51
|
Kadin ME, Morgan J, Wei W, Song Z, Yang Y. CD30 Regulation of IL-13-STAT6 Pathway in Breast Implant-Associated Anaplastic Large Cell Lymphoma. Aesthet Surg J 2023; 43:137-146. [PMID: 35999655 PMCID: PMC10208747 DOI: 10.1093/asj/sjac234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Breast implant-associated anaplastic large cell lymphoma (BIA-ALCL) is a rare, usually indolent CD30+ T-cell lymphoma with tumor cells, often surrounded by eosinophils, expressing IL-13 and pSTAT6. OBJECTIVES The aim of this study was to understand the unique tumor pathology and growth regulation of BIA-ALCL, leading to potential targeted therapies. METHODS We silenced CD30 and analyzed its effect on IL-13 signaling and tumor cell viability. IL-13 signaling receptors of BIA-ALCL cell lines were evaluated by flow cytometry and pSTAT6 detected by immunohistochemistry. CD30 was deleted by CRISPR/Cas9 editing. Effects of CD30 deletion on transcription of IL-13 and IL-4, and phosphorylation of STAT6 were determined by real-time polymerase chain reaction and western blotting. The effect of CD30 deletion on p38 mitogen-activated protein kinase (MAPK) phosphorylation was determined. Suppression of IL-13 transcription by a p38 MAPK inhibitor was tested. Tumor cell viability following CD30 deletion and treatment with a pSTAT6 inhibitor were measured in cytotoxicity assays. RESULTS BIA-ALCL lines TLBR1 and TLBR2 displayed signaling receptors IL-4Rα, IL-13Rα1 and downstream pSTAT6. Deletion of CD30 by CRISPR/Cas9 editing significantly decreased transcription of IL-13, less so Th2 cytokine IL-4, and phosphorylation of STAT6. Mechanistically, we found CD30 expression is required for p38 MAPK phosphorylation and activation, and IL-13-STAT6 signaling was reduced by an inhibitor of p38 MAPK in BIA-ALCL tumor cells. Tumor cell viability was decreased by silencing of CD30, and a specific inhibitor of STAT6, indicating STAT6 inhibition is cytotoxic to BIA-ALCL tumor cells. CONCLUSIONS These findings suggest reagents targeting the IL-13 pathway, pSTAT6 and p38 MAPK, may become useful for treating BIA-ALCL patients.
Collapse
Affiliation(s)
- Marshall E Kadin
- Department of Pathology and Laboratory Medicine, Brown University Alpert
School of Medicine, Providence, RI. USA
| | | | - Wei Wei
- Fox Chase Cancer Center, Philadelphia,
PA, USA
| | - Zhihui Song
- Fox Chase Cancer Center, Philadelphia,
PA, USA
| | - Yibin Yang
- Blood Cell Development and Function Program, Fox Chase Cancer
Center, Philadelphia, PA, USA
| |
Collapse
|
52
|
Huang J, Puente H, Wareing NE, Wu M, Mayes MD, Karmouty-Quintana H, Assassi S, Mills TW. STAT6 suppression prevents bleomycin-induced dermal fibrosis. FASEB J 2023; 37:e22761. [PMID: 36629780 PMCID: PMC10226134 DOI: 10.1096/fj.202200994r] [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: 06/22/2022] [Revised: 12/14/2022] [Accepted: 12/27/2022] [Indexed: 01/12/2023]
Abstract
Fibrosis of the skin and internal organs is a hallmark of systemic sclerosis (SSc). Although the pathogenesis of SSc is poorly understood, increasing evidence suggests that interleukins (IL)-4 and - 13 contribute to the pathogenesis of skin fibrosis by promoting collagen production and myofibroblast differentiation. Signal transducers and activators of transcription 6 (STAT6) is one of the most important downstream transcription factors activated by both IL-4 and IL-13. However, it is not completely understood whether STAT6 plays a role during the pathogenesis of skin fibrosis in SSc. In this study, we observed increased STAT6 phosphorylation in fibrotic skin samples collected from SSc patients as well as bleomycin-injected murine mice. Knockout of Stat6 in mice significantly (1) suppressed the expression of fibrotic cytokines including Il13, Il17, Il22, Ccl2, and the alternatively activated macrophage marker Cd206; (2) reduced the production of collagen and fibronectin, and (3) attenuated late-stage skin fibrosis and inflammation induced by bleomycin. Consistently, mice treated with STAT6 inhibitor AS1517499 also attenuated skin fibrosis on day 28. In addition, a co-culture experiment demonstrated that skin epithelial cells with STAT6 knockdown had reduced cytokine expression in response to IL-4/IL-13, and subsequently attenuated fibrotic protein expression in skin fibroblasts. On the other side, STAT6 depletion in skin fibroblasts attenuated IL-4/IL-13-induced cytokine and fibrotic marker expression, and reduced CXCL2 expression in co-cultured keratinocytes. In summary, our study highlighted an important yet not fully understood role of STAT6 in skin fibrosis by driving innate inflammation and differentiation of alternatively activated macrophages in response to injury.
