401
|
Dustin CM, Habibovic A, Hristova M, Schiffers C, Morris CR, Lin MCJ, Bauer RA, Heppner DE, Daphtary N, Aliyeva M, van der Vliet A. Oxidation-Dependent Activation of Src Kinase Mediates Epithelial IL-33 Production and Signaling during Acute Airway Allergen Challenge. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:2989-2999. [PMID: 34088769 PMCID: PMC8642476 DOI: 10.4049/jimmunol.2000995] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 04/06/2021] [Indexed: 11/19/2022]
Abstract
The respiratory epithelium forms the first line of defense against inhaled pathogens and acts as an important source of innate cytokine responses to environmental insults. One critical mediator of these responses is the IL-1 family cytokine IL-33, which is rapidly secreted upon acute epithelial injury as an alarmin and induces type 2 immune responses. Our recent work highlighted the importance of the NADPH oxidase dual oxidase 1 (DUOX1) in acute airway epithelial IL-33 secretion by various airborne allergens associated with H2O2 production and reduction-oxidation-dependent activation of Src kinases and epidermal growth factor receptor (EGFR) signaling. In this study, we show that IL-33 secretion in response to acute airway challenge with house dust mite (HDM) allergen critically depends on the activation of Src by a DUOX1-dependent oxidative mechanism. Intriguingly, HDM-induced epithelial IL-33 secretion was dramatically attenuated by small interfering RNA- or Ab-based approaches to block IL-33 signaling through its receptor IL1RL1 (ST2), indicating that HDM-induced IL-33 secretion includes a positive feed-forward mechanism involving ST2-dependent IL-33 signaling. Moreover, activation of type 2 cytokine responses by direct airway IL-33 administration was associated with ST2-dependent activation of DUOX1-mediated H2O2 production and reduction-oxidation-based activation of Src and EGFR and was attenuated in Duox1 -/- and Src +/- mice, indicating that IL-33-induced epithelial signaling and subsequent airway responses involve DUOX1/Src-dependent pathways. Collectively, our findings suggest an intricate relationship between DUOX1, Src, and IL-33 signaling in the activation of innate type 2 immune responses to allergens, involving DUOX1-dependent epithelial Src/EGFR activation in initial IL-33 secretion and in subsequent IL-33 signaling through ST2 activation.
Collapse
Affiliation(s)
- Christopher M Dustin
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - Aida Habibovic
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - Milena Hristova
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - Caspar Schiffers
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Carolyn R Morris
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - Miao-Chong Joy Lin
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - Robert A Bauer
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - David E Heppner
- Department of Chemistry, University at Buffalo, State University of New York, Buffalo, NY; and
| | - Nirav Daphtary
- Department of Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - Minara Aliyeva
- Department of Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT;
| |
Collapse
|
402
|
Zaini A, Fulford TS, Grumont RJ, Runting J, Rodrigues G, Ng J, Gerondakis S, Zaph C, Scheer S. c-Rel Is Required for IL-33-Dependent Activation of ILC2s. Front Immunol 2021; 12:667922. [PMID: 34194431 PMCID: PMC8236704 DOI: 10.3389/fimmu.2021.667922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/19/2021] [Indexed: 11/22/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are emerging as important cellular regulators of homeostatic and disease-associated immune processes. The cytokine interleukin-33 (IL-33) promotes ILC2-dependent inflammation and immunity, with IL-33 having been shown to activate NF-κB in a wide variety of cell types. However, it is currently unclear which NF-κB members play an important role in IL-33-dependent ILC2 biology. Here, we identify the NF-κB family member c-Rel as a critical component of the IL-33-dependent activation of ILC2s. Although c-Rel is dispensable for ILC2 development, it is critical for ILC2 function in the lung, with c-Rel-deficient (c-Rel-/- ) mice present a significantly reduced response to papain- and IL-33-induced lung inflammation. We also show that the absence of c-Rel reduces the IL-33-dependent expansion of ILC2 precursors and lower levels of IL-5 and IL-13 cytokine production by mature ILC2s in the lung. Together, these results identify the IL-33-c-Rel axis as a central control point of ILC2 activation and function.
Collapse
Affiliation(s)
- Aidil Zaini
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Thomas S. Fulford
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Raelene J. Grumont
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Jessica Runting
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Grace Rodrigues
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Judy Ng
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Steve Gerondakis
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Colby Zaph
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Sebastian Scheer
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
403
|
Plasma IL-33 levels are decreased in patients with high-risk myelodysplastic syndrome and show no correlation with pro-inflammatory IL-6 levels. Cytokine 2021; 148:155617. [PMID: 34130905 DOI: 10.1016/j.cyto.2021.155617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 11/21/2022]
Abstract
Aberrant inflammatory signaling has been shown to be a key pathogenic driver in myelodysplastic syndromes (MDS). Abnormal IL-33 expression has been implicated in inflammatory, immune-related disorders and, some tumors. However, its role in MDS remains widely unknown. This study aimed to evaluate the relationship between plasma levels of IL-33, clinical and prognostic data and, IL-6 levels in 101 patients with MDS. A comparative group of 59 healthy individuals was also evaluated. Plasma levels of cytokines were determined by enzyme-linked immunosorbent assay. Lower levels of IL-33 were found in patients with MDS when compared to the control group (p = 0.001), mainly in patients with more advanced stages of the disease and worse prognosis. No significant correlation between the levels of IL-33 and IL-6 was observed (r = 0.175; p = 0.081). These results reinforce the close association between immunological disorders and the pathogenesis of MDS. A greater understanding of the role of inflammatory cytokines in the disease can potentially provide new diagnosis and prognosis markers and new therapeutic targets.
Collapse
|
404
|
Iwaszko M, Wielińska J, Świerkot J, Kolossa K, Sokolik R, Bugaj B, Chaszczewska-Markowska M, Jeka S, Bogunia-Kubik K. IL-33 Gene Polymorphisms as Potential Biomarkers of Disease Susceptibility and Response to TNF Inhibitors in Rheumatoid Arthritis, Ankylosing Spondylitis, and Psoriatic Arthritis Patients. Front Immunol 2021; 12:631603. [PMID: 34177886 PMCID: PMC8226138 DOI: 10.3389/fimmu.2021.631603] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/20/2021] [Indexed: 12/31/2022] Open
Abstract
Objective Rheumatoid arthritis (RA), ankylosing spondylitis (AS), and psoriatic arthritis (PsA) belong to inflammatory rheumatic diseases, the group of conditions of unknown etiology. However, a strong genetic component in their pathogenesis has been well established. A dysregulation of cytokine networks plays an important role in the development of inflammatory arthritis. Interleukin 33 (IL-33) is a recently identified member of the IL-1 family. To date, the significance of IL-33 in inflammatory arthritis has been poorly studied. This research aimed to investigate the potential of IL-33 gene polymorphisms to serve as biomarkers for disease susceptibility and TNF inhibitor response in RA, AS, and PsA patients. Materials and Methods In total, 735 patients diagnosed with RA, AS, and PsA and 229 healthy individuals were enrolled in the study. Genotyping for three single nucleotide polymorphisms (SNPs) within the IL-33 gene, namely, rs16924159 (A/G), rs10975519 (T/C), and rs7044343 (C/T), was performed using polymerase chain reaction amplification employing LightSNiP assays. Results In the present study, the IL-33 rs10975519 CC genotype was associated with a decreased risk of developing RA in females, while the IL-33 rs16924159 polymorphism was associated with the efficacy of anti-TNF therapy and clinical parameters for RA and AS patients. The IL-33 rs16924159 AA genotype correlated with higher disease activity and worse clinical outcomes in RA patients treated with TNF inhibitors, and AS patients carrying the IL-33 rs16924159 AA genotype had higher disease activity and a worse response to anti-TNF therapy. That indicates a deleterious role of the IL-33 rs16924159 AA genotype in the context of RA, as well as AS. Conclusions The obtained results suggest that IL-33 gene polymorphisms might be potential candidate biomarkers of disease susceptibility and anti-TNF treatment response in patients with inflammatory rheumatic diseases.
Collapse
Affiliation(s)
- Milena Iwaszko
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Joanna Wielińska
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Jerzy Świerkot
- Department of Rheumatology and Internal Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Katarzyna Kolossa
- Department of Rheumatology and Connective Tissue Diseases, Jan Biziel University Hospital No. 2, Bydgoszcz, Poland
| | - Renata Sokolik
- Department of Rheumatology and Internal Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Bartosz Bugaj
- Department of Rheumatology and Internal Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Monika Chaszczewska-Markowska
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Sławomir Jeka
- Department of Rheumatology and Connective Tissue Diseases, Jan Biziel University Hospital No. 2, Bydgoszcz, Poland.,Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
405
|
Zhang Y, Guo J, Jia R. Treg: A Promising Immunotherapeutic Target in Oral Diseases. Front Immunol 2021; 12:667862. [PMID: 34177907 PMCID: PMC8222692 DOI: 10.3389/fimmu.2021.667862] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 05/24/2021] [Indexed: 12/24/2022] Open
Abstract
With the pandemic of COVID-19, maintenance of oral health has increasingly become the main challenge of global health. Various common oral diseases, such as periodontitis and oral cancer, are closely associated with immune disorders in the oral mucosa. Regulatory T cells (Treg) are essential for maintaining self-tolerance and immunosuppression. During the process of periodontitis and apical periodontitis, two typical chronic immune-inflammatory diseases, Treg contributes to maintain host immune homeostasis and minimize tissue damage. In contrast, in the development of oral precancerous lesions and oral cancer, Treg is expected to be depleted or down-regulated to enhance the anti-tumor immune response. Therefore, a deeper understanding of the distribution, function, and regulatory mechanisms of Treg cells may provide a prospect for the immunotherapy of oral diseases. In this review, we summarize the distribution and multiple roles of Treg in different oral diseases and discuss the possible mechanisms involved in Treg cell regulation, hope to provide a reference for future Treg-targeted immunotherapy in the treatment of oral diseases.
Collapse
Affiliation(s)
- Yujing Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jihua Guo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Endodontics, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Rong Jia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
406
|
Zheng H, Zhang Y, Pan J, Liu N, Qin Y, Qiu L, Liu M, Wang T. The Role of Type 2 Innate Lymphoid Cells in Allergic Diseases. Front Immunol 2021; 12:586078. [PMID: 34177881 PMCID: PMC8220221 DOI: 10.3389/fimmu.2021.586078] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 05/10/2021] [Indexed: 12/22/2022] Open
Abstract
Allergic diseases are significant diseases that affect many patients worldwide. In the past few decades, the incidence of allergic diseases has increased significantly due to environmental changes and social development, which has posed a substantial public health burden and even led to premature death. The understanding of the mechanism underlying allergic diseases has been substantially advanced, and the occurrence of allergic diseases and changes in the immune system state are known to be correlated. With the identification and in-depth understanding of innate lymphoid cells, researchers have gradually revealed that type 2 innate lymphoid cells (ILC2s) play important roles in many allergic diseases. However, our current studies of ILC2s are limited, and their status in allergic diseases remains unclear. This article provides an overview of the common phenotypes and activation pathways of ILC2s in different allergic diseases as well as potential research directions to improve the understanding of their roles in different allergic diseases and ultimately find new treatments for these diseases.
Collapse
Affiliation(s)
- Haocheng Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jiachuang Pan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Nannan Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Qin
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Linghui Qiu
- Journal Press of Global Traditional Chinese Medicine, Beijing, China
| | - Min Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
407
|
Veraar C, Koschutnik M, Nitsche C, Laggner M, Polak D, Bohle B, Mangold A, Moser B, Mascherbauer J, Ankersmit HJ. Inflammatory immune response in recipients of transcatheter aortic valves. JTCVS OPEN 2021; 6:85-96. [PMID: 36003560 PMCID: PMC9390500 DOI: 10.1016/j.xjon.2021.02.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 02/22/2021] [Indexed: 01/17/2023]
Abstract
Objective Transcatheter aortic valve implantation (TAVI) is rapidly replacing cardiac surgery due to its minimal invasiveness and practicality. Midterm immunological studies on the biocompatibility of galactose-alpha-1,3-galactose (α-Gal)–carrying bioprosthetic heart valves for TAVI are not available. In this study we investigated whether bioprosthetic heart valves employed for TAVI augment an α-Gal–specific antibody-dependent and antibody-independent immune response 3 months after TAVI implantation. Methods This prospective observational study included 27 patients with severe aortic valve stenosis undergoing TAVI and 10 patients with severe mitral valve regurgitation treated with a transcatheter MitraClip (Abbott Laboratories, Abbott Park, Ill) procedure. Blood samples were drawn before and 90 days after treatment at a routine checkup. Serum samples were analyzed using enzyme-linked immunosorbent assay. Serum concentrations of α-Gal–specific immunoglobulin (Ig) G, IgG subclasses and IgE, complement factor 3a, NETosis-specific citrullinated H3, and the systemic inflammation markers soluble suppression of tumorigenicity and interleukin 33 were evaluated. Results Three months after TAVI, we found significantly increased serum concentrations of α-Gal–specific IgG3, complement factor complement factor 3a, citrullinated H3 levels, and soluble suppression of tumorigenicity (P = .002, P = .001, P = .025, and P = .039, respectively). Sensitization of α-Gal–specific IgE antibodies occurred in 55% of all patients after TAVI. Conclusions Our results indicate that TAVI elicits a midterm, specific humoral immune response against α-Gal and causes an unspecific humoral inflammation compared with patients undergoing MitraClip implantation. This observation will lead to a better understanding of postintervention morbidity and the long-term durability of bioprostheses and indicates that caution is appropriate when designing implantation strategies for younger patients.
