1
|
Yu L, Shi H, Gao T, Xu W, Qian H, Jiang J, Yang X, Zhang X. Exomeres and supermeres: Current advances and perspectives. Bioact Mater 2025; 50:322-343. [PMID: 40276541 PMCID: PMC12020890 DOI: 10.1016/j.bioactmat.2025.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/26/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
Recent studies have revealed a great diversity and complexity in extracellular vesicles and particles (EVPs). The developments in techniques and the growing awareness of the particle heterogeneity have spurred active research on new particle subsets. Latest discoveries highlighted unique features and roles of non-vesicular extracellular nanoparticles (NVEPs) as promising biomarkers and targets for diseases. These nanoparticles are distinct from extracellular vesicles (EVs) in terms of their smaller particle sizes and lack of a bilayer membrane structure and they are enriched with diverse bioactive molecules particularly proteins and RNAs, which are widely reported to be delivered and packaged in exosomes. This review is focused on the two recently identified membraneless NVEPs, exomeres and supermeres, to provide an overview of their biogenesis and contents, particularly those bioactive substances linked to their bio-properties. This review also explains the concepts and characteristics of these nanoparticles, to compare them with other EVPs, especially EVs, as well as to discuss their isolation and identification methods, research interests, potential clinical applications and open questions.
Collapse
Affiliation(s)
- Li Yu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Hui Shi
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Tingxin Gao
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Wenrong Xu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
2
|
Zhou Z, Zhu R, Yang H, Deng W, Zhang Z, Li Y, Xu J, Yan Z, Wang R, Chang S, Yin Z, Wu Y, Zhang D, Fang M, Liu C, Que Y, Zhang J, Xia N, Wang Y, Xu L, Cheng T. Transgenic mice expressing the human CDHR3 receptor: A sensitive RV-C infection model for the evaluation of vaccines and therapeutics. Antiviral Res 2025; 235:106102. [PMID: 39922540 DOI: 10.1016/j.antiviral.2025.106102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/10/2025]
Abstract
Rhinovirus C (RV-C) is the primary causative agent of severe acute respiratory illnesses (ARTIs) in infants and young children. The limited availability of animal models complicates the development of prophylactic and therapeutic strategies targeting RV-C. Previous studies have identified human cadherin-related family member 3 (hCDHR3) as the cellular receptor for RV-C, with its expression enabling previously unsusceptible cells to support both viral entry and replication. Recently, an adult hCDHR3 transgenic mouse model was developed to investigate the role of human stimulator of interferon genes (hSTING) in RV-C15 infection in vivo. However, adult mice do not support efficient RV-C15 infection. Here, we report a transgenic mouse line expressing hCDHR3 constitutively that is highly susceptible to early-life infections by multiple serotypes of RV-C, including RV-C15, RV-C2, and RV-C41. Neonatal transgenic mice infected with various RV-C strains via the intraperitoneal (i.p.) route exhibit similar symptoms, such as severe inflammation, limb paralysis, and death. Moreover, passive immunization with antisera or therapeutic antibodies can protect against lethal RV-C infection in these transgenic mice. Overall, this study provides a valuable animal model for the in vivo antiviral evaluation against RV-C.
Collapse
Affiliation(s)
- Zhenhong Zhou
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Rui Zhu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Hongwei Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Weixi Deng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Zijie Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Yue Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Jiaxin Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Ziyang Yan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Ruoxi Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Sijia Chang
- Beijing Wantai Biological Pharmacy Enterprise Co., Ltd., Beijing, 102206, PR China
| | - Zhichao Yin
- Beijing Wantai Biological Pharmacy Enterprise Co., Ltd., Beijing, 102206, PR China
| | - Yuanyuan Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Dongqing Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Mujin Fang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Che Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Yuqiong Que
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China
| | - Yingbin Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China.
| | - Longfa Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China.
| | - Tong Cheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, 361102, PR China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, 361102, PR China.
| |
Collapse
|
3
|
Dai SZ, Wu RH, Chen H, Chen MH, Xie W, Zheng WP, Tan GH, Huang FY. Progesterone suppresses rhinovirus-induced airway inflammation by inhibiting neutrophil infiltration and extracellular traps formation. Int Immunopharmacol 2025; 144:113714. [PMID: 39626540 DOI: 10.1016/j.intimp.2024.113714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/13/2024] [Accepted: 11/23/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND The process of NETosis is observed in a range of inflammatory conditions. Progesterone (P4) has been shown to alleviate inflammation caused by viral infections such as influenza and SARS-CoV-2. However, the precise molecular mechanisms responsible for this effect are not yet fully understood. Therefore, the present investigation aims to explore whether P4 can exert its anti-inflammatory properties by inhibiting NETosis and the related molecular pathways. METHODS Airway inflammation caused by rhinovirus serotype-1b (RV-1b) was induced in male BALB/c mice. The inflammation was assessed through histological examination and calculation of inflammatory cells present in the bronchoalveolar lavage fluid. Flow cytometry was used to analyze the inflammatory cells and NETotic neutrophils. Western blotting analysis was conducted to detect proteins associated with NETosis, inflammasome activation, and signaling. Furthermore, confocal microscopy was utilized to observe neutrophil extracellular trap (NET) structures in vivo tissues and in vitro neutrophils, neutrophil infiltration, and inflammasome formation. RESULTS The administration of P4 proved to be an effective treatment for reducing airway inflammation and the production of NETs caused by RV-1b infection. The infection triggered the activation of NLRP3 inflammasomes in neutrophils, which led to the maturation of IL-1β and subsequent activation of both the NF-κB and p38 signaling pathways. The activation of NF-κB signaling resulted in the secretion of downstream chemokines CCL3 and IL-6, which led to an increase in neutrophil infiltration into the lung airways. Moreover, the activation of p38 signaling led to the generation of reactive oxygen species, resulting in NETosis. However, the administration of P4 inhibited the activation of the NLRP3 inflammasome, which subsequently led to the deactivation of both the IL-1β-NF-κB and IL-1β-p38 axes. As a result, there was a reduction in neutrophil infiltration and NETosis. Furthermore, TGF-β-activated kinase 1 (TAK1) was identified as an intermediary enzyme. P4 inhibits both the NF-κB and IL-1β-p38 pathways by suppressing the activity of TAK1. CONCLUSION The capacity of P4 to mitigate rhinovirus-induced airway inflammation is attributed to its ability to impede the infiltration of neutrophils and NETosis. As inflammation mediated by NETosis is widespread in diverse disorders, our findings propose that P4 could potentially function as a universal therapeutic agent in the management of such ailments.
Collapse
Affiliation(s)
- Shu-Zhen Dai
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China; Hainan Academy of Medical Sciences, Hainan Medical University, Hainan 571199, China
| | - Ri-Hong Wu
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Hengyu Chen
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Ming-Hui Chen
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Weijing Xie
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Wu-Ping Zheng
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Guang-Hong Tan
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China.
| | - Feng-Ying Huang
- NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine & The Second Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China.
| |
Collapse
|
4
|
Han M, Hershenson M. Infection of Immature Mice with Rhinovirus for the Study of Asthma Pathogenesis. Methods Mol Biol 2025; 2903:173-184. [PMID: 40016466 DOI: 10.1007/978-1-0716-4410-2_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Early-life wheezing-associated respiratory infection with rhinovirus (RV) is a risk factor for asthma development in combination with allergen sensitization and family history. RV infection of immature mice is a useful model to decipher potential immunological mechanisms underlying asthma development following RV infection. Increased type 2 immune response, expansion of group 2 innate lymphoid cells (ILC2s), eosinophilic inflammation, mucous metaplasia, and airway hyperresponsiveness have been observed in RV-infected six-day-old mice. Herein, we present several materials and methods used for propagating and partially purifying RV, measuring RV titers, infecting immature mice with RV, and harvesting lung tissue for whole lung mRNA extraction and flow cytometry.
Collapse
Affiliation(s)
- Mingyuan Han
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Marc Hershenson
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
5
|
Ganjian H, Sajjan U. Laboratory Protocol for Propagation and Purification of Rhinovirus A and B Suitable for In Vitro and In Vivo Infection. Methods Mol Biol 2025; 2903:9-19. [PMID: 40016454 DOI: 10.1007/978-1-0716-4410-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
H1HeLa cell line has become the most utilized cell culture system for in vitro studies of the rhinovirus life cycle. These cells are also widely used to propagate the rhinovirus to study host responses. Unlike other viruses, high-titer virus is required for studies focusing on understanding the host responses to rhinovirus infection in vivo and in vitro. Therefore, after propagation of rhinovirus, H1HeLa cells should be concentrated and purified. In this chapter, we describe the methods used for the propagation of rhinovirus A and B that are adapted to H1HeLa cells.
Collapse
Affiliation(s)
- Haleh Ganjian
- Centre for Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA
| | - Umadevi Sajjan
- Centre for Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA.
- Department of Microbiology, Immunology and Inflammation, Temple University, Philadelphia, PA, USA.
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Sajjan U. In Vivo Model to Assess the Nasal Infection by Minor Group Rhinovirus. Methods Mol Biol 2025; 2903:141-152. [PMID: 40016463 DOI: 10.1007/978-1-0716-4410-2_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Rhinovirus primarily infects humans via nasal epithelial cells and is spread through the inhalation of shed progeny virus via micro-droplets containing the virus. A handful of studies have investigated the nasal mucosal responses to rhinovirus in experimentally infected healthy volunteers. There are also studies that have investigated the effect of antiviral drugs in reducing symptoms induced by rhinovirus infection. It is extremely expensive to test the antiviral drugs in human volunteers, and therefore having preclinical in vivo models is necessary to confirm the antiviral effect of the drugs prior to conducting clinical trials. The existing in vivo models for RV infection are focused on lung inflammation, which may be relevant to patients with chronic lung diseases, in whom rhinovirus causes lower respiratory tract infections. In this chapter, we describe a mouse model of rhinovirus infection, which shows self-limiting inflammation of the sinonasal mucosa.
Collapse
Affiliation(s)
- Umadevi Sajjan
- Centre for Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA.
- Department of Microbiology, Immunology and Inflammation, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA.
- Department of Thoracic Medicine and Surgery, Lewis Katz Medical School, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Rajput C, Ganjian H, Muruganandam G, Weyer K, Dannenmaier J, Seilheimer B, Sajjan U. Euphorbium compositum SN improves the innate defenses of the airway mucosal barrier network during rhinovirus infection. Respir Res 2024; 25:407. [PMID: 39538325 PMCID: PMC11562495 DOI: 10.1186/s12931-024-03030-7] [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: 05/07/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Rhinoviruses (RV) are the major cause of common colds in healthy individuals and are associated with acute exacerbations in patients with chronic lung diseases. Yet, no vaccines or effective treatment against RV are available. This study investigated the effect of Euphorbium compositum SN (ECSN6), a multicomponent, multitarget medication made from natural ingredients, on the mucosal barrier network during RV infection. METHODS Mucociliary-differentiated airway epithelial cell cultures were infected with RV or sham, and treated with 20% ECSN6 or placebo twice daily. Barrier integrity was assessed by measuring transepithelial resistance (TER), permeability to inulin, and expression and localization of intercellular junctions proteins (IJ). Ciliary beat frequency (CBF), expression of pro-inflammatory cytokines, antiviral interferons and mucins, and viral load were also measured. C57BL/6 mice were infected intranasally with RV or sham and treated with 40% ECSN6 or placebo twice daily. Inflammation of sinunasal mucosa, localization of E-cadherin, viral load and mucin gene expression were determined. RESULTS ECSN6-treated, uninfected cell cultures showed small, but significant increase in TER over placebo, which was associated with enhanced localization of E-cadherin and ZO-1 to IJ. In RV-infected cultures, treatment with ECSN6, but not placebo prevented RV-induced (1) reduction in TER, (2) dissociation of E-cadherin and ZO-1 from the IJ, (3) mucin expression, and (4) CBF attenuation. ECSN6 also decreased RV-stimulated expression of pro-inflammatory cytokines and permeability to inulin. Although ECSN6 significantly increased the expression of some antiviral type I and type III interferons, it did not alter viral load. In vivo, ECSN6 reduced RV-A1-induced moderate inflammation of nasal mucosa, beneficially affected RV-A1-induced cytokine responses and Muc5ac mRNA expression and prevented RV-caused dissociation of E-cadherin from the IJ of nasal mucosa without an effect on viral clearance. CONCLUSIONS ECSN6 prevents RV-induced airway mucosal barrier dysfunction and improves the immunological and mucociliary barrier function. ECSN6 may maintain integrity of barrier function by promoting localization of tight and adherence junction proteins to the IJ. This in turn may lead to the observed decrease in RV-induced pro-inflammatory responses in vitro. By improving the innate defenses of the airway mucosal barrier network, ECSN6 may alleviate respiratory symptoms caused by RV infections.
Collapse
Affiliation(s)
- Charu Rajput
- Center for Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, PA, 19140, USA
| | - Haleh Ganjian
- Center for Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, PA, 19140, USA
| | - Ganesh Muruganandam
- Center for Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, PA, 19140, USA
| | | | | | | | - Umadevi Sajjan
- Center for Inflammation and Lung Research, Lewis Katz Medical School, Temple University, Philadelphia, PA, 19140, USA.
- Department of Microbiology, Immunology and Inflammation, Lewis Katz Medical School, Temple University, Philadelphia, PA, 19140, USA.
