionylleptin (rLeptin) were evaluated. We provide evidence that rLeptin exerts diverse pro-inflammatory effects on immune cell differentiation and function, including the metabolic reprogramming of immune cells and the induction of TNFα, ultimately aggravating Crohn’s disease in the AGLCD patient, which can be reversed by anti-TNFα therapy. Our results indicate that leptin is required for human immune homeostasis and contributes to autoimmunity in a TNFα-dependent manner. The adipokine leptin modulates intestinal inflammation in mice. Here the authors describe a patient with inflammatory bowel disease and lipodystrophy, providing evidence that leptin aggravates intestinal inflammation with proinflammatory effects on leukocytes that are reversible by TNFα blockade.
Collapse
Affiliation(s)
- Jörn F Ziegler
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Berlin, Germany
| | - Chotima Böttcher
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Laboratory of Molecular Psychiatry and Department of Neuropsychiatry, Berlin, Germany
| | - Marilena Letizia
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Berlin, Germany
| | - Cansu Yerinde
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Berlin, Germany
| | - Hao Wu
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Berlin, Germany
| | - Inka Freise
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Berlin, Germany
| | - Yasmina Rodriguez-Sillke
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Berlin, Germany
| | - Ani K Stoyanova
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Department of Visceral Surgery, Campus Benjamin Franklin, Berlin, Germany
| | - Martin E Kreis
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Department of Visceral Surgery, Campus Benjamin Franklin, Berlin, Germany
| | - Patrick Asbach
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Department of Radiology, Campus Benjamin Franklin, Berlin, Germany
| | - Desiree Kunkel
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,BIH Cytometry Core, Berlin Institute of Health, 10178, Berlin, Germany
| | - Josef Priller
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Laboratory of Molecular Psychiatry and Department of Neuropsychiatry, Berlin, Germany.,BIH Berlin, DZNE Berlin and University of Edinburgh and UK DRI, Edinburgh, UK
| | - Ioannis Anagnostopoulos
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Department of Pathology, Campus Charité Mitte, Berlin, Germany
| | - Anja A Kühl
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,iPATH.Berlin-Immunopathology for Experimental Models, Core Facility of the Charité, Berlin, Germany
| | - Konstanze Miehle
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Michael Stumvoll
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Florian Tran
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Broder Fredrich
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Michael Forster
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Christian Bojarski
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Berlin, Germany
| | - Rainer Glauben
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Berlin, Germany
| | - Britt-Sabina Löscher
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Britta Siegmund
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany. .,Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Berlin, Germany.
| | - Carl Weidinger
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany. .,Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Berlin, Germany. .,Clinician Scientist Program, Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
124
|
Mantani PT, Dunér P, Ljungcrantz I, Nilsson J, Björkbacka H, Fredrikson GN. ILC2 transfers to apolipoprotein E deficient mice reduce the lipid content of atherosclerotic lesions. BMC Immunol 2019; 20:47. [PMID: 31823769 PMCID: PMC6905041 DOI: 10.1186/s12865-019-0330-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 12/02/2019] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Expansion of type 2 innate lymphoid cells (ILC2s) in hypercholesterolaemic mice protects against atherosclerosis while different ILC2 subsets have been described (natural, inflammatory) based on their suppression of tumorigenicity 2 (ST2) and killer-cell lectin like receptor G1 (KLRG1) expression. The aim of the current study is to characterize the interleukin 25 (IL25)-induced splenic ILC2 population (Lin-CD45+IL17RB+ICOS+IL7raintermediate) and address its direct role in experimental atherosclerosis by its adoptive transfer to hypercholesterolaemic apolipoprotein E deficient (apoE-/-) mice. RESULTS Immunomagnetically enriched, FACS-sorted ILC2s from the spleens of IL-25 treated apoE-/- mice were stained for KLRG1 and ST2 directly upon cell obtainment or in vitro cell expansion for flow cytometric analysis. IL25-induced splenic ILC2s express high levels of both KLRG1 and ST2. However, both markers are downregulated upon in vitro cell expansion. In vitro expanded splenic ILC2s were intraperitoneally transferred to apoE-/- recipients on high fat diet. ApoE-/- mice that received in vitro expanded splenic ILC2s had decreased lipid content in subvalvular heart and brachiocephalic artery (BCA) plaques accompanied by increased peritoneal B1 cells, activated eosinophils and alternatively activated macrophages (AAMs) as well as anti-phosphorylcholine (PC) immunoglobulin (Ig) M in plasma. CONCLUSIONS With the current data we designate the IL25-induced ILC2 population to decrease the lipid content of atherosclerotic lesions in apoE-/- mice and we directly link the induction of B1 cells and the atheroprotective anti-PC IgM antibodies with ILC2s.
Collapse
Affiliation(s)
- Polyxeni T Mantani
- Department of Clinical Sciences, Skåne University Hospital Malmö, CRC, Building 91:12, Jan Waldenströms gata 35, 20502, Malmö, Sweden. .,Lund University, Lund, Sweden.
| | - Pontus Dunér
- Department of Clinical Sciences, Skåne University Hospital Malmö, CRC, Building 91:12, Jan Waldenströms gata 35, 20502, Malmö, Sweden.,Lund University, Lund, Sweden
| | - Irena Ljungcrantz
- Department of Clinical Sciences, Skåne University Hospital Malmö, CRC, Building 91:12, Jan Waldenströms gata 35, 20502, Malmö, Sweden.,Lund University, Lund, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences, Skåne University Hospital Malmö, CRC, Building 91:12, Jan Waldenströms gata 35, 20502, Malmö, Sweden.,Lund University, Lund, Sweden
| | - Harry Björkbacka
- Department of Clinical Sciences, Skåne University Hospital Malmö, CRC, Building 91:12, Jan Waldenströms gata 35, 20502, Malmö, Sweden.,Lund University, Lund, Sweden
| | - Gunilla Nordin Fredrikson
- Department of Clinical Sciences, Skåne University Hospital Malmö, CRC, Building 91:12, Jan Waldenströms gata 35, 20502, Malmö, Sweden. .,Lund University, Lund, Sweden.