Collapse
Affiliation(s)
- Cecilia Veraar
- Division of Cardiothoracic and Vascular Anaesthesia and Intensive Care Medicine, Department of Anaesthesiology, General Intensive Care, and Pain Medicine, Medical University of Vienna, Vienna, Austria
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Medical University of Vienna, Vienna, Austria
| | - Matthias Koschutnik
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Christian Nitsche
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Maria Laggner
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Medical University of Vienna, Vienna, Austria
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Dominika Polak
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Barbara Bohle
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Andreas Mangold
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Bernhard Moser
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Julia Mascherbauer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- Department of Internal Medicine III, University Hospital St. Pölten, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Hendrik J. Ankersmit
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Medical University of Vienna, Vienna, Austria
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
- Address for reprints: Hendrik J. Ankersmit, MD, MBA, Department of Thoracic Surgery, Laboratory for Cardiac and Thoracic Diagnosis, Regeneration, and Applied Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
408
|
Scala G, Delaval MN, Mukherjee SP, Federico A, Khaliullin TO, Yanamala N, Fatkhutdinova LM, Kisin ER, Greco D, Fadeel B, Shvedova AA. Multi-walled carbon nanotubes elicit concordant changes in DNA methylation and gene expression following long-term pulmonary exposure in mice. CARBON 2021; 178:563-572. [PMID: 37206955 PMCID: PMC10193301 DOI: 10.1016/j.carbon.2021.03.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Pulmonary exposure to multi-walled carbon nanotubes (MWCNTs) causes inflammation and fibrosis. Our previous work has shown that industrially produced MWCNTs trigger specific changes in gene expression in the lungs of exposed animals. To elucidate whether epigenetic effects play a role for these gene expression changes, we performed whole genome bisulphite sequencing to assess DNA methylation patterns in the lungs 56 days after exposure to MWCNTs. Lung tissues were also evaluated with respect to histopathological changes and cytokine profiling of bronchoalveolar lavage (BAL) fluid was conducted using a multi-plex array. Integrated analysis of transcriptomics data and DNA methylation data revealed concordant changes in gene expression. Functional analysis showed that the muscle contraction, immune system/inflammation, and extracellular matrix pathways were the most affected pathways. Taken together, the present study revealed that MWCNTs exert epigenetic effects in the lungs of exposed animals, potentially driving the subsequent gene expression changes.
Collapse
Affiliation(s)
- Giovanni Scala
- Department of Biology, University of Naples, Naples, Italy
| | - Mathilde N. Delaval
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sourav P. Mukherjee
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Naveena Yanamala
- Health Effects Laboratory Division, NIOSH, CDC, Morgantown, WV, USA
| | - Liliya M. Fatkhutdinova
- Department of Hygiene and Occupational Medicine, Kazan State Medical University, Kazan, Russia
| | - Elena R. Kisin
- Health Effects Laboratory Division, NIOSH, CDC, Morgantown, WV, USA
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Corresponding author. (D. Greco)
| | - Bengt Fadeel
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Corresponding author. (B. Fadeel)
| | - Anna A. Shvedova
- Health Effects Laboratory Division, NIOSH, CDC, Morgantown, WV, USA
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
- Corresponding author. Health Effects Laboratory Division, NIOSH, CDC, Morgantown, WV, USA. (A.A. Shvedova)
| |
Collapse
|
409
|
Wu MX, Wang SH, Xie Y, Chen ZT, Guo Q, Yuan WL, Guan C, Xu CZ, Huang YN, Wang JF, Zhang HF, Chen YX. Interleukin-33 alleviates diabetic cardiomyopathy through regulation of endoplasmic reticulum stress and autophagy via insulin-like growth factor-binding protein 3. J Cell Physiol 2021; 236:4403-4419. [PMID: 33184863 DOI: 10.1002/jcp.30158] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/27/2020] [Accepted: 11/02/2020] [Indexed: 12/29/2022]
Abstract
Prolonged endoplasmic reticulum (ER) stress is the key driving force behind diabetic cardiomyopathy (DCM). Autophagy is extensively implicated in adaptive mechanisms for cell survival. Interleukin-33 (IL-33) is known to be a potent cardiac protector, but its roles in DCM, ER stress, and autophagy are currently unknown. We aimed to explore the effects of IL-33 on DCM and characterize the roles that ER stress and autophagy play in DCM. The effects of IL-33 on DCM, ER stress, and autophagy were characterized both in db/db mice and in palmitic acid (PA)-treated cardiomyocytes. The manipulators of ER stress and autophagy were used to clarify their roles in DCM remittance conferred by IL-33. Gene expression analysis was used to identify IL-33-dependent regulators of ER stress and autophagy. Both db/db mice and PA-treated cells presented with enhanced levels of ER stress, apoptosis, and lipid deposition, as well as impaired autophagy, all of which could be reversed by IL-33. Treatment with IL-33 improved the cardiac diastolic function of diabetic mice. Nonselective autophagy inhibitors, such as 3-methyladenine (3-MA) or wortmannin, abolished the protective effects of IL-33, resulting in an increase in both ER stress and apoptosis. Strikingly, insulin-like growth factor-binding protein 3 (IGFBP3) was identified as the gene most significantly differentially expressed between IL-33 and control groups. Knockdown of IGFBP3 expression, similar to the effect of nonselective autophagy inhibitors, resulted in high levels of ER stress, impaired autophagy, and apoptosis that were not rescued upon treatment with IL-33. IL-33 abates DCM by alleviating ER stress and promoting autophagy. IGFBP3 is essential for IL-33-induced ER stress resolution and autophagic enhancement during DCM.
Collapse
MESH Headings
- Animals
- Apoptosis Regulatory Proteins/metabolism
- Autophagy/drug effects
- Autophagy-Related Proteins/metabolism
- Cells, Cultured
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/metabolism
- Diabetic Cardiomyopathies/pathology
- Diabetic Cardiomyopathies/prevention & control
- Disease Models, Animal
- Endoplasmic Reticulum Stress/drug effects
- Insulin-Like Growth Factor Binding Protein 3/genetics
- Insulin-Like Growth Factor Binding Protein 3/metabolism
- Interleukin-33/pharmacology
- Male
- Mice
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Palmitic Acid/toxicity
- Rats, Sprague-Dawley
- Signal Transduction
- Rats
Collapse
Affiliation(s)
- Mao-Xiong Wu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, Guangdong, China
| | - Shao-Hua Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, Guangdong, China
| | - Yong Xie
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, Guangdong, China
| | - Zhi-Teng Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, Guangdong, China
| | - Qi Guo
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, Guangdong, China
| | - Wo-Liang Yuan
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, Guangdong, China
| | - Chang Guan
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, Guangdong, China
| | - Cheng-Zhang Xu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, Guangdong, China
| | - Yu-Na Huang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, Guangdong, China
| | - Jing-Feng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, Guangdong, China
| | - Hai-Feng Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, Guangdong, China
| | - Yang-Xin Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, Guangdong, China
| |
Collapse
|
410
|
Transient IL-33 upregulation in neonatal mouse lung promotes acute but not chronic type 2 immune responses induced by allergen later in life. PLoS One 2021; 16:e0252199. [PMID: 34048460 PMCID: PMC8162637 DOI: 10.1371/journal.pone.0252199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/11/2021] [Indexed: 01/02/2023] Open
Abstract
Early life respiratory insults, such as viral infections or hyperoxia, often increase asthma susceptibility later in life. The mechanisms underlying this increased susceptibility are not fully understood. IL-33 has been shown to be critically involved in allergic airway diseases. IL-33 expression in the neonatal lung can be increased by various respiratory insults associated with asthma development. Therefore, we investigated whether and how early life increases in IL-33 impact allergic airway responses later in life. Using a novel IL-33 transgenic mouse model, in which full-length IL-33 was inducible overexpressed in lung epithelial cells, we transiently upregulated lung IL-33 expression in neonatal mice for one week. After resting for 4–6 weeks, mice were intranasally exposed to a single-dose of recombinant IL-33 or the airborne allergen Alternaria. Alternatively, mice were exposed to Alternaria and ovalbumin multiple times for one month. We found that a transient increase in IL-33 expression during the neonatal period promoted IL-5 and IL-13 production when mice were later exposed to a single-dose of IL-33 or Alternaria in adulthood. However, increased IL-33 expression during the neonatal period did not affect airway inflammation, type 2 cytokine production, lung mucus production, or antigen-specific antibody responses when adult mice were exposed to Alternaria and ovalbumin multiple times. These results suggest that transient increased IL-33 expression early in life may have differential effects on allergic airway responses in later life, preferentially affecting allergen-induced acute type 2 cytokine production.
Collapse
|
411
|
Milling S, Siebert S. T cells and cytokines in inflamed psoriatic skin. Who's in charge? Immunology 2021; 160:311-312. [PMID: 32686183 DOI: 10.1111/imm.13237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Inflammation in psoriasis is driven through the IL-23/IL-17 pathway. Monoclonal antibodies targeting cytokines in the pathway have proven highly effective and are widely used in clinical practice. There is still much to learn, however, about how these pathogenic responses are controlled, particularly with respect to the highly immunologically active molecules produced by the inflamed skin tissue itself. These tissue-derived molecules, which include IL-33, play important roles in modulating chronic inflammation that we are only beginning to understand. Here we highlight new research indicating a role for IL-33 in modulating the inflammatory Th17 response in psoriasis, which may provide avenues for developing new psoriasis treatments.
Collapse
Affiliation(s)
- Simon Milling
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Stefan Siebert
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
412
|
Mado H, Adamczyk-Sowa M, Bartman W, Wierzbicki K, Tadeusiak B, Sowa P. Plasma Interleukin-33 level in relapsing-remitting multiple sclerosis. Is it negatively correlated with central nervous system lesions in patients with mild disability? Clin Neurol Neurosurg 2021; 206:106700. [PMID: 34030079 DOI: 10.1016/j.clineuro.2021.106700] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/06/2021] [Accepted: 05/12/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND Cytokines and chemokines are undoubtedly involved in the pathogenesis of multiple sclerosis (MS). There are many reports that suggest a significant role for Interleukin-33 (IL-33) in the course of MS development, but it is not clear whether negative or positive. We therefore investigated plasma IL-33 levels in patients with relapsing-remitting MS (RRMS). METHODS The study consisted of RRMS patients (n = 73) and healthy subjects (n = 54). Blood samples were taken from all and plasma IL-33 levels were then determined using an enzyme-linked immunosorbent assay method. Patients also underwent laboratory and imaging tests and their disability status was assessed. RESULTS Plasma IL-33 levels were marginally significantly higher in patients with RRMS (p = 0.07). Higher IL-33 levels are significantly associated with higher age (p = 0.01). There was also a statistically significant negative correlation between plasma IL-33 levels and the number of high signal intensity lesions in T2-weighted MRI (p = 0.03). After dividing the number of lesions into groups < 9 and ≥ 9 T2-weighted lesions, the Student's t-test for unrelated variables showed a negative correlation, but not statistically significant (p = 0.22), while the Spearman's correlation showed a marginally significant correlation (p = 0.06) between IL-33 level and number of T2-weighted lesions. IL-33 was also shown to have a significant ability to differentiate RRMS patients from healthy subjects with a sensitivity of 99% and specificity of 70% (p = 0.00). CONCLUSIONS Patients with RRMS have elevated plasma IL-33 levels. In RRMS patients with mild disability, high plasma levels of IL-33 may have neuroprotective effects potentially by stimulating remyelination and/or suppressing autoimmune inflammation and damage. Further studies on this matter on a larger number of patients are needed.
Collapse
Affiliation(s)
- Hubert Mado
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland.
| | - Monika Adamczyk-Sowa
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Wojciech Bartman
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Krzysztof Wierzbicki
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Bartosz Tadeusiak
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| | - Paweł Sowa
- Department of Otorhinolaryngology and Oncological Laryngology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Poland
| |
Collapse
|
413
|
Planté-Bordeneuve T, Pilette C, Froidure A. The Epithelial-Immune Crosstalk in Pulmonary Fibrosis. Front Immunol 2021; 12:631235. [PMID: 34093523 PMCID: PMC8170303 DOI: 10.3389/fimmu.2021.631235] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 05/04/2021] [Indexed: 12/12/2022] Open
Abstract
Interactions between the lung epithelium and the immune system involve a tight regulation to prevent inappropriate reactions and have been connected to several pulmonary diseases. Although the distal lung epithelium and local immunity have been implicated in the pathogenesis and disease course of idiopathic pulmonary fibrosis (IPF), consequences of their abnormal interplay remain less well known. Recent data suggests a two-way process, as illustrated by the influence of epithelial-derived periplakin on the immune landscape or the effect of macrophage-derived IL-17B on epithelial cells. Additionally, damage associated molecular patterns (DAMPs), released by damaged or dying (epithelial) cells, are augmented in IPF. Next to “sterile inflammation”, pathogen-associated molecular patterns (PAMPs) are increased in IPF and have been linked with lung fibrosis, while outer membrane vesicles from bacteria are able to influence epithelial-macrophage crosstalk. Finally, the advent of high-throughput technologies such as microbiome-sequencing has allowed for the identification of a disease-specific microbial environment. In this review, we propose to discuss how the interplays between the altered distal airway and alveolar epithelium, the lung microbiome and immune cells may shape a pro-fibrotic environment. More specifically, it will highlight DAMPs-PAMPs pathways and the specificities of the IPF lung microbiome while discussing recent elements suggesting abnormal mucosal immunity in pulmonary fibrosis.