Collapse
Affiliation(s)
- Jingjing Huang
- Department of Geriatrics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Hydia Puente
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Internal Medicine, Division of Rheumatology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nancy E. Wareing
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Internal Medicine, Division of Rheumatology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Minghua Wu
- Department of Internal Medicine, Division of Rheumatology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Maureen D. Mayes
- Department of Internal Medicine, Division of Rheumatology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Shervin Assassi
- Department of Internal Medicine, Division of Rheumatology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Tingting W. Mills
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
53
|
Donlan AN, Mallawaarachchi I, Sasson JM, Preissner R, Loomba JJ, Petri WA. Dupilumab Use Is Associated With Protection From Coronavirus Disease 2019 Mortality: A Retrospective Analysis. Clin Infect Dis 2023; 76:148-151. [PMID: 36104868 PMCID: PMC9494491 DOI: 10.1093/cid/ciac745] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 08/22/2022] [Accepted: 09/08/2022] [Indexed: 01/13/2023] Open
Abstract
We previously found that type 2 immunity promotes coronavirus disease 2019 (COVID-19) pathogenesis in a mouse model. To test relevance to human disease, we used electronic health record databases and determined that patients on dupilumab (anti-interleukin [IL]-4R monoclonal antibody that blocks IL-13 and IL-4 signaling) at the time of COVID-19 infection had lower mortality.
Collapse
Affiliation(s)
- Alexandra N Donlan
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Indika Mallawaarachchi
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Jennifer M Sasson
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Robert Preissner
- Science-IT and Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Johanna J Loomba
- Integrated Translational Health Research Institute of Virginia (iTHRIV), Charlottesville, Virginia, USA
| | - William A Petri
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
54
|
Esnault S, Jarjour NN. Development of Adaptive Immunity and Its Role in Lung Remodeling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1426:287-351. [PMID: 37464127 DOI: 10.1007/978-3-031-32259-4_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Asthma is characterized by airflow limitations resulting from bronchial closure, which can be either reversible or fixed due to changes in airway tissue composition and structure, also known as remodeling. Airway remodeling is defined as increased presence of mucins-producing epithelial cells, increased thickness of airway smooth muscle cells, angiogenesis, increased number and activation state of fibroblasts, and extracellular matrix (ECM) deposition. Airway inflammation is believed to be the main cause of the development of airway remodeling in asthma. In this chapter, we will review the development of the adaptive immune response and the impact of its mediators and cells on the elements defining airway remodeling in asthma.
Collapse
|
55
|
Elarekibep (PRS-060/AZD1402), a new class of inhaled Anticalin medicine targeting IL-4Ra for type 2 endotype asthma. J Allergy Clin Immunol 2022; 151:966-975. [PMID: 36592703 DOI: 10.1016/j.jaci.2022.12.815] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/20/2022] [Accepted: 12/29/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND Type 2 endotype asthma is driven by IL-4 and IL-13 signaling via IL-4Ra, which is highly expressed on airway epithelium, airway smooth muscle, and immunocytes in the respiratory mucosa, suggesting potential advantages of an inhalable antagonist. Lipocalin 1 (Lcn1), a 16 kDa protein abundant in human periciliary fluid, has a robust drug-like structure well suited to protein engineering, but it has never been used to make an inhaled Anticalin protein therapeutic. OBJECTIVES We sought to reengineer Lcn1 into an inhalable IL-4Ra antagonist and assess its pharmacodynamic/kinetic profile. METHODS Lcn1 was systematically modified by directed protein mutagenesis yielding a high-affinity, slowly dissociating, long-acting full antagonist of IL-4Ra designated PRS-060 with properties analogous to dupilumab, competitively antagonizing IL-4Ra-dependent cell proliferation, mucus induction, and eotaxin expression in vitro. Because PRS-060 displayed exquisite specificity for human IL-4Ra, with no cross-reactivity to rodents or higher primates, we created a new triple-humanized mouse model substituting human IL-4Ra, IL-4, and IL-13 at their correct syntenic murine loci to model clinical dosing. RESULTS Inhaled PRS-060 strongly suppressed acute allergic inflammation indexes in triple-humanized mice with a duration of action longer than its bulk clearance, suggesting that it may act locally in the lung. CONCLUSION Lcn1 can be reengineered into the Anticalin antagonist PRS-060 (elarekibep), exemplifying a new class of inhaled topical, long-acting therapeutic drugs with the potential to treat type 2 endotype asthma.