- Department of Thoracic Medicine and Surgery, Temple University Health System, Philadelphia, PA, 19140, USA.
| |
Collapse
|
8
|
Bentley JK, Kreger JE, Breckenridge HA, Singh S, Lei J, Li Y, Baker SC, Lumeng CN, Hershenson MB. Developing a mouse model of human coronavirus NL63 infection: comparison with rhinovirus-A1B and effects of prior rhinovirus infection. Am J Physiol Lung Cell Mol Physiol 2024; 327:L557-L573. [PMID: 39189801 PMCID: PMC11888781 DOI: 10.1152/ajplung.00149.2023] [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: 05/09/2023] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 08/28/2024] Open
Abstract
Human coronavirus (HCoV)-NL63 causes respiratory tract infections in humans and uses angiotensin-converting enzyme 2 (ACE2) as a receptor. We sought to establish a mouse model of HCoV-NL63 and determine whether prior rhinovirus (RV)-A1B infection affected HCoV-NL63 replication. HCoV-NL63 was propagated in LLC-MK2 cells expressing human ACE2. RV-A1B was grown in HeLa-H1 cells. C57BL6/J or transgenic mice expressing human ACE2 were infected intranasally with sham LLC-MK2 cell supernatant or 1 × 105 tissue culture infectious dose (TCID50) units HCoV-NL63. Wild-type mice were infected with 1 × 106 plaque-forming units (PFU) RV-A1B. Lungs were assessed for vRNA, bronchoalveolar lavage (BAL) cells, histology, HCoV-NL63 nonstructural protein 3 (nsp3), and host gene expression by next-generation sequencing and qPCR. To evaluate sequential infections, mice were infected with RV-A1B followed by HCoV-NL63 infection 4 days later. We report that hACE2 mice infected with HCoV-NL63 showed evidence of replicative infection with increased levels of vRNA, BAL neutrophils and lymphocytes, peribronchial and perivascular infiltrates, and expression of nsp3. Viral replication peaked 3 days after infection and inflammation persisted 6 days after infection. HCoV-NL63-infected hACE2 mice showed increased mRNA expression of IFNs, IFN-stimulated proteins, and proinflammatory cytokines. Infection with RV-A1B 4 days before HCoV-NL63 significantly decreased both HCoV-NL63 vRNA levels and airway inflammation. Mice infected with RV-A1B prior to HCoV-NL63 showed increased expression of antiviral proteins compared with sham-treated mice. In conclusion, we established a mouse model of HCoV-NL63 replicative infection characterized by relatively persistent viral replication and inflammation. Prior infection with RV-A1B reduced HCoV-NL63 replication and airway inflammation, indicative of viral interference.NEW & NOTEWORTHY We describe a mouse model of human coronavirus (HCoV) infection. Infection of transgenic mice expressing human angiotensin-converting enzyme 2 (ACE2) with HCoV-NL63 produced a replicative infection with peribronchial inflammation and nonstructural protein 3 expression. Mice infected with RV-A1B 4 days before HCoV-NL63 showed decreased HCoV-NL63 replication and airway inflammation and increased expression of antiviral proteins compared with sham-treated mice. This research may shed light on human coronavirus infections, viral interference, and viral-induced asthma exacerbations.
Collapse
Affiliation(s)
- J Kelley Bentley
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Jordan E Kreger
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Haley A Breckenridge
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Shilpi Singh
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Jing Lei
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Yiran Li
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Susan C Baker
- Department of Microbiology and Immunology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, United States
| | - Carey N Lumeng
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
- Department Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Marc B Hershenson
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
- Department Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| |
Collapse
|
9
|
Berdnikovs S, Newcomb DC, Hartert TV. How early life respiratory viral infections impact airway epithelial development and may lead to asthma. Front Pediatr 2024; 12:1441293. [PMID: 39156016 PMCID: PMC11327159 DOI: 10.3389/fped.2024.1441293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/25/2024] [Indexed: 08/20/2024] Open
Abstract
Childhood asthma is a common chronic disease of the airways that results from host and environment interactions. Most risk factor studies of asthma point to the first year of life as a susceptibility window of mucosal exposure that directly impacts the airway epithelium and airway epithelial cell development. The development of the airway epithelium, which forms a competent barrier resulting from coordinated interactions of different specialized cell subsets, occurs during a critical time frame in normal postnatal development in the first year of life. Understanding the normal and aberrant developmental trajectory of airway epithelial cells is important in identifying pathways that may contribute to barrier dysfunction and asthma pathogenesis. Respiratory viruses make first contact with and infect the airway mucosa. Human rhinovirus (HRV) and respiratory syncytial virus (RSV) are mucosal pathogens that are consistently identified as asthma risk factors. Respiratory viruses represent a unique early life exposure, different from passive irritant exposures which injure the developing airway epithelium. To replicate, respiratory viruses take over the host cell transcriptional and translational processes and exploit host cell energy metabolism. This takeover impacts the development and differentiation processes of airway epithelial cells. Therefore, delineating the mechanisms through which early life respiratory viral infections alter airway epithelial cell development will allow us to understand the maturation and heterogeneity of asthma and develop tools tailored to prevent disease in specific children. This review will summarize what is understood about the impact of early life respiratory viruses on the developing airway epithelium and define critical gaps in our knowledge.
Collapse
Affiliation(s)
- Sergejs Berdnikovs
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Dawn C. Newcomb
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Tina V. Hartert
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
10
|
Bouzari B, Chugaeva UY, Karampoor S, Mirzaei R. Immunometabolites in viral infections: Action mechanism and function. J Med Virol 2024; 96:e29807. [PMID: 39037069 DOI: 10.1002/jmv.29807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/10/2024] [Accepted: 07/05/2024] [Indexed: 07/23/2024]
Abstract
The interplay between viral pathogens and host metabolism plays a pivotal role in determining the outcome of viral infections. Upon viral detection, the metabolic landscape of the host cell undergoes significant changes, shifting from oxidative respiration via the tricarboxylic acid (TCA) cycle to increased aerobic glycolysis. This metabolic shift is accompanied by elevated nutrient accessibility, which is vital for cell function, development, and proliferation. Furthermore, depositing metabolites derived from fatty acids, TCA intermediates, and amino acid catabolism accelerates the immunometabolic transition, facilitating pro-inflammatory and antimicrobial responses. Immunometabolites refer to small molecules involved in cellular metabolism regulating the immune response. These molecules include nutrients, such as glucose and amino acids, along with metabolic intermediates and signaling molecules adenosine, lactate, itaconate, succinate, kynurenine, and prostaglandins. Emerging evidence suggests that immunometabolites released by immune cells establish a complex interaction network within local niches, orchestrating and fine-tuning immune responses during viral diseases. However, our current understanding of the immense capacity of metabolites to convey essential cell signals from one cell to another or within cellular compartments remains incomplete. Unraveling these complexities would be crucial for harnessing the potential of immunometabolites in therapeutic interventions. In this review, we discuss specific immunometabolites and their mechanisms of action in viral infections, emphasizing recent findings and future directions in this rapidly evolving field.
Collapse
Affiliation(s)
- Behnaz Bouzari
- Department of Pathology, Firouzgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Uliana Y Chugaeva
- Department of Pediatric, Preventive Dentistry and Orthodontics, Institute of Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
11
|
Li Q, Jiang G, Lv Y. Inhibition of phosphoinositide 3-kinase activity attenuates neutrophilic airway inflammation and inhibits pyrin domain-containing 3 inflammasome activation in an ovalbumin-lipopolysaccharide-induced asthma murine model. Mol Biol Rep 2024; 51:698. [PMID: 38811424 PMCID: PMC11136729 DOI: 10.1007/s11033-024-09360-5] [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: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Existing investigations suggest that the blockade of phosphoinositide 3-kinase (PI3K) activity contributes to inflammatory solution in allergic asthma, but whether this inhibition directly attenuates neutrophilic airway inflammation in vivo is still unclear. We explored the pharmacological effects of LY294002, a specific inhibitor of PI3K on the progression of neutrophilic airway inflammation and investigated the underlying mechanism. METHODS AND RESULTS Female C57BL/6 mice were intranasally sensitized with ovalbumin (OVA) together with lipopolysaccharide (LPS) on days 0 and 6, and challenged with OVA on days 14-17 to establish a neutrophilic airway disease model. In the challenge phase, a subset of mice was treated intratracheally with LY294002. We found that treatment of LY294002 attenuates clinic symptoms of inflammatory mice. Histological studies showed that LY294002 significantly inhibited inflammatory cell infiltration and mucus production. The treatment also significantly inhibited OVA-LPS induced increases in inflammatory cell counts, especially neutrophil counts, and IL-17 levels in bronchoalveolar lavage fluid (BALF). LY294002 treated mice exhibited significantly increased IL-10 levels in BALF compared to the untreated mice. In addition, LY294002 reduced the plasma concentrations of IL-6 and IL-17. The anti-inflammatory effects of LY29402 were correlated with the downregulation of NLRP3 inflammasome. CONCLUSIONS Our findings suggested that LY294002 as a potential pharmacological target for neutrophilic airway inflammation.
Collapse
Affiliation(s)
- Qun Li
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Bengbu Medical University, Anhui, China
| | - Guiyun Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Yunxiang Lv
- Department of Pulmonary and Critical Care MedicineAnhui Clinical and Preclinical Key Laboratory of Respiratory DiseaseMolecular Diagnosis Center, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China.
| |
Collapse
|
12
|
Li Y, Hershenson MB. Remember the Airway Smooth Muscle! How Rhinovirus Impairs Bronchodilator Responses. Am J Respir Cell Mol Biol 2023; 69:121-122. [PMID: 37163760 PMCID: PMC10399143 DOI: 10.1165/rcmb.2023-0146ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Affiliation(s)
- Yiran Li
- Departments of Pediatrics and Molecular and Integrative Physiology University of Michigan Medical School Ann Arbor, Michigan
| | - Marc B Hershenson
- Departments of Pediatrics and Molecular and Integrative Physiology University of Michigan Medical School Ann Arbor, Michigan
| |
Collapse
|
13
|
Gandhi VD, Cephus JY, Norlander AE, Chowdhury NU, Zhang J, Ceneviva ZJ, Tannous E, Polosukhin VV, Putz ND, Wickersham N, Singh A, Ware LB, Bastarache JA, Shaver CM, Chu HW, Peebles RS, Newcomb DC. Androgen receptor signaling promotes Treg suppressive function during allergic airway inflammation. J Clin Invest 2022; 132:e153397. [PMID: 35025767 PMCID: PMC8843736 DOI: 10.1172/jci153397] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 01/04/2022] [Indexed: 11/23/2022] Open
Abstract
Women have higher prevalence of asthma compared with men. In asthma, allergic airway inflammation is initiated by IL-33 signaling through ST2, leading to increased IL-4, IL-5, and IL-13 production and eosinophil infiltration. Foxp3+ Tregs suppress and ST2+ Tregs promote allergic airway inflammation. Clinical studies showed that the androgen dehydroepiandrosterone (DHEA) reduced asthma symptoms in patients, and mouse studies showed that androgen receptor (AR) signaling decreased allergic airway inflammation. Yet the impact of AR signaling on lung Tregs remains unclear. Using AR-deficient and Foxp3 fate-mapping mice, we determined that AR signaling increased Treg suppression during Alternaria extract (Alt Ext; allergen) challenge by stabilizing Foxp3+ Tregs and limiting the number of ST2+ ex-Tregs and IL-13+ Th2 cells and ex-Tregs. AR signaling also decreased Alt Ext-induced ST2+ Tregs in mice by limiting expression of Gata2, a transcription factor for ST2, and by decreasing Alt Ext-induced IL-33 production from murine airway epithelial cells. We confirmed our findings in human cells where 5α-dihydrotestosterone (DHT), an androgen, decreased IL-33-induced ST2 expression in lung Tregs and decreased Alt Ext-induced IL-33 secretion in human bronchial epithelial cells. Our findings showed that AR signaling stabilized Treg suppressive function, providing a mechanism for the sex difference in asthma.
Collapse
Affiliation(s)
| | | | | | - Nowrin U. Chowdhury
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | | | | | | | | | - Amrit Singh
- Prevention of Organ Failure (PROOF) Centre of Excellence, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | - Hong Wei Chu
- National Jewish Medical Center, Denver, Colorado, USA
| | - R. Stokes Peebles
- Department of Medicine and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Dawn C. Newcomb
- Department of Medicine and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
14
|
Bentley JK. Modeling Asthma in Mice Using Rhinovirus Infection. Methods Mol Biol 2022; 2506:43-56. [PMID: 35771462 DOI: 10.1007/978-1-0716-2364-0_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Rhinovirus (RV) infection is linked to early life wheezing and exacerbation of adult asthma. RV infection can be modeled in adult and neonatal mice. This chapter outlines methods for the production of standardized human rhinovirus A1B and mouse infection. The chapter also describes methods to couple infections with allergen (ovalbumin and house dust mite) administrations. The production of the virus involves its amplification, purification, and concentration. In order to standardize the concentrated RV stock, a plaque assay on HeLa cells is outlined as a method of calibrating infectivity. Once the number of plaque-forming units is determined, the standardized virus is used for mouse infection.
