1
|
Zhou Y, Yan Z, Pang Y, Jiang Y, Zhuang R, Zhang S, Nurmamat A, Xiu M, Li D, Zhao L, Liu X, Li Q, Han Y. Exploring the Multiple Roles of Notch1 in Biological Development: An Analysis and Study Based on Phylogenetics and Transcriptomics. Int J Mol Sci 2024; 25:611. [PMID: 38203782 PMCID: PMC10778765 DOI: 10.3390/ijms25010611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
At present, there is a research gap concerning the specific functions and mechanisms of the Notch gene family and its signaling pathway in jawless vertebrates. In this study, we identified a Notch1 homologue (Lr. Notch1) in the Lethenteron reissneri database. Through bioinformatics analysis, we identified Lr. Notch1 as the likely common ancestor gene of the Notch gene family in higher vertebrates, indicating a high degree of conservation in the Notch gene family and its signaling pathways. To validate the biological function of Lr. Notch1, we conducted targeted silencing of Lr. Notch1 in L. reissneri and analyzed the resultant gene expression profile before and after silencing using transcriptome analysis. Our findings revealed that the silencing of Lr. Notch1 resulted in differential expression of pathways and genes associated with signal transduction, immune regulation, and metabolic regulation, mirroring the biological function of the Notch signaling pathway in higher vertebrates. This article systematically elucidated the origin and evolution of the Notch gene family while also validating the biological function of Lr. Notch1. These insights offer valuable clues for understanding the evolution of the Notch signaling pathway and establish a foundation for future research on the origin of the Notch signaling pathway, as well as its implications in human diseases and immunomodulation.
Collapse
Affiliation(s)
- Yuesi Zhou
- Key Research Base of Humanities and Social Sciences of Ministry of Education, Institute of Marine Sustainable Development, Liaoning Normal University, Dalian 116029, China;
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Zihao Yan
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Ya Pang
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Yao Jiang
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Ruyu Zhuang
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Shuyuan Zhang
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Ayqeqan Nurmamat
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Min Xiu
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Ding Li
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Liang Zhao
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
| | - Xin Liu
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Qingwei Li
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Yinglun Han
- Key Research Base of Humanities and Social Sciences of Ministry of Education, Institute of Marine Sustainable Development, Liaoning Normal University, Dalian 116029, China;
- Lamprey Research Center, College of Life Sciences, Liaoning Normal University, Dalian 116081, China; (Z.Y.)
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
2
|
Zhang C, Xu H, Netto KG, Sokulsky LA, Miao Y, Mo Z, Meng Y, Du Y, Wu C, Han L, Zhang L, Liu C, Zhang G, Li F, Yang M. Inhibition of γ-glutamyl transferase suppresses airway hyperresponsiveness and airway inflammation in a mouse model of steroid resistant asthma exacerbation. Front Immunol 2023; 14:1132939. [PMID: 37377967 PMCID: PMC10292800 DOI: 10.3389/fimmu.2023.1132939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Introduction Despite recent advances, there are limited treatments available for acute asthma exacerbations. Here, we investigated the therapeutic potential of GGsTop, a γ-glutamyl transferase inhibitor, on the disease with a murine model of asthma exacerbation. Methods GGsTop was administered to mice that received lipopolysaccharide (LPS) and ovalbumin (OVA) challenges. Airway hyperresponsiveness (AHR), lung histology, mucus hypersecretion, and collagen deposition were analyzed to evaluate the hallmark features of asthma exacerbation. The level of proinflammatory cytokines and glutathione were determined with/without GGsTop. The transcription profiles were also examined. Results GGsTop attenuates hallmark features of the disease with a murine model of LPS and OVA driven asthma exacerbation. Airway hyperresponsiveness (AHR), mucus hypersecretion, collagen deposition, and expression of inflammatory cytokines were dramatically inhibited by GGsTop treatment. Additionally, GGsTop restored the level of glutathione. Using RNA-sequencing and pathway analysis, we demonstrated that the activation of LPS/NFκB signaling pathway in airway was downregulated by GGsTop. Interestingly, further analysis revealed that GGsTop significantly inhibited not only IFNγ responses but also the expression of glucocorticoid-associated molecules, implicating that GGsTop profoundly attenuates inflammatory pathways. Conclusions Our study suggests that GGsTop is a viable treatment for asthma exacerbation by broadly inhibiting the activation of multiple inflammatory pathways.