| |
Collapse
|
125
|
Ercolano G, Falquet M, Vanoni G, Trabanelli S, Jandus C. ILC2s: New Actors in Tumor Immunity. Front Immunol 2019; 10:2801. [PMID: 31849977 PMCID: PMC6902088 DOI: 10.3389/fimmu.2019.02801] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022] Open
Abstract
Innate lymphoid cells (ILCs) represent the most recently identified family of innate lymphocytes that act as first responders, maintaining tissue homeostasis and protecting epithelial barriers. In the last few years, group 2 ILCs (ILC2s) have emerged as key regulators in several immunological processes such as asthma and allergy. Whilst ILC2s are currently being evaluated as novel targets for immunotherapy in these diseases, their involvement in tumor immunity has only recently begun to be deciphered. Here, we provide a comprehensive overview of the pleiotropic roles of ILC2s in different tumor settings. Furthermore, we discuss how different therapeutic approaches targeting ILC2s could improve the efficacy of current tumor immunotherapies.
Collapse
Affiliation(s)
- Giuseppe Ercolano
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Maryline Falquet
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Giulia Vanoni
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Sara Trabanelli
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Camilla Jandus
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
126
|
Michaeloudes C, Bhavsar PK, Mumby S, Xu B, Hui CKM, Chung KF, Adcock IM. Role of Metabolic Reprogramming in Pulmonary Innate Immunity and Its Impact on Lung Diseases. J Innate Immun 2019; 12:31-46. [PMID: 31786568 DOI: 10.1159/000504344] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022] Open
Abstract
Lung innate immunity is the first line of defence against inhaled allergens, pathogens and environmental pollutants. Cellular metabolism plays a key role in innate immunity. Catabolic pathways, including glycolysis and fatty acid oxidation (FAO), are interconnected with biosynthetic and redox pathways. Innate immune cell activation and differentiation trigger extensive metabolic changes that are required to support their function. Pro-inflammatory polarisation of macrophages and activation of dendritic cells, mast cells and neutrophils are associated with increased glycolysis and a shift towards the pentose phosphate pathway and fatty acid synthesis. These changes provide the macromolecules required for proliferation and inflammatory mediator production and reactive oxygen species for anti-microbial effects. Conversely, anti-inflammatory macrophages use primarily FAO and oxidative phosphorylation to ensure efficient energy production and redox balance required for prolonged survival. Deregulation of metabolic reprogramming in lung diseases, such as asthma and chronic obstructive pulmonary disease, may contribute to impaired innate immune cell function. Understanding how innate immune cell metabolism is altered in lung disease may lead to identification of new therapeutic targets. This is important as drugs targeting a number of metabolic pathways are already in clinical development for the treatment of other diseases such as cancer.
Collapse
Affiliation(s)
- Charalambos Michaeloudes
- Experimental Studies and Cell and Molecular Biology, Airway Disease Section, National Heart and Lung Institute, Imperial College London and Biomedical Research Unit, Royal Brompton Hospital, London, United Kingdom,
| | - Pankaj K Bhavsar
- Experimental Studies and Cell and Molecular Biology, Airway Disease Section, National Heart and Lung Institute, Imperial College London and Biomedical Research Unit, Royal Brompton Hospital, London, United Kingdom
| | - Sharon Mumby
- Experimental Studies and Cell and Molecular Biology, Airway Disease Section, National Heart and Lung Institute, Imperial College London and Biomedical Research Unit, Royal Brompton Hospital, London, United Kingdom
| | - Bingling Xu
- Respiratory and Critical Care Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Christopher Kim Ming Hui
- Respiratory and Critical Care Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Kian Fan Chung
- Experimental Studies and Cell and Molecular Biology, Airway Disease Section, National Heart and Lung Institute, Imperial College London and Biomedical Research Unit, Royal Brompton Hospital, London, United Kingdom
| | - Ian M Adcock
- Experimental Studies and Cell and Molecular Biology, Airway Disease Section, National Heart and Lung Institute, Imperial College London and Biomedical Research Unit, Royal Brompton Hospital, London, United Kingdom
| |
Collapse
|
127
|
Abstract
Arginine metabolism is linked to several important metabolic processes, and reprogramming of arginine metabolism occurs in various physiological and pathological conditions. Here we describe a method, using a LC-MS-based metabolomics and 15N4-arginine tracing approach, to quantitatively analyze arginine metabolism. This method can reliably quantify the abundance of important intermediates and fluxes of major metabolic reactions in arginine metabolism in a variety of cultured mammalian cell models.
Collapse
Affiliation(s)
- Gretchen L Seim
- Morgridge Institute for Research, Madison, WI, USA
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Emily C Britt
- Morgridge Institute for Research, Madison, WI, USA
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jing Fan
- Morgridge Institute for Research, Madison, WI, USA.
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
128
|
Huang X, Li Y, Guo X, Zhu Z, Kong X, Yu F, Wang Q. Identification of differentially expressed genes and signaling pathways in chronic obstructive pulmonary disease via bioinformatic analysis. FEBS Open Bio 2019; 9:1880-1899. [PMID: 31419078 PMCID: PMC6823288 DOI: 10.1002/2211-5463.12719] [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: 06/05/2019] [Revised: 08/07/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a multifactorial and heterogeneous disease that creates public health challenges worldwide. The underlying molecular mechanisms of COPD are not entirely clear. In this study, we aimed to identify the critical genes and potential molecular mechanisms of COPD by bioinformatic analysis. The gene expression profiles of lung tissues of COPD cases and healthy control subjects were obtained from the Gene Expression Omnibus. Differentially expressed genes were analyzed by integration with annotations from Gene Ontology and Kyoto Encyclopedia of Genes and Genomes, followed by construction of a protein‐protein interaction network and weighted gene coexpression analysis. We identified 139 differentially expressed genes associated with the progression of COPD, among which 14 Hub genes were identified and found to be enriched in certain categories, including immune and inflammatory response, response to lipopolysaccharide and receptor for advanced glycation end products binding; in addition, these Hub genes are involved in multiple signaling pathways, particularly hematopoietic cell lineage and cytokine‐cytokine receptor interaction. The 14 Hub genes were positively or negatively associated with COPD by wgcna analysis. The genes CX3CR1,PTGS2,FPR1,FPR2, S100A12,EGR1,CD163, S100A8 and S100A9 were identified to mediate inflammation and injury of the lung, and play critical roles in the pathogenesis of COPD. These findings improve our understanding of the underlying molecular mechanisms of COPD.