Collapse
Affiliation(s)
- Thomas Planté-Bordeneuve
- Pôle de pneumologie, O.R.L. et dermatologie, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Bruxelles, Belgium
| | - Charles Pilette
- Pôle de pneumologie, O.R.L. et dermatologie, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Bruxelles, Belgium.,Service de pneumologie, Cliniques universitaires Saint-Luc, Bruxelles, Belgium
| | - Antoine Froidure
- Pôle de pneumologie, O.R.L. et dermatologie, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Bruxelles, Belgium.,Service de pneumologie, Cliniques universitaires Saint-Luc, Bruxelles, Belgium
| |
Collapse
|
414
|
Sun Y, Wen Y, Wang L, Wen L, You W, Wei S, Mao L, Wang H, Chen Z, Yang X. Therapeutic Opportunities of Interleukin-33 in the Central Nervous System. Front Immunol 2021; 12:654626. [PMID: 34079543 PMCID: PMC8165230 DOI: 10.3389/fimmu.2021.654626] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 05/04/2021] [Indexed: 01/14/2023] Open
Abstract
Interleukin-33 (IL-33), a member of the IL-1 cytokine family, is involved in various diseases. IL-33 exerts its effects via its heterodimeric receptor complex, which comprises suppression of tumorigenicity 2 (ST2) and the IL-1 receptor accessory protein (IL-1RAP). Increasing evidence has demonstrated that IL-33/ST2 signaling plays diverse but crucial roles in the homeostasis of the central nervous system (CNS) and the pathogenesis of CNS diseases, including neurodegenerative diseases, cerebrovascular diseases, infection, trauma, and ischemic stroke. In the current review, we focus on the functional roles and cellular signaling mechanisms of IL-33 in the CNS and evaluate the potential for diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Yun Sun
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Yankai Wen
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Luxi Wang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liang Wen
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Wendong You
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Shuang Wei
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Lin Mao
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Hao Wang
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Zuobing Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Xiaofeng Yang
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| |
Collapse
|
415
|
Guo Q, Bartish M, Gonçalves C, Huang F, Smith-Voudouris J, Krisna SS, Preston SEJ, Emond A, Li VZ, Duerr CU, Gui Y, Cleret-Buhot A, Thebault P, Lefrère H, Lenaerts L, Plourde D, Su J, Mindt BC, Hewgill SA, Cotechini T, Hindmarch CCT, Yang W, Khoury E, Zhan Y, Narykina V, Wei Y, Floris G, Basik M, Amant F, Quail DF, Lapointe R, Fritz JH, Del Rincon SV, Miller WH. The MNK1/2-eIF4E Axis Supports Immune Suppression and Metastasis in Postpartum Breast Cancer. Cancer Res 2021; 81:3876-3889. [PMID: 33975880 DOI: 10.1158/0008-5472.can-20-3143] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 04/08/2021] [Accepted: 05/10/2021] [Indexed: 11/16/2022]
Abstract
Breast cancer diagnosed within 10 years following childbirth is defined as postpartum breast cancer (PPBC) and is highly metastatic. Interactions between immune cells and other stromal cells within the involuting mammary gland are fundamental in facilitating an aggressive tumor phenotype. The MNK1/2-eIF4E axis promotes translation of prometastatic mRNAs in tumor cells, but its role in modulating the function of nontumor cells in the PPBC microenvironment has not been explored. Here, we used a combination of in vivo PPBC models and in vitro assays to study the effects of inactivation of the MNK1/2-eIF4E axis on the protumor function of select cells of the tumor microenvironment. PPBC mice deficient for phospho-eIF4E (eIF4ES209A) were protected against lung metastasis and exhibited differences in the tumor and lung immune microenvironment compared with wild-type mice. Moreover, the expression of fibroblast-derived IL33, an alarmin known to induce invasion, was repressed upon MNK1/2-eIF4E axis inhibition. Imaging mass cytometry on PPBC and non-PPBC patient samples indicated that human PPBC contains phospho-eIF4E high-expressing tumor cells and CD8+ T cells displaying markers of an activated dysfunctional phenotype. Finally, inhibition of MNK1/2 combined with anti-PD-1 therapy blocked lung metastasis of PPBC. These findings implicate the involvement of the MNK1/2-eIF4E axis during PPBC metastasis and suggest a promising immunomodulatory route to enhance the efficacy of immunotherapy by blocking phospho-eIF4E. SIGNIFICANCE: This study investigates the MNK1/2-eIF4E signaling axis in tumor and stromal cells in metastatic breast cancer and reveals that MNK1/2 inhibition suppresses metastasis and sensitizes tumors to anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Qianyu Guo
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montréal, Québec, Canada.,Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada
| | - Margarita Bartish
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montréal, Québec, Canada.,Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada
| | - Christophe Gonçalves
- Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada
| | - Fan Huang
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montréal, Québec, Canada.,Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada
| | - Julian Smith-Voudouris
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montréal, Québec, Canada.,Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada
| | - Sai Sakktee Krisna
- Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada.,Department of Physiology, Faculty of Medicine, McGill University, Montréal, Québec, Canada.,McGill University Research Centre on Complex Traits, Montréal, Québec, Canada
| | - Samuel E J Preston
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montréal, Québec, Canada.,Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada
| | - Audrey Emond
- Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada
| | - Vivian Z Li
- Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada
| | - Claudia U Duerr
- Department of Microbiology & Immunology, Faculty of Medicine, McGill University, Montréal, Québec, Canada
| | - Yirui Gui
- Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada
| | - Aurélie Cleret-Buhot
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Pamela Thebault
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Institut du cancer de Montréal, Montreal, Canada.,Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada.,Clinical Immuno-Monitoring Core Facility, CRCHUM, Montréal, Québec, Canada
| | - Hanne Lefrère
- Department of Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Liesbeth Lenaerts
- Department of Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Dany Plourde
- Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada
| | - Jie Su
- Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada
| | - Barbara C Mindt
- McGill University Research Centre on Complex Traits, Montréal, Québec, Canada.,Department of Microbiology & Immunology, Faculty of Medicine, McGill University, Montréal, Québec, Canada
| | - Shannon A Hewgill
- McGill University Research Centre on Complex Traits, Montréal, Québec, Canada.,Department of Microbiology & Immunology, Faculty of Medicine, McGill University, Montréal, Québec, Canada
| | - Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - William Yang
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montréal, Québec, Canada
| | - Elie Khoury
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montréal, Québec, Canada
| | - Yao Zhan
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montréal, Québec, Canada
| | - Valeria Narykina
- Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada
| | - Yuhong Wei
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada
| | - Giuseppe Floris
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium.,Department of Imaging and Pathology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Mark Basik
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montréal, Québec, Canada.,Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada
| | - Frédéric Amant
- Department of Oncology, Katholieke Universiteit Leuven, Leuven, Belgium.,Center Gynaecologic Oncology Amsterdam at the Netherlands Cancer Institute and Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Daniela F Quail
- Department of Physiology, Faculty of Medicine, McGill University, Montréal, Québec, Canada.,Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada
| | - Réjean Lapointe
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Institut du cancer de Montréal, Montreal, Canada.,Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Jörg H Fritz
- McGill University Research Centre on Complex Traits, Montréal, Québec, Canada.,Department of Microbiology & Immunology, Faculty of Medicine, McGill University, Montréal, Québec, Canada.,Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada
| | - Sonia V Del Rincon
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montréal, Québec, Canada. .,Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada
| | - Wilson H Miller
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montréal, Québec, Canada. .,Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, Montréal, Québec, Canada.,Montreal Rossy Cancer Network, Montréal, Québec, Canada
| |
Collapse
|
416
|
Liang Y, Ge Y, Sun J. IL-33 in COVID-19: friend or foe? Cell Mol Immunol 2021; 18:1602-1604. [PMID: 33972738 PMCID: PMC8108013 DOI: 10.1038/s41423-021-00685-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 04/10/2021] [Indexed: 01/30/2023] Open
Affiliation(s)
- Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA. .,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| | - Yiyue Ge
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA. .,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
417
|
Guttenberg MA, Vose AT, Tighe RM. Role of Innate Immune System in Environmental Lung Diseases. Curr Allergy Asthma Rep 2021; 21:34. [PMID: 33970346 PMCID: PMC8311569 DOI: 10.1007/s11882-021-01011-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2021] [Indexed: 01/07/2023]
Abstract
The lung mucosa functions as a principal barrier between the body and inhaled environmental irritants and pathogens. Precise and targeted surveillance mechanisms are required at this lung-environment interface to maintain homeostasis and preserve gas exchange. This is performed by the innate immune system, a germline-encoded system that regulates initial responses to foreign irritants and pathogens. Environmental pollutants, such as particulate matter (PM), ozone (O3), and other products of combustion (NO2, SO3, etc.), both stimulate and disrupt the function of the innate immune system of the lung, leading to the potential for pathologic consequences. PURPOSE OF REVIEW: The purpose of this review is to explore recent discoveries and investigations into the role of the innate immune system in responding to environmental exposures. This focuses on mechanisms by which the normal function of the innate immune system is modified by environmental agents leading to disruptions in respiratory function. RECENT FINDINGS: This is a narrative review of mechanisms of pulmonary innate immunity and the impact of environmental exposures on these responses. Recent findings highlighted in this review are categorized by specific components of innate immunity including epithelial function, macrophages, pattern recognition receptors, and the microbiome. Overall, the review supports broad impacts of environmental exposures to alterations to normal innate immune functions and has important implications for incidence and exacerbations of lung disease. The innate immune system plays a critical role in maintaining pulmonary homeostasis in response to inhaled air pollutants. As many of these agents are unable to be mitigated, understanding their mechanistic impact is critical to develop future interventions to limit their pathologic consequences.
Collapse
Affiliation(s)
| | | | - Robert M. Tighe
- Department of Medicine, Duke University, Durham, NC,Corresponding Author: Robert M Tighe, MD, Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Box 2969, Durham, North Carolina 27710, Telephone: 919-684-4894, Fax: 919-684-5266,
| |
Collapse
|
418
|
Khan AUH, Almutairi SM, Ali AK, Salcedo R, Stewart CA, Wang L, Lee SH. Expression of Nutrient Transporters on NK Cells During Murine Cytomegalovirus Infection Is MyD88-Dependent. Front Immunol 2021; 12:654225. [PMID: 34093543 PMCID: PMC8177011 DOI: 10.3389/fimmu.2021.654225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/06/2021] [Indexed: 12/03/2022] Open
Abstract
Natural killer (NK) cells are the predominant innate lymphocytes that provide early defense against infections. In the inflammatory milieu, NK cells modify their metabolism to support high energy demands required for their proliferation, activation, and functional plasticity. This metabolic reprogramming is usually accompanied by the upregulation of nutrient transporter expression on the cell surface, leading to increased nutrient uptake required for intense proliferation. The interleukin-1 family members of inflammatory cytokines are critical in activating NK cells during infection; however, their underlying mechanism in NK cell metabolism is not fully elucidated. Previously, we have shown that IL-18 upregulates the expression of solute carrier transmembrane proteins and thereby induces a robust metabolic boost in NK cells. Unexpectedly, we found that IL-18 signaling is dispensable during viral infection in vivo, while the upregulation of nutrient transporters is primarily MyD88-dependent. NK cells from Myd88-/- mice displayed significantly reduced surface expression of nutrient receptors and mTOR activity during MCMV infection. We also identified that IL-33, another cytokine employing MyD88 signaling, induces the expression of nutrient transporters but requires a pre-exposure to IL-12. Moreover, signaling through the NK cell activating receptor, Ly49H, can also promote the expression of nutrient transporters. Collectively, our findings revealed multiple pathways that can induce the expression of nutrient transporters on NK cells while highlighting the imperative role of MyD88 in NK cell metabolism during infection.
Collapse
Affiliation(s)
- Abrar Ul Haq Khan
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Saeedah Musaed Almutairi
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Botany and Microbiology Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Alaa Kassim Ali
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Rosalba Salcedo
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, United States
| | - C. Andrew Stewart
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, United States
| | - Lisheng Wang
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- The University of Ottawa Centre for Infection, Immunity, and Inflammation, Ottawa, ON, Canada
| | - Seung-Hwan Lee
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- The University of Ottawa Centre for Infection, Immunity, and Inflammation, Ottawa, ON, Canada
| |
Collapse
|
419
|
Larsen S, Seidelin JB, Davidsen J, Dahlgaard K, Nielsen CH, Bennett EP, Pedersen OB, Coskun M, Troelsen JT. CDX2 regulates interleukin-33 gene expression in intestinal epithelial cells (LS174T). FEBS Open Bio 2021; 11:1638-1644. [PMID: 33838073 PMCID: PMC8167865 DOI: 10.1002/2211-5463.13161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/18/2021] [Accepted: 04/08/2021] [Indexed: 01/22/2023] Open
Abstract
Dysregulation of interleukin-33 (IL-33) has been implicated in the pathogenesis of several autoimmune and inflammatory diseases, but few studies have examined transcriptional regulation of the IL33 gene. In the intestines, gene regulation is controlled by a transcription factor network of which the intestinal-specific transcription factor CDX2 is a key component. In this study, we investigated whether CDX2 regulates IL33 mRNA expression. We examined IL33 mRNA expression in primary colonic epithelial cells from healthy humans and epithelial cell lines, revealing high expression levels in primary colonic and LS174T cells. Combining genomics data (ChIP-seq, RNA-seq) and IL33 promoter analyses in LS174T cells revealed intronic enhancer activity in the IL33 gene that is dependent on CDX2 expression. Western blotting and qRT-PCR confirmed that IL33 expression is upregulated in a CDX2 concentration-dependent manner, thereby providing the first evidence that CDX2 regulates the expression of IL33.