Collapse
Affiliation(s)
- Cancan Zhang
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Huisha Xu
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Keilah G. Netto
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Leon A. Sokulsky
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Yiyan Miao
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhongyuan Mo
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yan Meng
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yingying Du
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chengyong Wu
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Liyou Han
- Institute for Liberal Arts and Sciences, Kyoto University, Kyoto, Japan
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chi Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Guojun Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fuguang Li
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ming Yang
- Academy of Medical Sciences & Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Priority Research Centre for Healthy Lungs, School of Biomedical Sciences & Pharmacy, Faculty of Health and Hunter Medical Research Institute, The University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
3
|
Wang J, Zhou Y, Zhang H, Hu L, Liu J, Wang L, Wang T, Zhang H, Cong L, Wang Q. Pathogenesis of allergic diseases and implications for therapeutic interventions. Signal Transduct Target Ther 2023; 8:138. [PMID: 36964157 PMCID: PMC10039055 DOI: 10.1038/s41392-023-01344-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/20/2023] [Accepted: 02/03/2023] [Indexed: 03/26/2023] Open
Abstract
Allergic diseases such as allergic rhinitis (AR), allergic asthma (AAS), atopic dermatitis (AD), food allergy (FA), and eczema are systemic diseases caused by an impaired immune system. Accompanied by high recurrence rates, the steadily rising incidence rates of these diseases are attracting increasing attention. The pathogenesis of allergic diseases is complex and involves many factors, including maternal-fetal environment, living environment, genetics, epigenetics, and the body's immune status. The pathogenesis of allergic diseases exhibits a marked heterogeneity, with phenotype and endotype defining visible features and associated molecular mechanisms, respectively. With the rapid development of immunology, molecular biology, and biotechnology, many new biological drugs have been designed for the treatment of allergic diseases, including anti-immunoglobulin E (IgE), anti-interleukin (IL)-5, and anti-thymic stromal lymphopoietin (TSLP)/IL-4, to control symptoms. For doctors and scientists, it is becoming more and more important to understand the influencing factors, pathogenesis, and treatment progress of allergic diseases. This review aimed to assess the epidemiology, pathogenesis, and therapeutic interventions of allergic diseases, including AR, AAS, AD, and FA. We hope to help doctors and scientists understand allergic diseases systematically.
Collapse
Affiliation(s)
- Ji Wang
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Yumei Zhou
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Honglei Zhang
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Linhan Hu
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Juntong Liu
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Lei Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 1000210, China
| | - Tianyi Wang
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Haiyun Zhang
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Linpeng Cong
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Qi Wang
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China.
| |
Collapse
|
4
|
NOTCH Signaling in Osteosarcoma. Curr Issues Mol Biol 2023; 45:2266-2283. [PMID: 36975516 PMCID: PMC10047431 DOI: 10.3390/cimb45030146] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
The combination of neoadjuvant chemotherapy and surgery has been promoted for the treatment of osteosarcoma; however, the local recurrence and lung metastasis rates remain high. Therefore, it is crucial to explore new therapeutic targets and strategies that are more effective. The NOTCH pathway is not only involved in normal embryonic development but also plays an important role in the development of cancers. The expression level and signaling functional status of the NOTCH pathway vary in different histological types of cancer as well as in the same type of cancer from different patients, reflecting the distinct roles of the Notch pathway in tumorigenesis. Studies have reported abnormal activation of the NOTCH signaling pathway in most clinical specimens of osteosarcoma, which is closely related to a poor prognosis. Similarly, studies have reported that NOTCH signaling affected the biological behavior of osteosarcoma through various molecular mechanisms. NOTCH-targeted therapy has shown potential for the treatment of osteosarcoma in clinical research. After the introduction of the composition and biological functions of the NOTCH signaling pathway, the review paper discussed the clinical significance of dysfunction in osteosarcoma. Then the paper reviewed the recent relevant research progress made both in the cell lines and in the animal models of osteosarcoma. Finally, the paper explored the potential of the clinical application of NOTCH-targeted therapy for the treatment of osteosarcoma.