Collapse
Affiliation(s)
- Xinwei Huang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, China.,Medical School, Kunming University of Science and Technology, China
| | - Yunwei Li
- Medical School, Kunming University of Science and Technology, China.,Department of Pharmacy, Kunming Children's Hospital, China
| | - Xiaoran Guo
- Medical School, Kunming University of Science and Technology, China
| | - Zongxin Zhu
- Medical School, Kunming University of Science and Technology, China
| | - Xiangyang Kong
- Medical School, Kunming University of Science and Technology, China
| | - Fubing Yu
- Department of Gastroenterology, Fourth Affiliated Hospital of Kunming Medical University, China
| | - Qiang Wang
- Physical Examination Center, Second People's Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
129
|
Lewis G, Wang B, Shafiei Jahani P, Hurrell BP, Banie H, Aleman Muench GR, Maazi H, Helou DG, Howard E, Galle-Treger L, Lo R, Santosh S, Baltus A, Bongers G, San-Mateo L, Gilliland FD, Rehan VK, Soroosh P, Akbari O. Dietary Fiber-Induced Microbial Short Chain Fatty Acids Suppress ILC2-Dependent Airway Inflammation. Front Immunol 2019; 10:2051. [PMID: 31620118 PMCID: PMC6760365 DOI: 10.3389/fimmu.2019.02051] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/14/2019] [Indexed: 12/21/2022] Open
Abstract
Group 2 Innate lymphoid cells (ILC2) contribute significantly to allergic inflammation. However, the role of microbiota on ILC2s remains to be unraveled. Here we show that short chain fatty acids (SCFAs), such as butyrate, derived from fermentation of dietary fibers by the gut microbiota inhibit pulmonary ILC2 functions and subsequent development of airway hyperreactivity (AHR). We further show that SCFAs modulate GATA3, oxidative phosphorylation, and glycolytic metabolic pathways in pulmonary ILC2s. The observed phenotype is associated with increased IL-17a secretion by lung ILC2s and linked to enhanced neutrophil recruitment to the airways. Finally, we show that butyrate-producing gut bacteria in germ-free mice effectively suppress ILC2-driven AHR. Collectively, our results demonstrate a previously unrecognized role for microbial-derived SCFAs on pulmonary ILC2s in the context of AHR. The data suggest strategies aimed at modulating metabolomics and microbiota in the gut, not only to treat, but to prevent lung inflammation and asthma.
Collapse
Affiliation(s)
- Gavin Lewis
- Janssen Research and Development, San Diego, CA, United States
| | - Bowen Wang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Pedram Shafiei Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Benjamin P. Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Homayon Banie
- Janssen Research and Development, San Diego, CA, United States
| | | | - Hadi Maazi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Doumet Georges Helou
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Emily Howard
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Lauriane Galle-Treger
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Richard Lo
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Swetha Santosh
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Andrew Baltus
- Janssen Research and Development, Spring House, PA, United States
| | - Gerrold Bongers
- Janssen Research and Development, Spring House, PA, United States
| | - Lani San-Mateo
- Janssen Research and Development, Spring House, PA, United States
| | - Frank D. Gilliland
- Division of Environmental Health, Department of Preventive Medicine, University of Southern California, Los Angeles, CA, United States
| | - Virender K. Rehan
- Division of Neonatology, Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Pejman Soroosh
- Janssen Research and Development, San Diego, CA, United States
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
130
|
Riera-Domingo C, Audigé A, Granja S, Cheng WC, Ho PC, Baltazar F, Stockmann C, Mazzone M. Immunity, Hypoxia, and Metabolism-the Ménage à Trois of Cancer: Implications for Immunotherapy. Physiol Rev 2019; 100:1-102. [PMID: 31414610 DOI: 10.1152/physrev.00018.2019] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
It is generally accepted that metabolism is able to shape the immune response. Only recently we are gaining awareness that the metabolic crosstalk between different tumor compartments strongly contributes to the harsh tumor microenvironment (TME) and ultimately impairs immune cell fitness and effector functions. The major aims of this review are to provide an overview on the immune system in cancer; to position oxygen shortage and metabolic competition as the ground of a restrictive TME and as important players in the anti-tumor immune response; to define how immunotherapies affect hypoxia/oxygen delivery and the metabolic landscape of the tumor; and vice versa, how oxygen and metabolites within the TME impinge on the success of immunotherapies. By analyzing preclinical and clinical endeavors, we will discuss how a metabolic characterization of the TME can identify novel targets and signatures that could be exploited in combination with standard immunotherapies and can help to predict the benefit of new and traditional immunotherapeutic drugs.