Collapse
Affiliation(s)
- Sylvester Larsen
- Department of Science and Environment, Roskilde University, Denmark.,Department of Clinical Immunology, Naestved Hospital, Denmark
| | - Jakob Benedict Seidelin
- Department of Gastroenterology, Medical Section, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Johanne Davidsen
- Department of Science and Environment, Roskilde University, Denmark.,Department of Surgical Gastroenterology, Zealand University Hospital, Køge, Denmark
| | - Katja Dahlgaard
- Department of Science and Environment, Roskilde University, Denmark
| | - Claus Henrik Nielsen
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Section 7521, Copenhagen University Hospital Rigshospitalet, Denmark
| | - Eric Paul Bennett
- Department of Odontology, Copenhagen Center for Glycomics, Faculty of Health Sciences, University of Copenhagen, Denmark
| | | | - Mehmet Coskun
- Department of Gastroenterology, Medical Section, Herlev Hospital, University of Copenhagen, Herlev, Denmark.,Department of Biology, The Bioinformatics Centre, Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Denmark
| | | |
Collapse
|
420
|
Lau SF, Fu AKY, Ip NY. Cytokine signaling convergence regulates the microglial state transition in Alzheimer's disease. Cell Mol Life Sci 2021; 78:4703-4712. [PMID: 33847763 PMCID: PMC8195901 DOI: 10.1007/s00018-021-03810-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 12/13/2022]
Abstract
Genetic analyses have revealed the pivotal contribution of microglial dysfunctions to the pathogenesis of Alzheimer's disease (AD). Along AD progression, the accumulation of danger-associated molecular patterns (DAMPs) including beta-amyloid and hyperphosphorylated tau continuously stimulates microglia, which results in their chronic activation. Chronically activated microglia secrete excessive pro-inflammatory cytokines, which further regulate microglial responses towards DAMPs. This has spurred longstanding interest in targeting cytokine-induced microglial responses for AD therapeutic development. However, the cytokine-induced microglial state transition is not comprehensively understood. Cytokines are assumed to induce microglial state transition from a resting state to an activated state. However, recent evidence indicate that this microglial state transition involves multiple sequential functional states. Moreover, the mechanisms by which different functional states within the cytokine-induced microglial state transition regulate AD pathology remain unclear. In this review, we summarize how different cytokine signaling pathways, including those of IL-33 (interleukin-33), NLRP3 inflammasome-IL-1β, IL-10, and IL-12/IL-23, regulate microglial functions in AD. Furthermore, we discuss how the modulation of these cytokine signaling pathways can result in beneficial outcomes in AD. Finally, we describe a stepwise functional state transition of microglia induced by cytokine signaling that can provide insights into the molecular basis of the beneficial effects of cytokine modulation in AD and potentially aid therapeutic development.
Collapse
Affiliation(s)
- Shun-Fat Lau
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Amy K Y Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, 518057, Guangdong, China
| | - Nancy Y Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, 518057, Guangdong, China.
| |
Collapse
|
421
|
Clark JT, Christian DA, Gullicksrud JA, Perry JA, Park J, Jacquet M, Tarrant JC, Radaelli E, Silver J, Hunter CA. IL-33 promotes innate lymphoid cell-dependent IFN-γ production required for innate immunity to Toxoplasma gondii. eLife 2021; 10:e65614. [PMID: 33929319 PMCID: PMC8121546 DOI: 10.7554/elife.65614] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/29/2021] [Indexed: 12/29/2022] Open
Abstract
IL-33 is an alarmin required for resistance to the parasite Toxoplasma gondii, but its role in innate resistance to this organism is unclear. Infection with T. gondii promotes increased stromal cell expression of IL-33, and levels of parasite replication correlate with release of IL-33 in affected tissues. In response to infection, a subset of innate lymphoid cells (ILC) emerges composed of IL-33R+ NK cells and ILC1s. In Rag1-/-mice, where NK cells and ILC1 production of IFN-γ mediate innate resistance to T. gondii, the loss of the IL-33R resulted in reduced ILC responses and increased parasite replication. Furthermore, administration of IL-33 to Rag1-/- mice resulted in a marked decrease in parasite burden, increased production of IFN-γ, and the recruitment and expansion of inflammatory monocytes associated with parasite control. These protective effects of exogenous IL-33 were dependent on endogenous IL-12p40 and the ability of IL-33 to enhance ILC production of IFN-γ. These results highlight that IL-33 synergizes with IL-12 to promote ILC-mediated resistance to T. gondii.
Collapse
Affiliation(s)
- Joseph T Clark
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| | - David A Christian
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| | - Jodi A Gullicksrud
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| | - Joseph A Perry
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| | - Jeongho Park
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
- Kangwon National University College of Veterinary Medicine and Institute of Veterinary ScienceChuncheonRepublic of Korea
| | - Maxime Jacquet
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
- Liver Immunology, Department of Biomedicine, University Hospital of Basel and University of BaselBaselSwitzerland
| | - James C Tarrant
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| | - Enrico Radaelli
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| | - Jonathan Silver
- Department of Respiratory Inflammation and Autoimmunity, AstraZenecaGaithersburgUnited States
| | - Christopher A Hunter
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| |
Collapse
|
422
|
Wang Y, Fu AKY, Ip NY. Instructive roles of astrocytes in hippocampal synaptic plasticity: neuronal activity-dependent regulatory mechanisms. FEBS J 2021; 289:2202-2218. [PMID: 33864430 PMCID: PMC9290076 DOI: 10.1111/febs.15878] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/02/2021] [Accepted: 04/12/2021] [Indexed: 12/22/2022]
Abstract
In the adult hippocampus, synaptic plasticity is important for information processing, learning, and memory encoding. Astrocytes, the most common glial cells, play a pivotal role in the regulation of hippocampal synaptic plasticity. While astrocytes were initially described as a homogenous cell population, emerging evidence indicates that in the adult hippocampus, astrocytes are highly heterogeneous and can differentially respond to changes in neuronal activity in a subregion‐dependent manner to actively modulate synaptic plasticity. In this review, we summarize how local neuronal activity changes regulate the interactions between astrocytes and synapses, either by modulating the secretion of gliotransmitters and synaptogenic proteins or via contact‐mediated signaling pathways. In turn, these specific responses induced in astrocytes mediate the interactions between astrocytes and neurons, thus shaping synaptic communication in the adult hippocampus. Importantly, the activation of astrocytic signaling is required for memory performance including memory acquisition and recall. Meanwhile, the dysregulation of this signaling can cause hippocampal circuit dysfunction in pathological conditions, resulting in cognitive impairment and neurodegeneration. Indeed, reactive astrocytes, which have dysregulated signaling associated with memory, are induced in the brains of patients with Alzheimer's disease (AD) and transgenic mouse model of AD. Emerging technologies that can precisely manipulate and monitor astrocytic signaling in vivo enable the examination of the specific actions of astrocytes in response to neuronal activity changes as well as how they modulate synaptic connections and circuit activity. Such findings will clarify the roles of astrocytes in hippocampal synaptic plasticity and memory in health and disease.
Collapse
Affiliation(s)
- Ye Wang
- Division of Life Science, The Hong Kong University of Science and Technology, China.,Molecular Neuroscience Center, The Hong Kong University of Science and Technology, China.,State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, China.,Hong Kong Center for Neurodegenerative Diseases, China
| | - Amy K Y Fu
- Division of Life Science, The Hong Kong University of Science and Technology, China.,Molecular Neuroscience Center, The Hong Kong University of Science and Technology, China.,State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, China.,Hong Kong Center for Neurodegenerative Diseases, China.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, China
| | - Nancy Y Ip
- Division of Life Science, The Hong Kong University of Science and Technology, China.,Molecular Neuroscience Center, The Hong Kong University of Science and Technology, China.,State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, China.,Hong Kong Center for Neurodegenerative Diseases, China.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, China
| |
Collapse
|
423
|
Kelsen SG, Agache IO, Soong W, Israel E, Chupp GL, Cheung DS, Theess W, Yang X, Staton TL, Choy DF, Fong A, Dash A, Dolton M, Pappu R, Brightling CE. Astegolimab (anti-ST2) efficacy and safety in adults with severe asthma: A randomized clinical trial. J Allergy Clin Immunol 2021; 148:790-798. [PMID: 33872652 DOI: 10.1016/j.jaci.2021.03.044] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 02/20/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND The IL-33/ST2 pathway is linked with asthma susceptibility. Inhaled allergens, pollutants, and respiratory viruses, which trigger asthma exacerbations, induce release of IL-33, an epithelial-derived "alarmin." Astegolimab, a human IgG2 mAb, selectively inhibits the IL-33 receptor, ST2. Approved biologic therapies for severe asthma mainly benefit patients with elevated blood eosinophils (type 2-high), but limited options are available for patients with low blood eosinophils (type 2-low). Inhibiting IL-33 signaling may target pathogenic pathways in a wider spectrum of asthmatics. OBJECTIVES This study evaluated astegolimab efficacy and safety in patients with severe asthma. METHODS This double-blind, placebo-controlled, dose-ranging study (ZENYATTA [A Study to Assess the Efficacy and Safety of MSTT1041A in Participants With Uncontrolled Severe Asthma]) randomized 502 adults with severe asthma to subcutaneous placebo or 70-mg, 210-mg, or 490-mg doses of astegolimab every 4 weeks. The primary endpoint was the annualized asthma exacerbation rate (AER) at week 54. Enrollment caps ensured ∼30 patients who were eosinophil-high (≥300 cells/μL) and ∼95 patients who were eosinophil-low (<300 cells/μL) per arm. RESULTS Overall, adjusted AER reductions relative to placebo were 43% (P = .005), 22% (P = .18), and 37% (P = .01) for 490-mg, 210-mg, and 70-mg doses of astegolimab, respectively. Adjusted AER reductions for patients who were eosinophil-low were comparable to reductions in the overall population: 54% (P = .002), 14% (P = .48), and 35% (P = .05) for 490-mg, 210-mg, and 70-mg doses of astegolimab. Adverse events were similar in astegolimab- and placebo-treated groups. CONCLUSIONS Astegolimab reduced AER in a broad population of patients, including those who were eosinophil-low, with inadequately controlled, severe asthma. Astegolimab was safe and well tolerated.
