1
|
Wu XM, Shi CN, Liu K, Hu XY, He QL, Yao H, Fan D, Ma DQ, Yang JJ, Shen JC, Ji MH. Decreased excitatory and increased inhibitory transmission in the hippocampal CA1 drive neuroinflammation-induced cognitive impairments in mice. Brain Behav Immun 2025; 128:S0889-1591(25)00164-3. [PMID: 40286992 DOI: 10.1016/j.bbi.2025.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
Neuroinflammation is one of crucial pathogenic mechanisms underlying Alzheimer's disease, sepsis-associated encephalopathy, and postoperative cognitive dysfunction. These diseases or conditions are often accompanied by typical clinical manifestations of cognitive impairments, including impaired learning and memory but underlying mechanisms are unknown. Hence, effective treatments are not available. In the current study, mice received intraperitoneal administrations of LPS (0.5 mg/kg, daily, Escherichia coliO55:B5) for seven consecutive days and after which, different cohorts were used for behavioral assessments with open field, Y maze, and novel object recognition test or for electrophysiology recordings of mEPSC, mIPSC or LTP in ex vivo preparations. Their hippocampi were harvested for immunostaining or Western blotting of PSD95, vGLUT1, vGAT, gephyrin, PV, and SST. In vivo optical fiber calcium recording was used to evaluate the neuronal excitability. During the early stage of neuroinflammation induced by LPS, there was a decrease of excitatory afferent synapses and transmission in the CA1. During the later stage of neuroinflammation, there was an increase of inhibitory afferent synapses and transmission in the CA1, resulting in excessive inhibition on excitatory neurons. Both of them contributed to the decreased hippocampal neuronal excitability and impaired LTP, ultimately leading to cognitive impairments. Overexpression of CREB in the early stage or inactivation of PV-positive interneurons in the later stage in the CA1 both improved cognitive impairments. Our work suggests that negating decreased excitatory and increased inhibitory afferent in the hippocampus may improve cognitive impairments relate to neuroinflammation associated with neurological diseases.
Collapse
Affiliation(s)
- Xin-Miao Wu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cui-Na Shi
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kai Liu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao-Yi Hu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiu-Li He
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Yao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Di Fan
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Da-Qing Ma
- Perioperative and Systems Medicine Laboratory and Department of Anesthesiology, National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Jin-Chun Shen
- Department of Anesthesiology, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, China.
| | - Mu-Huo Ji
- Department of Anesthesiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Zhao L, Ding X, Zhou L, Song C, Kang T, Xu Y, Liu Y, Han Y, Zhao W, Zhang B, Xu D, Guo J. Effect of PM 2.5 exposure on susceptibility to allergic asthma in elderly rats treated with allergens. Sci Rep 2025; 15:5594. [PMID: 39955443 PMCID: PMC11830082 DOI: 10.1038/s41598-025-90261-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 02/11/2025] [Indexed: 02/17/2025] Open
Abstract
Fine particulate matter 2.5 (PM2.5) is a prevalent atmospheric pollutant that is closely associated with asthma. Elderly patients have a high incidence of asthma with a long course of illness. Our previous studies revealed that exposure to PM2.5 diminishes lung function and exacerbates lung damage in elderly rats. In the present study, we investigated whether PM2.5 exposure influences susceptibility to allergic asthma in elderly rats. Brown-Norway elderly rats were treated with ovalbumin (OVA) for different durations before and after PM2.5 exposure. The results from pulmonary function tests and histopathology indicated that early exposure to allergens prior to PM2.5 exposure increased susceptibility to airway hyperresponsiveness and led to severe lung injury in elderly asthmatic rats. Cytokine microarray analysis demonstrated that the majority of cytokines and chemokines were upregulated in OVA-treated rats before and after PM2.5 exposure. Cytological examination showed no change in eosinophil (EOS) counts, yet the amounts of neutrophils (NEU), white blood cells (WBC), lymphocytes (LYM), and monocytes (MON) in the lung lavage fluid of OVA-treated rats were significantly higher than those in control rats before and after PM2.5 exposure, suggesting that PM2.5 affects noneosinophilic asthma in elderly rats. ELISA results from the plasma and lung lavage fluid revealed that the levels of IgG1, IgE, IgG2a and IgG2b were significantly elevated in OVA-treated rats, whereas the level of IgG2b in the lung lavage fluid was significantly lower in rats treated with OVA prior to PM2.5 exposure compared to those treated afterward. A non-targeted metabolomic analysis of plasma identified 202 metabolites, among which 31 metabolites were differentially abundant. Ten metabolites and 11 metabolic pathways were uniquely detected in OVA-treated rats before PM2.5 exposure. Specifically, there were positive or negative correlations between the levels of Th2-associated cytokines (IL-4, IL-5, and IL-13) and six metabolites in the OVA-treated group before PM2.5 exposure, whereas the levels of IL-4 and IL-5 were negatively correlated with five metabolites in the OVA-treated group after PM2.5 exposure. Our findings suggest that PM2.5 exposure could influence the susceptibility of allergic asthma in response to allergens in elderly rats, potentially through changes in plasma metabolites.