Collapse
|
5
|
Notch signaling pathway: architecture, disease, and therapeutics. Signal Transduct Target Ther 2022; 7:95. [PMID: 35332121 PMCID: PMC8948217 DOI: 10.1038/s41392-022-00934-y] [Citation(s) in RCA: 354] [Impact Index Per Article: 177.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
The NOTCH gene was identified approximately 110 years ago. Classical studies have revealed that NOTCH signaling is an evolutionarily conserved pathway. NOTCH receptors undergo three cleavages and translocate into the nucleus to regulate the transcription of target genes. NOTCH signaling deeply participates in the development and homeostasis of multiple tissues and organs, the aberration of which results in cancerous and noncancerous diseases. However, recent studies indicate that the outcomes of NOTCH signaling are changeable and highly dependent on context. In terms of cancers, NOTCH signaling can both promote and inhibit tumor development in various types of cancer. The overall performance of NOTCH-targeted therapies in clinical trials has failed to meet expectations. Additionally, NOTCH mutation has been proposed as a predictive biomarker for immune checkpoint blockade therapy in many cancers. Collectively, the NOTCH pathway needs to be integrally assessed with new perspectives to inspire discoveries and applications. In this review, we focus on both classical and the latest findings related to NOTCH signaling to illustrate the history, architecture, regulatory mechanisms, contributions to physiological development, related diseases, and therapeutic applications of the NOTCH pathway. The contributions of NOTCH signaling to the tumor immune microenvironment and cancer immunotherapy are also highlighted. We hope this review will help not only beginners but also experts to systematically and thoroughly understand the NOTCH signaling pathway.
Collapse
|
6
|
Allen F, Maillard I. Therapeutic Targeting of Notch Signaling: From Cancer to Inflammatory Disorders. Front Cell Dev Biol 2021; 9:649205. [PMID: 34124039 PMCID: PMC8194077 DOI: 10.3389/fcell.2021.649205] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Over the past two decades, the Notch signaling pathway has been investigated as a therapeutic target for the treatment of cancers, and more recently in the context of immune and inflammatory disorders. Notch is an evolutionary conserved pathway found in all metazoans that is critical for proper embryonic development and for the postnatal maintenance of selected tissues. Through cell-to-cell contacts, Notch orchestrates cell fate decisions and differentiation in non-hematopoietic and hematopoietic cell types, regulates immune cell development, and is integral to shaping the amplitude as well as the quality of different types of immune responses. Depriving some cancer types of Notch signals has been shown in preclinical studies to stunt tumor growth, consistent with an oncogenic function of Notch signaling. In addition, therapeutically antagonizing Notch signals showed preclinical potential to prevent or reverse inflammatory disorders, including autoimmune diseases, allergic inflammation and immune complications of life-saving procedures such allogeneic bone marrow and solid organ transplantation (graft-versus-host disease and graft rejection). In this review, we discuss some of these unique approaches, along with the successes and challenges encountered so far to target Notch signaling in preclinical and early clinical studies. Our goal is to emphasize lessons learned to provide guidance about emerging strategies of Notch-based therapeutics that could be deployed safely and efficiently in patients with immune and inflammatory disorders.
Collapse
Affiliation(s)
- Frederick Allen
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Ivan Maillard
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
7
|
Involvement of Dendritic Cells and Th17 Cells in Induced Tertiary Lymphoid Structures in a Chronic Beryllium Disease Mouse Model. Mediators Inflamm 2021; 2021:8845966. [PMID: 34054347 PMCID: PMC8123089 DOI: 10.1155/2021/8845966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/27/2021] [Accepted: 04/20/2021] [Indexed: 11/18/2022] Open
Abstract
Objective To study airway pathophysiology and the role of dendritic cells (DCs) and IL-17 receptor (IL-17R) signals in a mouse model for CBD. Methods Here, we present a CBD mouse model in which mice were exposed to beryllium during three weeks. We also exposed IL-17R-deficient mice and mice in which DCs were depleted. Results Eight weeks after the initial beryllium exposure, an inflammatory response was detected in the lungs. Mice displayed inflammation of the lower airways that included focal dense infiltrates, granuloma-like foci, and tertiary lymphoid structure (TLS) containing T cells, B cells, and germinal centers. Alveolar cell analysis showed significantly increased numbers of CD4+ T cells expressing IFNγ, IL-17, or both cytokines. The pathogenic role of IL-17R signals was demonstrated in IL-17R-deficient mice, which had strongly reduced lung inflammation and TLS development following beryllium exposure. In CBD mice, pulmonary DC subsets including CD103+ conventional DCs (cDCs), CD11b+ cDCs, and monocyte-derived DCs (moDCs) were also prominently increased. We used diphtheria toxin receptor-mediated targeted cell ablation to conditionally deplete DCs and found that DCs are essential for the maintenance of TLS in CBD. Furthermore, the presence of antinuclear autoantibodies in the serum of CBD mice showed that CBD had characteristics of autoimmune disease. Conclusions We generated a translational model of sarcoidosis driven by beryllium and show that DCs and IL-17R signals play a pathophysiological role in CBD development as well as in established CBD in vivo.