Collapse
Affiliation(s)
- Steven G Kelsen
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pa
| | - Ioana O Agache
- Allergy and Clinical Immunology, Faculty of Medicine, Transylvania University, Brasov, Romania
| | - Weily Soong
- Alabama Allergy and Asthma Center and Clinical Research Center of Alabama, Birmingham, Ala
| | - Elliot Israel
- Divisions of Pulmonary and Critical Care Medicine and Allergy and Immunology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Mass
| | - Geoffrey L Chupp
- Division of Pulmonary and Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn
| | | | | | | | | | | | - Alice Fong
- Genentech, Inc., South San Francisco, Calif
| | - Ajit Dash
- Genentech, Inc., South San Francisco, Calif
| | | | | | | |
Collapse
|
424
|
Liu O, Xu J, Wang F, Jin W, Zanvit P, Wang D, Goldberg N, Cain A, Guo N, Han Y, Bynum A, Ma G, Wang S, Tang Z, Chen W. Adipose-mesenchymal stromal cells suppress experimental Sjögren syndrome by IL-33-driven expansion of ST2 + regulatory T cells. iScience 2021; 24:102446. [PMID: 33997712 PMCID: PMC8105666 DOI: 10.1016/j.isci.2021.102446] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/11/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Adipose-derived mesenchymal stromal cells (ADSCs) play important roles in the alleviation of inflammation and autoimmune diseases. Interleukin-33 (IL-33), a member of the IL-1 family, has been shown to regulate innate and adaptive immunity. However, it is still unknown whether ADSCs regulate immune responses via IL-33. We show here that ADSCs produced IL-33 in response to IL-1β stimulation, which depended on TAK1, ERK, and p38 pathways. ADSCs-derived IL-33 drove the proliferation of CD4+Foxp3+ST2+ regulatory T cells (Tregs) and alleviated experimental autoimmune Sjögren syndrome in mice. Importantly, human ADSCs also produced IL-33 in response to IL-1β. Thus, we have revealed a previously unrecognized immunoregulatory function of ADSCs by IL-33 production in experimental autoimmunity, which may have clinical applications for human immunopathology. Human and mouse ADSCs express IL-33 in response to IL-β stimulation mADSC-derived IL-33 inhibits inflammation in salivary glands in SS model mADSC-derived IL-33 expand ST2+ Tregs in vitro and in SS model
Collapse
Affiliation(s)
- Ousheng Liu
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Junji Xu
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Fu Wang
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
- Dalian Medical University, School of Stomatology, Dalian 116044, China
| | - Wenwen Jin
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Peter Zanvit
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Dandan Wang
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Nathan Goldberg
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Alexander Cain
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Nancy Guo
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Yichen Han
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Andrew Bynum
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Guowu Ma
- Dalian Medical University, School of Stomatology, Dalian 116044, China
| | - Songlin Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Zhangui Tang
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Wanjun Chen
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD 20892, USA
- Corresponding author
| |
Collapse
|
425
|
Oncel S, Gupta R, Wang Q, Basson MD. ZINC40099027 Promotes Gastric Mucosal Repair in Ongoing Aspirin-Associated Gastric Injury by Activating Focal Adhesion Kinase. Cells 2021; 10:908. [PMID: 33920786 PMCID: PMC8071155 DOI: 10.3390/cells10040908] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/06/2021] [Accepted: 04/13/2021] [Indexed: 12/23/2022] Open
Abstract
Nonsteroidal anti-inflammatory drugs cause gastric ulcers and gastritis. No drug that treats GI injury directly stimulates mucosal healing. ZINC40099027 (ZN27) activates focal adhesion kinase (FAK) and heals acute indomethacin-induced small bowel injury. We investigated the efficacy of ZN27 in rat and human gastric epithelial cells and ongoing aspirin-associated gastric injury. ZN27 (10 nM) stimulated FAK activation and wound closure in rat and human gastric cell lines. C57BL/6J mice were treated with 300 mg/kg/day aspirin for five days to induce ongoing gastric injury. One day after the initial injury, mice received 900 µg/kg/6 h ZN27, 10 mg/kg/day omeprazole, or 900 µg/kg/6 h ZN27 plus 10 mg/kg/day omeprazole. Like omeprazole, ZN27 reduced gastric injury vs. vehicle controls. ZN27-treated mice displayed better gastric architecture, with thicker mucosa and less hyperemia, inflammation, and submucosal edema, and lost less weight than vehicle controls. Gastric pH, serum creatinine, serum alanine aminotransferase (ALT), and renal and hepatic histology were unaffected by ZN27. Blinded scoring of pFAK-Y-397 immunoreactivity at the edge of ZN27-treated lesions demonstrated increased FAK activation, compared to vehicle-treated lesions, confirming target activation in vivo. These results suggest that ZN27 ameliorates ongoing aspirin-associated gastric mucosal injury by a pathway involving FAK activation. ZN27-derivatives may be useful to promote gastric mucosal repair.
Collapse
Affiliation(s)
- Sema Oncel
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58203, USA;
| | - Rashmi Gupta
- Department of Surgery, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58203, USA; (R.G.); (Q.W.)
| | - Qinggang Wang
- Department of Surgery, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58203, USA; (R.G.); (Q.W.)
| | - Marc D. Basson
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58203, USA;
- Department of Surgery, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58203, USA; (R.G.); (Q.W.)
- Department of Pathology, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND 58203, USA
| |
Collapse
|
426
|
Saikumar Jayalatha AK, Hesse L, Ketelaar ME, Koppelman GH, Nawijn MC. The central role of IL-33/IL-1RL1 pathway in asthma: From pathogenesis to intervention. Pharmacol Ther 2021; 225:107847. [PMID: 33819560 DOI: 10.1016/j.pharmthera.2021.107847] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/18/2021] [Indexed: 02/06/2023]
Abstract
Interleukin-33 (IL-33), a member of the IL-1 family, and its cognate receptor, Interleukin-1 receptor like-1 (IL-1RL1 or ST2), are susceptibility genes for childhood asthma. In response to cellular damage, IL-33 is released from barrier tissues as an 'alarmin' to activate the innate immune response. IL-33 drives type 2 responses by inducing signalling through its receptor IL-1RL1 in several immune and structural cells, thereby leading to type 2 cytokine and chemokine production. IL-1RL1 gene transcript encodes different isoforms generated through alternative splicing. Its soluble isoform, IL-1RL1-a or sST2, acts as a decoy receptor by sequestering IL-33, thereby inhibiting IL1RL1-b/IL-33 signalling. IL-33 and its receptor IL-1RL1 are therefore considered as putative biomarkers or targets for pharmacological intervention in asthma. This review will provide an overview of the genetics and biology of the IL-33/IL-1RL1 pathway in the context of asthma pathogenesis. It will discuss the potential and complexities of targeting the cytokine or its receptor, how genetics or biomarkers may inform precision medicine for asthma targeting this pathway, and the possible positioning of therapeutics targeting IL-33 or its receptor in the expanding landscape of novel biologicals applied in asthma management.
Collapse
Affiliation(s)
- A K Saikumar Jayalatha
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
| | - L Hesse
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
| | - M E Ketelaar
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Beatrix Children's Hospital, Department of Paediatric Pulmonology and Paediatric Allergology, Groningen, the Netherlands
| | - G H Koppelman
- University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Beatrix Children's Hospital, Department of Paediatric Pulmonology and Paediatric Allergology, Groningen, the Netherlands
| | - M C Nawijn
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Laboratory of Experimental Pulmonology and Inflammation Research (EXPIRE), Groningen, the Netherlands; University of Groningen University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands.
| |
Collapse
|
427
|
Norlander AE, Bloodworth MH, Toki S, Zhang J, Zhou W, Boyd K, Polosukhin VV, Cephus JY, Ceneviva ZJ, Gandhi VD, Chowdhury NU, Charbonnier LM, Rogers LM, Wang J, Aronoff DM, Bastarache L, Newcomb DC, Chatila TA, Peebles RS. Prostaglandin I2 signaling licenses Treg suppressive function and prevents pathogenic reprogramming. J Clin Invest 2021; 131:140690. [PMID: 33529171 PMCID: PMC8011897 DOI: 10.1172/jci140690] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 01/27/2021] [Indexed: 12/29/2022] Open
Abstract
Tregs restrain both the innate and adaptive immune systems to maintain homeostasis. Allergic airway inflammation, characterized by a Th2 response that results from a breakdown of tolerance to innocuous environmental antigens, is negatively regulated by Tregs. We previously reported that prostaglandin I2 (PGI2) promoted immune tolerance in models of allergic inflammation; however, the effect of PGI2 on Treg function was not investigated. Tregs from mice deficient in the PGI2 receptor IP (IP KO) had impaired suppressive capabilities during allergic airway inflammatory responses compared with mice in which PGI2 signaling was intact. IP KO Tregs had significantly enhanced expression of immunoglobulin-like transcript 3 (ILT3) compared with WT Tregs, which may contribute to the impairment of the IP KO Treg's ability to suppress Th2 responses. Using fate-mapping mice, we reported that PGI2 signaling prevents Treg reprogramming toward a pathogenic phenotype. PGI2 analogs promoted the differentiation of naive T cells to Tregs in both mice and humans via repression of β-catenin signaling. Finally, a missense variant in IP in humans was strongly associated with chronic obstructive asthma. Together, these data support that PGI2 signaling licenses Treg suppressive function and that PGI2 is a therapeutic target for enhancing Treg function.
Collapse
Affiliation(s)
| | | | - Shinji Toki
- Division of Allergy, Pulmonary, and Critical Care Medicine and
| | - Jian Zhang
- Division of Allergy, Pulmonary, and Critical Care Medicine and
| | - Weisong Zhou
- Division of Allergy, Pulmonary, and Critical Care Medicine and
| | - Kelli Boyd
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | | | | - Vivek D. Gandhi
- Division of Allergy, Pulmonary, and Critical Care Medicine and
| | - Nowrin U. Chowdhury
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Louis-Marie Charbonnier
- Division of Immunology, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Janey Wang
- Department of Biomedical Informatics, and
| | - David M. Aronoff
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Division of Infectious Diseases, Department of Medicine
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | | | - Dawn C. Newcomb
- Division of Allergy, Pulmonary, and Critical Care Medicine and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Talal A. Chatila
- Division of Immunology, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - R. Stokes Peebles
- Division of Allergy, Pulmonary, and Critical Care Medicine and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- United States Department of Veterans Affairs, Nashville, Tennessee, USA
| |
Collapse
|
428
|
Wang J, Tang W, Yang M, Yin Y, Li H, Hu F, Tang L, Ma X, Zhang Y, Wang Y. Inflammatory tumor microenvironment responsive neutrophil exosomes-based drug delivery system for targeted glioma therapy. Biomaterials 2021; 273:120784. [PMID: 33848731 DOI: 10.1016/j.biomaterials.2021.120784] [Citation(s) in RCA: 164] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022]
Abstract
Clinical treatment of malignant glioma remains a major challenge due to high infiltrative growth and chemotherapeutic resistance of tumors and the presence of the blood brain barrier (BBB). Advanced nanoplatforms that can efficiently cross the BBB and target to brain tumor are urgently needed. Encouraged by the intrinsic inflammatory chemotaxis and excellent BBB-crossing capability of neutrophils, a bioinspired neutrophil-exosomes (NEs-Exos) system for delivering loaded doxorubicin (DOX) drug for glioma treatment is proposed and systematically investigated. In vivo zebrafish and C6-Luc glioma-bearing mice models show that NEs-Exos carrying the drug rapidly penetrate the BBB and migrate into the brain. Additionally, a transwell BBB model and mouse brain inflammatory study show that NEs-Exos can respond chemotactically to inflammatory stimuli and target infiltrating tumor cells in inflamed brain tumors. Moreover, intravenous injection of NEs-Exos/DOX efficiently suppress tumor growth and prolong survival time in a glioma mouse model. On the basis of these results, NEs-Exos are confirmed to have neutrophil-like chemotactic function and BBB penetration. This novel NEs-Exos/DOX delivery platform represents a promising chemotherapeutic approach for clinical treatment of glioma and other solid tumor or brain diseases.
Collapse
Affiliation(s)
- Jun Wang
- School of Medicine, Chongqing University, Chongqing, 400044, China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Wei Tang
- Key Laboratory of Human-Machine-Intelligence Synergic Systems, Research Center for Neural Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Meng Yang
- School of Medicine, Chongqing University, Chongqing, 400044, China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Ying Yin
- School of Medicine, Chongqing University, Chongqing, 400044, China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Hui Li
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Fangfang Hu
- School of Medicine, Chongqing University, Chongqing, 400044, China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Lin Tang
- School of Medicine, Chongqing University, Chongqing, 400044, China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Xiaoyue Ma
- School of Medicine, Chongqing University, Chongqing, 400044, China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yu Zhang
- School of Medicine, Chongqing University, Chongqing, 400044, China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yazhou Wang
- School of Medicine, Chongqing University, Chongqing, 400044, China; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
429
|
Thomas CN, Sim DA, Lee WH, Alfahad N, Dick AD, Denniston AK, Hill LJ. Emerging therapies and their delivery for treating age-related macular degeneration. Br J Pharmacol 2021; 179:1908-1937. [PMID: 33769566 DOI: 10.1111/bph.15459] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/11/2021] [Accepted: 03/14/2021] [Indexed: 12/13/2022] Open
Abstract
Age-related macular degeneration (AMD) is the most common cause of blindness in the Western world and is characterised in its latter stages by retinal cell death and neovascularisation and earlier stages with the loss of parainflammatory homeostasis. Patients with neovascular AMD (nAMD) are treated with frequent intraocular injections of anti-vascular endothelial growth factor (VEGF) therapies, which are not only unpopular with patients but carry risks of sight-threatening complications. A minority of patients are unresponsive with no alternative treatment available, and some patients who respond initially eventually develop a tolerance to treatment. New therapeutics with improved delivery methods and sustainability of clinical effects are required, in particular for non-neovascular AMD (90% of cases and no current approved treatments). There are age-related and disease-related changes that occur which can affect ocular drug delivery. Here, we review the latest emerging therapies for AMD, their delivery routes and implications for translating to clinical practice.
Collapse
Affiliation(s)
- Chloe N Thomas
- School of Biomedical Sciences, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Dawn A Sim
- Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK.,National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital and University College London Institute of Ophthalmology, London, UK
| | - Wen Hwa Lee
- Action Against AMD, London, UK.,Affordable Medicines Programme, Oxford Martin School, University of Oxford, Oxford, UK
| | - Nada Alfahad
- School of Biomedical Sciences, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Andrew D Dick
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital and University College London Institute of Ophthalmology, London, UK.,Academic Unit of Ophthalmology, Bristol Medical School and School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Alastair K Denniston
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital and University College London Institute of Ophthalmology, London, UK.,Academic Unit of Ophthalmology, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Department of Ophthalmology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.,Centre for Patient Reported Outcome Research, Institute of Applied Health Research, University of Birmingham, Birmingham, UK.,Birmingham Health Partners Centre for Regulatory Science and Innovation, University of Birmingham, Birmingham, UK.,Health Data Research UK, London, UK
| | - Lisa J Hill
- School of Biomedical Sciences, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
430
|
Ochayon DE, Waggoner SN. The Effect of Unconventional Cytokine Combinations on NK-Cell Responses to Viral Infection. Front Immunol 2021; 12:645850. [PMID: 33815404 PMCID: PMC8017335 DOI: 10.3389/fimmu.2021.645850] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 03/01/2021] [Indexed: 12/30/2022] Open
Abstract
Cytokines are soluble and membrane-bound factors that dictate immune responses. Dogmatically, cytokines are divided into families that promote type 1 cell-mediated immune responses (e.g., IL-12) or type 2 humoral responses (e.g., IL-4), each capable of antagonizing the opposing family of cytokines. The discovery of additional families of cytokines (e.g., IL-17) has added complexity to this model, but it was the realization that immune responses frequently comprise mixtures of different types of cytokines that dismantled this black-and-white paradigm. In some cases, one type of response may dominate these mixed milieus in disease pathogenesis and thereby present a clear therapeutic target. Alternatively, synergistic or blended cytokine responses may obfuscate the origins of disease and perplex clinical decision making. Most immune cells express receptors for many types of cytokines and can mediate a myriad of functions important for tolerance, immunity, tissue damage, and repair. In this review, we will describe the unconventional effects of a variety of cytokines on the activity of a prototypical type 1 effector, the natural killer (NK) cell, and discuss how this may impact the contributions of these cells to health and disease.