Collapse
Affiliation(s)
- Lianlian Zhao
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Dalian, 116026, China
- National Human Diseases Animal Model Resource Center, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, National Center of Technology Innovation for Animal Model, CAMS & PUMC, Beijing, China
| | - Xiaolin Ding
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Dalian, 116026, China
| | - Li Zhou
- National Human Diseases Animal Model Resource Center, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, National Center of Technology Innovation for Animal Model, CAMS & PUMC, Beijing, China
| | - Chenchen Song
- National Human Diseases Animal Model Resource Center, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, National Center of Technology Innovation for Animal Model, CAMS & PUMC, Beijing, China
| | - Taisheng Kang
- National Human Diseases Animal Model Resource Center, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, National Center of Technology Innovation for Animal Model, CAMS & PUMC, Beijing, China
| | - Yanfeng Xu
- National Human Diseases Animal Model Resource Center, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, National Center of Technology Innovation for Animal Model, CAMS & PUMC, Beijing, China
| | - Yunpeng Liu
- National Human Diseases Animal Model Resource Center, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, National Center of Technology Innovation for Animal Model, CAMS & PUMC, Beijing, China
| | - Yunlin Han
- National Human Diseases Animal Model Resource Center, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, National Center of Technology Innovation for Animal Model, CAMS & PUMC, Beijing, China
| | - Wenjie Zhao
- National Human Diseases Animal Model Resource Center, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, National Center of Technology Innovation for Animal Model, CAMS & PUMC, Beijing, China
| | - Boxiang Zhang
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Dalian, 116026, China
| | - Dan Xu
- Institute of Environmental Systems Biology, Environment Science and Engineering College, Dalian Maritime University, Dalian, 116026, China.
| | - Jianguo Guo
- National Human Diseases Animal Model Resource Center, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, National Center of Technology Innovation for Animal Model, CAMS & PUMC, Beijing, China.
| |
Collapse
|
3
|
Manell H, Tsolakis N, Janson C, Malinovschi A, Alving K. Multiarray screening identifies plasma proteins associated with Th17 cell differentiation and viral defense in coincident asthma and obesity. Pediatr Allergy Immunol 2024; 35:e14187. [PMID: 38967090 DOI: 10.1111/pai.14187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND The immunological mechanisms behind the clinical association between asthma and obesity in adolescence are not fully understood. This study aimed to find new plasma protein biomarkers associated specifically with coincident asthma and obesity in adolescents. METHODS This was a cross-sectional study in children and adolescents 10-19 years old (N = 390). Relative plasma concentrations of 113 protein biomarkers related to inflammation and immune response were determined by proximity extension assay (Target 96; Olink, Uppsala, Sweden). Differences in protein concentrations between healthy controls (n = 84), subjects with asthma (n = 138), subjects with obesity (n = 107), and subjects with both asthma and obesity (AO; n = 58) were analyzed by ANCOVA, adjusting for age and sex, and in a separate model adjusting also for the sum of specific IgE antibody concentrations to a mix of food allergens (fx5) and aeroallergens (Phadiatop). Proteins elevated in the AO group but not in the obesity or asthma groups were considered specifically elevated in asthma and obesity. RESULTS Five proteins were elevated specifically in the AO group compared to controls (here sorted from largest to smallest effect of asthma and obesity combined): CCL8, IL-33, IL-17C, FGF-23, and CLEC7A. The effects of adjusting also for specific IgE were small but IL-33, IL-17C, and FGF-23 were no longer statistically significant. CONCLUSION We identified several new potential plasma biomarkers specifically elevated in coincident asthma and obesity in adolescents. Four of the proteins, CCL8, IL-33, IL-17C, and CLEC7A, have previously been associated with viral mucosal host defense and Th17 cell differentiation.