Collapse
Affiliation(s)
- J Kelley Bentley
- Departments of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
15
|
Moradi S, Jarrahi E, Ahmadi A, Salimian J, Karimi M, Zarei A, Azimzadeh Jamalkandi S, Ghanei M. PI3K signalling in chronic obstructive pulmonary disease and opportunities for therapy. J Pathol 2021; 254:505-518. [PMID: 33959951 DOI: 10.1002/path.5696] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 04/01/2021] [Accepted: 04/26/2021] [Indexed: 11/08/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic lung disease characterised by airway inflammation and progressive obstruction of the lung airflow. Current pharmacological treatments include bronchodilators, alone or in combination with steroids, or other anti-inflammatory agents, which have only partially contributed to the inhibition of disease progression and mortality. Therefore, further research unravelling the underlying mechanisms is necessary to develop new anti-COPD drugs with both lower toxicity and higher efficacy. Extrinsic signalling pathways play crucial roles in COPD development and exacerbations. In particular, phosphoinositide 3-kinase (PI3K) signalling has recently been shown to be a major driver of the COPD phenotype. Therefore, several small-molecule inhibitors have been identified to block the hyperactivation of this signalling pathway in COPD patients, many of them showing promising outcomes in both preclinical animal models of COPD and human clinical trials. In this review, we discuss the critically important roles played by hyperactivated PI3K signalling in the pathogenesis of COPD. We also critically review current therapeutics based on PI3K inhibition, and provide suggestions focusing on PI3K signalling for the further improvement of the COPD phenotype. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Sharif Moradi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Esmaeil Jarrahi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ali Ahmadi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Salimian
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehrdad Karimi
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadeh Zarei
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadegh Azimzadeh Jamalkandi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Rajput C, Han M, Ishikawa T, Lei J, Goldsmith AM, Jazaeri S, Stroupe CC, Bentley JK, Hershenson MB. Rhinovirus C Infection Induces Type 2 Innate Lymphoid Cell Expansion and Eosinophilic Airway Inflammation. Front Immunol 2021; 12:649520. [PMID: 33968043 PMCID: PMC8100319 DOI: 10.3389/fimmu.2021.649520] [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: 01/04/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
Rhinovirus C (RV-C) infection is associated with severe asthma exacerbations. Since type 2 inflammation is an important disease mechanism in asthma, we hypothesized that RV-C infection, in contrast to RV-A, preferentially stimulates type 2 inflammation, leading to exacerbated eosinophilic inflammation. To test this, we developed a mouse model of RV-C15 airways disease. RV-C15 was generated from the full-length cDNA clone and grown in HeLa-E8 cells expressing human CDHR3. BALB/c mice were inoculated intranasally with 5 x 106 ePFU RV-C15, RV-A1B or sham. Mice inoculated with RV-C15 showed lung viral titers of 1 x 105 TCID50 units 24 h after infection, with levels declining thereafter. IFN-α, β, γ and λ2 mRNAs peaked 24-72 hrs post-infection. Immunofluorescence verified colocalization of RV-C15, CDHR3 and acetyl-α-tubulin in mouse ciliated airway epithelial cells. Compared to RV-A1B, mice infected with RV-C15 demonstrated higher bronchoalveolar eosinophils, mRNA expression of IL-5, IL-13, IL-25, Muc5ac and Gob5/Clca, protein production of IL-5, IL-13, IL-25, IL-33 and TSLP, and expansion of type 2 innate lymphoid cells. Analogous results were found in mice treated with house dust mite before infection, including increased airway responsiveness. In contrast to Rorafl/fl littermates, RV-C-infected Rorafl/flIl7rcre mice deficient in ILC2s failed to show eosinophilic inflammation or mRNA expression of IL-13, Muc5ac and Muc5b. We conclude that, compared to RV-A1B, RV-C15 infection induces ILC2-dependent type 2 airway inflammation, providing insight into the mechanism of RV-C-induced asthma exacerbations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Marc B. Hershenson
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
17
|
Besednova NN, Andryukov BG, Zaporozhets TS, Kryzhanovsky SP, Fedyanina LN, Kuznetsova TA, Zvyagintseva TN, Shchelkanov MY. Antiviral Effects of Polyphenols from Marine Algae. Biomedicines 2021; 9:200. [PMID: 33671278 PMCID: PMC7921925 DOI: 10.3390/biomedicines9020200] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 02/07/2023] Open
Abstract
The disease-preventive and medicinal properties of plant polyphenolic compounds have long been known. As active ingredients, they are used to prevent and treat many noncommunicable diseases. In recent decades, marine macroalgae have attracted the attention of biotechnologists and pharmacologists as a promising and almost inexhaustible source of polyphenols. This heterogeneous group of compounds contains many biopolymers with unique structure and biological properties that exhibit high anti-infective activity. In the present review, the authors focus on the antiviral potential of polyphenolic compounds (phlorotannins) from marine algae and consider the mechanisms of their action as well as other biological properties of these compounds that have effects on the progress and outcome of viral infections. Effective nutraceuticals, to be potentially developed on the basis of algal polyphenols, can also be used in the complex therapy of viral diseases. It is necessary to extend in vivo studies on laboratory animals, which subsequently will allow proceeding to clinical tests. Polyphenolic compounds have a great potential as active ingredients to be used for the creation of new antiviral pharmaceutical substances.
Collapse
Affiliation(s)
- Natalya N. Besednova
- G.P. Somov Institute of Epidemiology and Microbiology, Russian Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (B.G.A.); (T.S.Z.); (T.A.K.); (M.Y.S.)
| | - Boris G. Andryukov
- G.P. Somov Institute of Epidemiology and Microbiology, Russian Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (B.G.A.); (T.S.Z.); (T.A.K.); (M.Y.S.)
- School of Biomedicine, Far Eastern Federal University (FEFU), 690091 Vladivostok, Russia;
| | - Tatyana S. Zaporozhets
- G.P. Somov Institute of Epidemiology and Microbiology, Russian Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (B.G.A.); (T.S.Z.); (T.A.K.); (M.Y.S.)
| | - Sergey P. Kryzhanovsky
- Medical Association of the Far Eastern Branch of the Russian Academy of Sciences, 690022 Vladivostok, Russia;
| | - Ludmila N. Fedyanina
- School of Biomedicine, Far Eastern Federal University (FEFU), 690091 Vladivostok, Russia;
| | - Tatyana A. Kuznetsova
- G.P. Somov Institute of Epidemiology and Microbiology, Russian Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (B.G.A.); (T.S.Z.); (T.A.K.); (M.Y.S.)
| | | | - Mikhail Yu. Shchelkanov
- G.P. Somov Institute of Epidemiology and Microbiology, Russian Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (B.G.A.); (T.S.Z.); (T.A.K.); (M.Y.S.)
- School of Biomedicine, Far Eastern Federal University (FEFU), 690091 Vladivostok, Russia;
- Federal Scientific Center of the Eastern Asia Terrestrial Biodiversity, Far Eastern Branch of Russian Academy of Sciences, 690091 Vladivostok, Russia
- National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, 690091 Vladivostok, Russia
| |
Collapse
|
18
|
Marsh EK, Prestwich EC, Williams L, Hart AR, Muir CF, Parker LC, Jonker MR, Heijink IH, Timens W, Fife M, Hussell T, Hershenson MB, Bentley JK, Sun SC, Barksby BS, Borthwick LA, Stewart JP, Sabroe I, Dockrell DH, Marriott HM. Pellino-1 Regulates the Responses of the Airway to Viral Infection. Front Cell Infect Microbiol 2020; 10:456. [PMID: 32984077 PMCID: PMC7488214 DOI: 10.3389/fcimb.2020.00456] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/24/2020] [Indexed: 01/02/2023] Open
Abstract
Exposure to respiratory pathogens is a leading cause of exacerbations of airway diseases such as asthma and chronic obstructive pulmonary disease (COPD). Pellino-1 is an E3 ubiquitin ligase known to regulate virally-induced inflammation. We wished to determine the role of Pellino-1 in the host response to respiratory viruses in health and disease. Pellino-1 expression was examined in bronchial sections from patients with GOLD stage two COPD and healthy controls. Primary bronchial epithelial cells (PBECs) in which Pellino-1 expression had been knocked down were extracellularly challenged with the TLR3 agonist poly(I:C). C57BL/6 Peli1-/- mice and wild type littermates were subjected to intranasal infection with clinically-relevant respiratory viruses: rhinovirus (RV1B) and influenza A. We found that Pellino-1 is expressed in the airways of normal subjects and those with COPD, and that Pellino-1 regulates TLR3 signaling and responses to airways viruses. In particular we observed that knockout of Pellino-1 in the murine lung resulted in increased production of proinflammatory cytokines IL-6 and TNFα upon viral infection, accompanied by enhanced recruitment of immune cells to the airways, without any change in viral replication. Pellino-1 therefore regulates inflammatory airway responses without altering replication of respiratory viruses.
Collapse
Affiliation(s)
- Elizabeth K. Marsh
- Department of Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom,Human Sciences Research Centre, College of Life and Natural Sciences, University of Derby, Derby, United Kingdom
| | - Elizabeth C. Prestwich
- Department of Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom
| | - Lynne Williams
- Department of Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom
| | - Amber R. Hart
- Department of Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom
| | - Clare F. Muir
- Department of Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom
| | - Lisa C. Parker
- Department of Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom
| | - Marnix R. Jonker
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Irene H. Heijink
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Mark Fife
- Manchester Collaborative Centre for Inflammation Research, Core Technology Facility, University of Manchester, Manchester, United Kingdom
| | - Tracy Hussell
- Manchester Collaborative Centre for Inflammation Research, Core Technology Facility, University of Manchester, Manchester, United Kingdom
| | - Marc B. Hershenson
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, United States
| | - J. Kelley Bentley
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Shao-Cong Sun
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ben S. Barksby
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lee A. Borthwick
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - James P. Stewart
- Department of Infection Biology, University of Liverpool, Liverpool, United Kingdom
| | - Ian Sabroe
- Department of Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom
| | - David H. Dockrell
- Department of Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom,MRC/UoE Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Helen M. Marriott
- Department of Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom,*Correspondence: Helen M. Marriott
| |
Collapse
|
19
|
Han M, Ishikawa T, Bermick JR, Rajput C, Lei J, Goldsmith AM, Jarman CR, Lee J, Bentley JK, Hershenson MB. IL-1β prevents ILC2 expansion, type 2 cytokine secretion, and mucus metaplasia in response to early-life rhinovirus infection in mice. Allergy 2020; 75:2005-2019. [PMID: 32086822 DOI: 10.1111/all.14241] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 01/15/2020] [Accepted: 01/18/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Early-life wheezing-associated respiratory infection with human rhinovirus (RV) is associated with asthma development. RV infection of 6-day-old immature mice causes mucous metaplasia and airway hyperresponsiveness which is associated with the expansion of IL-13-producing type 2 innate lymphoid cells (ILC2s) and dependent on IL-25 and IL-33. We examined regulation of this asthma-like phenotype by IL-1β. METHODS Six-day-old wild-type or NRLP3-/- mice were inoculated with sham or RV-A1B. Selected mice were treated with IL-1 receptor antagonist (IL-1RA), anti-IL-1β, or recombinant IL-1β. RESULTS Rhinovirus infection induced Il25, Il33, Il4, Il5, Il13, muc5ac, and gob5 mRNA expression, ILC2 expansion, mucus metaplasia, and airway hyperresponsiveness. RV also induced lung mRNA and protein expression of pro-IL-1β and NLRP3 as well as cleavage of caspase-1 and pro-IL-1β, indicating inflammasome priming and activation. Lung macrophages were a major source of IL-1β. Inhibition of IL-1β signaling with IL-1RA, anti-IL-1β, or NLRP3 KO increased RV-induced type 2 cytokine immune responses, ILC2 number, and mucus metaplasia, while decreasing IL-17 mRNA expression. Treatment with IL-1β had the opposite effect, decreasing IL-25, IL-33, and mucous metaplasia while increasing IL-17 expression. IL-1β and IL-17 each suppressed Il25, Il33, and muc5ac mRNA expression in cultured airway epithelial cells. Finally, RV-infected 6-day-old mice showed reduced IL-1β mRNA and protein expression compared to mature mice. CONCLUSION Macrophage IL-1β limits type 2 inflammation and mucous metaplasia following RV infection by suppressing epithelial cell innate cytokine expression. Reduced IL-1β production in immature animals provides a mechanism permitting asthma development after early-life viral infection.
Collapse
Affiliation(s)
- Mingyuan Han
- Departments of Pediatrics University of Michigan Medical School Ann Arbor Michigan
| | - Tomoko Ishikawa
- Departments of Pediatrics University of Michigan Medical School Ann Arbor Michigan
| | - Jennifer R. Bermick
- Departments of Pediatrics University of Michigan Medical School Ann Arbor Michigan
| | - Charu Rajput
- Departments of Pediatrics University of Michigan Medical School Ann Arbor Michigan
| | - Jing Lei
- Departments of Pediatrics University of Michigan Medical School Ann Arbor Michigan
| | - Adam M. Goldsmith
- Departments of Pediatrics University of Michigan Medical School Ann Arbor Michigan
| | - Caitlin R. Jarman
- Departments of Pediatrics University of Michigan Medical School Ann Arbor Michigan
| | - Julie Lee
- Departments of Pediatrics University of Michigan Medical School Ann Arbor Michigan
| | - J. Kelley Bentley
- Departments of Pediatrics University of Michigan Medical School Ann Arbor Michigan
| | - Marc B. Hershenson
- Departments of Pediatrics University of Michigan Medical School Ann Arbor Michigan
- Departments of Molecular and Integrative Physiology University of Michigan Medical School Ann Arbor Michigan
| |
Collapse
|
20
|
Early-life heterologous rhinovirus infections induce an exaggerated asthma-like phenotype. J Allergy Clin Immunol 2020; 146:571-582.e3. [PMID: 32344055 DOI: 10.1016/j.jaci.2020.03.039] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 03/11/2020] [Accepted: 03/26/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Early-life wheezing-associated respiratory tract infection by rhinovirus (RV) is a risk factor for asthma development. Infants are infected with many different RV strains per year. OBJECTIVE We previously showed that RV infection of 6-day-old BALB/c mice induces a mucous metaplasia phenotype that is dependent on type 2 innate lymphoid cells (ILC2s). We hypothesized that early-life RV infection alters the response to subsequent heterologous infection, inducing an exaggerated asthma-like phenotype. METHODS Wild-type BALB/c mice and Rorafl/flIl7rcre mice lacking ILC2s were treated as follows: (1) sham on day 6 of life plus sham on day 13 of life, (2) RV-A1B on day 6 plus sham on day 13, (3) sham on day 6 plus RV-A2 on day 13, and (4) RV-A1B on day 6 plus RV-A2 on day 13. RESULTS Mice infected with RV-A1B at day 6 and sham at day 13 showed an increased number of bronchoalveolar lavage eosinophils and increased expression of IL-13 mRNA but not expression of IFN-γ mRNA (which is indicative of a type 2 immune response), whereas mice infected with sham on day 6 and RV-A2 on day 13 of life demonstrated increased IFN-γ expression (which is a mature antiviral response). In contrast, mice infected with RV-A1B on day 6 before RV-A2 infection on day 13 showed increased expression of IL-13, IL-5, Gob5, Muc5b, and Muc5ac mRNA; increased numbers of eosinophils and IL-13-producing ILC2s; and exaggerated mucus metaplasia and airway hyperresponsiveness. Compared with Rorafl/fl mice, Rorafl/flIl7rcre mice showed complete suppression of bronchoalveolar lavage eosinophils and mucous metaplasia. CONCLUSION Early-life RV infection alters the response to subsequent heterologous infection, inducing an intensified asthma-like phenotype that is dependent on ILC2s.