Collapse
Affiliation(s)
- David E. Ochayon
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Stephen N. Waggoner
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
431
|
Oshimori N, Guo Y, Taniguchi S. An emerging role for cellular crosstalk in the cancer stem cell niche. J Pathol 2021; 254:384-394. [DOI: 10.1002/path.5655] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/12/2021] [Accepted: 02/23/2021] [Indexed: 12/17/2022]
Affiliation(s)
- Naoki Oshimori
- Department of Cell, Developmental and Cancer Biology Oregon Health & Science University Portland OR USA
- Department of Dermatology Oregon Health & Science University Portland OR USA
- Department of Otolaryngology – Head & Neck Surgery Oregon Health & Science University Portland OR USA
- Knight Cancer Institute Oregon Health & Science University Portland OR USA
| | - Yifei Guo
- Department of Cell, Developmental and Cancer Biology Oregon Health & Science University Portland OR USA
| | - Sachiko Taniguchi
- Department of Cell, Developmental and Cancer Biology Oregon Health & Science University Portland OR USA
| |
Collapse
|
432
|
Chu C, Parkhurst CN, Zhang W, Zhou L, Yano H, Arifuzzaman M, Artis D. The ChAT-acetylcholine pathway promotes group 2 innate lymphoid cell responses and anti-helminth immunity. Sci Immunol 2021; 6:6/57/eabe3218. [PMID: 33674322 DOI: 10.1126/sciimmunol.abe3218] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 12/13/2020] [Accepted: 02/01/2021] [Indexed: 12/11/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) reside in multiple tissues, including lymphoid organs and barrier surfaces, and secrete type 2 cytokines including interleukin-5 (IL-5), IL-9, and IL-13. These cells participate in multiple physiological processes including allergic inflammation, tissue repair, metabolic homeostasis, and host defense against helminth infections. Recent studies indicate that neurotransmitters and neuropeptides can play an important role in regulating ILC2 responses; however, the mechanisms that underlie these processes in vivo remain incompletely defined. Here, we identify that activated ILC2s up-regulate choline acetyltransferase (ChAT)-the enzyme responsible for the biosynthesis of acetylcholine (ACh)-after infection with the helminth parasite Nippostrongylus brasiliensis or treatment with alarmins or cytokines including IL-25, IL-33, and thymic stromal lymphopoietin (TSLP). ILC2s also express acetylcholine receptors (AChRs), and ACh administration promotes ILC2 cytokine production and elicits expulsion of helminth infection. In accordance with this, ChAT deficiency in ILC2s leads to defective ILC2 responses and impaired immunity against helminth infection. Together, these results reveal a previously unrecognized role of the ChAT-ACh pathway in promoting type 2 innate immunity to helminth infection.
Collapse
Affiliation(s)
- Coco Chu
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Christopher N Parkhurst
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Wen Zhang
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Lei Zhou
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Hiroshi Yano
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Mohammad Arifuzzaman
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA. .,Friedman Center for Nutrition and Inflammation, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| |
Collapse
|
433
|
Carrero YN, Callejas DE, Mosquera JA. In situ immunopathological events in human cervical intraepithelial neoplasia and cervical cancer: Review. Transl Oncol 2021; 14:101058. [PMID: 33677234 PMCID: PMC7937982 DOI: 10.1016/j.tranon.2021.101058] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
Neoplasia of the cervix represents one of the most common cancers in women. Clinical and molecular research has identified immunological impairment in squamous intraepithelial cervical lesions and cervical cancer patients. The in-situ expression of several cytokines by uterine epithelial cells and by infiltrating leukocytes occurs during the cervical intraepithelial neoplasia and cervical cancer. Some of these cytokines can prevent and others can induce the progression of the neoplasm. The infiltrating leukocytes also produce cytokines and growth factors relate to angiogenesis, chemotaxis, and apoptosis capable of modulating the dysplasia progression. In this review we analyzed several interleukins with an inductive effect or blocking effect on the neoplastic progression. We also analyze the genetic polymorphism of some cytokines and their relationship with the risk of developing cervical neoplasia. In addition, we describe the leukocyte cells that infiltrate the cervical uterine tissue during the neoplasia and their effects on neoplasia progression.
Collapse
Affiliation(s)
- Yenddy N Carrero
- Facultad de Ciencias de la Salud. Carrera de Medicina, Universidad Técnica de Ambato, Ambato, Ecuador.
| | - Diana E Callejas
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Salud, Universidad Técnica de Manabí, Portoviejo, Ecuador.
| | - Jesús A Mosquera
- Instituto de Investigaciones Clínicas Dr. Américo Negrette. Facultad de Medicina, Universidad del Zulia. Maracaibo, Venezuela.
| |
Collapse
|
434
|
Swann JW, Koneva LA, Regan-Komito D, Sansom SN, Powrie F, Griseri T. IL-33 promotes anemia during chronic inflammation by inhibiting differentiation of erythroid progenitors. J Exp Med 2021; 217:151849. [PMID: 32520308 PMCID: PMC7478740 DOI: 10.1084/jem.20200164] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/10/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022] Open
Abstract
An important comorbidity of chronic inflammation is anemia, which may be related to dysregulated activity of hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM). Among HSPCs, we found that the receptor for IL-33, ST2, is expressed preferentially and highly on erythroid progenitors. Induction of inflammatory spondyloarthritis in mice increased IL-33 in BM plasma, and IL-33 was required for inflammation-dependent suppression of erythropoiesis in BM. Conversely, administration of IL-33 in healthy mice suppressed erythropoiesis, decreased hemoglobin expression, and caused anemia. Using purified erythroid progenitors in vitro, we show that IL-33 directly inhibited terminal maturation. This effect was dependent on NF-κB activation and associated with altered signaling events downstream of the erythropoietin receptor. Accordingly, IL-33 also suppressed erythropoietin-accelerated erythropoiesis in vivo. These results reveal a role for IL-33 in pathogenesis of anemia during inflammatory disease and define a new target for its treatment.
Collapse
Affiliation(s)
- James W Swann
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Lada A Koneva
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | - Stephen N Sansom
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Thibault Griseri
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
435
|
Griffiths JS, Camilli G, Kotowicz NK, Ho J, Richardson JP, Naglik JR. Role for IL-1 Family Cytokines in Fungal Infections. Front Microbiol 2021; 12:633047. [PMID: 33643264 PMCID: PMC7902786 DOI: 10.3389/fmicb.2021.633047] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
Fungal pathogens kill approximately 1.5 million individuals per year and represent a severe disease burden worldwide. It is estimated over 150 million people have serious fungal disease such as recurrent mucosal infections or life-threatening systemic infections. Disease can ensue from commensal fungi or new infection and involves different fungal morphologies and the expression of virulence factors. Therefore, anti-fungal immunity is complex and requires coordination between multiple facets of the immune system. IL-1 family cytokines are associated with acute and chronic inflammation and are essential for the innate response to infection. Recent research indicates IL-1 cytokines play a key role mediating immunity against different fungal infections. During mucosal disease, IL-1R and IL-36R are required for neutrophil recruitment and protective Th17 responses, but function through different mechanisms. During systemic disease, IL-18 drives protective Th1 responses, while IL-33 promotes Th2 and suppresses Th1 immunity. The IL-1 family represents an attractive anti-fungal immunotherapy target. There is a need for novel anti-fungal therapeutics, as current therapies are ineffective, toxic and encounter resistance, and no anti-fungal vaccine exists. Furthering our understanding of the IL-1 family cytokines and their complex role during fungal infection may aid the development of novel therapies. As such, this review will discuss the role for IL-1 family cytokines in fungal infections.
Collapse
Affiliation(s)
- James S Griffiths
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Giorgio Camilli
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Natalia K Kotowicz
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Jemima Ho
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Jonathan P Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
436
|
Ogawa K, Asano K, Yotsumoto S, Yamane T, Arita M, Hayashi Y, Harada H, Makino-Okamura C, Fukuyama H, Kondo K, Yamasoba T, Tanaka M. Frontline Science: Conversion of neutrophils into atypical Ly6G + SiglecF + immune cells with neurosupportive potential in olfactory neuroepithelium. J Leukoc Biol 2021; 109:481-496. [PMID: 32725843 DOI: 10.1002/jlb.1hi0620-190rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
Neutrophils are generally considered as short-lived, homogenous, and terminally differentiated phagocytes that play crucial roles in conquering infection, although they occasionally cause severe collateral tissue damage or chronic inflammation. Recent reports have indicated that neutrophils also play a protective role in inflammation resolution and tissue repair. However, how terminally differentiated neutrophils have diverse functions remains unclear. Here, we show that neutrophils undergo conversion into Ly6G+ SiglecF+ double-positive cells expressing neurosupportive genes in the olfactory neuroepithelium (OE) under an inflammatory state. Through comprehensive flow cytometric analysis of murine nose, we identified Ly6G+ SiglecF+ double-positive cells that reside only in the OE under steady-state conditions. Double-positive cells were neutrophil-derived cells and increased by more than 10-fold during inflammation or tissue injury. We found that neutrophils infiltrate into the nose to express proinflammatory genes in the acute phase of inflammatory state, and they gradually change their surface markers and gene expression, expressing some neurogenesis-related genes in addition to inflammation related genes in the later phase. As the OE is known to have exceptionally high regeneration capacity as a nervous system, these findings suggest that neutrophils have the potential to contribute neurogenesis after conversion in peripheral nervous tissues, providing a challenge on a classic view of neutrophils as terminally differentiated leukocytes.
Collapse
Affiliation(s)
- Kei Ogawa
- Laboratory of Immune Regulation, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
- Department of Otolaryngology - Head and Neck Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Kenichi Asano
- Laboratory of Immune Regulation, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Satoshi Yotsumoto
- Laboratory of Immune Regulation, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Tsuyoshi Yamane
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Division of Gastroenterology & Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Yoshihiro Hayashi
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Hironori Harada
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Chieko Makino-Okamura
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hidehiro Fukuyama
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kenji Kondo
- Department of Otolaryngology - Head and Neck Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tatsuya Yamasoba
- Department of Otolaryngology - Head and Neck Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Masato Tanaka
- Laboratory of Immune Regulation, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| |
Collapse
|
437
|
Turning off the alarm - Targeting alarmins and other epithelial mediators of allergic inflammation with biologics. Allergol Select 2021; 5:82-88. [PMID: 33615121 PMCID: PMC7890934 DOI: 10.5414/alx02194e] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/28/2021] [Indexed: 01/15/2023] Open
Abstract
Besides the major players IL-4, IL-13, IL-5, and IgE as targets for biologics, other mediators have been identified that are secreted by epithelial cells and act upstream in the cascade of allergic inflammation. Such are the alarmin IL-33 as well as TSLP and IL-5. The role of each cytokine in sensitization and effector phase of allergic inflammation and how development of biologics is ongoing in order to inhibit this pathomechanism will be described in the following article.
Collapse
|
438
|
Dwyer GK, Turnquist HR. Untangling Local Pro-Inflammatory, Reparative, and Regulatory Damage-Associated Molecular-Patterns (DAMPs) Pathways to Improve Transplant Outcomes. Front Immunol 2021; 12:611910. [PMID: 33708206 PMCID: PMC7940545 DOI: 10.3389/fimmu.2021.611910] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/05/2021] [Indexed: 12/28/2022] Open
Abstract
Detrimental inflammatory responses after solid organ transplantation are initiated when immune cells sense pathogen-associated molecular patterns (PAMPs) and certain damage-associated molecular patterns (DAMPs) released or exposed during transplant-associated processes, such as ischemia/reperfusion injury (IRI), surgical trauma, and recipient conditioning. These inflammatory responses initiate and propagate anti-alloantigen (AlloAg) responses and targeting DAMPs and PAMPs, or the signaling cascades they activate, reduce alloimmunity, and contribute to improved outcomes after allogeneic solid organ transplantation in experimental studies. However, DAMPs have also been implicated in initiating essential anti-inflammatory and reparative functions of specific immune cells, particularly Treg and macrophages. Interestingly, DAMP signaling is also involved in local and systemic homeostasis. Herein, we describe the emerging literature defining how poor outcomes after transplantation may result, not from just an over-abundance of DAMP-driven inflammation, but instead an inadequate presence of a subset of DAMPs or related molecules needed to repair tissue successfully or re-establish tissue homeostasis. Adverse outcomes may also arise when these homeostatic or reparative signals become dysregulated or hijacked by alloreactive immune cells in transplant niches. A complete understanding of the critical pathways controlling tissue repair and homeostasis, and how alloimmune responses or transplant-related processes disrupt these will lead to new immunotherapeutics that can prevent or reverse the tissue pathology leading to lost grafts due to chronic rejection.