Collapse
Affiliation(s)
- Carla Riera-Domingo
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Annette Audigé
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Sara Granja
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Wan-Chen Cheng
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Ping-Chih Ho
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Fátima Baltazar
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Christian Stockmann
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium; Institute of Anatomy, University of Zurich, Zurich, Switzerland; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland; and Ludwig Cancer Research Institute, Epalinges, Switzerland
| |
Collapse
|
131
|
Puleston DJ, Buck MD, Klein Geltink RI, Kyle RL, Caputa G, O'Sullivan D, Cameron AM, Castoldi A, Musa Y, Kabat AM, Zhang Y, Flachsmann LJ, Field CS, Patterson AE, Scherer S, Alfei F, Baixauli F, Austin SK, Kelly B, Matsushita M, Curtis JD, Grzes KM, Villa M, Corrado M, Sanin DE, Qiu J, Pällman N, Paz K, Maccari ME, Blazar BR, Mittler G, Buescher JM, Zehn D, Rospert S, Pearce EJ, Balabanov S, Pearce EL. Polyamines and eIF5A Hypusination Modulate Mitochondrial Respiration and Macrophage Activation. Cell Metab 2019; 30:352-363.e8. [PMID: 31130465 PMCID: PMC6688828 DOI: 10.1016/j.cmet.2019.05.003] [Citation(s) in RCA: 231] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/05/2019] [Accepted: 04/30/2019] [Indexed: 12/26/2022]
Abstract
How cells adapt metabolism to meet demands is an active area of interest across biology. Among a broad range of functions, the polyamine spermidine is needed to hypusinate the translation factor eukaryotic initiation factor 5A (eIF5A). We show here that hypusinated eIF5A (eIF5AH) promotes the efficient expression of a subset of mitochondrial proteins involved in the TCA cycle and oxidative phosphorylation (OXPHOS). Several of these proteins have mitochondrial targeting sequences (MTSs) that in part confer an increased dependency on eIF5AH. In macrophages, metabolic switching between OXPHOS and glycolysis supports divergent functional fates stimulated by activation signals. In these cells, hypusination of eIF5A appears to be dynamically regulated after activation. Using in vivo and in vitro models, we show that acute inhibition of this pathway blunts OXPHOS-dependent alternative activation, while leaving aerobic glycolysis-dependent classical activation intact. These results might have implications for therapeutically controlling macrophage activation by targeting the polyamine-eIF5A-hypusine axis.
Collapse
Affiliation(s)
- Daniel J Puleston
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Michael D Buck
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | | | - Ryan L Kyle
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - George Caputa
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - David O'Sullivan
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Alanna M Cameron
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Angela Castoldi
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Yaarub Musa
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Agnieszka M Kabat
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Ying Zhang
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, and BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg 79104, Germany
| | - Lea J Flachsmann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Cameron S Field
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Annette E Patterson
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Stefanie Scherer
- Department of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany
| | - Francesca Alfei
- Department of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany
| | - Francesc Baixauli
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - S Kyle Austin
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Beth Kelly
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Mai Matsushita
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Jonathan D Curtis
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Katarzyna M Grzes
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Matteo Villa
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Mauro Corrado
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - David E Sanin
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Jing Qiu
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Nora Pällman
- Division of Haematology, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland
| | - Katelyn Paz
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Maria Elena Maccari
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Center for Pediatrics, and Faculty of Medicine, Medical Center - University of Freiburg, Freiburg 79106, Germany
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Gerhard Mittler
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Joerg M Buescher
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Dietmar Zehn
- Department of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany
| | - Sabine Rospert
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, and BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg 79104, Germany
| | - Edward J Pearce
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Faculty of Biology, University of Freiburg, Freiburg 79104, Germany
| | - Stefan Balabanov
- Division of Haematology, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland
| | - Erika L Pearce
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany.
| |
Collapse
|
132
|
Functional interactions between innate lymphoid cells and adaptive immunity. Nat Rev Immunol 2019; 19:599-613. [PMID: 31350531 PMCID: PMC6982279 DOI: 10.1038/s41577-019-0194-8] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 12/19/2022]
Abstract
Innate lymphoid cells (ILCs) are enriched at barrier surfaces of the mammalian body where they rapidly respond to host, microbial or environmental stimuli to promote immunity or tissue homeostasis. Furthermore, ILCs are dysregulated in multiple human diseases. Over the past decade, substantial advances have been made in identifying the heterogeneity and functional diversity of ILCs, which have revealed striking similarities to T cell subsets. However, emerging evidence indicates that ILCs also have a complex role in directly influencing the adaptive immune response in the context of development, homeostasis, infection or inflammation. In turn, adaptive immunity reciprocally regulates ILCs, which indicates that these interactions are a crucial determinant of immune responses within tissues. Here, we summarize our current understanding of functional interactions between ILCs and the adaptive immune system, discuss limitations and future areas of investigation, and consider the potential for these interactions to be therapeutically harnessed to benefit human health.
Collapse
|
133
|
Borger JG, Lau M, Hibbs ML. The Influence of Innate Lymphoid Cells and Unconventional T Cells in Chronic Inflammatory Lung Disease. Front Immunol 2019; 10:1597. [PMID: 31354734 PMCID: PMC6637857 DOI: 10.3389/fimmu.2019.01597] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/26/2019] [Indexed: 12/11/2022] Open
Abstract
The lungs are continuously subjected to environmental insults making them susceptible to infection and injury. They are protected by the respiratory epithelium, which not only serves as a physical barrier but also a reactive one that can release cytokines, chemokines, and other defense proteins in response to danger signals, and can undergo conversion to protective mucus-producing goblet cells. The lungs are also guarded by a complex network of highly specialized immune cells and their mediators to support tissue homeostasis and resolve integrity deviation. This review focuses on specialized innate-like lymphocytes present in the lung that act as key sensors of lung insults and direct the pulmonary immune response. Included amongst these tissue-resident lymphocytes are innate lymphoid cells (ILCs), which are classified into five distinct subsets (natural killer, ILC1, ILC2, ILC3, lymphoid tissue-inducer cells), and unconventional T cells including natural killer T (NKT) cells, mucosal-associated invariant T (MAIT) cells, and γδ-T cells. While ILCs and unconventional T cells together comprise only a small proportion of the total immune cells in the lung, they have been found to promote lung homeostasis and are emerging as contributors to a variety of chronic lung diseases including pulmonary fibrosis, allergic airway inflammation, and chronic obstructive pulmonary disease (COPD). A particularly intriguing trait of ILCs that has recently emerged is their plasticity and ability to alter their gene expression profiles and adapt their function in response to environmental cues. The malleable nature of these cells may aid in rapid responses to pathogen but may also have downstream pathological consequences. The role of ILC2s in Th2 allergic airway responses is becoming apparent but the contribution of other ILCs and unconventional T cells during chronic lung inflammation is poorly described. This review presents an overview of our current understanding of the involvement of ILCs and unconventional T cells in chronic pulmonary diseases.