Collapse
Affiliation(s)
- Hannes Manell
- Department of Women's and Children's Health, Paediatrics, Uppsala University, Uppsala, Sweden
| | - Nikolaos Tsolakis
- Department of Women's and Children's Health, Paediatrics, Uppsala University, Uppsala, Sweden
| | - Christer Janson
- Department of Medical Sciences, Respiratory-, Allergy- and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Andrei Malinovschi
- Department of Medical Sciences, Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Kjell Alving
- Department of Women's and Children's Health, Paediatrics, Uppsala University, Uppsala, Sweden
| |
Collapse
|
4
|
Kaur S, Mishra J, Sehrawat A, Bhatti GK, Navik U, Reddy PH, Bhatti JS. Epigenetic Regulators of Inflammatory Gene Expression. TARGETING EPIGENETICS IN INFLAMMATORY LUNG DISEASES 2023:57-88. [DOI: https:/doi.org/10.1007/978-981-99-4780-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
|
5
|
Alobaidi A, Alsamarai A, Alsamarai MA. Inflammation in Asthma Pathogenesis: Role of T cells, Macrophages, Epithelial Cells and Type 2 Inflammation. Antiinflamm Antiallergy Agents Med Chem 2021; 20:317-332. [PMID: 34544350 DOI: 10.2174/1871523020666210920100707] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/06/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
Asthma is a chronic disease with abnormal inflammatory and immunological responses. The disease initiated by antigens in subjects with genetic susceptibility. However, environmental factors play a role in the initiation and exacerbation of asthma attack. Asthma is T helper 2 (Th2)-cell-mediated disease. Recent studies indicated that asthma is not a single disease entity, but it is with multiple phenotypes and endotypes. The pathophysiological changes in asthma included a series of subsequent continuous vicious circle of cellular activation contributed to induction of chemokines and cytokines that potentiate inflammation. The heterogeneity of asthma influenced the treatment response. The asthma pathogenesis driven by varied set of cells such as eosinophils, basophils, neutrophils, mast cells, macrophages, epithelial cells and T cells. In this review the role of T cells, macrophage, and epithelial cells are discussed.
Collapse
Affiliation(s)
- Amina Alobaidi
- Kirkuk University College of Veterinary Medicine, Kirkuk. Iraq
| | - Abdulghani Alsamarai
- Aalborg Academy College of Medicine [AACOM], Denmark. Tikrit University College of Medicine, [TUCOM], Tikrit. Iraq
| | | |
Collapse
|
6
|
The Role of T Cells and Macrophages in Asthma Pathogenesis: A New Perspective on Mutual Crosstalk. Mediators Inflamm 2020; 2020:7835284. [PMID: 32922208 PMCID: PMC7453253 DOI: 10.1155/2020/7835284] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 12/11/2022] Open
Abstract
Asthma is associated with innate and adaptive immunity mediated by immune cells. T cell or macrophage dysfunction plays a particularly significant role in asthma pathogenesis. Furthermore, crosstalk between them continuously transmits proinflammatory or anti-inflammatory signals, causing the immune cell activation or repression in the immune response. Consequently, the imbalanced immune microenvironment is the major cause of the exacerbation of asthma. Here, we discuss the role of T cells, macrophages, and their interactions in asthma pathogenesis.