Collapse
Affiliation(s)
- Gaelen K Dwyer
- Departments of Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Hēth R Turnquist
- Departments of Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
439
|
Pinto DO, Al Sharif S, Mensah G, Cowen M, Khatkar P, Erickson J, Branscome H, Lattanze T, DeMarino C, Alem F, Magni R, Zhou W, Alais S, Dutartre H, El-Hage N, Mahieux R, Liotta LA, Kashanchi F. Extracellular vesicles from HTLV-1 infected cells modulate target cells and viral spread. Retrovirology 2021; 18:6. [PMID: 33622348 PMCID: PMC7901226 DOI: 10.1186/s12977-021-00550-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The Human T-cell Lymphotropic Virus Type-1 (HTLV-1) is a blood-borne pathogen and etiological agent of Adult T-cell Leukemia/Lymphoma (ATLL) and HTLV-1 Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP). HTLV-1 has currently infected up to 10 million globally with highly endemic areas in Japan, Africa, the Caribbean and South America. We have previously shown that Extracellular Vesicles (EVs) enhance HTLV-1 transmission by promoting cell-cell contact. RESULTS Here, we separated EVs into subpopulations using differential ultracentrifugation (DUC) at speeds of 2 k (2000×g), 10 k (10,000×g), and 100 k (100,000×g) from infected cell supernatants. Proteomic analysis revealed that EVs contain the highest viral/host protein abundance in the 2 k subpopulation (2 k > 10 k > 100 k). The 2 k and 10 k populations contained viral proteins (i.e., p19 and Tax), and autophagy proteins (i.e., LC3 and p62) suggesting presence of autophagosomes as well as core histones. Interestingly, the use of 2 k EVs in an angiogenesis assay (mesenchymal stem cells + endothelial cells) caused deterioration of vascular-like-tubules. Cells commonly associated with the neurovascular unit (i.e., astrocytes, neurons, and macrophages) in the blood-brain barrier (BBB) showed that HTLV-1 EVs may induce expression of cytokines involved in migration (i.e., IL-8; 100 k > 2 k > 10 k) from astrocytes and monocyte-derived macrophages (i.e., IL-8; 2 k > 10 k). Finally, we found that EVs were able to promote cell-cell contact and viral transmission in monocytic cell-derived dendritic cell. The EVs from both 2 k and 10 k increased HTLV-1 spread in a humanized mouse model, as evidenced by an increase in proviral DNA and RNA in the Blood, Lymph Node, and Spleen. CONCLUSIONS Altogether, these data suggest that various EV subpopulations induce cytokine expression, tissue damage, and viral spread.
Collapse
Affiliation(s)
- Daniel O Pinto
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Sarah Al Sharif
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Gifty Mensah
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Maria Cowen
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Pooja Khatkar
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - James Erickson
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Heather Branscome
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Thomas Lattanze
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Catherine DeMarino
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Farhang Alem
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Ruben Magni
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Weidong Zhou
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Sandrine Alais
- International Center for Research in Infectiology, Retroviral Oncogenesis Laboratory, INSERM U1111-Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université de Lyon, Fondation Pour La Recherche Médicale, Labex Ecofect, Lyon, France
| | - Hélène Dutartre
- International Center for Research in Infectiology, Retroviral Oncogenesis Laboratory, INSERM U1111-Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université de Lyon, Fondation Pour La Recherche Médicale, Labex Ecofect, Lyon, France
| | - Nazira El-Hage
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Renaud Mahieux
- International Center for Research in Infectiology, Retroviral Oncogenesis Laboratory, INSERM U1111-Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université de Lyon, Fondation Pour La Recherche Médicale, Labex Ecofect, Lyon, France
| | - Lance A Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, USA
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA.
| |
Collapse
|
440
|
Katz-Kiriakos E, Steinberg DF, Kluender CE, Osorio OA, Newsom-Stewart C, Baronia A, Byers DE, Holtzman MJ, Katafiasz D, Bailey KL, Brody SL, Miller MJ, Alexander-Brett J. Epithelial IL-33 appropriates exosome trafficking for secretion in chronic airway disease. JCI Insight 2021; 6:136166. [PMID: 33507882 PMCID: PMC7934940 DOI: 10.1172/jci.insight.136166] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 01/15/2021] [Indexed: 02/06/2023] Open
Abstract
IL-33 is a key mediator of chronic airway disease driven by type 2 immune pathways, yet the nonclassical secretory mechanism for this cytokine remains undefined. We performed a comprehensive analysis in human airway epithelial cells, which revealed that tonic IL-33 secretion is dependent on the ceramide biosynthetic enzyme neutral sphingomyelinase 2 (nSMase2). IL-33 is cosecreted with exosomes by the nSMase2-regulated multivesicular endosome (MVE) pathway as surface-bound cargo. In support of these findings, human chronic obstructive pulmonary disease (COPD) specimens exhibited increased epithelial expression of the abundantly secreted IL33Δ34 isoform and augmented nSMase2 expression compared with non-COPD specimens. Using an Alternaria-induced airway disease model, we found that the nSMase2 inhibitor GW4869 abrogated both IL-33 and exosome secretion as well as downstream inflammatory pathways. This work elucidates a potentially novel aspect of IL-33 biology that may be targeted for therapeutic benefit in chronic airway diseases driven by type 2 inflammation.
Collapse
Affiliation(s)
- Ella Katz-Kiriakos
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, and
| | - Deborah F Steinberg
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, and
| | - Colin E Kluender
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, and
| | - Omar A Osorio
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, and
| | | | - Arjun Baronia
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, and
| | - Derek E Byers
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, and
| | - Michael J Holtzman
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, and.,Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dawn Katafiasz
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Kristina L Bailey
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Steven L Brody
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, and
| | - Mark J Miller
- Department of Medicine, Division of Infectious Diseases, and
| | - Jennifer Alexander-Brett
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, and.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
441
|
Li M, Duan L, Cai Y, Hao B, Chen J, Li H, Liu H. Prognostic value of soluble suppression of tumorigenesis-2 (sST2) for cardiovascular events in coronary artery disease patients with and without diabetes mellitus. Cardiovasc Diabetol 2021; 20:49. [PMID: 33608010 PMCID: PMC7896409 DOI: 10.1186/s12933-021-01244-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/13/2021] [Indexed: 11/10/2022] Open
Abstract
Background Soluble suppression of tumorigenesis-2 (sST2) is implicated in myocardial overload and has long been recognized as an inflammatory marker related to heart failure and acute coronary syndrome, but data on the prognostic value of sST2 in patients with coronary artery disease (CAD) remain limited. This study sought to investigate the prognostic value of sST2 in patients with established CAD and its predictive value in CAD patients with and without type 2 diabetes mellitus (T2DM). Methods A total of 3641 consecutive patients were included in this prospective cohort study. The primary end point was major adverse cardiovascular events (MACEs). The secondary end point was all-cause death. The association between sST2 and outcomes was investigated using multivariable Cox regression. Results During a median follow-up of 6.4 years, MACEs occurred in 775 patients, and 275 patients died. Multiple Cox regression models showed that a higher level of sST2 was an independent predictor of MACEs development (HR = 1.36, 95% CI 1.17–1.56, p < 0.001) and all-cause death (HR = 2.01, 95% CI 1.56–2.59, p < 0.001). The addition of sST2 to established risk factors significantly improved risk prediction of the composite outcome of MACEs and all-cause death (C-index, net reclassification index, and integrated discrimination improvement, all p < 0.05). In subgroup analysis depending on diabetes status, the diabetes group had a significantly higher level of sST2, which remained a significant predictor of MACEs and all-cause death in patients with and without T2DM in multivariable models. The area under the curve (AUC) of CAD patients with diabetes mellitus was significantly higher than that of those without T2DM. For MACEs, the AUC was 0.737 (patients with T2DM) vs 0.620 (patients without T2DM). For all-cause death, the AUC was 0.923 (patients with T2DM) vs 0.789 (patients without T2DM). Conclusions A higher level of sST2 is significantly associated with long-term MACEs and all-cause death in CAD patients with and without T2DM. sST2 has strong predictive value for cardiovascular adverse events in CAD patients with T2DM, and these results provide new evidence for the role of sST2.
Collapse
Affiliation(s)
- Man Li
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, No. 28, Fu Xing Road, Hai Dian, Beijing, China
| | - Lei Duan
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, No. 28, Fu Xing Road, Hai Dian, Beijing, China
| | - Yulun Cai
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, No. 28, Fu Xing Road, Hai Dian, Beijing, China
| | - Benchuan Hao
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, No. 28, Fu Xing Road, Hai Dian, Beijing, China
| | - Jianqiao Chen
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, No. 28, Fu Xing Road, Hai Dian, Beijing, China
| | - Huiying Li
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, No. 28, Fu Xing Road, Hai Dian, Beijing, China
| | - Hongbin Liu
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, No. 28, Fu Xing Road, Hai Dian, Beijing, China. .,Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Beijing, China.
| |
Collapse
|
442
|
Peng L, Sun W, Wei F, Chen L, Wen W. Interleukin-33 modulates immune responses in cutaneous melanoma in a context-specific way. Aging (Albany NY) 2021; 13:6740-6751. [PMID: 33621202 PMCID: PMC7993738 DOI: 10.18632/aging.202531] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/29/2020] [Indexed: 06/12/2023]
Abstract
Controversial roles of interleukin-33 (IL-33) have been reported in melanoma from animal studies. We aimed to investigate the role of IL-33 in human cutaneous melanoma. RNA-seq data of 471 cases of cutaneous melanoma were retrieved from The Cancer Genome Atlas. The tumor microenvironment (TME) was deconstructed by the xCell algorithm using RNA-seq data. We evaluated the prognostic value of IL-33 and the relationship between IL-33 and immune components in TME. We also inferred the potential cellular sources of IL-33. All the analyses were conducted separately in three sub-cohorts, which are based on the biopsy sites of samples: primary melanoma; lymph node (LN) metastases; other metastases, including metastases to skin/soft tissue, or visceral sites. In the two metastasis sub-cohorts, IL-33 is associated with better prognosis and more active immune responses in the tumor. However, IL-33 is not a prognostic factor in the primary melanoma sub-cohort. Furthermore, we found that IL-33 is mainly derived from stromal cells in the metastasis sub-cohorts, and from epithelial cells/keratinocytes in the primary melanoma sub-cohort. These findings provide evidence for the context-specific anti-tumor effects of IL-33 in melanoma. And the distinct effects of IL-33 may be determined by the cellular sources of IL-33.
Collapse
Affiliation(s)
- Liang Peng
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Wei Sun
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Fanqin Wei
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Lin Chen
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Weiping Wen
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- Institute of Otorhinolaryngology Head and Neck Surgery, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
- Department of Otorhinolaryngology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong, China
| |
Collapse
|
443
|
Leon G, Hussey S, Walsh PT. The Diverse Roles of the IL-36 Family in Gastrointestinal Inflammation and Resolution. Inflamm Bowel Dis 2021; 27:440-450. [PMID: 32860042 DOI: 10.1093/ibd/izaa232] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Indexed: 12/16/2022]
Abstract
The interleukin (IL)-36 family is a member of the IL-1 superfamily of cytokines and, in common with other IL-1 family members, has been shown to exhibit pleiotropic effects in homeostasis and inflammation. Although the important role these cytokines play in the skin has been widely reported, recent evidence suggests that IL-36 family members are expressed and can also exert significant influence at the intestinal mucosa. In this review, we summarize current knowledge surrounding the role of the IL-36 in the intestines. In particular, we examine its likely dichotomous role as a mediator of both inflammation and resolution, highlighting its overlapping roles in innate and adaptive inflammation at the mucosa and its contribution to pathophysiology of inflammatory bowel disease. We also summarize the complexities of targeting this cytokine family in a clinical setting.
Collapse
Affiliation(s)
- Gemma Leon
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland.,National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | - Seamus Hussey
- National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | - Patrick T Walsh
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland.,National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| |
Collapse
|
444
|
Huang Q, Li CD, Yang YR, Qin XF, Wang JJ, Zhang X, Du XN, Yang X, Wang Y, Li L, Mu M, Lv Z, Cui Y, Huang K, Corrigan CJ, Wang W, Ying S. Role of the IL-33/ST2 axis in cigarette smoke-induced airways remodelling in chronic obstructive pulmonary disease. Thorax 2021; 76:thoraxjnl-2020-214712. [PMID: 33589512 DOI: 10.1136/thoraxjnl-2020-214712] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 01/17/2021] [Accepted: 01/23/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Efficient therapy and potential prophylaxis are confounded by current ignorance of the pathogenesis of airway remodelling and blockade in COPD. OBJECTIVE To explore the role of the IL-33/ST2 axis in cigarette smoke (CS) exposure-induced airways remodelling. METHODS C57BL/6, BALB/c and IL-1RL1 -/- mice exposed to CS were used to establish an animal surrogate of COPD (air-exposed=5~8, CS-exposed=6~12). Hallmarks of remodelling were measured in mice. Cigarette smoke extract (CSE)-induced proliferation and protein production in vitro by fibroblasts in the presence of anti-interleukin-33 (anti-IL-33) or hST2 antibodies were measured. Expression of IL-33 and ST2 and other remodelling hallmarks were measured, respectively, in bronchoalveolar lavage fluid (BALF) (controls=20, COPD=20), serum (controls=59, COPD=90) and lung tissue sections (controls=11, COPD=7) from patients with COPD and controls. RESULTS Wild-type mice exposed to CS elevated expression of hallmarks of tissue remodelling in the lungs and also in the heart, spleen and kidneys, which were significantly abrogated in the IL-1RL1 -/- mice. Fibroblasts exposed to CSE, compared with control, exhibited early cellular translocation of IL-33, accompanied by proliferation and elevated protein synthesis, all inhabitable by blockade of IL-33/ST2 signalling. Expression of IL-33 and ST2 and hallmarks of tissue remodelling were significantly and proportionally elevated in BALF, serum and tissue samples from patients with COPD. CONCLUSIONS Exposure to CS induces remodelling changes in multiple organs. The data support the hypothesis that CS-induced lung collagen deposition is at least partly a result of CS-induced IL-33 translocation and release from local fibroblasts.