Collapse
|
56
|
Zhong Y, Hu L, Chen W, Wang B, Sun J, Dong J. Exploring the comorbidity mechanisms between asthma and idiopathic pulmonary fibrosis and the pharmacological mechanisms of Bu-Shen-Yi-Qi decoction therapy via network pharmacology. BMC Complement Med Ther 2022; 22:151. [PMID: 35672815 PMCID: PMC9175349 DOI: 10.1186/s12906-022-03637-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 05/30/2022] [Indexed: 11/10/2022] Open
Abstract
Backgrounds Asthma and idiopathic pulmonary fibrosis (IPF) are common chronic diseases of the respiratory system in clinical practice. However, the relationship and molecular links remain unclear, and the current treatment’s efficacy is disappointing. Bu-Shen-Yi-Qi (BSYQ) decoction has proven effective in treating various chronic airway inflammatory diseases, including asthma and IPF. But the underlying pharmacological mechanisms are still to be elucidated. Methods This study searched the proteins related to asthma and IPF via TTD, CTD, and DisGeNET databases and then submitted to the STRING to establish the protein–protein interaction (PPI) network. The co-bioinformatics analysis was conducted by Metascape. The active ingredients of BSYQ decoction were screened from TCMSP, ETCM, BATMAN-TCM databases, and HPLC/MS experiment. The corresponding targets were predicted based on TCMSP, ETCM, and BATMAN-TCM databases. The shared targets for asthma and IPF treatment were recognized, and further GO and KEGG analyses were conducted with the DAVID platform. Finally, molecule docking via Autodock Vina was employed to predict the potential binding mode between core potential compounds and targets. Results Finally, 1333 asthma-related targets and 404 IPF-related proteins were retrieved, 120 were overlapped between them, and many of the asthma-related proteins fall into the same statistically significant GO terms with IPF. Moreover, 116 active ingredients of BSYQ decoction were acquired, and 1535 corresponding targets were retrieved. Eighty-three potential compounds and 56 potential targets were recognized for both asthma and IPF treatment. GO and KEGG analysis indicated that the inflammation response, cytokine production, leukocyte differentiation, oxygen level response, etc., were the common pathological processes in asthma and IPF, which were regulated by BSYQ decoction. Molecule docking further predicted the potential binding modes between the core potential compounds and targets. Conclusion The current study successfully clarified the complex molecule links between asthma and IPF and found the potential common targets. Then we demonstrated the efficacy of BSYQ decoction for asthma and IPF treatment from the angle of network pharmacology, which may provide valuable references for further studies and clinical use. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03637-7.
Collapse
|
57
|
Bai F, Chen Z, Xu S, Han L, Zeng X, Huang S, Zhu Z, Zhou L. Wogonin attenuates neutrophilic inflammation and airway smooth muscle proliferation through inducing caspase-dependent apoptosis and inhibiting MAPK/Akt signaling in allergic airways. Int Immunopharmacol 2022; 113:109410. [DOI: 10.1016/j.intimp.2022.109410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 11/13/2022]
|
58
|
Currie C, Framroze B, Singh D, Lea S, Bjerknes C, Hermansen E. Assessing the Anti-Inflammatory Effects of an Orally Dosed Enzymatically Liberated Fish Oil in a House Dust Model of Allergic Asthma. Biomedicines 2022; 10:biomedicines10102574. [PMID: 36289834 PMCID: PMC9599594 DOI: 10.3390/biomedicines10102574] [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: 08/28/2022] [Revised: 10/03/2022] [Accepted: 10/11/2022] [Indexed: 11/20/2022] Open
Abstract
Eosinophils are a major driver of inflammation in a number of human diseases, including asthma. Biologic therapies targeting IL-5 have enabled better control of severe eosinophilic asthma, but no such advances have been made for enhancing the control of moderate asthma. However, a number of moderate asthma sufferers remain troubled by unresolved symptoms, treatment side effects, or both. OmeGo, an enzymatically liberated fish oil, has demonstrated antioxidant and anti-inflammatory properties including the reduction of eosinophilia. A house dust mite model of induced asthma in mice was utilized in this study, and OmeGo showed a significant reduction in eosinophilic lung and systemic inflammation and reduced lung remodelling compared to cod liver oil. The CRTH2 antagonist fevipiprant showed an anti-inflammatory profile similar to that of OmeGo. OmeGo has the potential to be a pragmatic, cost-effective co-treatment for less severe forms of eosinophilic asthma. Proof-of-concept studies are planned.