Collapse
|
8
|
Christopoulos PF, Gjølberg TT, Krüger S, Haraldsen G, Andersen JT, Sundlisæter E. Targeting the Notch Signaling Pathway in Chronic Inflammatory Diseases. Front Immunol 2021; 12:668207. [PMID: 33912195 PMCID: PMC8071949 DOI: 10.3389/fimmu.2021.668207] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
The Notch signaling pathway regulates developmental cell-fate decisions and has recently also been linked to inflammatory diseases. Although therapies targeting Notch signaling in inflammation in theory are attractive, their design and implementation have proven difficult, at least partly due to the broad involvement of Notch signaling in regenerative and homeostatic processes. In this review, we summarize the supporting role of Notch signaling in various inflammation-driven diseases, and highlight efforts to intervene with this pathway by targeting Notch ligands and/or receptors with distinct therapeutic strategies, including antibody designs. We discuss this in light of lessons learned from Notch targeting in cancer treatment. Finally, we elaborate on the impact of individual Notch members in inflammation, which may lay the foundation for development of therapeutic strategies in chronic inflammatory diseases.
Collapse
Affiliation(s)
| | - Torleif T. Gjølberg
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Centre for Eye Research and Department of Ophthalmology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Stig Krüger
- Department of Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Guttorm Haraldsen
- Department of Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Jan Terje Andersen
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Eirik Sundlisæter
- Department of Pathology, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
9
|
Tindemans I, van Schoonhoven A, KleinJan A, de Bruijn MJ, Lukkes M, van Nimwegen M, van den Branden A, Bergen IM, Corneth OB, van IJcken WF, Stadhouders R, Hendriks RW. Notch signaling licenses allergic airway inflammation by promoting Th2 cell lymph node egress. J Clin Invest 2021; 130:3576-3591. [PMID: 32255764 DOI: 10.1172/jci128310] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 03/24/2020] [Indexed: 12/27/2022] Open
Abstract
Allergic asthma is mediated by Th2 responses to inhaled allergens. Although previous experiments indicated that Notch signaling activates expression of the key Th2 transcription factor Gata3, it remains controversial how Notch promotes allergic airway inflammation. Here we show that T cell-specific Notch deficiency in mice prevented house dust mite-driven eosinophilic airway inflammation and significantly reduced Th2 cytokine production, serum IgE levels, and airway hyperreactivity. However, transgenic Gata3 overexpression in Notch-deficient T cells only partially rescued this phenotype. We found that Notch signaling was not required for T cell proliferation or Th2 polarization. Instead, Notch-deficient in vitro-polarized Th2 cells showed reduced accumulation in the lungs upon in vivo transfer and allergen challenge, as Notch-deficient Th2 cells were retained in the lung-draining lymph nodes. Transcriptome analyses and sequential adoptive transfer experiments revealed that while Notch-deficient lymph node Th2 cells established competence for lung migration, they failed to upregulate sphingosine-1-phosphate receptor 1 (S1PR1) and its critical upstream transcriptional activator Krüppel-like factor 2 (KLF2). As this KLF2/S1PR1 axis represents the essential cell-intrinsic regulator of T cell lymph node egress, we conclude that the druggable Notch signaling pathway licenses the Th2 response in allergic airway inflammation via promoting lymph node egress.