Collapse
Affiliation(s)
- Jessica G Borger
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Maverick Lau
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, Lung Health Research Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Margaret L Hibbs
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
134
|
Xiong J, Wang H, He J, Wang Q. Functions of Group 2 Innate Lymphoid Cells in Tumor Microenvironment. Front Immunol 2019; 10:1615. [PMID: 31354745 PMCID: PMC6635601 DOI: 10.3389/fimmu.2019.01615] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/28/2019] [Indexed: 12/13/2022] Open
Abstract
Innate lymphoid cells (ILCs), defined as a heterogeneous population of lymphocytes, have received much attention over recent years. They can be categorized into three subsets according to the expression profiles of transcription factors and differing levels of cytokine production. These cells are widely distributed in human organs and tissues, especially in mucosal tissue. The ILCs are involved in various physiological and pathological processes, including inflammation, worm expulsion, autoimmune disease and tumor progression, many of which have been investigated and clarified in recent studies. In the tumor microenvironment, group 2 innate lymphoid cells (ILC2s) have been proved to be able to either promote or inhibit tumor progression by producing different cytokines, recruiting diverse types of immune cells, expressing immunosuppressive molecules and by regulating the expression of certain inflammatory factors. This review summarizes recent research progress on the immunomodulatory functions of ILC2s in the tumor microenvironment and puts forward some perspectives for future study.
Collapse
Affiliation(s)
- Jia Xiong
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Haofei Wang
- Department of Pharmacology, China Medical University School of Pharmacy, Shenyang, China
| | - Jia He
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
135
|
Krishnan S, Lawrence CB. Old Dog New Tricks; Revisiting How Stroke Modulates the Systemic Immune Landscape. Front Neurol 2019; 10:718. [PMID: 31312180 PMCID: PMC6614437 DOI: 10.3389/fneur.2019.00718] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/18/2019] [Indexed: 12/27/2022] Open
Abstract
Infections in the post-acute phase of cerebral ischaemia impede optimal recovery by exacerbating morbidity and mortality. Our review aims to reconcile the increased infection susceptibility of patients post-stroke by consolidating our understanding of compartmentalised alterations to systemic immunity. Mounting evidence has catalogued alterations to numerous immune cell populations but an understanding of the mechanisms of long-range communication between the immune system, nervous system and other organs beyond the involvement of autonomic signalling is lacking. By taking our cues from established and emerging concepts of neuro-immune interactions, immune-mediated inter-organ cross-talk, innate immune training and the role of microbiota-derived signals in central nervous system (CNS) function we will explore mechanisms of how cerebral ischaemia could shape systemic immune function. In this context, we will also discuss a key question: how are immune requirements critical for mediating repair of the ischaemic insult balanced by the need for anti-microbial immunity post-stroke, given that they are mediated by mutually exclusive immune networks? Our reformed understanding of the immune landscape post-stroke and novel mechanisms at play could guide targeted therapeutic interventions and initiate a step-change in the clinical management of these infectious complications post-stroke.
Collapse
Affiliation(s)
- Siddharth Krishnan
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.,Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom.,Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Catherine B Lawrence
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
136
|
Abstract
The lungs, a special site that is frequently challenged by tumors, pathogens and other environmental insults, are populated by large numbers of innate immune cells. Among these, natural killer (NK) cells are gaining increasing attention. Recent studies have revealed that NK cells are heterogeneous populations consisting of distinct subpopulations with diverse characteristics, some of which are determined by their local tissue microenvironment. Most current information about NK cells comes from studies of NK cells from the peripheral blood of humans and NK cells from the spleen and bone marrow of mice. However, the functions and phenotypes of lung NK cells differ from those of NK cells in other tissues. Here, we provide an overview of human and mouse lung NK cells in the context of homeostasis, pathogenic infections, asthma, chronic obstructive pulmonary disease (COPD) and lung cancer, mainly focusing on their phenotype, function, frequency, and their potential role in pathogenesis or immune defense. A comprehensive understanding of the biology of NK cells in the lungs will aid the development of NK cell-based immunotherapies for the treatment of lung diseases.
Collapse
Affiliation(s)
- Jingjing Cong
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Molecular Medicine, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Institue of Immunology, University of Science and Technology of China, Hefei, China
- Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Molecular Medicine, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Institue of Immunology, University of Science and Technology of China, Hefei, China
| |
Collapse
|
137
|
Stehle C, Hernández DC, Romagnani C. Innate lymphoid cells in lung infection and immunity. Immunol Rev 2019; 286:102-119. [PMID: 30294964 DOI: 10.1111/imr.12712] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/24/2018] [Indexed: 12/30/2022]
Abstract
In recent years, innate lymphoid cells (ILCs) have emerged as key mediators of protection and repair of mucosal surfaces during infection. The lung, a dynamic mucosal tissue that is exposed to a plethora of microbes, is a playground for respiratory infection-causing pathogens which are not only a major cause of fatalities worldwide, but are also associated with comorbidities and decreased quality of life. The lung provides a rich microenvironment to study ILCs in the context of innate protection mechanisms within the airways, unraveling their distinct functions not only in health but also in disease. In this review, we discuss how pulmonary ILCs play a role in protection against viral, parasitic, bacterial, and fungal challenge, along with the mechanisms underlying this ILC-mediated immunity.