Collapse
Affiliation(s)
- Crawford Currie
- Hofseth BioCare, Kipervikgata 13, 6003 Ålesund, Norway
- Correspondence:
| | - Bomi Framroze
- Hofseth BioCare, Kipervikgata 13, 6003 Ålesund, Norway
| | - Dave Singh
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PL, UK
- The Medicines Evaluation Unit, Manchester University NHS Foundation Trust, Manchester M23 9QZ, UK
| | - Simon Lea
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PL, UK
- The Medicines Evaluation Unit, Manchester University NHS Foundation Trust, Manchester M23 9QZ, UK
| | | | - Erland Hermansen
- Hofseth BioCare, Kipervikgata 13, 6003 Ålesund, Norway
- Department of Clinical Medicine, University of Bergen, 5007 Bergen, Norway
| |
Collapse
|
59
|
Masanam HB, Perumal G, Krishnan S, Singh SK, Jha NK, Chellappan DK, Dua K, Gupta PK, Narasimhan AK. Advances and opportunities in nanoimaging agents for the diagnosis of inflammatory lung diseases. Nanomedicine (Lond) 2022; 17:1981-2005. [PMID: 36695290 DOI: 10.2217/nnm-2021-0427] [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: 01/26/2023] Open
Abstract
The development of rapid, noninvasive diagnostics to detect lung diseases is a great need after the COVID-2019 outbreak. The nanotechnology-based approach has improved imaging and facilitates the early diagnosis of inflammatory lung diseases. The multifunctional properties of nanoprobes enable better spatial-temporal resolution and a high signal-to-noise ratio in imaging. Targeted nanoimaging agents have been used to bind specific tissues in inflammatory lungs for early-stage diagnosis. However, nanobased imaging approaches for inflammatory lung diseases are still in their infancy. This review provides a solution-focused approach to exploring medical imaging technologies and nanoprobes for the detection of inflammatory lung diseases. Prospects for the development of contrast agents for lung disease detection are also discussed.
Collapse
Affiliation(s)
- Hema Brindha Masanam
- Advanced Nano-Theranostics (ANTs), Biomaterials Lab, Department of Biomedical Engineering, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu, 603 203, India
| | - Govindaraj Perumal
- Department of Conservative Dentistry & Endodontics, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Velappanchavadi, Chennai, 600 077, India.,Department of Biomedical Engineering, Rajalakshmi Engineering College, Thandalam, Chennai, 602 105, India
| | | | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201310, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Bukit Jalil, Kuala Lumpur, 57000, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Sciences & Research (SBSR), Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh, 201310, India.,Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, 248002, India.,Faculty of Health and Life Sciences, INTI International University, Nilai 71800, Malaysia
| | - Ashwin Kumar Narasimhan
- Advanced Nano-Theranostics (ANTs), Biomaterials Lab, Department of Biomedical Engineering, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu, 603 203, India
| |
Collapse
|
60
|
Rui Y, Han X, Jiang A, Hu J, Li M, Liu B, Qian F, Huang L. Eucalyptol prevents bleomycin-induced pulmonary fibrosis and M2 macrophage polarization. Eur J Pharmacol 2022; 931:175184. [PMID: 35964659 DOI: 10.1016/j.ejphar.2022.175184] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 07/23/2022] [Accepted: 08/01/2022] [Indexed: 11/03/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrosing interstitial pneumonia with limited therapeutic options. Eucalyptol, a terpenoid oxide isolated from eucalyptus species, reportedly exhibits various biological activities such as anti-inflammatory and antioxidant effects. In the present study, we aimed to determine whether eucalyptol could alleviate bleomycin (BLM)-induced pulmonary fibrosis and inhibit interleukin (IL)-13-induced M2 macrophage polarization. Upon treatment with eucalyptol, BLM-induced pulmonary fibrosis and lung inflammation were significantly reduced. The pulmonary neutrophil accumulation and pulmonary permeability were inhibited and the expression of hydroxyproline, alpha-smooth muscle actin, and fibronectin was significantly down-regulated. Eucalyptol also markedly inhibited the expression of arginase-1, Ym-1, IL-13, and transforming growth factor (TGF)-β1, reduced the production of IL-13, IL-6, tumor necrosis factor (TNF)-α, and attenuated the activity of TGF-β1 in bronchoalveolar lavage fluid (BALF). Furthermore, the in vitro assay revealed that eucalyptol disturbed M2 macrophage polarization and reduced the macrophage-mediated secretion of the profibrotic factor TGF-β1. Eucalyptol inhibited the nuclear location of signal transducer and activator of transcription 6 (STAT6) and the phosphorylation of STAT6 and p38 mitogen-activated protein kinase (p38 MAPK), and reduced the expression of their downstream transcription factors, krupple-like factor 4 (KLF4) and peroxisome proliferator-activated receptor gamma (PPAR-γ). These findings indicated that eucalyptol alleviates BLM-induced pulmonary fibrosis by regulating M2 macrophage polarization, which, in turn, inhibits the activation of signaling molecules (e.g., STAT6 and p38 MAPK) and the expression of transcription factors (e.g., KLF4 and PPAR-γ). Thus, eucalyptol might be a potential therapeutic agent for IPF.