Collapse
|
21
|
FOXO3a regulates rhinovirus-induced innate immune responses in airway epithelial cells. Sci Rep 2019; 9:18180. [PMID: 31796819 PMCID: PMC6890790 DOI: 10.1038/s41598-019-54567-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/15/2019] [Indexed: 12/27/2022] Open
Abstract
Forkhead transcription factor class O (FOXO)3a, which plays a critical role in a wide variety of cellular processes, was also found to regulate cell-type-specific antiviral responses. Airway epithelial cells express FOXO3a and play an important role in clearing rhinovirus (RV) by mounting antiviral type I and type III interferon (IFN) responses. To elucidate the role of FOXO3a in regulating antiviral responses, we generated airway epithelial cell-specific Foxo3a knockout (Scga1b1-Foxo3a−/−) mice and a stable FOXO3a knockout human airway epithelial cell line. Compared to wild-type, Scga1b1-Foxo3a−/− mice show reduced IFN-α, IFN-β, IFN-λ2/3 in response to challenge with RV or double-stranded (ds)RNA mimic, Poly Inosinic-polycytidylic acid (Poly I:C) indicating defective dsRNA receptor signaling. RV-infected Scga1b1-Foxo3a−/− mice also show viral persistence, enhanced lung inflammation and elevated pro-inflammatory cytokine levels. FOXO3a K/O airway epithelial cells show attenuated IFN responses to RV infection and this was associated with conformational change in mitochondrial antiviral signaling protein (MAVS) but not with a reduction in the expression of dsRNA receptors under unstimulated conditions. Pretreatment with MitoTEMPO, a mitochondrial-specific antioxidant corrects MAVS conformation and restores antiviral IFN responses to subsequent RV infection in FOXO3a K/O cells. Inhibition of oxidative stress also reduces pro-inflammatory cytokine responses to RV in FOXO3a K/O cells. Together, our results indicate that FOXO3a plays a critical role in regulating antiviral responses as well as limiting pro-inflammatory cytokine expression. Based on these results, we conclude that FOXO3a contributes to optimal viral clearance and prevents excessive lung inflammation following RV infection.
Collapse
|
22
|
Xander N, Reddy Vari H, Eskandar R, Li W, Bolla S, Marchetti N, Sajjan US. Rhinovirus-Induced SIRT-1 via TLR2 Regulates Subsequent Type I and Type III IFN Responses in Airway Epithelial Cells. THE JOURNAL OF IMMUNOLOGY 2019; 203:2508-2519. [PMID: 31548332 DOI: 10.4049/jimmunol.1900165] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 08/21/2019] [Indexed: 01/10/2023]
Abstract
IFN responses to viral infection are necessary to establish intrinsic antiviral state, but if unchecked can lead to heightened inflammation. Recently, we showed that TLR2 activation contributes to limitation of rhinovirus (RV)-induced IFN response in the airway epithelial cells. We also demonstrated that compared with normal airway epithelial cells, those from patients with chronic obstructive pulmonary disease (COPD) show higher IFN responses to RV, but the underlying mechanisms are not known. Initially, RV-induced IFN responses depend on dsRNA receptor activation and then are amplified via IFN-stimulated activation of JAK/STAT signaling. In this study, we show that in normal cells, TLR2 limits RV-induced IFN responses by attenuating STAT1 and STAT2 phosphorylation and this was associated with TLR2-dependent SIRT-1 expression. Further, inhibition of SIRT-1 enhanced RV-induced IFN responses, and this was accompanied by increased STAT1/STAT2 phosphorylation, indicating that TLR2 may limit RV-induced IFN responses via SIRT-1. COPD airway epithelial cells showed attenuated IL-8 responses to TLR2 agonist despite expressing TLR2 similar to normal, indicating dysregulation in TLR2 signaling pathway. Unlike normal, COPD cells failed to show RV-induced TLR2-dependent SIRT-1 expression. Pretreatment with quercetin, which increases SIRT-1 expression, normalized RV-induced IFN levels in COPD airway epithelial cells. Inhibition of SIRT-1 in quercetin-pretreated COPD cells abolished the normalizing effects of quercetin on RV-induced IFN expression in these cells, confirming that quercetin exerts its effect via SIRT-1. In summary, we show that TLR2 is required for limiting RV-induced IFNs, and this pathway is dysregulated in COPD airway epithelial cells, leading to exaggerated IFN production.
Collapse
Affiliation(s)
- Nathaniel Xander
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA 19140; and
| | - Hymavathi Reddy Vari
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA 19140; and
| | - Rewees Eskandar
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA 19140; and
| | - Wuyan Li
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA 19140; and
| | - Sudhir Bolla
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA 19140; and
| | - Nathaniel Marchetti
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA 19140; and
| | - Umadevi S Sajjan
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA 19140; and .,Department of Physiology, Temple University, Philadelphia, PA 19140
| |
Collapse
|
23
|
Menzel M, Kosinski J, Uller L, Akbarshahi H. Rhinovirus-induced IFNβ expression is NFκB-dependent and regulated by the macrophage microenvironment. Sci Rep 2019; 9:13394. [PMID: 31527772 PMCID: PMC6746757 DOI: 10.1038/s41598-019-50034-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 09/04/2019] [Indexed: 12/18/2022] Open
Abstract
Macrophages play an important role in asthma pathogenesis both in the inflammatory and resolution phase of the disease. Macrophages can acquire different polarisation states dependent on their microenvironment. It is yet unclear through which mechanism the microenvironment affects the anti-viral response in macrophages. We hypothesized that the macrophage microenvironment regulates rhinovirus-induced IFNβ expression. Murine bone marrow-derived monocytes and human differentiated THP-1 cells were stimulated with M-CSF or GM-CSF and IFNγ or IL-4/IL-13, respectively, to mimic a Th1 or Th2 environment. Macrophages were infected with rhinovirus and gene and protein levels of IFNβ and pattern recognition receptor expression were measured. In subsequent experiments an IκB kinase inhibitor was used to study the involvement of NFκB. Both murine and human M1-like macrophages exhibited higher levels of IFNβ and pattern recognition receptors after rhinovirus infection than M2-like macrophages. Blockage of NFκB resulted in a lower expression of rhinovirus-induced IFNβ in human M1-like macrophages while inducing a higher expression in M2-like macrophages, suggesting that the interferon response towards viral infection was mediated by NFκB. These findings could contribute to a better understanding of mechanisms causing reduced anti-viral responses at viral-induced exacerbations in asthma.
Collapse
Affiliation(s)
- Mandy Menzel
- Respiratory Immunopharmacology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Joakim Kosinski
- Respiratory Immunopharmacology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Lena Uller
- Respiratory Immunopharmacology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Hamid Akbarshahi
- Respiratory Immunopharmacology, Department of Experimental Medical Science, Lund University, Lund, Sweden. .,Respiratory Medicine and Allergology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden.
| |
Collapse
|
24
|
Han M, Rajput C, Hershenson MB. Rhinovirus Attributes that Contribute to Asthma Development. Immunol Allergy Clin North Am 2019; 39:345-359. [PMID: 31284925 PMCID: PMC6624084 DOI: 10.1016/j.iac.2019.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Early-life wheezing-associated infections with human rhinovirus (HRV) are strongly associated with the inception of asthma. The immune system of immature mice and humans is skewed toward a type 2 cytokine response. Thus, HRV-infected 6-day-old mice but not adult mice develop augmented type 2 cytokine expression, eosinophilic inflammation, mucous metaplasia, and airway hyperresponsiveness. This asthma phenotype depends on interleukin (IL)-13-producing type 2 innate lymphoid cells, the expansion of which in turn depends on release of the innate cytokines IL-25, IL-33, and thymic stromal lymphopoietin from the airway epithelium. In humans, certain genetic variants may predispose to HRV-induced childhood asthma.
Collapse
Affiliation(s)
- Mingyuan Han
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Medical Sciences Research Building II, 1150 West Medical Center Drive, Ann Arbor, MI, USA
| | - Charu Rajput
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Medical Sciences Research Building II, 1150 West Medical Center Drive, Ann Arbor, MI, USA
| | - Marc B Hershenson
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Medical Sciences Research Building II, 1150 West Medical Center Drive, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Medical Sciences Research Building II, 1150 West Medical Center Drive, Ann Arbor, MI, USA.
| |
Collapse
|
25
|
Han M, Bentley JK, Rajput C, Lei J, Ishikawa T, Jarman CR, Lee J, Goldsmith AM, Jackson WT, Hoenerhoff MJ, Lewis TC, Hershenson MB. Inflammasome activation is required for human rhinovirus-induced airway inflammation in naive and allergen-sensitized mice. Mucosal Immunol 2019; 12:958-968. [PMID: 31089187 PMCID: PMC6668626 DOI: 10.1038/s41385-019-0172-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/09/2019] [Accepted: 04/29/2019] [Indexed: 02/04/2023]
Abstract
Activation of the inflammasome is a key function of the innate immune response that regulates inflammation in response to microbial substances. Inflammasome activation by human rhinovirus (RV), a major cause of asthma exacerbations, has not been well studied. We examined whether RV induces inflammasome activation in vivo, molecular mechanisms underlying RV-stimulated inflammasome priming and activation, and the contribution of inflammasome activation to RV-induced airway inflammation and exacerbation. RV infection triggered lung mRNA and protein expression of pro-IL-1β and NLRP3, indicative of inflammasome priming, as well as cleavage of caspase-1 and pro-IL-1β, completing inflammasome activation. Immunofluorescence staining showed IL-1β in lung macrophages. Depletion with clodronate liposomes and adoptive transfer experiments showed macrophages to be required and sufficient for RV-induced inflammasome activation. TLR2 was required for RV-induced inflammasome priming in vivo. UV irradiation blocked inflammasome activation and RV genome was sufficient for inflammasome activation in primed cells. Naive and house dust mite-treated NLRP3-/- and IL-1β-/- mice, as well as IL-1 receptor antagonist-treated mice, showed attenuated airway inflammation and responsiveness following RV infection. We conclude that RV-induced inflammasome activation is required for maximal airway inflammation and hyperresponsiveness in naive and allergic mice. The inflammasome represents a molecular target for RV-induced asthma exacerbations.
Collapse
Affiliation(s)
- Mingyuan Han
- Department of Pediatrics,University of Michigan Medical School, Ann Arbor, MI 48109
| | - J. Kelley Bentley
- Department of Pediatrics,University of Michigan Medical School, Ann Arbor, MI 48109
| | - Charu Rajput
- Department of Pediatrics,University of Michigan Medical School, Ann Arbor, MI 48109
| | - Jing Lei
- Department of Pediatrics,University of Michigan Medical School, Ann Arbor, MI 48109
| | - Tomoko Ishikawa
- Department of Pediatrics,University of Michigan Medical School, Ann Arbor, MI 48109
| | - Caitlin R. Jarman
- Department of Pediatrics,University of Michigan Medical School, Ann Arbor, MI 48109
| | - Julie Lee
- Department of Pediatrics,University of Michigan Medical School, Ann Arbor, MI 48109
| | - Adam M. Goldsmith
- Department of Pediatrics,University of Michigan Medical School, Ann Arbor, MI 48109
| | - William T. Jackson
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Mark J. Hoenerhoff
- Department of Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Toby C. Lewis
- Department of Pediatrics,University of Michigan Medical School, Ann Arbor, MI 48109
| | - Marc B. Hershenson
- Department of Pediatrics,University of Michigan Medical School, Ann Arbor, MI 48109;,Department of Molecular and Integrative Physiology,University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
26
|
Transcriptomic Analysis Reveals Priming of The Host Antiviral Interferon Signaling Pathway by Bronchobini ® Resulting in Balanced Immune Response to Rhinovirus Infection in Mouse Lung Tissue Slices. Int J Mol Sci 2019; 20:ijms20092242. [PMID: 31067687 PMCID: PMC6540047 DOI: 10.3390/ijms20092242] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 12/22/2022] Open
Abstract
Rhinovirus (RV) is the predominant virus causing respiratory tract infections. Bronchobini® is a low dose multi component, multi target preparation used to treat inflammatory respiratory diseases such as the common cold, described to ease severity of symptoms such as cough and viscous mucus production. The aim of the study was to assess the efficacy of Bronchobini® in RV infection and to elucidate its mode of action. Therefore, Bronchobini®’s ingredients (BRO) were assessed in an ex vivo model of RV infection using mouse precision-cut lung slices, an organotypic tissue capable to reflect the host immune response to RV infection. Cytokine profiles were assessed using enzyme-linked immunosorbent assay (ELISA) and mesoscale discovery (MSD). Gene expression analysis was performed using Affymetrix microarrays and ingenuity pathway analysis. BRO treatment resulted in the significant suppression of RV-induced antiviral and pro-inflammatory cytokine release. Transcriptome analysis revealed a multifactorial mode of action of BRO, with a strong inhibition of the RV-induced pro-inflammatory and antiviral host response mediated by nuclear factor kappa B (NFkB) and interferon signaling pathways. Interestingly, this was due to priming of these pathways in the absence of virus. Overall, BRO exerted its beneficial anti-inflammatory effect by priming the antiviral host response resulting in a reduced inflammatory response to RV infection, thereby balancing an otherwise excessive inflammatory response.