Collapse
|
10
|
Context Matters: NOTCH Signatures and Pathway in Cancer Progression and Metastasis. Cells 2021; 10:cells10010094. [PMID: 33430387 PMCID: PMC7827494 DOI: 10.3390/cells10010094] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023] Open
Abstract
The Notch signaling pathway is a critical player in embryogenesis but also plays various roles in tumorigenesis, with both tumor suppressor and oncogenic activities. Mutations, deletions, amplifications, or over-expression of Notch receptors, ligands, and a growing list of downstream Notch-activated genes have by now been described for most human cancer types. Yet, it often remains unclear what may be the functional impact of these changes for tumor biology, initiation, and progression, for cancer therapy, and for personalized medicine. Emerging data indicate that Notch signaling can also contribute to increased aggressive properties such as invasion, tumor heterogeneity, angiogenesis, or tumor cell dormancy within solid cancer tissues; especially in epithelial cancers, which are in the center of this review. Notch further supports the “stemness” of cancer cells and helps define the stem cell niche for their long-term survival, by integrating the interaction between cancer cells and the cells of the tumor microenvironment (TME). The complexity of Notch crosstalk with other signaling pathways and its roles in cell fate and trans-differentiation processes such as epithelial-to-mesenchymal transition (EMT) point to this pathway as a decisive player that may tip the balance between tumor suppression and promotion, differentiation and invasion. Here we not only review the literature, but also explore genomic databases with a specific focus on Notch signatures, and how they relate to different stages in tumor development. Altered Notch signaling hereby plays a key role for tumor cell survival and coping with a broad spectrum of vital issues, contributing to failed therapies, poor patient outcome, and loss of lives.
Collapse
|
11
|
Pu Z, Yang F, Wang L, Diao Y, Chen D. Advancements of compounds targeting Wnt and Notch signalling pathways in the treatment of inflammatory bowel disease and colon cancer. J Drug Target 2020; 29:507-519. [PMID: 33307848 DOI: 10.1080/1061186x.2020.1864741] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Wnt and Notch signalling pathways are important for maintenance of intestinal epithelial barrier integrity by intestinal stem cells (ISCs). Dysfunction of these pathways is implicated in inflammatory bowel disease (IBD) and colon cancer. The objective of this review is to summarise advancements of drugs that regulate Wnt and Notch in the treatment of IBD and colon cancer. The compositions and biological effects of Wnt and Notch modulators in both ISCs and non-ISCs are discussed. The drugs, including phytochemicals, plant extracts, probiotics and synthetic compounds, have been found to regulate Wnt and Notch signalling pathways by targeting regulatory factors (including secreted frizzled-related proteins or pathway proteins such as β-catenin and γ-secretase) to alleviate IBD and colon cancer. This review highlights the potential for targeting Wnt and Notch pathways to treat IBD and colon cancer.
Collapse
Affiliation(s)
- Zhuonan Pu
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Fang Yang
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Liang Wang
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| | - Yunpeng Diao
- Colleage of Pharmacy, Dalian Medical University, Dalian, Liaoning, China
| | - Dapeng Chen
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
12
|
Yao YE, Zhang JH, Chen XJ, Huang JL, Sun QX, Liu WW, Zeng H, Li CQ. Regulation of γδT17 cells by Mycobacterium vaccae through interference with Notch/Jagged1 signaling pathway. ACTA ACUST UNITED AC 2020; 53:e9551. [PMID: 33053115 PMCID: PMC7552905 DOI: 10.1590/1414-431x20209551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 07/23/2020] [Indexed: 11/22/2022]
Abstract
The objective of this study was to investigate the effect of Mycobacterium vaccae on Jagged 1 and gamma delta T17 (γδT17) cells in asthmatic mice. An asthma mouse model was established through immunization with ovalbumin (OVA). Gamma-secretase inhibitor (DAPT) was used to block the Notch signaling pathway. M. vaccae was used to treat asthma, and related indicators were measured. Blocking Notch signaling inhibited the production of γδT17 cells and secretion of cytokine interleukin (IL)-17, which was accompanied by a decrease in Jagged1 mRNA and protein expression in the treated asthma group compared with the untreated asthma group. Similarly, treatment with M. vaccae inhibited Jagged1 expression and γδT17 cell production, which was associated with decreased airway inflammation and reactivity. The Notch signaling pathway may play a role in the pathogenesis of asthma through the induction of Jagged1 receptor. On the other hand, the inhibitory effect of M. vaccae on Jagged1 receptor in γδT17 cells could be used for the prevention and treatment of asthma.