Collapse
Affiliation(s)
- Christina Stehle
- Innate Immunity, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | | | - Chiara Romagnani
- Innate Immunity, Deutsches Rheuma-Forschungszentrum, Berlin, Germany.,Medical Department I, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
138
|
Wallrapp A, Riesenfeld SJ, Burkett PR, Kuchroo VK. Type 2 innate lymphoid cells in the induction and resolution of tissue inflammation. Immunol Rev 2019; 286:53-73. [PMID: 30294962 DOI: 10.1111/imr.12702] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/08/2018] [Indexed: 12/14/2022]
Abstract
Type 2 immunity against pathogens is tightly regulated to ensure appropriate inflammatory responses that clear infection and prevent excessive tissue damage. Recent research has shown that type 2 innate lymphoid cells (ILC2s) contribute to steady-state tissue integrity and exert tissue-specific functions. However, upon exposure to inflammatory stimuli, they also initiate and amplify type 2 inflammation by inducing mucus production, eosinophilia, and Th2 differentiation. In this review, we discuss the regulation of ILC2 activation by transcription factors and metabolic pathways, as well as by extrinsic signals such as cytokines, lipid mediators, hormones, and neuropeptides. We also review recent discoveries about ILC2 plasticity and heterogeneity in different tissues, as revealed partly through single-cell RNA sequencing of transcriptional responses to various stimuli. Understanding the tissue-specific pathways that regulate ILC2 diversity and function is a critical step in the development of potential therapies for allergic diseases.
Collapse
Affiliation(s)
- Antonia Wallrapp
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham & Women's Hospital, Boston, Massachusetts
| | - Samantha J Riesenfeld
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Patrick R Burkett
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham & Women's Hospital, Boston, Massachusetts.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham & Women's Hospital, Boston, Massachusetts.,Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Department of Neurology, Brigham & Women's Hospital, Boston, Massachusetts
| |
Collapse
|
139
|
Schneider C, Lee J, Koga S, Ricardo-Gonzalez RR, Nussbaum JC, Smith LK, Villeda SA, Liang HE, Locksley RM. Tissue-Resident Group 2 Innate Lymphoid Cells Differentiate by Layered Ontogeny and In Situ Perinatal Priming. Immunity 2019; 50:1425-1438.e5. [PMID: 31128962 PMCID: PMC6645687 DOI: 10.1016/j.immuni.2019.04.019] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/18/2019] [Accepted: 04/28/2019] [Indexed: 01/21/2023]
Abstract
The perinatal period is a critical window for distribution of innate tissue-resident immune cells within developing organs. Despite epidemiologic evidence implicating the early-life environment in the risk for allergy, temporally controlled lineage tracing of group 2 innate lymphoid cells (ILC2s) during this period remains unstudied. Using complementary fate-mapping approaches and reporters for ILC2 activation, we show that ILC2s appeared in multiple organs during late gestation like tissue macrophages, but, unlike the latter, a majority of peripheral ILC2 pools were generated de novo during the postnatal window. This period was accompanied by systemic ILC2 priming and acquisition of tissue-specific transcriptomes. Although perinatal ILC2s were variably replaced across tissues with age, the dramatic increases in tissue ILC2s following helminth infection were mediated through local expansion independent of de novo generation by bone marrow hematopoiesis. We provide comprehensive temporally controlled fate mapping of an innate lymphocyte subset with notable nuances as compared to tissue macrophage ontogeny.
Collapse
Affiliation(s)
- Christoph Schneider
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jinwoo Lee
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Satoshi Koga
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Jesse C Nussbaum
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lucas K Smith
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Saul A Villeda
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hong-Erh Liang
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Richard M Locksley
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
140
|
Castellanos JG, Longman RS. The balance of power: innate lymphoid cells in tissue inflammation and repair. J Clin Invest 2019; 129:2640-2650. [PMID: 31180335 DOI: 10.1172/jci124617] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Over the last ten years, immunologists have recognized the central importance of an emerging group of innate lymphoid cells (ILCs) in health and disease. Characterization of these cells has provided a molecular definition of ILCs and their tissue-specific functions. Although the lineage-defining transcription factors, cytokine production, and nomenclature parallel those of T helper cells, ILCs do not require adaptive immune programming. Both environmental and host-derived signals shape the function of these evolutionarily ancient cells, which provide pathogen protection and promote tissue restoration. As such, ILCs function as a double-edged sword, balancing the inflammatory and reparative responses that arise during injury and disease. This Review highlights our recent understanding of tissue-resident ILCs and the signals that regulate their contribution to inflammation and tissue repair in health and disease.
Collapse
|
141
|
Helfrich S, Mindt BC, Fritz JH, Duerr CU. Group 2 Innate Lymphoid Cells in Respiratory Allergic Inflammation. Front Immunol 2019; 10:930. [PMID: 31231357 PMCID: PMC6566538 DOI: 10.3389/fimmu.2019.00930] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/11/2019] [Indexed: 12/14/2022] Open
Abstract
Millions of people worldwide are suffering from allergic inflammatory airway disorders. These conditions are regarded as a consequence of multiple imbalanced immune events resulting in an inadequate response with the exact underlying mechanisms still being a subject of ongoing research. Several cell populations have been proposed to be involved but it is becoming increasingly evident that group 2 innate lymphoid cells (ILC2s) play a key role in the initiation and orchestration of respiratory allergic inflammation. ILC2s are important mediators of inflammation but also tissue remodeling by secreting large amounts of signature cytokines within a short time period. Thereby, ILC2s instruct innate but also adaptive immune responses. Here, we will discuss the recent literature on allergic inflammation of the respiratory tract with a focus on ILC2 biology. Furthermore, we will highlight different therapeutic strategies to treat pulmonary allergic inflammation and their potential influence on ILC2 function as well as discuss the perspective of using human ILC2s for diagnostic purposes.