Collapse
|
27
|
Bentley JK, Han M, Jaipalli S, Hinde JL, Lei J, Ishikawa T, Goldsmith AM, Rajput C, Hershenson MB. Myristoylated rhinovirus VP4 protein activates TLR2-dependent proinflammatory gene expression. Am J Physiol Lung Cell Mol Physiol 2019; 317:L57-L70. [PMID: 30908938 DOI: 10.1152/ajplung.00365.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Asthma exacerbations are often caused by rhinovirus (RV). We and others have shown that Toll-like receptor 2 (TLR2), a membrane surface receptor that recognizes bacterial lipopeptides and lipoteichoic acid, is required and sufficient for RV-induced proinflammatory responses in vitro and in vivo. We hypothesized that viral protein-4 (VP4), an internal capsid protein that is myristoylated upon viral replication and externalized upon viral binding, is a ligand for TLR2. Recombinant VP4 and myristoylated VP4 (MyrVP4) were purified by Ni-affinity chromatography. MyrVP4 was also purified from RV-A1B-infected HeLa cells by urea solubilization and anti-VP4 affinity chromatography. Finally, synthetic MyrVP4 was produced by chemical peptide synthesis. MyrVP4-TLR2 interactions were assessed by confocal fluorescence microscopy, fluorescence resonance energy transfer (FRET), and monitoring VP4-induced cytokine mRNA expression in the presence of anti-TLR2 and anti-VP4. MyrVP4 and TLR2 colocalized in TLR2-expressing HEK-293 cells, mouse bone marrow-derived macrophages, human bronchoalveolar macrophages, and human airway epithelial cells. Colocalization was absent in TLR2-null HEK-293 cells and blocked by anti-TLR2 and anti-VP4. Cy3-labeled MyrVP4 and Cy5-labeled anti-TLR2 showed an average fractional FRET efficiency of 0.24 ± 0.05, and Cy5-labeled anti-TLR2 increased and unlabeled MyrVP4 decreased FRET efficiency. MyrVP4-induced chemokine mRNA expression was higher than that elicited by VP4 alone and was attenuated by anti-TLR2 and anti-VP4. Cytokine expression was similarly increased by MyrVP4 purified from RV-infected HeLa cells and synthetic MyrVP4. We conclude that, during RV infection, MyrVP4 and TLR2 interact to generate a proinflammatory response.
Collapse
Affiliation(s)
- J Kelley Bentley
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Mingyuan Han
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Suraj Jaipalli
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Joanna L Hinde
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Jing Lei
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Tomoko Ishikawa
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Adam M Goldsmith
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Charu Rajput
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Marc B Hershenson
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan.,Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
28
|
Zhang Y, Mao D, Keeler SP, Wang X, Wu K, Gerovac BJ, Shornick LL, Agapov EV, Holtzman MJ. Respiratory Enterovirus (like Parainfluenza Virus) Can Cause Chronic Lung Disease if Protection by Airway Epithelial STAT1 Is Lost. THE JOURNAL OF IMMUNOLOGY 2019; 202:2332-2347. [PMID: 30804041 DOI: 10.4049/jimmunol.1801491] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 02/11/2019] [Indexed: 12/11/2022]
Abstract
Epithelial barrier cells are proposed to be critical for host defense, and airway epithelial cell capacity for IFN signal transduction is presumed to protect against respiratory viral infection. However, it has been difficult to fully test these concepts given the absence of tools to analyze IFN signaling specific to airway epithelial cells in vivo. To address these issues, we generated a new line of transgenic mice with Cre-driver genes (Foxj1 and Scgb1a1) for a floxed-Stat1 allele (designated Foxj1-Scgb1a1-Cre-Stat1f/f mice) to target the master IFN signal regulator STAT1 in airway epithelial cells and tested these mice for control of infection because of mouse parainfluenza (Sendai) virus and human enterovirus D68 (EV-D68). Indeed, both types of infections showed increases in viral titers and severity of acute illness in Foxj1-Scgb1a1-Cre-Stat1f/f mice and conventional Stat1-/- mice compared with wild-type mice. In concert, the chronic lung disease that develops after Sendai virus infection was also increased in Foxj1-Scgb1a1-Cre-Stat1f/f and Stat1-/ - mice, marked by airway and adjacent parenchymal immune cell infiltration and mucus production for at least 7 wk postinfection. Unexpectedly, relatively mild EV-D68 infection also progressed to chronic lung disease in Foxj1-Scgb1a1-Cre-Stat1f/f and Stat1 -/- mice but was limited (like viral replication) to airways. The results thereby provide proof-of-concept for a critical role of barrier epithelial cells in protection from acute illness and chronic disease after viral infection and suggest a specific role for airway epithelial cells given the limitation of EV-D68 replication and acute and chronic manifestations of disease primarily to airway tissue.
Collapse
Affiliation(s)
- Yong Zhang
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Dailing Mao
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Shamus P Keeler
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Xinyu Wang
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kangyun Wu
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Benjamin J Gerovac
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Laurie L Shornick
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Eugene V Agapov
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
29
|
DUSP10 Negatively Regulates the Inflammatory Response to Rhinovirus through Interleukin-1β Signaling. J Virol 2019; 93:JVI.01659-18. [PMID: 30333178 PMCID: PMC6321923 DOI: 10.1128/jvi.01659-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/12/2018] [Indexed: 12/21/2022] Open
Abstract
Rhinoviruses are one of the causes of the common cold. In patients with asthma or chronic obstructive pulmonary disease, viral infections, including those with rhinovirus, are the commonest cause of exacerbations. Novel therapeutics to limit viral inflammation are clearly required. The work presented here identifies DUSP10 as an important protein involved in limiting the inflammatory response in the airway without affecting immune control of the virus. Rhinoviral infection is a common trigger of the excessive inflammation observed during exacerbations of asthma and chronic obstructive pulmonary disease. Rhinovirus (RV) recognition by pattern recognition receptors activates the mitogen-activated protein kinase (MAPK) pathways, which are common inducers of inflammatory gene production. A family of dual-specificity phosphatases (DUSPs) can regulate MAPK function, but their roles in rhinoviral infection are not known. We hypothesized that DUSPs would negatively regulate the inflammatory response to RV infection. Our results revealed that the p38 and c-Jun N-terminal kinase (JNK) MAPKs play key roles in the inflammatory response of epithelial cells to RV infection. Three DUSPs previously shown to have roles in innate immunity (DUSPs 1, 4, and 10) were expressed in primary bronchial epithelial cells, and one of them, DUSP10, was downregulated by RV infection. Small interfering RNA-mediated knockdown of DUSP10 identified a role for the protein in negatively regulating inflammatory cytokine production in response to interleukin-1β (IL-1β), alone and in combination with RV, without any effect on RV replication. This study identifies DUSP10 as an important regulator of airway inflammation in respiratory viral infection. IMPORTANCE Rhinoviruses are one of the causes of the common cold. In patients with asthma or chronic obstructive pulmonary disease, viral infections, including those with rhinovirus, are the commonest cause of exacerbations. Novel therapeutics to limit viral inflammation are clearly required. The work presented here identifies DUSP10 as an important protein involved in limiting the inflammatory response in the airway without affecting immune control of the virus.
Collapse
|
30
|
Girkin J, Maltby S, Singanayagam A, Bartlett N, Mallia P. In vivo experimental models of infection and disease. RHINOVIRUS INFECTIONS 2019. [PMCID: PMC7149593 DOI: 10.1016/b978-0-12-816417-4.00008-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Human and animal models continue to play a crucial role in research to understand host immunity to rhinovirus (RV) and identify disease mechanisms. Human models have provided direct evidence that RV infection is capable of exacerbating chronic respiratory diseases and identified immunological processes that correlate with clinical disease outcomes. Mice are the most commonly used nonhuman experimental RV infection model. Although semipermissive, under defined experimental conditions sufficient replication occurs to induce host immune responses that recapitulate immunity and disease during human infection. The capacity to use genetically modified mouse strains and drug interventions has shown the mouse model to be an invaluable research tool defining causal relationships between host immunity and disease and supporting development of new treatments. Used in combination the insights achieved from human and animal experimental infection models provide complementary insights into RV biology and yield novel therapeutic options to reduce the burden of RV-induced disease.
Collapse
|
31
|
Han M, Rajput C, Ishikawa T, Jarman CR, Lee J, Hershenson MB. Small Animal Models of Respiratory Viral Infection Related to Asthma. Viruses 2018; 10:E682. [PMID: 30513770 PMCID: PMC6316391 DOI: 10.3390/v10120682] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/21/2018] [Accepted: 11/29/2018] [Indexed: 12/20/2022] Open
Abstract
Respiratory viral infections are strongly associated with asthma exacerbations. Rhinovirus is most frequently-detected pathogen; followed by respiratory syncytial virus; metapneumovirus; parainfluenza virus; enterovirus and coronavirus. In addition; viral infection; in combination with genetics; allergen exposure; microbiome and other pathogens; may play a role in asthma development. In particular; asthma development has been linked to wheezing-associated respiratory viral infections in early life. To understand underlying mechanisms of viral-induced airways disease; investigators have studied respiratory viral infections in small animals. This report reviews animal models of human respiratory viral infection employing mice; rats; guinea pigs; hamsters and ferrets. Investigators have modeled asthma exacerbations by infecting mice with allergic airways disease. Asthma development has been modeled by administration of virus to immature animals. Small animal models of respiratory viral infection will identify cell and molecular targets for the treatment of asthma.
Collapse
Affiliation(s)
- Mingyuan Han
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Charu Rajput
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Tomoko Ishikawa
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Caitlin R Jarman
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Julie Lee
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Marc B Hershenson
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
32
|
Keeler SP, Agapov EV, Hinojosa ME, Letvin AN, Wu K, Holtzman MJ. Influenza A Virus Infection Causes Chronic Lung Disease Linked to Sites of Active Viral RNA Remnants. THE JOURNAL OF IMMUNOLOGY 2018; 201:2354-2368. [PMID: 30209189 DOI: 10.4049/jimmunol.1800671] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/14/2018] [Indexed: 12/18/2022]
Abstract
Clinical and experimental observations suggest that chronic lung disease is linked to respiratory viral infection. However, the long-term aspect of this relationship is not yet defined using a virus that replicates at properly high levels in humans and a corresponding animal model. In this study, we show that influenza A virus infection achieves 1 × 106-fold increases in viral load in the lung and dose-dependent severity of acute illness in mice. Moreover, these events are followed by persistence of negative- and positive-strand viral RNA remnants for 15 wk and chronic lung disease for at least 26 wk postinfection. The disease is manifested by focal areas of bronchiolization and mucus production that contain increased levels of viral RNA remnants along with mucin Muc5ac and Il13 mRNA compared with uninvolved areas of the lung. Excess mucus production and associated airway hyperreactivity (but not fibrosis or emphysema) are partially attenuated with loss of IL-13 production or signaling (using mice with IL-13 or STAT6 deficiency). These deficiencies cause reciprocal increases in l17a mRNA and neutrophils in the lung; however, none of these disease endpoints are changed with IL-13/IL-17a compared with IL-13 deficiency or STAT6/IL-17a compared with STAT6 deficiency. The results establish the capacity of a potent human respiratory virus to produce chronic lung disease focally at sites of active viral RNA remnants, likely reflecting locations of viral replication that reprogram the region. Viral dose dependency of disease also implicates high-level viral replication and severity of acute infection as determinants of chronic lung diseases such as asthma and COPD with IL-13-dependent and IL-13/IL-17-independent mechanisms.
Collapse
Affiliation(s)
- Shamus P Keeler
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Eugene V Agapov
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael E Hinojosa
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Adam N Letvin
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kangyun Wu
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
33
|
Han M, Rajput C, Hinde JL, Wu Q, Lei J, Ishikawa T, Bentley JK, Hershenson MB. Construction of a recombinant rhinovirus accommodating fluorescent marker expression. Influenza Other Respir Viruses 2018; 12:717-727. [PMID: 30120824 PMCID: PMC6185886 DOI: 10.1111/irv.12602] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/02/2018] [Accepted: 08/15/2018] [Indexed: 12/28/2022] Open
Abstract
Background Rhinovirus (RV) causes the common cold and asthma exacerbations. The RV genome is a 7.3 kb single‐strand positive‐sense RNA. Objective Using minor group RV1A as a backbone, we sought to design and generate a recombinant RV1A accommodating fluorescent marker expression, thereby allowing tracking of viral infection. Method Recombinant RV1A infectious cDNA clones harboring the coding sequence of green fluorescent protein (GFP), Renilla luciferase, or iLOV (for light, oxygen, or voltage sensing) were engineered and constructed. RV‐infected cells were determined by flow cytometry, immunohistochemistry, and immunofluorescence microscopy. Results RV1A‐GFP showed a cytopathic effect in HeLa cells but failed to express GFP or Renilla luciferase due to deletion. The smaller fluorescent protein construct, RV1A‐iLOV, was stably expressed in infected cells. RV1A‐iLOV expression was used to examine the antiviral effect of bafilomycin in HeLa cells. Compared to parental virus, RV1A‐iLOV infection of BALB/c mice yielded a similar viral load and level of cytokine mRNA expression. However, imaging of fixed lung tissue failed to reveal a fluorescent signal, likely due to the oxidation and bleaching of iLOV‐bound flavin mononucleotide. We therefore employed an anti‐iLOV antibody for immunohistochemical and immunofluorescence imaging. The iLOV signal was identified in airway epithelial cells and CD45+ CD11b+ lung macrophages. Conclusions These results suggest that RV1A‐iLOV is a useful molecular tool for studying RV pathogenesis. The construction strategy for RV1A‐iLOV could be applied to other RV serotypes. However, the detection of iLOV‐expressing RV in fixed tissue required the use of an anti‐iLOV antibody, limiting the value of this construct.