Collapse
Affiliation(s)
- Yi En Yao
- Department of Respiratory Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jing Hong Zhang
- Department of Internal Medicine, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiao Ju Chen
- Department of Critical Care, First People's Hospital of Yulin City, Nanning, Guangxi, China
| | - Jian Lin Huang
- Department of Respiratory Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qi Xiang Sun
- Department of Respiratory Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Wei Wei Liu
- Department of Emergency Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Huan Zeng
- The Second Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Chao Qian Li
- Department of Respiratory Medicine, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
13
|
Goruganthu MUL, Shanker A, Dikov MM, Carbone DP. Specific Targeting of Notch Ligand-Receptor Interactions to Modulate Immune Responses: A Review of Clinical and Preclinical Findings. Front Immunol 2020; 11:1958. [PMID: 32922403 PMCID: PMC7456812 DOI: 10.3389/fimmu.2020.01958] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022] Open
Abstract
Understanding and targeting Notch signaling effectively has long been valued in the field of cancer and other immune disorders. Here, we discuss key discoveries at the intersection of Notch signaling, cancer and immunology. While there is a plethora of Notch targeting agents tested in vitro, in vivo and in clinic, undesirable off-target effects and therapy-related toxicities have been significant obstacles. We make a case for the clinical application of ligand-derived and affinity modifying compounds as novel therapeutic agents and discuss major research findings with an emphasis on Notch ligand-specific modulation of immune responses.
Collapse
Affiliation(s)
- Mounika U. L. Goruganthu
- Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Anil Shanker
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College School of Medicine, Nashville, TN, United States
- Vanderbilt-Ingram Cancer Center, Nashville, TN, United States
| | - Mikhail M. Dikov
- Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - David P. Carbone
- Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
14
|
Moore G, Annett S, McClements L, Robson T. Top Notch Targeting Strategies in Cancer: A Detailed Overview of Recent Insights and Current Perspectives. Cells 2020; 9:cells9061503. [PMID: 32575680 PMCID: PMC7349363 DOI: 10.3390/cells9061503] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/05/2020] [Accepted: 06/11/2020] [Indexed: 12/17/2022] Open
Abstract
Evolutionarily conserved Notch plays a critical role in embryonic development and cellular self-renewal. It has both tumour suppressor and oncogenic activity, the latter of which is widely described. Notch-activating mutations are associated with haematological malignancies and several solid tumours including breast, lung and adenoid cystic carcinoma. Moreover, upregulation of Notch receptors and ligands and aberrant Notch signalling is frequently observed in cancer. It is involved in cancer hallmarks including proliferation, survival, migration, angiogenesis, cancer stem cell renewal, metastasis and drug resistance. It is a key component of cell-to-cell interactions between cancer cells and cells of the tumour microenvironment, such as endothelial cells, immune cells and fibroblasts. Notch displays diverse crosstalk with many other oncogenic signalling pathways, and may drive acquired resistance to targeted therapies as well as resistance to standard chemo/radiation therapy. The past 10 years have seen the emergence of different classes of drugs therapeutically targeting Notch including receptor/ligand antibodies, gamma secretase inhibitors (GSI) and most recently, the development of Notch transcription complex inhibitors. It is an exciting time for Notch research with over 70 cancer clinical trials registered and the first-ever Phase III trial of a Notch GSI, nirogacestat, currently at the recruitment stage.
Collapse
Affiliation(s)
- Gillian Moore
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons, D02 YN77 Dublin, Ireland; (G.M.); (S.A.)
| | - Stephanie Annett
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons, D02 YN77 Dublin, Ireland; (G.M.); (S.A.)
| | - Lana McClements
- The School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia;
| | - Tracy Robson
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons, D02 YN77 Dublin, Ireland; (G.M.); (S.A.)