Collapse
|
7
|
Finney LJ, Belchamber KBR, Fenwick PS, Kemp SV, Edwards MR, Mallia P, Donaldson G, Johnston SL, Donnelly LE, Wedzicha JA. Human Rhinovirus Impairs the Innate Immune Response to Bacteria in Alveolar Macrophages in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2020; 199:1496-1507. [PMID: 30562053 DOI: 10.1164/rccm.201806-1095oc] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Rationale: Human rhinovirus (HRV) is a common cause of chronic obstructive pulmonary disease (COPD) exacerbations. Secondary bacterial infection is associated with more severe symptoms and delayed recovery. Alveolar macrophages clear bacteria from the lung and maintain lung homeostasis through cytokine secretion. These processes are defective in COPD. The effect of HRV on macrophage function is unknown. Objectives: To investigate the effect of HRV on phagocytosis and cytokine response to bacteria by alveolar macrophages and monocyte-derived macrophages (MDM) in COPD and healthy control subjects. Methods: Alveolar macrophages were obtained by bronchoscopy and MDM by adherence. Macrophages were exposed to HRV16 (multiplicity of infection 5), polyinosinic:polycytidylic acid (poly I:C) 30 μg/ml, IFN-β 10 μg/ml, IFN-γ 10 μg/ml, or medium control for 24 hours. Phagocytosis of fluorescently labeled Haemophilus influenzae or Streptococcus pneumoniae was assessed by fluorimetry. CXCL8 (IL-8), IL-6, TNF-α (tumor necrosis factor-α), and IL-10 release was measured by ELISA. Measurements and Main Results: HRV significantly impaired phagocytosis of H. influenzae by 23% in MDM (n = 37; P = 0.004) and 18% in alveolar macrophages (n = 20; P < 0.0001) in COPD. HRV also significantly reduced phagocytosis of S. pneumoniae by 33% in COPD MDM (n = 20; P = 0.0192). There was no effect in healthy control subjects. Phagocytosis of H. influenzae was also impaired by poly I:C but not IFN-β or IFN-γ in COPD MDM. HRV significantly reduced cytokine responses to H. influenzae. The IL-10 response to H. influenzae was significantly impaired by poly I:C, IFN-β, and IFN-γ in COPD cells. Conclusions: HRV impairs phagocytosis of bacteria in COPD, which may lead to an outgrowth of bacteria. HRV also impairs cytokine responses to bacteria via the TLR3/IFN pathway, which may prevent resolution of inflammation leading to prolonged exacerbations in COPD.
Collapse
Affiliation(s)
- Lydia J Finney
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Kylie B R Belchamber
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Peter S Fenwick
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Samuel V Kemp
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and.,2 Royal Brompton Hospital, London, United Kingdom
| | - Michael R Edwards
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Patrick Mallia
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Gavin Donaldson
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Sebastian L Johnston
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Louise E Donnelly
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Jadwiga A Wedzicha
- 1 COPD and Asthma Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| |
Collapse
|
8
|
Nelwan SC, Nugraha RA, Endaryanto A, Dewi F, Nuraini P, Tedjosasongko U, Utomo DH. Effect of scaling and root planing on level of immunoglobulin E and immunoglobulin G 4 in children with gingivitis and house-dust mite allergy: A pilot randomised controlled trial. ACTA ACUST UNITED AC 2020; 39:21-31. [PMID: 32054426 DOI: 10.1142/s2214607519500020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background and Objective: There is a pressing need for developing innovative strategies to prevent allergic diseases among children. As house-dust mite (HDM) allergy is often seen in children with gingivitis, strategies should be derived from a conceptual framework of allergen elimination and pathogen eradication; one such strategy is dental scaling and root planing (SRP) to remove dental plaque and periodontal pathogens. The study aimed to evaluate the beneficial effects of comprehensive 6-months dental SRP to reduce the level of immunoglobulin E (IgE) and immunoglobulin G4 (IgG4) in children with gingivitis and HDM allergy. IgE and IgG4, whose production is controlled mainly by Th-2 cells and B cells, are proven biomarkers for atopic inflammatory responses. Methods: The present study conducted a non-blinded randomised controlled trial with superiority design. A total of 10 subjects (age range 6-16 years) with gingivitis and positive skin-prick test to HDM from Pediatric Allergy Outpatient Clinic, Dr. Soetomo General Hospital were enrolled in the present study. Of the 10 subjects, only five received dental SRP. We further evaluated total serum IgE and IgG4 level before and 6 months after treatment. Results and Discussion: Subjects in the standard treatment group showed a slight decrease in the IgE level ([Formula: see text]) but no change in the IgG4 level ([Formula: see text]), while subjects in the intervention group showed a significant decrease in IgE ([Formula: see text]) and IgG4 levels ([Formula: see text]). Conclusion: The study results suggest that 6-month comprehensive dental scaling combined with root planing may help to reduce IgE and IgG4 levels in children with gingivitis and HDM allergy. Furthermore, untreated or undertreated gingivitis is often associated with worsening allergic manifestation and thus should be avoided. Trial Registration: ISRCTN31416107, retrospectively registered on 17 April 2018.