Collapse
Affiliation(s)
- Mingyuan Han
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Charu Rajput
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Joanna L Hinde
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Qian Wu
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jing Lei
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Tomoko Ishikawa
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - J Kelley Bentley
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Marc B Hershenson
- Department of Pediatrics & Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
34
|
Rajput C, Han M, Bentley JK, Lei J, Ishikawa T, Wu Q, Hinde JL, Callear AP, Stillwell TL, Jackson WT, Martin ET, Hershenson MB. Enterovirus D68 infection induces IL-17-dependent neutrophilic airway inflammation and hyperresponsiveness. JCI Insight 2018; 3:121882. [PMID: 30135310 PMCID: PMC6141171 DOI: 10.1172/jci.insight.121882] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/03/2018] [Indexed: 12/15/2022] Open
Abstract
Enterovirus D68 (EV-D68) shares biologic features with rhinovirus (RV). In 2014, a nationwide outbreak of EV-D68 was associated with severe asthma-like symptoms. We sought to develop a mouse model of EV-D68 infection and determine the mechanisms underlying airway disease. BALB/c mice were inoculated intranasally with EV-D68 (2014 isolate), RV-A1B, or sham, alone or in combination with anti-IL-17A or house dust mite (HDM) treatment. Like RV-A1B, lung EV-D68 viral RNA peaked 12 hours after infection. EV-D68 induced airway inflammation, expression of cytokines (TNF-α, IL-6, IL-12b, IL-17A, CXCL1, CXCL2, CXCL10, and CCL2), and airway hyperresponsiveness, which were suppressed by anti-IL-17A antibody. Neutrophilic inflammation and airway responsiveness were significantly higher after EV-D68 compared with RV-A1B infection. Flow cytometry showed increased lineage-, NKp46-, RORγt+ IL-17+ILC3s and γδ T cells in the lungs of EV-D68-treated mice compared with those in RV-treated mice. EV-D68 infection of HDM-exposed mice induced additive or synergistic increases in BAL neutrophils and eosinophils and expression of IL-17, CCL11, IL-5, and Muc5AC. Finally, patients from the 2014 epidemic period with EV-D68 showed significantly higher nasopharyngeal IL-17 mRNA levels compared with patients with RV-A infection. EV-D68 infection induces IL-17-dependent airway inflammation and hyperresponsiveness, which is greater than that generated by RV-A1B, consistent with the clinical picture of severe asthma-like symptoms.
Collapse
Affiliation(s)
- Charu Rajput
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Mingyuan Han
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - J. Kelley Bentley
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jing Lei
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Tomoko Ishikawa
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Qian Wu
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Joanna L. Hinde
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Amy P. Callear
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Terri L. Stillwell
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - William T. Jackson
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Emily T. Martin
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Marc B. Hershenson
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
35
|
Lauzon-Joset JF, Jones AC, Mincham KT, Thomas JA, Rosenthal LA, Bosco A, Holt PG, Strickland DH. Atopy-Dependent and Independent Immune Responses in the Heightened Severity of Atopics to Respiratory Viral Infections: Rat Model Studies. Front Immunol 2018; 9:1805. [PMID: 30150981 PMCID: PMC6099265 DOI: 10.3389/fimmu.2018.01805] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/23/2018] [Indexed: 12/04/2022] Open
Abstract
Allergic (Th2high immunophenotype) asthmatics have a heightened susceptibility to common respiratory viral infections such as human rhinovirus. Evidence suggests that the innate interferon response is deficient in asthmatic/atopic individuals, while other studies show no differences in antiviral response pathways. Unsensitized and OVA-sensitized/challenged Th2high (BN rats) and Th2low immunophenotype (PVG rats) animals were inoculated intranasally with attenuated mengovirus (vMC0). Sensitized animals were exposed/unexposed during the acute viral response phase. Cellular and transcriptomic profiling was performed on bronchoalveolar lavage cells. In unsensitized PVG rats, vMC0 elicits a prototypical antiviral response (neutrophilic airways inflammation, upregulation of Th1/type I interferon-related pathways). In contrast, response to infection in the Th2high BN rats was associated with a radically altered intrinsic host response to respiratory viral infection, characterized by macrophage influx/Th2-associated pathways. In sensitized animals, response to virus infection alone was not altered compared to unsensitized animals. However, allergen exposure of sensitized animals during viral infection unleashes a notably exaggerated airways inflammatory response profile orders of magnitude higher in BN versus PVG rats despite similar viral loads. The co-exposure responses in the Th2high BN incorporated type I interferon/Th1, alternative macrophage activation/Th2 and Th17 signatures. Similar factors may underlie the hyper-susceptibility to infection-associated airways inflammation characteristic of the human Th2high immunophenotype.
Collapse
Affiliation(s)
| | - Anya C Jones
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia.,School of Medicine, University of Western Australia, Perth, WA, Australia
| | - Kyle T Mincham
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia.,School of Medicine, University of Western Australia, Perth, WA, Australia
| | - Jenny A Thomas
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Louis A Rosenthal
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Anthony Bosco
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Patrick G Holt
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | | |
Collapse
|
36
|
Gualdoni GA, Mayer KA, Kapsch AM, Kreuzberg K, Puck A, Kienzl P, Oberndorfer F, Frühwirth K, Winkler S, Blaas D, Zlabinger GJ, Stöckl J. Rhinovirus induces an anabolic reprogramming in host cell metabolism essential for viral replication. Proc Natl Acad Sci U S A 2018; 115:E7158-E7165. [PMID: 29987044 PMCID: PMC6065033 DOI: 10.1073/pnas.1800525115] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Rhinoviruses (RVs) are responsible for the majority of upper airway infections; despite their high prevalence and the resulting economic burden, effective treatment is lacking. We report here that RV induces metabolic alterations in host cells, which offer an efficient target for antiviral intervention. We show that RV-infected cells rapidly up-regulate glucose uptake in a PI3K-dependent manner. In parallel, infected cells enhance the expression of the PI3K-regulated glucose transporter GLUT1. In-depth metabolomic analysis of RV-infected cells revealed a critical role of glucose mobilization from extracellular and intracellular pools via glycogenolysis for viral replication. Infection resulted in a highly anabolic state, including enhanced nucleotide synthesis and lipogenesis. Consistently, we observed that glucose deprivation from medium and via glycolysis inhibition by 2-deoxyglucose (2-DG) potently impairs viral replication. Metabolomic analysis showed that 2-DG specifically reverts the RV-induced anabolic reprogramming. In addition, treatment with 2-DG inhibited RV infection and inflammation in a murine model. Thus, we demonstrate that the specific metabolic fingerprint of RV infection can be used to identify new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Guido A Gualdoni
- Institute of Immunology, Center of Pathophysiology, Immunology & Infectiology, Medical University of Vienna, 1090 Vienna, Austria;
- Division of Nephrology and Dialysis, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria
| | - Katharina A Mayer
- Institute of Immunology, Center of Pathophysiology, Immunology & Infectiology, Medical University of Vienna, 1090 Vienna, Austria
| | - Anna-Maria Kapsch
- Institute of Immunology, Center of Pathophysiology, Immunology & Infectiology, Medical University of Vienna, 1090 Vienna, Austria
- Global Pathogen Safety, Shire, 1090 Vienna, Austria
| | - Katharina Kreuzberg
- Institute of Immunology, Center of Pathophysiology, Immunology & Infectiology, Medical University of Vienna, 1090 Vienna, Austria
| | - Alexander Puck
- Institute of Immunology, Center of Pathophysiology, Immunology & Infectiology, Medical University of Vienna, 1090 Vienna, Austria
| | - Philip Kienzl
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Karin Frühwirth
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine 1, Medical University of Vienna, 1090 Vienna, Austria
| | - Stefan Winkler
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine 1, Medical University of Vienna, 1090 Vienna, Austria
| | - Dieter Blaas
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Vienna Biocenter, Medical University of Vienna, 1090 Vienna, Austria
| | - Gerhard J Zlabinger
- Institute of Immunology, Center of Pathophysiology, Immunology & Infectiology, Medical University of Vienna, 1090 Vienna, Austria
| | - Johannes Stöckl
- Institute of Immunology, Center of Pathophysiology, Immunology & Infectiology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
37
|
Jing Y, Gimenes JA, Mishra R, Pham D, Comstock AT, Yu D, Sajjan U. NOTCH3 contributes to rhinovirus-induced goblet cell hyperplasia in COPD airway epithelial cells. Thorax 2018; 74:18-32. [PMID: 29991510 DOI: 10.1136/thoraxjnl-2017-210593] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 05/15/2018] [Accepted: 06/11/2018] [Indexed: 11/04/2022]
Abstract
RATIONALE Goblet cell hyperplasia (GCH) is one of the cardinal features of chronic obstructive pulmonary disease (COPD) and contributes to airways obstruction. Rhinovirus (RV), which causes acute exacerbations in patients with COPD, also causes prolonged airways obstruction. Previously, we showed that RV enhances mucin gene expression and increases goblet cell number in a COPD mouse model. This study examines whether RV causes sustained GCH in relevant models of COPD. METHODS Mucociliary-differentiated COPD and normal airway epithelial cell cultures and mice with normal or COPD phenotype were infected with RV or sham and examined for GCH by immunofluorescence and/or mucin gene expression. In some experiments, RV-infected COPD cells and mice with COPD phenotype were treated with γ-secretase inhibitor or interleukin-13 neutralising antibody and assessed for GCH. To determine the contribution of NOTCH1/3 in RV-induced GCH, COPD cells transduced with NOTCH1/3 shRNA were used. RESULTS RV-infected COPD, but not normal cell cultures, showed sustained GCH and increased mucin genes expression. Microarray analysis indicated increased expression of NOTCH1, NOTCH3 and HEY1 only in RV-infected COPD cells. Blocking NOTCH3, but not NOTCH1, attenuated RV-induced GCH in vitro. Inhibition of NOTCH signalling by γ-secretase inhibitor, but not neutralising antibody to IL-13, abrogated RV-induced GCH and mucin gene expression. CONCLUSIONS RV induces sustained GCH via NOTCH3 particularly in COPD cells or mice with COPD phenotype. This may be one of the mechanisms that may contribute to RV-induced prolonged airways obstruction in COPD.
Collapse
Affiliation(s)
- Yaxun Jing
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Joao Antonio Gimenes
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Rahul Mishra
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Duc Pham
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Adam T Comstock
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Daohai Yu
- Department of Clinical Sciences, Temple University, Philadelphia, Pennsylvania, USA
| | - Umadevi Sajjan
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA.,Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, Pennsylvania, USA.,Department of Physiology, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
38
|
Farazuddin M, Mishra R, Jing Y, Srivastava V, Comstock AT, Sajjan US. Quercetin prevents rhinovirus-induced progression of lung disease in mice with COPD phenotype. PLoS One 2018; 13:e0199612. [PMID: 29975735 PMCID: PMC6033397 DOI: 10.1371/journal.pone.0199612] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/27/2018] [Indexed: 12/14/2022] Open
Abstract
Acute exacerbations are the major cause of morbidity and mortality in patients with chronic obstructive pulmonary disease (COPD). Rhinovirus, which causes acute exacerbations may also accelerate progression of lung disease in these patients. Current therapies reduces the respiratory symptoms and does not treat the root cause of exacerbations effectively. We hypothesized that quercetin, a potent antioxidant and anti-inflammatory agent with antiviral properties may be useful in treating rhinovirus-induced changes in COPD. Mice with COPD phenotype maintained on control or quercetin diet and normal mice were infected with sham or rhinovirus, and after 14 days mice were examined for changes in lung mechanics and lung inflammation. Rhinovirus-infected normal mice showed no changes in lung mechanics or histology. In contrast, rhinovirus-infected mice with COPD phenotype showed reduction in elastic recoiling and increase in lung inflammation, goblet cell metaplasia, and airways cholinergic responsiveness compared to sham-infected mice. Interestingly, rhinovirus-infected mice with COPD phenotype also showed accumulation of neutrophils, CD11b+/CD11c+ macrophages and CD8+ T cells in the lungs. Quercetin supplementation attenuated rhinovirus-induced all the pathologic changes in mice with COPD phenotype. Together these results indicate that quercetin effectively mitigates rhinovirus-induced progression of lung disease in a mouse model of COPD. Therefore, quercetin may be beneficial in the treatment of rhinovirus-associated exacerbations and preventing progression of lung disease in COPD.