- Correspondence:
| |
Collapse
|
15
|
Qu SY, Ti XY, Zhang J, Wu CG. Disruption of the Notch pathway aggravates airway inflammation by inhibiting regulatory T cell differentiation via regulation of plasmacytoid dendritic cells. Scand J Immunol 2020; 91:e12865. [PMID: 32185817 DOI: 10.1111/sji.12865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 12/20/2019] [Accepted: 01/03/2020] [Indexed: 12/29/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) regulate immunity and promote tolerance in asthma. Notch signalling is a highly conserved pathway that regulates the immune response; however, its role in pDC-mediated asthmatic airway inflammation is unclear. This study clarified the effects of Notch signalling on pDC-mediated airway inflammation using murine models of ovalbumin-sensitized allergic asthma. RBP-J-deficient pDCs (RBP-J-/- pDCs) were co-cultured with naïve CD4+ T cells and supernatants and T cell subtypes were analysed. RBP-J-/- pDCs were intranasally transferred to the airways of ovalbumin-sensitized recipient mice. Lung samples of all mice were subjected to tests for histopathology, cytokine profile of bronchoalveolar lavage fluid, airway hyperactivity and expression of T helper type 1 (Th1)/Th2 cells, regulatory T cells and type 2 innate lymphoid cells (ILC2s). The results showed that pDCs with and without RBP-J deficiency significantly differed in expression levels of cluster of differentiation 83 (CD83), but not CD80, CD86 and major histocompatibility complex class II. Co-culturing pDCs with naïve T cells revealed a poorer immunosuppressive effect of RBP-J-/- pDCs. This may be attributed to the lower expression levels of inducible co-stimulator ligand and lower production of interleukin 10 in RBP-J-/- pDCs than in control pDCs, which impeded T cell activation and Treg suppression. RBP-J-/- pDCs were associated with high ILC2 expression and severe Th2 immune responses and airway inflammation. Therefore, Notch signalling is critical for pDC-dependent immunoregulation, and RBP-J deficiency reduces pDC-based immunosuppression via T cell activation and Th cell differentiation. Thus, this pathway may be a therapeutic target for pDC-based anti-asthma immunotherapy.
Collapse
Affiliation(s)
- Shuo-Yao Qu
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xin-Yu Ti
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jian Zhang
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chang-Gui Wu
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
16
|
Xu S, Kong YG, Jiao WE, Yang R, Qiao YL, Xu Y, Tao ZZ, Chen SM. Tangeretin promotes regulatory T cell differentiation by inhibiting Notch1/Jagged1 signaling in allergic rhinitis. Int Immunopharmacol 2019; 72:402-412. [PMID: 31030096 DOI: 10.1016/j.intimp.2019.04.039] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVE Tangeretin demonstrates broad anti-inflammatory effects. The present study aimed to assess whether tangeretin functions in regulating T-regulatory cells (Tregs) and alleviating allergic rhinitis (AR). METHODS An ovalbumin (OVA)-induced AR animal model was constructed to monitor the changes in the allergic symptom score, OVA-specific IgE titers, histopathological characteristics and T-helper cell (Th1, Th2, and Th17)-related cytokine levels under tangeretin or dexamethasone (DXM) administration. The expression levels of Notch1/Jagged1 and FOXP3, and the proportion of Tregs in the spleens of these animals, were also detected. Furthermore, purified naive CD4 + T cells were utilized to assess the effects of tangeretin on Notch1 expression and their differentiation in vitro. RESULTS Both tangeretin and DXM administration alleviated airway inflammation, decreased the production of serum OVA-induced IgE, but only tangeretin administration restored the balance of cytokine profiles compared with those in the AR group. The abundance of splenic CD4 + CD25 + FOXP3 + Treg cells and the transcription factor FOXP3 were significantly increased under tangeretin treatment, either in AR mice or in naïve CD4 + T-cell differentiation, followed by a concomitant reduction in Notch1/Jagged1 expression. However, as a positive control, the treatment of allergic rhinitis with dexamethasone was not related to the expression of Notch1/Jagged1 or the differentiation of Treg cells. CONCLUSION Tangeretin could promote regulatory T cell responses by inhibiting Notch1/Jagged1 expression, followed by promoting FOXP3/Treg cell differentiation and thus could serve as a novel curative therapeutic for AR.
Collapse
Affiliation(s)
- Shan Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan 430060, Hubei, PR China
| | - Yong-Gang Kong
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan 430060, Hubei, PR China
| | - Wo-Er Jiao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan 430060, Hubei, PR China
| | - Rui Yang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan 430060, Hubei, PR China
| | - Yue-Long Qiao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan 430060, Hubei, PR China
| | - Yu Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan 430060, Hubei, PR China; Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan 430060, Hubei, PR China
| | - Ze-Zhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan 430060, Hubei, PR China; Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan 430060, Hubei, PR China
| | - Shi-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan 430060, Hubei, PR China; Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan 430060, Hubei, PR China.