Collapse
Affiliation(s)
- Yan Rui
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, China; The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Department of Respiration and Critical Care Medicine, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Xiaojing Han
- Department of Clinical Laboratory Diagnostics, School of Laboratory Medicine, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Anbang Jiang
- Department of Respiration and Critical Care Medicine, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Junfeng Hu
- Department of Respiration and Critical Care Medicine, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Miao Li
- Department of General Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China
| | - Bangzhu Liu
- Department of Respiratory Medicine, The Second People's Hospital of Anhui, Wuhu, Anhui, 233000, China
| | - Feng Qian
- Department of Clinical Laboratory Diagnostics, School of Laboratory Medicine, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui, 233000, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 201100, China
| | - Linian Huang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, China; The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Department of Respiration and Critical Care Medicine, Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233000, China.
| |
Collapse
|
61
|
Singh S, Allwood BW, Chiyaka TL, Kleyhans L, Naidoo CC, Moodley S, Theron G, Segal LN. Immunologic and imaging signatures in post tuberculosis lung disease. Tuberculosis (Edinb) 2022; 136:102244. [PMID: 36007338 PMCID: PMC10061373 DOI: 10.1016/j.tube.2022.102244] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/24/2022] [Accepted: 07/31/2022] [Indexed: 11/25/2022]
Abstract
Post Tuberculosis Lung Disease (PTLD) affects millions of tuberculosis survivors and is a global health burden. The immune mechanisms that drive PTLD are complex and have historically been under investigated. Here, we discuss two immune-mediated paradigms that could drive human PTLD. We review the characteristics of a fibrotic granuloma that favors the development of PTLD via an abundance of T-helper-2 and T-regulatory cells and an upregulation of TGF-β mediated collagen deposition. Next, we discuss the post-primary tuberculosis paradigm and the complex mixture of caseous pneumonia, cavity formation and fibrosis that can also lead to PTLD. We review the delicate balance between cellular subsets and cytokines of the innate and adaptive immune system in conjunction with host-derived proteases that can perpetuate the parenchymal lung damage seen in PTLD. Next, we discuss the role of novel host directed therapies (HDT) to limit the development of PTLD and in particular, the recent repurposing of established medications such as statins, metformin and doxycycline. Finally, we review the emerging role of novel imaging techniques as a non-invasive modality for the early recognition of PTLD. While access to computed tomography imaging is unlikely to be available widely in countries with a high TB burden, its use in research settings can help phenotype PTLD. Due to a lack of disease-specific biomarkers and controlled clinical trials, there are currently no evidence-based recommendations for the management of PTLD. It is likely that an integrated antifibrotic strategy that could simultaneously target inflammatory and pro-fibrotic pathways will probably emerge as a successful way to treat this complex condition. In a disease spectrum as wide as PTLD, a single immunologic or radiographic marker may not be sufficient and a combination is more likely to be a successful surrogate that could aid in the development of successful HDTs.
Collapse
Affiliation(s)
- S Singh
- NYU Langone Translational Lung Biology Laboratory, Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York University School of Medicine, NYU Langone Health, 550 First Avenue, MSB 594, New York, NY, USA.
| | - B W Allwood
- Division of Pulmonology, Department of Medicine, Stellenbosch University & Tygerberg Hospital, South Africa.
| | - T L Chiyaka
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa.
| | - L Kleyhans
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa.
| | - C C Naidoo
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa.
| | - S Moodley
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa.
| | - G Theron
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa.
| | - L N Segal
- NYU Langone Translational Lung Biology Laboratory, Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York University School of Medicine, NYU Langone Health, 550 First Avenue, MSB 594, New York, NY, USA.
| |
Collapse
|
62
|
Nii T, Fukushima K, Kida H. Specific targeting of lung ILC2s via NRP1 in pulmonary fibrosis. Cell Mol Immunol 2022; 19:869-871. [PMID: 35459852 PMCID: PMC9338289 DOI: 10.1038/s41423-022-00867-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 11/09/2022] Open
Affiliation(s)
- Takuro Nii
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, 5-1-1 Toneyama, Osaka, Japan
| | - Kiyoharu Fukushima
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, 5-1-1 Toneyama, Osaka, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, Japan
- Laboratory of Host Defense, World Premier Institute Immunology Frontier Research Center (WPI-IFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka, Japan
- Department of Host Defense, Research Institute for Microbial Diseases (RIMD), Osaka University, Osaka, 565-0871, Japan
- Global Center for Medical Engineering and Informatics, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Hiroshi Kida
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, 5-1-1 Toneyama, Osaka, Japan.