Collapse
Affiliation(s)
- Mohammad Farazuddin
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Rahul Mishra
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yaxun Jing
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Vikram Srivastava
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Adam T. Comstock
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Umadevi S. Sajjan
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, Pennsylvania, United States of America
- Department of Physiology, Temple University, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
39
|
Restori KH, Srinivasa BT, Ward BJ, Fixman ED. Neonatal Immunity, Respiratory Virus Infections, and the Development of Asthma. Front Immunol 2018; 9:1249. [PMID: 29915592 PMCID: PMC5994399 DOI: 10.3389/fimmu.2018.01249] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/18/2018] [Indexed: 12/27/2022] Open
Abstract
Infants are exposed to a wide range of potential pathogens in the first months of life. Although maternal antibodies acquired transplacentally protect full-term neonates from many systemic pathogens, infections at mucosal surfaces still occur with great frequency, causing significant morbidity and mortality. At least part of this elevated risk is attributable to the neonatal immune system that tends to favor T regulatory and Th2 type responses when microbes are first encountered. Early-life infection with respiratory viruses is of particular interest because such exposures can disrupt normal lung development and increase the risk of chronic respiratory conditions, such as asthma. The immunologic mechanisms that underlie neonatal host-virus interactions that contribute to the subsequent development of asthma have not yet been fully defined. The goals of this review are (1) to outline the differences between the neonatal and adult immune systems and (2) to present murine and human data that support the hypothesis that early-life interactions between the immune system and respiratory viruses can create a lung environment conducive to the development of asthma.
Collapse
Affiliation(s)
- Katherine H Restori
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Bharat T Srinivasa
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Brian J Ward
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Elizabeth D Fixman
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada.,Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
40
|
Rajput C, Walsh MP, Eder BN, Metitiri EE, Popova AP, Hershenson MB. Rhinovirus infection induces distinct transcriptome profiles in polarized human macrophages. Physiol Genomics 2018. [PMID: 29521598 DOI: 10.1152/physiolgenomics.00122.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Infections with rhinovirus (RV) cause asthma exacerbations. Recent studies suggest that macrophages play a role in asthmatic airway inflammation and the innate immune response to RV infection. Macrophages exhibit phenotypes based on surface markers and gene expression. We hypothesized that macrophage polarization state alters gene expression in response to RV infection. Cells were derived from human peripheral blood derived monocytes. M1 and M2 polarization was carried out by using IFN-γ and IL-4, respectively, and RNA was extracted for Affymetrix Human Gene ST2.1 exon arrays. Selected genes were validated by quantitative (q)PCR. Treatment of nonactivated (M0) macrophages with IFN-γ and IL-4 induced the expression of 252 and 153 distinct genes, respectively, including previously-identified M1 and M2 markers. RV infection of M0 macrophages induced upregulation of 232 genes; pathway analysis showed significant overrepresentation of genes involved in IFN-α/β signaling and cytokine signaling in the immune system. RV infection induced differential expression of 195 distinct genes in M1-like macrophages but only seven distinct genes in M2-like-polarized cells. In a secondary analysis, comparison between M0-, RV-infected, and M1-like-polarized, RV-infected macrophages revealed differential expression of 227 genes including those associated with asthma and its exacerbation. qPCR demonstrated increased expression of CCL8, CXCL10, TNFSF10, TNFSF18, IL6, NOD2, and GSDMD and reduced expression of VNN1, AGO1, and AGO2. Together, these data show that, in contrast to M2-like-polarized macrophages, gene expression of M1-like macrophages is highly regulated by RV.
Collapse
Affiliation(s)
- Charu Rajput
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School , Ann Arbor, Michigan
| | - Megan P Walsh
- Department of Epidemiology, University of Michigan School of Public Health , Ann Arbor, MI
| | - Breanna N Eder
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School , Ann Arbor, Michigan
| | - Ediri E Metitiri
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School , Ann Arbor, Michigan
| | - Antonia P Popova
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School , Ann Arbor, Michigan
| | - Marc B Hershenson
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School , Ann Arbor, Michigan.,Department of Molecular and Integrative Physiology, University of Michigan Medical School , Ann Arbor, Michigan
| |
Collapse
|
41
|
Song DJ, Miller M, Beppu A, Rosenthal P, Das S, Karta M, Vuong C, Mehta AK, Croft M, Broide DH. Rhinovirus Infection of ORMDL3 Transgenic Mice Is Associated with Reduced Rhinovirus Viral Load and Airway Inflammation. THE JOURNAL OF IMMUNOLOGY 2017; 199:2215-2224. [PMID: 28827284 DOI: 10.4049/jimmunol.1601412] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 07/27/2017] [Indexed: 01/29/2023]
Abstract
Orosomucoid like 3 (ORMDL3), a gene localized to chromosome 17q21, has been linked in epidemiologic studies to childhood asthma and rhinovirus (RV) infections. As the single nucleotide polymorphisms linking ORMDL3 to asthma are associated with increased expression of ORMDL3, we have used hORMDL3zp3-Cre mice (which have universal increased expression of human ORMDL3) to determine whether infection of these transgenic mice with RV influences levels of airway inflammation or RV viral load. RV infection of hORMDL3zp3-Cre mice resulted in reduced RV viral load assessed by quantitative real-time PCR (lung and airway epithelium), as well as reduced airway inflammation (total bronchoalveolar lavage cells, neutrophils, macrophages, and lymphocytes) compared with RV-infected wild-type mice. Levels of the antiviral pathways including IFNs (IFN-α, IFN-β, IFN-λ) and RNAse L were significantly increased in the lungs of RV-infected hORMDL3zp3-Cre mice. Levels of the antiviral mouse oligoadenylate synthetase (mOas)1g pathway and RNAse L were upregulated in the lungs of unchallenged hORMDL3zp3-Cre mice. In addition, levels of mOas2, but not mOas1 (mOas1a, mOas1b, mOas1g), or mOas3 pathways were significantly more upregulated by IFNs (IFN-α, IFN-β, IFN-λ) in epithelial cells from hORMDL3zp3-Cre mice compared with RV-infected wild-type mouse epithelial cells. RNAse L-deficient mice infected with RV had increased RV viral load. Overall, these studies suggest that increased levels of ORMDL3 contribute to antiviral defense to RV infection in mice through pathways that may include IFNs (IFN-α, IFN-β, IFN-λ), OAS, and RNAse L.
Collapse
Affiliation(s)
- Dae Jin Song
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093.,Department of Pediatrics, Korea University College of Medicine, Seoul 03080, Korea; and
| | - Marina Miller
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Andrew Beppu
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Peter Rosenthal
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Sudipta Das
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Maya Karta
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Christine Vuong
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Amit Kumar Mehta
- Division of Immune Regulation, La Jolla Institute, La Jolla, CA 92037
| | - Michael Croft
- Division of Immune Regulation, La Jolla Institute, La Jolla, CA 92037
| | - David H Broide
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093;
| |
Collapse
|
42
|
Zhou X, Zhu L, Lizarraga R, Chen Y. Human Airway Epithelial Cells Direct Significant Rhinovirus Replication in Monocytic Cells by Enhancing ICAM1 Expression. Am J Respir Cell Mol Biol 2017; 57:216-225. [PMID: 28328242 DOI: 10.1165/rcmb.2016-0271oc] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Human rhinovirus (RV) is the major cause of common cold, and it also plays a significant role in asthma and asthma exacerbation. The airway epithelium is the primary site of RV infection and production. In contrast, monocytic cells (e.g., monocytes and macrophages) are believed to be nonpermissive for RV replication. Instead, RV has been shown to modulate inflammatory gene expressions in these cells via a replication-independent mechanism. In the study presented here, replication of RV16 (a major-group RV) was found to be significantly enhanced in monocytes when it was cocultivated with airway epithelial cells. This effect appeared to be mediated by secretory components from epithelial cells, which stimulated RV16 replication and significantly elevated the expression of a number of proinflammatory cytokines. The lack of such an effect on RV1A, a minor-group RV that enters the cell by a different receptor, suggests that intercellular adhesion molecule 1 (ICAM1), the receptor for major-group RVs, may be involved. Indeed, conditioned media from epithelial cells significantly increased ICAM1 expression in monocytes. Consistently, ICAM1 overexpression and ICAM1 knockdown enhanced and blocked RV production, respectively, confirming the role of ICAM1 in this process. Thus, this is the first report demonstrating that airway epithelial cells direct significant RV16 replication in monocytic cells via an ICAM1-dependent mechanism. This finding will open a new avenue for the study of RV infection in airway disease and its exacerbation.
Collapse
Affiliation(s)
- Xu Zhou
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona
| | - Lingxiang Zhu
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona
| | - Rosa Lizarraga
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona
| | - Yin Chen
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona
| |
Collapse
|
43
|
Han M, Rajput C, Hong JY, Lei J, Hinde JL, Wu Q, Bentley JK, Hershenson MB. The Innate Cytokines IL-25, IL-33, and TSLP Cooperate in the Induction of Type 2 Innate Lymphoid Cell Expansion and Mucous Metaplasia in Rhinovirus-Infected Immature Mice. THE JOURNAL OF IMMUNOLOGY 2017; 199:1308-1318. [PMID: 28701507 DOI: 10.4049/jimmunol.1700216] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 06/14/2017] [Indexed: 12/31/2022]
Abstract
Early-life respiratory viral infection is a risk factor for asthma development. Rhinovirus (RV) infection of 6-d-old mice, but not mature mice, causes mucous metaplasia and airway hyperresponsiveness that are associated with the expansion of lung type 2 innate lymphoid cells (ILC2s) and are dependent on IL-13 and the innate cytokine IL-25. However, contributions of the other innate cytokines, IL-33 and thymic stromal lymphopoietin (TSLP), to the observed asthma-like phenotype have not been examined. We reasoned that IL-33 and TSLP expression are also induced by RV infection in immature mice and are required for maximum ILC2 expansion and mucous metaplasia. We inoculated 6-d-old BALB/c (wild-type) and TSLP receptor-knockout mice with sham HeLa cell lysate or RV. Selected mice were treated with neutralizing Abs to IL-33 or recombinant IL-33, IL-25, or TSLP. ILC2s were isolated from RV-infected immature mice and treated with innate cytokines ex vivo. RV infection of 6-d-old mice increased IL-33 and TSLP protein abundance. TSLP expression was localized to the airway epithelium, whereas IL-33 was expressed in epithelial and subepithelial cells. RV-induced mucous metaplasia, ILC2 expansion, airway hyperresponsiveness, and epithelial cell IL-25 expression were attenuated by anti-IL-33 treatment and in TSLP receptor-knockout mice. Administration of intranasal IL-33 and TSLP was sufficient for mucous metaplasia. Finally, TSLP was required for maximal ILC2 gene expression in response to IL-25 and IL-33. The generation of mucous metaplasia in immature RV-infected mice involves a complex interplay among the innate cytokines IL-25, IL-33, and TSLP.
Collapse
Affiliation(s)
- Mingyuan Han
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Charu Rajput
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Jun Y Hong
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Jing Lei
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Joanna L Hinde
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Qian Wu
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - J Kelley Bentley
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Marc B Hershenson
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109; and .,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
44
|
Song JH, Kim SR, Heo EY, Lee JY, Kim DE, Cho S, Chang SY, Yoon BI, Seong J, Ko HJ. Antiviral activity of gemcitabine against human rhinovirus in vitro and in vivo. Antiviral Res 2017; 145:6-13. [PMID: 28705625 DOI: 10.1016/j.antiviral.2017.07.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/06/2017] [Accepted: 07/06/2017] [Indexed: 12/30/2022]
Abstract
Rhinovirus, a major causative agent of the common cold, is associated with exacerbation of asthma and chronic obstructive pulmonary disease. Currently, there is no antiviral treatment or vaccine for human rhinovirus (HRV). Gemcitabine (2',2'-difluorodeoxycytidine, dFdC) is a deoxycytidine analog with antiviral activity against rhinovirus, as well as enterovirus 71, in vitro. However, the antiviral effects of gemcitabine in vivo have not been investigated. In the current study, we assessed whether gemcitabine mediated antiviral effects in the murine HRV infection model. Intranasal administration of gemcitabine significantly lowered pulmonary viral load and inflammation by decreasing proinflammatory cytokines, including TNF-α and IL-1β, and reduction in the number of lung-infiltrating lymphocytes. Interestingly, we found that the addition of UTP and CTP significantly attenuated the antiviral activity of gemcitabine. Thus the limitation of UTP and CTP by the addition of gemcitabine may inhibit the viral RNA synthesis. These results suggest that gemcitabine, an antineoplastic drug, can be repositioned as an antiviral drug to inhibit HRV infection.
Collapse
Affiliation(s)
- Jae-Hyoung Song
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, South Korea
| | - Seong-Ryeol Kim
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, South Korea
| | - Eun-Young Heo
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, South Korea
| | - Jae-Young Lee
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, South Korea
| | - Dong-Eun Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience & Biotechnology, Ochang, South Korea
| | - Sungchan Cho
- Anticancer Agent Research Center, Korea Research Institute of Bioscience & Biotechnology, Ochang, South Korea
| | - Sun-Young Chang
- Laboratory of Microbiology and Immunology, College of Pharmacy, Ajou University, South Korea
| | - Byung-Il Yoon
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, South Korea
| | - Jeongmin Seong
- Department of Dental Hygiene, College of Health Science, Kangwon National University, South Korea.
| | - Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, South Korea; Convergence Research Center for Functional Plant Products, Advanced Institutes of Convergence Technology, Yeongtong-gu, Suwon-si, Gyeonggi-do, South Korea.
| |
Collapse
|
45
|
Han M, Hong JY, Jaipalli S, Rajput C, Lei J, Hinde JL, Chen Q, Hershenson NM, Bentley JK, Hershenson MB. IFN-γ Blocks Development of an Asthma Phenotype in Rhinovirus-Infected Baby Mice by Inhibiting Type 2 Innate Lymphoid Cells. Am J Respir Cell Mol Biol 2017; 56:242-251. [PMID: 27679954 DOI: 10.1165/rcmb.2016-0056oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Early-life wheezing-associated infections with rhinovirus (RV) have been associated with asthma development in children. We have shown that RV infection of 6-day-old mice induces mucous metaplasia and airways hyperresponsiveness, which is dependent on IL-13, IL-25, and type 2 innate lymphoid cells (ILC2s). Infection of immature mice fails to induce lung IFN-γ expression, in contrast to mature 8-week-old mice with a robust IFN-γ response, consistent with the notion that deficient IFN-γ production in immature mice permits RV-induced type 2 immune responses. We therefore examined the effects of intranasal IFN-γ administration on RV-induced ILC2 expansion and IL-13 expression in 6-day-old BALB/c and IL-13 reporter mice. Airway responses were assessed by histology, immunofluorescence microscopy, quantitative polymerase chain reaction, ELISA, and flow cytometry. Lung ILC2s were also treated with IFN-γ ex vivo. We found that, compared with untreated RV-infected immature mice, IFN-γ treatment attenuated RV-induced IL-13 and Muc5ac mRNA expression and mucous metaplasia. IFN-γ also reduced ILC2 expansion and the percentage of IL-13-secreting ILC2s. IFN-γ had no effect on the mRNA or protein expression of IL-25, IL-33, or thymic stromal lymphoprotein. Finally, IFN-γ treatment of sorted ILC2s reduced IL-5, IL-13, IL-17RB, ST2, and GATA-3 mRNA expression. We conclude that, in immature mice, IFN-γ inhibits ILC2 expansion and IL-13 expression in vivo and ex vivo, thereby attenuating RV-induced mucous metaplasia. These findings demonstrate the antagonistic function of IFN-γ on ILC2 expansion and gene expression, the absence of which may contribute to the development of an asthma-like phenotype after early-life RV infection.