Collapse
Affiliation(s)
- Sindy Cornelia Nelwan
- Department of Pediatric Dentistry, Universitas Airlangga, Surabaya, 60135, Indonesia
| | | | - Anang Endaryanto
- Department of Child Health, Universitas Airlangga, Surabaya, 60135, Indonesia
| | - Frisma Dewi
- Department of Pediatric Dentistry, Universitas Airlangga, Surabaya, 60135, Indonesia
| | - Prawati Nuraini
- Department of Pediatric Dentistry, Universitas Airlangga, Surabaya, 60135, Indonesia
| | | | | |
Collapse
|
9
|
Huoman J, Papapavlou G, Pap A, Alm J, Nilsson LJ, Jenmalm MC. Sublingual immunotherapy alters salivary IgA and systemic immune mediators in timothy allergic children. Pediatr Allergy Immunol 2019; 30:522-530. [PMID: 30803044 DOI: 10.1111/pai.13047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND Immunomodulatory effects of sublingual immunotherapy on systemic and mucosal mediators in allergic children are largely unexplored. The aim of this study was to investigate allergy-related cytokine and chemokine levels, as well as IgA-responses upon a 3-year treatment with timothy grass pollen sublingual immunotherapy in children with allergic rhinoconjunctivitis. METHODS From children included in the GRAZAX® Asthma Prevention study, blood and saliva samples were analyzed at inclusion, after 3 years of treatment, and 2 years after treatment ending. By means of Luminex and ELISA methodologies, allergy-related cytokines and chemokines were measured in plasma samples and allergen-stimulated peripheral blood mononuclear cell supernatants. Furthermore, studies of total, secretory, and Phl p 1-specific salivary IgA antibodies were performed using the same methods. RESULTS GRAZAX® -treated children exhibited significantly higher levels of Phl p 1-specific salivary IgA and serum IgG4 , along with significantly lower skin prick test positivity, after 3 years of treatment and 2 years after treatment cessation. Additionally, plasma levels of the Th1-associated chemokines CXCL10 and CXCL11 were significantly higher in treated than untreated children at these time points. Timothy-induced ratios of IL-5/IL-13 over IFN-γ were significantly decreased after 3 years with active treatment, as were symptoms of allergic rhinitis in terms of both severity and visual analogue scale scores. However, no consistent correlations were found between the clinical outcomes and immunologic parameters. CONCLUSION Phleum pratense sublingual immunotherapy in grass pollen allergic children modulates the immune response in the oral mucosa as well as systemically-by increasing Th1-responses, decreasing Th2-responses, and inducing immunoregulatory responses-all signs of tolerance induction.
Collapse
Affiliation(s)
- Johanna Huoman
- Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Georgia Papapavlou
- Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Anna Pap
- Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Johan Alm
- Karolinska Institutet, Department of Clinical Science and Education, Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Lennart J Nilsson
- Allergy Center, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Maria C Jenmalm
- Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
10
|
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
|
11
|
Abstract
The mucosal surfaces of the human body are typically colonized by polymicrobial communities seeded in infancy and are continuously shaped by environmental exposures. These communities interact with the mucosal immune system to maintain homeostasis in health, but perturbations in their composition and function are associated with lower airway diseases, including asthma, a developmental and heterogeneous chronic disease with various degrees and types of airway inflammation. This review will summarize recent studies examining airway microbiota dysbioses associated with asthma and their relationship with the pathophysiology of this disease.
Collapse
|
12
|
Lee H, Ko G. Antiviral effect of vitamin A on norovirus infection via modulation of the gut microbiome. Sci Rep 2016; 6:25835. [PMID: 27180604 PMCID: PMC4867650 DOI: 10.1038/srep25835] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 04/22/2016] [Indexed: 12/04/2022] Open
Abstract
The effect and underlying mechanism of vitamin A on norovirus infection are largely unknown. This study aimed to investigate how vitamin A administration affects the gut microbiome after norovirus infection. Here, we demonstrate that treatment with either retinol or retinoic acid (RA) inhibits murine norovirus (MNV) replication using both in vitro and in vivo models. Compositional changes in the gut microbiome associated with RA administration and/or norovirus infection were also investigated. Oral administration of RA and/or MNV significantly altered intestinal microbiome profiles. Particularly, bacterial species belonging to the Lactobacillaceae families were remarkably increased by MNV inoculation and RA administration, suggesting that the antiviral effects of RA occur via the modulation of specific microbiota. The antiviral causal effect of Lactobacillus was identified and demonstrated using in vitro models in RAW264.7 cells. The antiviral immune response to MNV was mediated by IFN-β upregulation. This study represents the first comprehensive profiling of gut microbiota in response to RA treatment against norovirus infection. Moreover, we conclude that the abundance of Lactobacillus through gut microbiota modulation by RA is at least partially responsible for norovirus inhibition.