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, Japan.
| |
Collapse
|
63
|
Kang Q, Li L, Pang Y, Zhu W, Meng L. An update on Ym1 and its immunoregulatory role in diseases. Front Immunol 2022; 13:891220. [PMID: 35967383 PMCID: PMC9366555 DOI: 10.3389/fimmu.2022.891220] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 07/06/2022] [Indexed: 11/23/2022] Open
Abstract
Ym1 is a rodent-specific chitinase-like protein (CLP) lacking catalytic activity, whose cellular origins are mainly macrophages, neutrophils and other cells. Although the detailed function of Ym1 remains poorly understood, Ym1 has been generally recognized as a fundamental feature of alternative activation of macrophages in mice and hence one of the prevalent detecting targets in macrophage phenotype distinguishment. Studies have pointed out that Ym1 may have regulatory effects, which are multifaceted and even contradictory, far more than just a mere marker. Allergic lung inflammation, parasite infection, autoimmune diseases, and central nervous system diseases have been found associations with Ym1 to varying degrees. Thus, insights into Ym1’s role in diseases would help us understand the pathogenesis of different diseases and clarify the genuine roles of CLPs in mammals. This review summarizes the information on Ym1 from the gene to its expression and regulation and focuses on the association between Ym1 and diseases.
Collapse
Affiliation(s)
- Qi Kang
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, China
- Department of Clinical Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Luyao Li
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, China
- Department of Clinical Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yucheng Pang
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, China
- Department of Clinical Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Wenhua Zhu
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, China
- *Correspondence: Wenhua Zhu, ; Liesu Meng,
| | - Liesu Meng
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, China
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Wenhua Zhu, ; Liesu Meng,
| |
Collapse
|
64
|
Kratzer B, Schlax LC, Gattinger P, Waidhofer‐Söllner P, Trapin D, Tauber PA, Sehgal ANA, Körmöczi U, Rottal A, Feichter M, Oberhofer T, Grabmeier‐Pfistershammer K, Borochova K, Dorofeeva Y, Tulaeva I, Weber M, Mühl B, Kropfmüller A, Negrin B, Kundi M, Valenta R, Pickl WF. Combined assessment of S- and N-specific IL-2 and IL-13 secretion and CD69 neo-expression for discrimination of post-infection and post-vaccination cellular SARS-CoV-2-specific immune response. Allergy 2022; 77:3408-3425. [PMID: 35690994 PMCID: PMC9348018 DOI: 10.1111/all.15406] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/20/2022] [Accepted: 05/30/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND Antibody-based tests are available for measuring SARS-CoV-2-specific immune responses but fast T-cell assays remain scarce. Robust T cell-based tests are needed to differentiate specific cellular immune responses after infection from those after vaccination. METHODS One hundred seventeen individuals (COVID-19 convalescent patients: n = 40; SARS-CoV-2 vaccinees: n = 41; healthy controls: n = 36) were evaluated for SARS-CoV-2-specific cellular immune responses (proliferation, Th1, Th2, Th17, and inflammatory cytokines, activation-induced marker [AIM] expression) by incubating purified peripheral blood mononuclear cells (PBMC) or whole blood (WB) with SARS-CoV-2 peptides (S, N, or M), vaccine antigens (tetanus toxoid, tick borne encephalitis virus) or polyclonal stimuli (Staphylococcal enterotoxin, phytohemagglutinin). RESULTS N-peptide mix stimulation of WB identified the combination of IL-2 and IL-13 secretion as superior to IFN-γ secretion to discriminate between COVID-19-convalescent patients and healthy controls (p < .0001). Comparable results were obtained with M- or S-peptides, the latter almost comparably recalled IL-2, IFN-γ, and IL-13 responses in WB of vaccinees. Analysis 10 months as opposed to 10 weeks after COVID-19, but not allergic disease status, positively correlated with IL-13 recall responses. WB cytokine responses correlated with cytokine and proliferation responses of PBMC. Antigen-induced neo-expression of the C-type lectin CD69 on CD4+ (p < .0001) and CD8+ (p = .0002) T cells informed best about the SARS-CoV-2 exposure status with additional benefit coming from CD25 upregulation. CONCLUSION Along with N- and S-peptide-induced IL-2 and CD69 neo-expression, we suggest to include the type 2 cytokine IL-13 as T-cellular recall marker for SARS-CoV-2 specific T-cellular immune responses after infection and vaccination.