| |
Collapse
|
17
|
Increased surface expression of NOTCH on memory T cells in peripheral blood from patients with asthma. J Allergy Clin Immunol 2018; 143:769-771.e3. [PMID: 30296524 DOI: 10.1016/j.jaci.2018.09.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/31/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022]
|
18
|
Grayson MH, Feldman S, Prince BT, Patel PJ, Matsui EC, Apter AJ. Advances in asthma in 2017: Mechanisms, biologics, and genetics. J Allergy Clin Immunol 2018; 142:1423-1436. [PMID: 30213625 DOI: 10.1016/j.jaci.2018.08.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/22/2018] [Accepted: 08/31/2018] [Indexed: 02/07/2023]
Abstract
This review summarizes some of the most significant advances in asthma research over the past year. We first focus on novel discoveries in the mechanism of asthma development and exacerbation. This is followed by a discussion of potential new biomarkers, including the use of radiographic markers of disease. Several new biologics have become available to the clinician in the past year, and we summarize these advances and how they can influence the clinical delivery of asthma care. After this, important findings in the genetics of asthma and heterogeneity in phenotypes of the disease are explored, as is the role the environment plays in shaping the development and exacerbation of asthma. Finally, we conclude with a discussion of advances in health literacy and how they will affect asthma care.
Collapse
Affiliation(s)
- Mitchell H Grayson
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children's Hospital, Ohio State University College of Medicine, Columbus, Ohio.
| | - Scott Feldman
- Section of Allergy and Immunology, Division of Pulmonary Allergy Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pa
| | - Benjamin T Prince
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children's Hospital, Ohio State University College of Medicine, Columbus, Ohio
| | - Priya J Patel
- Section of Allergy and Immunology, Division of Pulmonary Allergy Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pa
| | - Elizabeth C Matsui
- Department of Population Health, Dell Medical School, University of Texas-Austin, Austin, Tex
| | - Andrea J Apter
- Section of Allergy and Immunology, Division of Pulmonary Allergy Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pa
| |
Collapse
|
19
|
Beute J, Lukkes M, Koekoek EP, Nastiti H, Ganesh K, de Bruijn MJ, Hockman S, van Nimwegen M, Braunstahl GJ, Boon L, Lambrecht BN, Manganiello VC, Hendriks RW, KleinJan A. A pathophysiological role of PDE3 in allergic airway inflammation. JCI Insight 2018; 3:94888. [PMID: 29367458 DOI: 10.1172/jci.insight.94888] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 12/12/2017] [Indexed: 01/04/2023] Open
Abstract
Phosphodiesterase 3 (PDE3) and PDE4 regulate levels of cyclic AMP, which are critical in various cell types involved in allergic airway inflammation. Although PDE4 inhibition attenuates allergic airway inflammation, reported side effects preclude its application as an antiasthma drug in humans. Case reports showed that enoximone, which is a smooth muscle relaxant that inhibits PDE3, is beneficial and lifesaving in status asthmaticus and is well tolerated. However, clinical observations also showed antiinflammatory effects of PDE3 inhibition. In this study, we investigated the role of PDE3 in a house dust mite-driven (HDM-driven) allergic airway inflammation (AAI) model that is characterized by T helper 2 cell activation, eosinophilia, and reduced mucosal barrier function. Compared with wild-type (WT) littermates, mice with a targeted deletion of the PDE3A or PDE3B gene showed significantly reduced HDM-driven AAI. Therapeutic intervention in WT mice showed that all hallmarks of HDM-driven AAI were abrogated by the PDE3 inhibitors enoximone and milrinone. Importantly, we found that enoximone also reduced the upregulation of the CD11b integrin on mouse and human eosinophils in vitro, which is crucial for their recruitment during allergic inflammation. This study provides evidence for a hitherto unknown antiinflammatory role of PDE3 inhibition in allergic airway inflammation and offers a potentially novel treatment approach.
Collapse
Affiliation(s)
- Jan Beute
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| | - Melanie Lukkes
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| | - Ewout P Koekoek
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| | - Hedwika Nastiti
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| | - Keerthana Ganesh
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| | | | - Steve Hockman
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland USA
| | - Menno van Nimwegen
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| | | | - Louis Boon
- Epirus Biopharmaceuticals Netherlands Yalelaan, Utrecht, Netherlands
| | - Bart N Lambrecht
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands.,VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
| | - Vince C Manganiello
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland USA
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| | - Alex KleinJan
- Department of Pulmonary Medicine, Erasmus MC, 's-Gravendijkwal, Rotterdam, Netherlands
| |
Collapse
|