Collapse
Affiliation(s)
- Heetae Lee
- Center for Human and Environmental Microbiome, Institute of Health and Environment, School of Public Health, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea
| | - GwangPyo Ko
- Center for Human and Environmental Microbiome, Institute of Health and Environment, School of Public Health, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea.,N-BIO, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea
| |
Collapse
|
13
|
Hewitt R, Farne H, Ritchie A, Luke E, Johnston SL, Mallia P. The role of viral infections in exacerbations of chronic obstructive pulmonary disease and asthma. Ther Adv Respir Dis 2016; 10:158-74. [PMID: 26611907 PMCID: PMC5933560 DOI: 10.1177/1753465815618113] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are major causes of global morbidity and mortality worldwide. The clinical course of both asthma and COPD are punctuated by the occurrence of exacerbations, acute events characterized by increased symptoms and airflow obstruction. Exacerbations contribute most of the morbidity, mortality and excess healthcare costs associated with both asthma and COPD. COPD and asthma exacerbations are frequently associated with respiratory virus infections and this has led to an intense research focus into the mechanisms of virus-induced exacerbations over the past decade. Current therapies are effective in reducing chronic symptoms but are less effective in preventing exacerbations, particularly in COPD. Understanding the mechanisms of virus-induced exacerbation will lead to the development of new targeted therapies that can reduce the burden of virus-induced exacerbations. In this review we discuss current knowledge of virus-induced exacerbations of asthma and COPD with a particular focus on mechanisms, human studies, virus-bacteria interactions and therapeutic advances.
Collapse
Affiliation(s)
- Richard Hewitt
- National Heart and Lung Institute, Imperial College London, UK
| | - Hugo Farne
- National Heart and Lung Institute, Imperial College London, UK
| | - Andrew Ritchie
- National Heart and Lung Institute, Imperial College London, UK
| | - Emma Luke
- Imperial Healthcare NHS Trust, London, UK
| | | | - Patrick Mallia
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
14
|
Halvorsen EC, Hamilton MJ, Young A, Wadsworth BJ, LePard NE, Lee HN, Firmino N, Collier JL, Bennewith KL. Maraviroc decreases CCL8-mediated migration of CCR5(+) regulatory T cells and reduces metastatic tumor growth in the lungs. Oncoimmunology 2016; 5:e1150398. [PMID: 27471618 DOI: 10.1080/2162402x.2016.1150398] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 01/21/2016] [Accepted: 01/31/2016] [Indexed: 02/08/2023] Open
Abstract
Regulatory T cells (Tregs) play a crucial physiological role in the regulation of immune homeostasis, although recent data suggest Tregs can contribute to primary tumor growth by suppressing antitumor immune responses. Tregs may also influence the development of tumor metastases, although there is a paucity of information regarding the phenotype and function of Tregs in metastatic target organs. Herein, we demonstrate that orthotopically implanted metastatic mammary tumors induce significant Treg accumulation in the lungs, which is a site of mammary tumor metastasis. Tregs in the primary tumor and metastatic lungs express high levels of C-C chemokine receptor type 5 (CCR5) relative to Tregs in the mammary fat pad and lungs of tumor-free mice, and Tregs in the metastatic lungs are enriched for CCR5 expression in comparison to other immune cell populations. We also identify that C-C chemokine ligand 8 (CCL8), an endogenous ligand of CCR5, is produced by F4/80(+) macrophages in the lungs of mice with metastatic primary tumors. Migration of Tregs toward CCL8 ex vivo is reduced in the presence of the CCR5 inhibitor Maraviroc. Importantly, treatment of mice with Maraviroc (MVC) reduces the level of CCR5(+) Tregs and metastatic tumor burden in the lungs. This work provides evidence of a CCL8/CCR5 signaling axis driving Treg recruitment to the lungs of mice bearing metastatic primary tumors, representing a potential therapeutic target to decrease Treg accumulation and metastatic tumor growth.