Collapse
Affiliation(s)
- Bernhard Kratzer
- Medical University of ViennaCenter for PathophysiologyInfectiology and ImmunologyInstitute of ImmunologyViennaAustria
| | - Larissa C. Schlax
- Medical University of ViennaCenter for PathophysiologyInfectiology and ImmunologyInstitute of ImmunologyViennaAustria
| | - Pia Gattinger
- Medical University of ViennaCenter for Pathophysiology, Infectiology and ImmunologyDepartment of Pathophysiology and Allergy ResearchViennaAustria
| | - Petra Waidhofer‐Söllner
- Medical University of ViennaCenter for PathophysiologyInfectiology and ImmunologyInstitute of ImmunologyViennaAustria
| | - Doris Trapin
- Medical University of ViennaCenter for PathophysiologyInfectiology and ImmunologyInstitute of ImmunologyViennaAustria
| | - Peter A. Tauber
- Medical University of ViennaCenter for PathophysiologyInfectiology and ImmunologyInstitute of ImmunologyViennaAustria
| | - Al Nasar Ahmed Sehgal
- Medical University of ViennaCenter for PathophysiologyInfectiology and ImmunologyInstitute of ImmunologyViennaAustria
| | - Ulrike Körmöczi
- Medical University of ViennaCenter for PathophysiologyInfectiology and ImmunologyInstitute of ImmunologyViennaAustria
| | - Arno Rottal
- Medical University of ViennaCenter for PathophysiologyInfectiology and ImmunologyInstitute of ImmunologyViennaAustria
| | - Melanie Feichter
- Medical University of ViennaCenter for PathophysiologyInfectiology and ImmunologyInstitute of ImmunologyViennaAustria
| | - Teresa Oberhofer
- Medical University of ViennaCenter for PathophysiologyInfectiology and ImmunologyInstitute of ImmunologyViennaAustria
| | | | - Kristina Borochova
- Medical University of ViennaCenter for Pathophysiology, Infectiology and ImmunologyDepartment of Pathophysiology and Allergy ResearchViennaAustria
| | - Yulia Dorofeeva
- Medical University of ViennaCenter for Pathophysiology, Infectiology and ImmunologyDepartment of Pathophysiology and Allergy ResearchViennaAustria
| | - Inna Tulaeva
- Medical University of ViennaCenter for Pathophysiology, Infectiology and ImmunologyDepartment of Pathophysiology and Allergy ResearchViennaAustria,I. M. Sechenov First Moscow State Medical University (Sechenov University)Department of Clinical Immunology and AllergologyLaboratory for ImmunopathologyMoscowRussia
| | - Milena Weber
- Medical University of ViennaCenter for Pathophysiology, Infectiology and ImmunologyDepartment of Pathophysiology and Allergy ResearchViennaAustria
| | | | | | - Bettina Negrin
- Österreichische Gesundheitskasse, Klinikum PeterhofBadenAustria
| | - Michael Kundi
- Medical University of ViennaCenter for Public HealthDepartment for Environmental HealthViennaAustria
| | - Rudolf Valenta
- Medical University of ViennaCenter for Pathophysiology, Infectiology and ImmunologyDepartment of Pathophysiology and Allergy ResearchViennaAustria,I. M. Sechenov First Moscow State Medical University (Sechenov University)Department of Clinical Immunology and AllergologyLaboratory for ImmunopathologyMoscowRussia,NRC Institute of Immunology FMBA of RussiaMoscowRussia,Karl Landsteiner University of Health SciencesKremsAustria
| | - Winfried F. Pickl
- Medical University of ViennaCenter for PathophysiologyInfectiology and ImmunologyInstitute of ImmunologyViennaAustria,Karl Landsteiner University of Health SciencesKremsAustria
| |
Collapse
|
65
|
Sun YB, Ni Y, Fan XS, Zhou LP, Yue QF, Shang EX. Effect of Houpo-Mahuang Decoction on aggravated asthma induced by cigarette smoke and the expression of TRPA1 and tight junctions in mice. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115217. [PMID: 35337920 DOI: 10.1016/j.jep.2022.115217] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/07/2022] [Accepted: 03/19/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cigarette smoke (CS) is a common environmental irritant and a risk factor for asthma, as it induces as well as aggravates asthmatic attacks. The injured airway epithelial tight junctions (TJs) aggravate asthma. CS can aggravate asthma by activating the transient receptor potential ankyrin A1 (TRPA1) channel and enhancing TJs destruction. Houpo Mahuang decoction (HPMHD) is a classic traditional Chinese prescription for the treatment of asthma. However, its underlying action mechanism is unclear. AIM OF THE STUDY The present study aimed to evaluate the effect of HPMHD on the asthma phenotype and the regulation of TRPA1 and TJs in a CS-induced mouse model of aggravated asthma. MATERIALS AND METHODS Under optimized chromatographic and mass spectrometry conditions, the ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS) technique was used to detect and analyze the major chemical components of HPMHD. C57BL/6 female mice were randomly divided into seven groups, viz, normal saline (NS) group, ovalbumin (OVA) + CS group, dexamethasone group, HPMHD