Collapse
Affiliation(s)
- Sofia Helfrich
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Barbara C Mindt
- Department of Microbiology & Immunology, McGill University, Montréal, QC, Canada.,McGill University Research Centre on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada.,FOCiS Centre of Excellence in Translational Immunology (CETI), McGill University, Montréal, QC, Canada
| | - Jörg H Fritz
- Department of Microbiology & Immunology, McGill University, Montréal, QC, Canada.,McGill University Research Centre on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada.,FOCiS Centre of Excellence in Translational Immunology (CETI), McGill University, Montréal, QC, Canada.,Department of Physiology, McGill University, Montréal, QC, Canada
| | - Claudia U Duerr
- Institute of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
142
|
Ren J, Hou C, Shi C, Lin Z, Liao W, Yuan E. A polysaccharide isolated and purified from Platycladus orientalis (L.) Franco leaves, characterization, bioactivity and its regulation on macrophage polarization. Carbohydr Polym 2019; 213:276-285. [DOI: 10.1016/j.carbpol.2019.03.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 02/23/2019] [Accepted: 03/03/2019] [Indexed: 01/10/2023]
|
143
|
Chapman NM, Shrestha S, Chi H. Metabolism in Immune Cell Differentiation and Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1011:1-85. [PMID: 28875486 DOI: 10.1007/978-94-024-1170-6_1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The immune system is a central determinant of organismal health. Functional immune responses require quiescent immune cells to rapidly grow, proliferate, and acquire effector functions when they sense infectious agents or other insults. Specialized metabolic programs are critical regulators of immune responses, and alterations in immune metabolism can cause immunological disorders. There has thus been growing interest in understanding how metabolic processes control immune cell functions under normal and pathophysiological conditions. In this chapter, we summarize how metabolic programs are tuned and what the physiological consequences of metabolic reprogramming are as they relate to immune cell homeostasis, differentiation, and function.
Collapse
Affiliation(s)
- Nicole M Chapman
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Sharad Shrestha
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
144
|
Xu W, Cherrier DE, Chea S, Vosshenrich C, Serafini N, Petit M, Liu P, Golub R, Di Santo JP. An Id2 RFP-Reporter Mouse Redefines Innate Lymphoid Cell Precursor Potentials. Immunity 2019; 50:1054-1068.e3. [PMID: 30926235 PMCID: PMC6477155 DOI: 10.1016/j.immuni.2019.02.022] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 01/03/2019] [Accepted: 02/25/2019] [Indexed: 12/31/2022]
Abstract
Innate lymphoid cell (ILC) development proposes that ILC precursors (ILCPs) segregate along natural killer (NK) cell versus helper cell (ILC1, ILC2, ILC3) pathways, the latter depending on expression of Id2, Zbtb16, and Gata3. We have developed an Id2-reporter strain expressing red fluorescent protein (RFP) in the context of normal Id2 expression to re-examine ILCP phenotype and function. We show that bone-marrow ILCPs were heterogeneous and harbored extensive NK-cell potential in vivo and in vitro. By multiplexing Id2RFP with Zbtb16CreGFP and Bcl11btdTomato strains, we made a single-cell dissection of the ILCP compartment. In contrast with the current model, we have demonstrated that Id2+Zbtb16+ ILCPs included multi-potent ILCPs that retained NK-cell potential. Late-stage ILC2P and ILC3P compartments could be defined by differential Zbtb16 and Bcl11b expression. We suggest a revised model for ILC differentiation that redefines the cell-fate potential of helper-ILC-restricted Zbtb16+ ILCPs.
Collapse
Affiliation(s)
- Wei Xu
- Innate Immunity Unit, Institut Pasteur, Paris 75724, France; Inserm U1223, Institut Pasteur, Paris 75724, France; Department of Immunology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Dylan E Cherrier
- Innate Immunity Unit, Institut Pasteur, Paris 75724, France; Inserm U1223, Institut Pasteur, Paris 75724, France; Paris Diderot University, Sorbonne Paris Cité, Paris 75013, France
| | - Sylvestre Chea
- Inserm U1223, Institut Pasteur, Paris 75724, France; Lymphopoiesis Unit, Institut Pasteur, Paris 75724, France
| | - Christian Vosshenrich
- Innate Immunity Unit, Institut Pasteur, Paris 75724, France; Inserm U1223, Institut Pasteur, Paris 75724, France
| | - Nicolas Serafini
- Innate Immunity Unit, Institut Pasteur, Paris 75724, France; Inserm U1223, Institut Pasteur, Paris 75724, France
| | - Maxime Petit
- Inserm U1223, Institut Pasteur, Paris 75724, France; Paris Diderot University, Sorbonne Paris Cité, Paris 75013, France; Lymphopoiesis Unit, Institut Pasteur, Paris 75724, France
| | - Pentao Liu
- Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Rachel Golub
- Inserm U1223, Institut Pasteur, Paris 75724, France; Lymphopoiesis Unit, Institut Pasteur, Paris 75724, France
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, Paris 75724, France; Inserm U1223, Institut Pasteur, Paris 75724, France.
| |
Collapse
|
145
|
Bouchery T, Le Gros G, Harris N. ILC2s-Trailblazers in the Host Response Against Intestinal Helminths. Front Immunol 2019; 10:623. [PMID: 31019505 PMCID: PMC6458269 DOI: 10.3389/fimmu.2019.00623] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/08/2019] [Indexed: 12/18/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) were first discovered in experimental studies of intestinal helminth infection—and much of our current knowledge of ILC2 activation and function is based on the use of these models. It is perhaps not surprising therefore that these cells have also been found to play a key role in mediating protection against these large multicellular parasites. ILC2s have been intensively studied over the last decade, and are known to respond quickly and robustly to the presence of helminths—both by increasing in number and producing type 2 cytokines. These mediators function to activate and repair epithelial barriers, to recruit other innate cells such as eosinophils, and to help activate T helper 2 cells. More recent investigations have focused on the mechanisms by which the host senses helminth parasites to activate ILC2s. Such studies have identified novel stromal cell types as being involved in this process—including intestinal tuft cells and enteric neurons, which respond to the presence of helminths and activate ILC2s by producing IL-25 and Neuromedin, respectively. In the current review, we will outline the latest insights into ILC2 activation and discuss the requirement for—or redundancy of—ILC2s in providing protective immunity against intestinal helminth parasites.