Collapse
Affiliation(s)
- E C Halvorsen
- Integrative Oncology Department, British Columbia Cancer Agency, Vancouver, BC, Canada; Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - M J Hamilton
- Integrative Oncology Department, British Columbia Cancer Agency , Vancouver, BC, Canada
| | - A Young
- Integrative Oncology Department, British Columbia Cancer Agency, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - B J Wadsworth
- Integrative Oncology Department, British Columbia Cancer Agency, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - N E LePard
- Integrative Oncology Department, British Columbia Cancer Agency , Vancouver, BC, Canada
| | - H N Lee
- Integrative Oncology Department, British Columbia Cancer Agency , Vancouver, BC, Canada
| | - N Firmino
- Integrative Oncology Department, British Columbia Cancer Agency, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - J L Collier
- Integrative Oncology Department, British Columbia Cancer Agency, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - K L Bennewith
- Integrative Oncology Department, British Columbia Cancer Agency, Vancouver, BC, Canada; Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
15
|
Matrine Attenuates COX-2 and ICAM-1 Expressions in Human Lung Epithelial Cells and Prevents Acute Lung Injury in LPS-Induced Mice. Mediators Inflamm 2016; 2016:3630485. [PMID: 26880863 PMCID: PMC4736390 DOI: 10.1155/2016/3630485] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/08/2015] [Accepted: 11/10/2015] [Indexed: 01/07/2023] Open
Abstract
Matrine is isolated from Sophora flavescens and shows anti-inflammatory effects in macrophages. Here we evaluated matrine's suppressive effects on cyclooxygenase 2 (COX-2) and intercellular adhesion molecule-1 (ICAM-1) expressions in lipopolysaccharide- (LPS-) stimulated human lung epithelial A549 cells. Additionally, BALB/c mice were given various matrine doses by intraperitoneal injection, and then lung injury was induced via intratracheal instillation of LPS. In LPS-stimulated A549 cells, matrine inhibited the productions of interleukin-8 (IL-8), monocyte chemotactic protein-1, and IL-6 and decreased COX-2 expression. Matrine treatment also decreased ICAM-1 protein expression and suppressed the adhesion of neutrophil-like cells to inflammatory A549 cells. In vitro results demonstrated that matrine significantly inhibited mitogen-activated protein kinase phosphorylation and decreased nuclear transcription factor kappa-B subunit p65 protein translocation into the nucleus. In vivo data indicated that matrine significantly inhibited neutrophil infiltration and suppressed productions of tumor necrosis factor-α and IL-6 in mouse bronchoalveolar lavage fluid and serum. Analysis of lung tissue showed that matrine decreased the gene expression of proinflammatory cytokines, chemokines, COX-2, and ICAM-1. Our findings suggest that matrine improved lung injury in mice and decreased the inflammatory response in human lung epithelial cells.
Collapse
|
16
|
A short-term mouse model that reproduces the immunopathological features of rhinovirus-induced exacerbation of COPD. Clin Sci (Lond) 2015; 129:245-58. [PMID: 25783022 PMCID: PMC4557402 DOI: 10.1042/cs20140654] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Viral exacerbations of chronic obstructive pulmonary disease (COPD), commonly caused by rhinovirus (RV) infections, are poorly controlled by current therapies. This is due to a lack of understanding of the underlying immunopathological mechanisms. Human studies have identified a number of key immune responses that are associated with RV-induced exacerbations including neutrophilic inflammation, expression of inflammatory cytokines and deficiencies in innate anti-viral interferon. Animal models of COPD exacerbation are required to determine the contribution of these responses to disease pathogenesis. We aimed to develop a short-term mouse model that reproduced the hallmark features of RV-induced exacerbation of COPD. Evaluation of complex protocols involving multiple dose elastase and lipopolysaccharide (LPS) administration combined with RV1B infection showed suppression rather than enhancement of inflammatory parameters compared with control mice infected with RV1B alone. Therefore, these approaches did not accurately model the enhanced inflammation associated with RV infection in patients with COPD compared with healthy subjects. In contrast, a single elastase treatment followed by RV infection led to heightened airway neutrophilic and lymphocytic inflammation, increased expression of tumour necrosis factor (TNF)-α, C-X-C motif chemokine 10 (CXCL10)/IP-10 (interferon γ-induced protein 10) and CCL5 [chemokine (C-C motif) ligand 5]/RANTES (regulated on activation, normal T-cell expressed and secreted), mucus hypersecretion and preliminary evidence for increased airway hyper-responsiveness compared with mice treated with elastase or RV infection alone. In summary, we have developed a new mouse model of RV-induced COPD exacerbation that mimics many of the inflammatory features of human disease. This model, in conjunction with human models of disease, will provide an essential tool for studying disease mechanisms and allow testing of novel therapies with potential to be translated into clinical practice.