Collapse
Affiliation(s)
- Mingyuan Han
- Departments of 1 Pediatrics and Communicable Diseases, and
| | - Jun Young Hong
- 2 Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Suraj Jaipalli
- Departments of 1 Pediatrics and Communicable Diseases, and
| | - Charu Rajput
- Departments of 1 Pediatrics and Communicable Diseases, and
| | - Jing Lei
- Departments of 1 Pediatrics and Communicable Diseases, and
| | - Joanna L Hinde
- Departments of 1 Pediatrics and Communicable Diseases, and
| | - Qiang Chen
- Departments of 1 Pediatrics and Communicable Diseases, and
| | | | | | - Marc B Hershenson
- Departments of 1 Pediatrics and Communicable Diseases, and.,2 Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
46
|
VanLeuven JT, Ridenhour BJ, Gonzalez AJ, Miller CR, Miura TA. Lung epithelial cells have virus-specific and shared gene expression responses to infection by diverse respiratory viruses. PLoS One 2017; 12:e0178408. [PMID: 28575086 PMCID: PMC5456070 DOI: 10.1371/journal.pone.0178408] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 05/13/2017] [Indexed: 12/28/2022] Open
Abstract
The severity of respiratory viral infections is partially determined by the cellular response mounted by infected lung epithelial cells. Disease prevention and treatment is dependent on our understanding of the shared and unique responses elicited by diverse viruses, yet few studies compare host responses to viruses from different families while controlling other experimental parameters. Murine models are commonly used to study the pathogenesis of respiratory viral infections, and in vitro studies using murine cells provide mechanistic insight into the pathogenesis observed in vivo. We used microarray analysis to compare changes in gene expression of murine lung epithelial cells infected individually by three respiratory viruses causing mild (rhinovirus, RV1B), moderate (coronavirus, MHV-1), and severe (influenza A virus, PR8) disease in mice. RV1B infection caused numerous gene expression changes, but the differential effect peaked at 12 hours post-infection. PR8 altered an intermediate number of genes whose expression continued to change through 24 hours. MHV-1 had comparatively few effects on host gene expression. The viruses elicited highly overlapping responses in antiviral genes, though MHV-1 induced a lower type I interferon response than the other two viruses. Signature genes were identified for each virus and included host defense genes for PR8, tissue remodeling genes for RV1B, and transcription factors for MHV-1. Our comparative approach identified universal and specific transcriptional signatures of virus infection that can be used to distinguish shared and virus-specific mechanisms of pathogenesis in the respiratory tract.
Collapse
Affiliation(s)
- James T. VanLeuven
- Center for Modeling Complex Interactions, University of Idaho, Moscow, Idaho, United States of America
| | - Benjamin J. Ridenhour
- Center for Modeling Complex Interactions, University of Idaho, Moscow, Idaho, United States of America
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Andres J. Gonzalez
- Center for Modeling Complex Interactions, University of Idaho, Moscow, Idaho, United States of America
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Craig R. Miller
- Center for Modeling Complex Interactions, University of Idaho, Moscow, Idaho, United States of America
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
- Department of Mathematics, University of Idaho, Moscow, Idaho, United States of America
| | - Tanya A. Miura
- Center for Modeling Complex Interactions, University of Idaho, Moscow, Idaho, United States of America
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
- * E-mail:
| |
Collapse
|
47
|
Advanced Role of Neutrophils in Common Respiratory Diseases. J Immunol Res 2017; 2017:6710278. [PMID: 28589151 PMCID: PMC5447318 DOI: 10.1155/2017/6710278] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/22/2017] [Accepted: 04/16/2017] [Indexed: 12/18/2022] Open
Abstract
Respiratory diseases, always being a threat towards the health of people all over the world, are most tightly associated with immune system. Neutrophils serve as an important component of immune defense barrier linking innate and adaptive immunity. They participate in the clearance of exogenous pathogens and endogenous cell debris and play an essential role in the pathogenesis of many respiratory diseases. However, the pathological mechanism of neutrophils remains complex and obscure. The traditional roles of neutrophils in severe asthma, chronic obstructive pulmonary diseases (COPD), pneumonia, lung cancer, pulmonary fibrosis, bronchitis, and bronchiolitis had already been reviewed. With the development of scientific research, the involvement of neutrophils in respiratory diseases is being brought to light with emerging data on neutrophil subsets, trafficking, and cell death mechanism (e.g., NETosis, apoptosis) in diseases. We reviewed all these recent studies here to provide you with the latest advances about the role of neutrophils in respiratory diseases.
Collapse
|
48
|
Jiang D, Berman R, Wu Q, Stevenson C, Chu HW. The Anti-inflammatory Effect of Alpha-1 Antitrypsin in Rhinovirus-infected Human Airway Epithelial Cells. ACTA ACUST UNITED AC 2016; 7. [PMID: 28191362 DOI: 10.4172/2155-9899.1000475] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Excessive airway inflammation is seen in chronic obstructive pulmonary disease (COPD) patients experiencing acute exacerbations, which are often associated with human rhinovirus (HRV) infection. Alpha-1 antitrypsin (A1AT) has anti-inflammatory function in endothelial cells and monocytes, but its anti-inflammatory effect has not been investigated in COPD airway epithelial cells. We determined A1AT's anti-inflammatory function in COPD airway epithelial cells and the underlying mechanisms such as the role of caspase-1. METHODS Brushed bronchial epithelial cells from COPD and normal subjects were cultured at air-liquid interface and treated with A1AT or bovine serum albumin (BSA, control) two hours prior to whole cigarette smoke (WCS) or air exposure, followed by HRV-16 infection. After 24 hours of viral infection, cell supernatants were collected for measuring IL-8, and cells were examined for caspase-1. The in vivo anti-inflammatory function of A1AT was determined by infecting mice intranasally with HRV-1B followed by aerosolized A1AT or BSA. RESULTS A1AT significantly reduced WCS and HRV-16-induced IL-8 production in normal and COPD airway epithelial cells. COPD cells are less sensitive to A1AT's anti-inflammatory effect than normal cells. A1AT exerted the anti-inflammatory function in part via reducing caspase-1 in normal cells, but not in COPD cells. In mice, A1AT significantly reduced HRV-1B induced lung neutrophilic inflammation. CONCLUSIONS A1AT exerts an anti-inflammatory effect in cigarette smoke-exposed and HRV-infected human airway epithelial cells, which may be related to its inhibitory effect on caspase-1 activity.
Collapse
Affiliation(s)
- Di Jiang
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Reena Berman
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Qun Wu
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | | | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, CO, USA; Department of Immunology and Microbiology, University of Colorado Denver School of Medicine, Aurora, CO, USA
| |
Collapse
|
49
|
Kaduwal S, Jeong WJ, Park JC, Lee KH, Lee YM, Jeon SH, Lim YB, Min DS, Choi KY. Sur8/Shoc2 promotes cell motility and metastasis through activation of Ras-PI3K signaling. Oncotarget 2016; 6:33091-105. [PMID: 26384305 PMCID: PMC4741751 DOI: 10.18632/oncotarget.5173] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/26/2015] [Indexed: 12/12/2022] Open
Abstract
Sur8 (also known as Shoc2) is a Ras-Raf scaffold protein that modulates signaling through extracellular signal-regulated kinase (ERK) pathway. Although Sur8 has been shown to be a scaffold protein of the Ras-ERK pathway, its interaction with other signaling pathways and its involvement in tumor malignancy has not been reported. We identified that Sur8 interacts with the p110α subunit of phosphatidylinositol 3-kinase (PI3K), as well as with Ras and Raf, and these interactions are increased in an epidermal growth factor (EGF)- and oncogenic Ras-dependent manner. Sur8 regulates cell migration and invasion via activation of Rac and matrix metalloproteinases (MMPs). Interestingly, using inhibitors of MEK and PI3K we found Sur8 mediates these cellular behaviors predominantly through PI3K pathway. We further found that human metastatic melanoma tissues had higher Sur8 content followed by activations of Akt, ERK, and Rac. Lentivirus-mediated Sur8-knockdown attenuated metastatic potential of highly invasive B16-F10 melanoma cells indicating the role of Sur8 in melanoma metastasis. This is the first report to identify the role of scaffold protein Sur8 in regulating cell motility, invasion, and metastasis through activation of both ERK and PI3K pathways.
Collapse
Affiliation(s)
- Saluja Kaduwal
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, 120-749, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749, Korea
| | - Woo-Jeong Jeong
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, 120-749, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749, Korea
| | - Jong-Chan Park
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, 120-749, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749, Korea
| | - Kug Hwa Lee
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, 120-749, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749, Korea
| | - Young-Mi Lee
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, 120-749, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749, Korea.,Current address: Division of Pharmacology and Translational Research, Hanmi Research Center, Hwaseong-si Gyeonggi-do, 445-813, Korea
| | - Soung-Hoo Jeon
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, 120-749, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749, Korea.,Current address: Department of Microbiology and Immunology, Xenotransplantation Research Center, Medical Research Center, Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, 110-799, Korea
| | - Yong-Beom Lim
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, 120-749, Korea.,Department of Materials Science and Engineering, Yonsei University, Seoul, 120-749, Korea
| | - Do Sik Min
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, 120-749, Korea.,Department of Molecular Biology, College of Natural Science, Pusan National University, Pusan, 609-735, Korea
| | - Kang-Yell Choi
- Translational Research Center for Protein Function Control, Yonsei University, Seoul, 120-749, Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749, Korea
| |
Collapse
|
50
|
Ganesan S, Pham D, Jing Y, Farazuddin M, Hudy MH, Unger B, Comstock AT, Proud D, Lauring AS, Sajjan US. TLR2 Activation Limits Rhinovirus-Stimulated CXCL-10 by Attenuating IRAK-1-Dependent IL-33 Receptor Signaling in Human Bronchial Epithelial Cells. THE JOURNAL OF IMMUNOLOGY 2016; 197:2409-20. [PMID: 27503209 DOI: 10.4049/jimmunol.1502702] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 07/07/2016] [Indexed: 12/15/2022]
Abstract
Airway epithelial cells are the major target for rhinovirus (RV) infection and express proinflammatory chemokines and antiviral cytokines that play a role in innate immunity. Previously, we demonstrated that RV interaction with TLR2 causes ILR-associated kinase-1 (IRAK-1) depletion in both airway epithelial cells and macrophages. Further, IRAK-1 degradation caused by TLR2 activation was shown to inhibit ssRNA-induced IFN expression in dendritic cells. Therefore, in this study, we examined the role of TLR2 and IRAK-1 in RV-induced IFN-β, IFN-λ1, and CXCL-10, which require signaling by viral RNA. In airway epithelial cells, blocking TLR2 enhanced RV-induced expression of IFNs and CXCL-10. By contrast, IRAK-1 inhibition abrogated RV-induced expression of CXCL-10, but not IFNs in these cells. Neutralization of IL-33 or its receptor, ST2, which requires IRAK-1 for signaling, inhibited RV-stimulated CXCL-10 expression. In addition, RV induced expression of both ST2 and IL-33 in airway epithelial cells. In macrophages, however, RV-stimulated CXCL-10 expression was primarily dependent on TLR2/IL-1R. Interestingly, in a mouse model of RV infection, blocking ST2 not only attenuated RV-induced CXCL-10, but also lung inflammation. Finally, influenza- and respiratory syncytial virus-induced CXCL-10 was also found to be partially dependent on IL-33/ST2/IRAK-1 signaling in airway epithelial cells. Together, our results indicate that RV stimulates CXCL-10 expression via the IL-33/ST2 signaling axis, and that TLR2 signaling limits RV-induced CXCL-10 via IRAK-1 depletion at least in airway epithelial cells. To our knowledge, this is the first report to demonstrate the role of respiratory virus-induced IL-33 in the induction of CXCL-10 in airway epithelial cells.
Collapse
Affiliation(s)
- Shyamala Ganesan
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Duc Pham
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Yaxun Jing
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Mohammad Farazuddin
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Magdalena H Hudy
- Department of Physiology & Pharmacology, University of Calgary Faculty of Medicine, Calgary, Alberta T2N 4N1, Canada
| | - Benjamin Unger
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Adam T Comstock
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109
| | - David Proud
- Department of Physiology & Pharmacology, University of Calgary Faculty of Medicine, Calgary, Alberta T2N 4N1, Canada
| | - Adam S Lauring
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109; and Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109
| | - Uma S Sajjan
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109;
| |
Collapse
|