Collapse
Affiliation(s)
- Qiong Huang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chen Duo Li
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yi Ran Yang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiao Feng Qin
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jing Jing Wang
- Department of Laboratory Animal Sciences, Capital Medical University, Beijing, China
| | - Xin Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiao Nan Du
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xia Yang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Ying Wang
- Department of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lun Li
- Department of Respiratory Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mi Mu
- Department of Respiratory Medicine, the Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Kewu Huang
- Department of Pulmonary and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Chris J Corrigan
- Faculty of Life Sciences & Medicine, School of Immunology & Microbial Sciences, Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Sun Ying
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
445
|
Peng L, Sun W, Chen L, Wen WP. The Role of Interleukin-33 in Head and Neck Squamous Cell Carcinoma Is Determined by Its Cellular Sources in the Tumor Microenvironment. Front Oncol 2021; 10:588454. [PMID: 33634017 PMCID: PMC7902021 DOI: 10.3389/fonc.2020.588454] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/24/2020] [Indexed: 12/09/2022] Open
Abstract
Objectives To investigate the role of interleukin-33 (IL-33) in head and neck squamous cell carcinoma (HNSCC). Materials and Methods RNA-seq data of 520 cases of HNSCC were retrieved from The Cancer Genome Atlas. The tumor microenvironment was deconstructed by xCell using bulk RNA-seq data. The cohort was dichotomized by the median IL-33 expression level. Immune cell components and molecular markers were compared between the high and low IL-33 groups. The prognostic value of IL-33 was evaluated by the log-rank test. Differential gene expression analysis and KEGG pathway enrichment analysis were also conducted. The relationship between the IL-33 expression level and the abundance of its potential cellular sources was evaluated by Pearson’s partial correlation test. Subgroup analysis was conducted in laryngeal, oropharyngeal, and oral cavity squamous cell carcinoma (LSCC, OPSCC, and OCSCC). Results The role of IL-33 in HNSCC was heterogeneous among tumors at different sites. In LSCC, IL-33 may increase the extent of malignancy of tumor cells and act as a pro-tumor factor. In OCSCC, IL-33 may play a role in orchestrating the immune responses against tumor cells and act as an antitumor factor. The role of IL-33 in OPSCC was undetermined. IL-33 in LSCC was mainly derived from endothelial cells, while IL-33 in OCSCC was mainly derived from endothelial and epithelial cells. Conclusion According to the different sources of IL-33 in LSCC and OCSCC, we propose a hypothesis that stroma-derived IL-33 could favor tumor progression, while epithelial-derived IL-33 could favor antitumor immune responses in HNSCC.
Collapse
Affiliation(s)
- Liang Peng
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Wei Sun
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Lin Chen
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China
| | - Wei-Ping Wen
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, China.,Department of Otolaryngology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
446
|
Amôr NG, Santos PSDS, Campanelli AP. The Tumor Microenvironment in SCC: Mechanisms and Therapeutic Opportunities. Front Cell Dev Biol 2021; 9:636544. [PMID: 33634137 PMCID: PMC7900131 DOI: 10.3389/fcell.2021.636544] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/18/2021] [Indexed: 12/14/2022] Open
Abstract
Squamous cell carcinoma (SCC) is the second most common skin cancer worldwide and, despite the relatively easy visualization of the tumor in the clinic, a sizeable number of SCC patients are diagnosed at advanced stages with local invasion and distant metastatic lesions. In the last decade, immunotherapy has emerged as the fourth pillar in cancer therapy via the targeting of immune checkpoint molecules such as programmed cell-death protein-1 (PD-1), programmed cell death ligand-1 (PD-L1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). FDA-approved monoclonal antibodies directed against these immune targets have provide survival benefit in a growing list of cancer types. Currently, there are two immunotherapy drugs available for cutaneous SCC: cemiplimab and pembrolizumab; both monoclonal antibodies (mAb) that block PD-1 thereby promoting T-cell activation and/or function. However, the success rate of these checkpoint inhibitors currently remains around 50%, which means that half of the patients with advanced SCC experience no benefit from this treatment. This review will highlight the mechanisms by which the immune checkpoint molecules regulate the tumor microenvironment (TME), as well as the ongoing clinical trials that are employing single or combinatory therapeutic approaches for SCC immunotherapy. We also discuss the regulation of additional pathways that might promote superior therapeutic efficacy, and consequently provide increased survival for those patients that do not benefit from the current checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Nádia Ghinelli Amôr
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Paulo Sérgio da Silva Santos
- Department of Surgery, Stomatology, Pathology, and Radiology, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Ana Paula Campanelli
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| |
Collapse
|
447
|
She L, Barrera GD, Yan L, Alanazi HH, Brooks EG, Dube PH, Sun Y, Zan H, Chupp DP, Zhang N, Zhang X, Liu Y, Li XD. STING activation in alveolar macrophages and group 2 innate lymphoid cells suppresses IL-33-driven type 2 immunopathology. JCI Insight 2021; 6:143509. [PMID: 33400692 PMCID: PMC7934858 DOI: 10.1172/jci.insight.143509] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/23/2020] [Indexed: 12/27/2022] Open
Abstract
2'3'-cGAMP is known as a nonclassical second messenger and small immune modulator that possesses potent antitumor and antiviral activities via inducing the stimulator of IFN genes-mediated (STING-mediated) signaling pathway. However, its function in regulating type 2 immune responses remains unknown. Therefore, we sought to determine a role of STING activation by 2'3'-cGAMP in type 2 inflammatory reactions in multiple mouse models of eosinophilic asthma. We discovered that 2'3'-cGAMP administration strongly attenuated type 2 lung immunopathology and airway hyperreactivity induced by IL-33 and a fungal allergen, Aspergillus flavus. Mechanistically, upon the respiratory delivery, 2'3'-cGAMP was mainly internalized by alveolar macrophages, in which it activated the STING/IFN regulatory factor 3/type I IFN signaling axis to induce the production of inhibitory factors containing IFN-α, which blocked the IL-33-mediated activation of group 2 innate lymphoid (ILC2) cells in vivo. We further demonstrated that 2'3'-cGAMP directly suppressed the proliferation and function of both human and mouse ILC2 cells in vitro. Taken together, our findings suggest that STING activation by 2'3'-cGAMP in alveolar macrophages and ILC2 cells can negatively regulate type 2 immune responses, implying that the respiratory delivery of 2'3'-cGAMP might be further developed as an alternative strategy for treating type 2 immunopathologic diseases such as eosinophilic asthma.
Collapse
Affiliation(s)
- Li She
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Gema D. Barrera
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Liping Yan
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Hamad H. Alanazi
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Edward G. Brooks
- Division of Immunology and Infectious Disease, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Peter H. Dube
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Yilun Sun
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Hong Zan
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Daniel P. Chupp
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Xin Zhang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, and
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yong Liu
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, and
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Dong Li
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| |
Collapse
|
448
|
El-Naccache DW, Haskó G, Gause WC. Early Events Triggering the Initiation of a Type 2 Immune Response. Trends Immunol 2021; 42:151-164. [PMID: 33386241 PMCID: PMC9813923 DOI: 10.1016/j.it.2020.11.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/11/2020] [Accepted: 11/16/2020] [Indexed: 01/11/2023]
Abstract
Type 2 immune responses are typically associated with protection against helminth infections and also with harmful inflammation in response to allergens. Recent advances have revealed that type 2 immunity also contributes to sterile inflammation, cancer, and microbial infections. However, the early events that initiate type 2 immune responses remain poorly defined. New insights reveal major contributions from danger-associated molecular patterns (DAMPs) in the initiation of type 2 immune responses. In this review, we examine the molecules released by the host and pathogens and the role they play in mediating the initiation of mammalian innate type 2 immune responses under a variety of conditions.
Collapse
Affiliation(s)
- Darine W El-Naccache
- Center for Immunity and Inflammation, Rutgers - New Jersey Medical School, Newark, NJ, USA; Department of Medicine, Rutgers - New Jersey Medical School, Newark, NJ, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY, 10032, USA
| | - William C Gause
- Center for Immunity and Inflammation, Rutgers - New Jersey Medical School, Newark, NJ, USA; Department of Medicine, Rutgers - New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
449
|
Ramezani F, Babaie F, Aslani S, Hemmatzadeh M, Mohammadi FS, Gowhari-Shabgah A, Jadidi-Niaragh F, Ezzatifar F, Mohammadi H. The Role of the IL-33/ST2 Immune Pathway in Autoimmunity: New Insights and Perspectives. Immunol Invest 2021; 51:1060-1086. [PMID: 33522348 DOI: 10.1080/08820139.2021.1878212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Interleukin (IL)-33, a member of IL-1 cytokine family, is produced by various immune cells and acts as an alarm to alert the immune system after epithelial or endothelial cell damage during cell necrosis, infection, stress, and trauma. The biological functions of IL-33 largely depend on its ligation to the corresponding receptor, suppression of tumorigenicity 2 (ST2). The pathogenic roles of this cytokine have been implicated in several disorders, including allergic disease, cardiovascular disease, autoimmune disease, infectious disease, and cancers. However, alerted levels of IL-33 may result in either disease amelioration or progression. Genetic variations of IL33 gene may confer protective or susceptibility risk in the onset of autoimmune diseases. The purpose of this review is to discuss the involvement of IL-33 and ST2 in the pathogenesis of a variety of autoimmune disorders, such as autoimmune rheumatic, neurodegenerative, and endocrine diseases.
Collapse
Affiliation(s)
- Faezeh Ramezani
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
| | - Farhad Babaie
- Department of Immunology and Genetic, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Saeed Aslani
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Hemmatzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sadat Mohammadi
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Medical School, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Farhad Jadidi-Niaragh
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Ezzatifar
- Molecular and Cell Biology Research Center, Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.,Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
450
|
Alonso-Curbelo D, Ho YJ, Burdziak C, Maag JLV, Morris JP, Chandwani R, Chen HA, Tsanov KM, Barriga FM, Luan W, Tasdemir N, Livshits G, Azizi E, Chun J, Wilkinson JE, Mazutis L, Leach SD, Koche R, Pe'er D, Lowe SW. A gene-environment-induced epigenetic program initiates tumorigenesis. Nature 2021; 590:642-648. [PMID: 33536616 PMCID: PMC8482641 DOI: 10.1038/s41586-020-03147-x] [Citation(s) in RCA: 176] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023]
Abstract
Tissue damage increases the risk of cancer through poorly understood mechanisms1. In mouse models of pancreatic cancer, pancreatitis associated with tissue injury collaborates with activating mutations in the Kras oncogene to markedly accelerate the formation of early neoplastic lesions and, ultimately, adenocarcinoma2,3. Here, by integrating genomics, single-cell chromatin assays and spatiotemporally controlled functional perturbations in autochthonous mouse models, we show that the combination of Kras mutation and tissue damage promotes a unique chromatin state in the pancreatic epithelium that distinguishes neoplastic transformation from normal regeneration and is selected for throughout malignant evolution. This cancer-associated epigenetic state emerges within 48 hours of pancreatic injury, and involves an 'acinar-to-neoplasia' chromatin switch that contributes to the early dysregulation of genes that define human pancreatic cancer. Among the factors that are most rapidly activated after tissue damage in the pre-malignant pancreatic epithelium is the alarmin cytokine interleukin 33, which recapitulates the effects of injury in cooperating with mutant Kras to unleash the epigenetic remodelling program of early neoplasia and neoplastic transformation. Collectively, our study demonstrates how gene-environment interactions can rapidly produce gene-regulatory programs that dictate early neoplastic commitment, and provides a molecular framework for understanding the interplay between genetic and environmental cues in the initiation of cancer.
Collapse
Affiliation(s)
- Direna Alonso-Curbelo
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu-Jui Ho
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cassandra Burdziak
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Tri-Institutional Training Program in Computational Biology and Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jesper L V Maag
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John P Morris
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rohit Chandwani
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, Weill Cornell Medical College, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
| | - Hsuan-An Chen
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kaloyan M Tsanov
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Francisco M Barriga
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wei Luan
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nilgun Tasdemir
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Geulah Livshits
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elham Azizi
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jaeyoung Chun
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Alan and Sandra Gerry Metastasis and Tumor Ecosystems Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John E Wilkinson
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Linas Mazutis
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Alan and Sandra Gerry Metastasis and Tumor Ecosystems Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Steven D Leach
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Dartmouth Norris Cotton Cancer Center, Hanover, NH, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dana Pe'er
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|