Collapse
|
17
|
Sesamol suppresses the inflammatory response by inhibiting NF-κB/MAPK activation and upregulating AMP kinase signaling in RAW 264.7 macrophages. Inflamm Res 2015; 64:577-88. [PMID: 26059394 DOI: 10.1007/s00011-015-0836-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/12/2015] [Accepted: 05/29/2015] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES AND DESIGN Sesamol is a lignan isolated from sesame seed oil. In recent years, it was found that sesamol could decrease lung inflammation and lipopolysaccharide (LPS)-induced lung injury in rats. In this study, we investigated whether sesamol exhibited anti-inflammatory activity in LPS-stimulated macrophages. MATERIALS AND METHODS RAW 264.7 cells were treated with sesamol, then treated with LPS to induce inflammation. The levels of proinflammatory cytokines were analyzed with ELISA. The gene and protein expression of cyclooxygenase (COX)-2, inducible nitric oxide synthase (iNOS), and nuclear factor erythroid-2-related factor 2 (Nrf2) were evaluated with real-time PCR and Western blots, respectively. We also examined inflammatory signaling pathways, including nuclear transcription factor kappa-B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways. RESULTS Sesamol inhibited production of nitric oxide, prostaglandin E2 (PGE2), and proinflammatory cytokines. Sesamol markedly suppressed mRNA and protein expression of iNOS and COX-2. Sesamol enhanced the protective antioxidant pathway represented by Nrf2 and HO-1. Moreover, sesamol suppressed NF-κB transport into the nucleus and decreased MAPK activation, but it promoted adenosine monophosphate-activated protein kinase (AMPK) activation. CONCLUSIONS These data suggested that sesamol ameliorated inflammatory and oxidative damage by upregulating AMPK activation and Nrf2 signaling and blocking the NF-κB and MAPK signaling pathways.
Collapse
|
18
|
Schuler BA, Schreiber MT, Li L, Mokry M, Kingdon ML, Raugi DN, Smith C, Hameister C, Racaniello VR, Hall DJ. Major and minor group rhinoviruses elicit differential signaling and cytokine responses as a function of receptor-mediated signal transduction. PLoS One 2014; 9:e93897. [PMID: 24736642 PMCID: PMC3988043 DOI: 10.1371/journal.pone.0093897] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 03/08/2014] [Indexed: 11/18/2022] Open
Abstract
Major- and minor-group human rhinoviruses (HRV) enter their host by binding to the cell surface molecules ICAM-1 and LDL-R, respectively, which are present on both macrophages and epithelial cells. Although epithelial cells are the primary site of productive HRV infection, previous studies have implicated macrophages in establishing the cytokine dysregulation that occurs during rhinovirus-induced asthma exacerbations. Analysis of the transcriptome of primary human macrophages exposed to major- and minor-group HRV demonstrated differential gene expression. Alterations in gene expression were traced to differential mitochondrial activity and signaling pathway activation between two rhinovirus serotypes, HRV16 (major-group) and HRV1A (minor-group), upon initial HRV binding. Variances in phosphorylation of kinases (p38, JNK, ERK5) and transcription factors (ATF-2, CREB, CEBP-alpha) were observed between the major- and minor-group HRV treatments. Differential activation of signaling pathways led to changes in the production of the asthma-relevant cytokines CCL20, CCL2, and IL-10. This is the first report of genetically similar viruses eliciting dissimilar cytokine release, transcription factor phosphorylation, and MAPK activation from macrophages, suggesting that receptor use is a mechanism for establishing the inflammatory microenvironment in the human airway upon exposure to rhinovirus.
Collapse
Affiliation(s)
- Bryce A. Schuler
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
| | - Michael T. Schreiber
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
- Department of Microbiology & Immunology, Columbia University College of Physicians and Surgeons, New York, New York, United States of America
| | - LuYuan Li
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
| | - Michal Mokry
- Division of Pediatrics, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Megan L. Kingdon
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
| | - Dana N. Raugi
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
| | - Cosonya Smith
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
| | - Chelsea Hameister
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
| | - Vincent R. Racaniello
- Department of Microbiology & Immunology, Columbia University College of Physicians and Surgeons, New York, New York, United States of America
| | - David J. Hall
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|