Collapse
Affiliation(s)
- Tiffany Bouchery
- Department of Immunology and Pathology, Monash University, AMREP, Melbourne, VIC, Australia
| | - Graham Le Gros
- Allergic & Parasitic Diseases Programme, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Nicola Harris
- Department of Immunology and Pathology, Monash University, AMREP, Melbourne, VIC, Australia
| |
Collapse
|
146
|
Innate lymphoid cells: A potential link between microbiota and immune responses against cancer. Semin Immunol 2019; 41:101271. [PMID: 30902413 DOI: 10.1016/j.smim.2019.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 03/10/2019] [Accepted: 03/14/2019] [Indexed: 01/05/2023]
Abstract
The adaptive immune system plays a crucial role in anti-tumor surveillance. Enhancement of T cell responses through checkpoint blockade has become a major therapeutic avenue of intervention for several tumors. Because it shapes immune responses and regulates their amplitude and duration, the microbiota has a substantial impact on anti-tumor immunity. Innate lymphoid cells (ILCs) comprise a heterogeneous population of lymphocytes devoid of antigen-specific receptors that mirror T helper cells in their ability to secrete cytokines that activate immune responses. Ongoing studies suggest that ILCs contribute to anti-tumor responses. Moreover, since ILCs are present at barrier surfaces, they are stimulated by the microbiota and, reciprocally, influence the composition of the microbiota by regulating the surface barrier microenvironment. Thus, ILC-microbiota cross-talk may in part underpin the effects of the microbiota on anti-tumor responses. In this article, we review current evidence linking ILCs to cancer and discuss the potential impact of ILC-microbiota cross-talk in anti-tumor immune responses.
Collapse
|
147
|
Jacquelot N, Luong K, Seillet C. Physiological Regulation of Innate Lymphoid Cells. Front Immunol 2019; 10:405. [PMID: 30915072 PMCID: PMC6422966 DOI: 10.3389/fimmu.2019.00405] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/15/2019] [Indexed: 12/17/2022] Open
Abstract
Discovery of innate lymphoid cells (ILCs) have provoked a paradigm shift in our understanding of the immune protection. Their constitutive presence and activity at the body's barrier surfaces ensure the maintenance of the tissue homeostasis and immune protection. This complex family has distinct and non-redundant functions that can have both beneficial and detrimental effects on disease outcome. The capacity of ILCs to perform their function effectively relies on their ability to sense and integrate intrinsic and extrinsic signals. Recent studies have shown that ILCs are not only sensitive to pathogen-derived stimuli but are also very well equipped to sense host-derived signals such as neuropeptides, hormones, and metabolites. The integration of these signals represents a complex and constant cross-talk between the immune system and the physiological systems of the body, including the nervous, endocrine, digestive, and reproductive systems. The physiological regulation of ILCs constitutes an important step in our understanding of the events leading to the protective and pathological properties of these cells. This review summarizes the recent advances in the understanding of the regulation of ILCs by physiological signals and their consequences on the maintenance of tissue homeostasis.
Collapse
Affiliation(s)
- Nicolas Jacquelot
- Molecular Immunology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Kylie Luong
- Molecular Immunology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Cyril Seillet
- Molecular Immunology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
148
|
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: 55] [Impact Index Per Article: 11.0] [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
|
149
|
Trindade R, Albrektsson T, Galli S, Prgomet Z, Tengvall P, Wennerberg A. Bone Immune Response to Materials, Part I: Titanium, PEEK and Copper in Comparison to Sham at 10 Days in Rabbit Tibia. J Clin Med 2018; 7:E526. [PMID: 30544551 PMCID: PMC6307090 DOI: 10.3390/jcm7120526] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/02/2018] [Accepted: 12/05/2018] [Indexed: 11/16/2022] Open
Abstract
Bone anchored biomaterials have become an indispensable solution for the restoration of lost dental elements and for skeletal joint replacements. However, a thorough understanding is still lacking in terms of the biological mechanisms leading to osseointegration and its contrast, unwanted peri-implant bone loss. We have previously hypothesized on the participation of immune mechanisms in such processes, and later demonstrated enhanced bone immune activation up to 4 weeks around titanium implants. The current experimental study explored and compared in a rabbit tibia model after 10 days of healing time, the bone inflammation/immunological reaction at mRNA level towards titanium, polyether ether ketone (PEEK) and copper compared to a Sham control. Samples from the test and control sites were, after a healing period, processed for gene expression analysis (polymerase chain reaction, (qPCR)) and decalcified histology tissue analysis. All materials displayed immune activation and suppression of bone resorption, when compared to sham. The M1 (inflammatory)/M2 (reparative) -macrophage phenotype balance was correlated to the proximity and volume of bone growth at the implant vicinity, with titanium demonstrating a M2-phenotype at 10 days, whereas copper and PEEK were still dealing with a mixed M1- and M2-phenotype environment. Titanium was the only material showing adequate bone growth and proximity inside the implant threads. There was a consistent upregulation of (T-cell surface glycoprotein CD4) CD4 and downregulation of (T-cell transmembrane glycoprotein CD8) CD8, indicating a CD4-lymphocyte phenotype driven reaction around all materials at 10 days.
Collapse
Affiliation(s)
- Ricardo Trindade
- Department of Prosthodontics, Faculty of Odontology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden.
| | - Tomas Albrektsson
- Department of Biomaterials, Institute of Clinical Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden.
- Department of Prosthodontics, Faculty of Odontology, Malmö University, 205 06 Malmö, Sweden.
| | - Silvia Galli
- Department of Prosthodontics, Faculty of Odontology, Malmö University, 205 06 Malmö, Sweden.
| | - Zdenka Prgomet
- Department of Oral Pathology, Faculty of Odontology, Malmö University, 205 06 Malmö, Sweden.
| | - Pentti Tengvall
- Department of Biomaterials, Institute of Clinical Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden.
| | - Ann Wennerberg
- Department of Prosthodontics, Faculty of Odontology, The Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden.
| |
Collapse
|
150
|
Epigenome analysis links gene regulatory elements in group 2 innate lymphocytes to asthma susceptibility. J Allergy Clin Immunol 2018; 142:1793-1807. [DOI: 10.1016/j.jaci.2017.12.1006] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 12/12/2017] [Accepted: 12/15/2017] [Indexed: 12/19/2022]
|