high-dose group and low-dose groups, n-butanol extract group, and ethyl acetate extract group, with 10 mice in each group. OVA sensitization and challenge, and CS exposure were used to establish the aggravated asthma model. As the main indices to evaluate the protective effect of HPMHD, the eosinophils count in peripheral blood, percentages of inflammatory cells classified and the levels of interleukin (IL)-4, IL-5, IL-13 in the bronchoalveolar lavage fluid (BALF), airway responsiveness enhanced pause (Penh), and changes in lung histopathology were determined and compared among the groups. The mRNA and protein expression of TRPA1 and TJs in lung tissue was also examined. RESULTS Using UPLC-QTOF-MS, the chemical components of HPMHD, including ephedrine, pseudoephedrine, laetrile, and amygdalin amide, were identified by 51 signal peaks. Compared with those in the NS group, the eosinophil number in the peripheral blood and the eosinophils and neutrophils percentages in BALF of the OVA + CS group were remarkably increased. Following the inhalation of 50 μl of acetylcholine chloride (ACH) at doses of 25 and 50 mg/mL, the Penh increased significantly (p < 0.01). Moreover, in the OVA + CS group, hematoxylin and eosin (H&E) staining of lung tissue showed a significant number of infiltrated inflammatory cells, increased mucus secretion in the lumen, damaged bronchial mucosa, increased thickness of tracheal wall, and increased score of lung damage (p < 0.01). The IL-4/5/13 levels were also remarkably increased (p < 0.01). The protein as well as gene expression of both ZO-1 and occludin decreased markedly in the lung tissue, while the expression of TRPA1 and claudin-2 was increased (p < 0.05, p < 0.01). Next, the OVA + CS group and the treatment groups were compared. The inflammatory cells, Penh value, and levels of IL-4/5/13 were significantly reduced, and less lung injury was observed in the treatment groups. The gene and protein levels of TRPA1 and TJs were corrected (p < 0.05, p < 0.01); the effects on the HPMHD high-dose and ethyl acetate extract groups were particularly remarkable. CONCLUSIONS HPMHD reduced airway hyperresponsiveness, inflammatory cell recruitment and Th2 cytokine secretion in CS-induced aggravated asthma mice, in a manner potentially dependent on regulation of the expression of TRPA1 and TJ proteins. Both the n-butanol and ethyl acetate extracts contained the active ingredients, especially the ethyl acetate extract.
Collapse
Affiliation(s)
- Yu-Bo Sun
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ying Ni
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xin-Sheng Fan
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Li-Ping Zhou
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qin-Fei Yue
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Er-Xin Shang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| |
Collapse
|
66
|
Codonopsis laceolata Water Extract Ameliorates Asthma Severity by Inducing Th2 Cells’ and Pulmonary Epithelial Cells’ Apoptosis via NF-κB/COX-2 Pathway. Processes (Basel) 2022. [DOI: 10.3390/pr10071249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2022] Open
Abstract
Asthma is an incurable pulmonary disease with several symptoms, including abnormal breathing, coughing, and sleep apnea, which can lead to death, and the population of asthma patients has been increasing worldwide. There are many adverse effects in current drugs, and thus, we have tried to develop anti-asthmatic agents from natural products such as Codonopsis laceolata. To define the anti-asthmatic effect and the mechanism of Codonopsis laceolata, an animal study was conducted considering different cell counts of BALF, serum IgE levels, morphological changes in the pulmonary system, the Th2 cell transcription factor (GATA-3), and the apoptotic pathway (NF-κB/COX-2). Codonopsis laceolata significantly suppressed the representative asthmatic changes, such as airway remodeling, mucous hypersecretion, epithelial hyperplasia, and inflammatory cell infiltration, in the respiratory system. It suppressed the levels of GATA-3, IL-4, and IL-13. The down-regulation of Th2-related factors, such as GATA-3, IL-4, and IL-13, results from the stimulated apoptosis of Th2 cells and epithelial cells via a decrease in the levels of NF-κB and COX-2. We concluded that Codonopsis laceolata might be a promising anti-asthmatic drug.
Collapse
|
67
|
Rossi GA, Ballarini S, Salvati P, Sacco O, Colin AA. Alarmins and innate lymphoid cells 2 activation: A common pathogenetic link connecting respiratory syncytial virus bronchiolitis and later wheezing/asthma? Pediatr Allergy Immunol 2022; 33:e13803. [PMID: 35754131 DOI: 10.1111/pai.13803] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 12/21/2022]
Abstract
Severe respiratory syncytial virus (RSV) infection in infancy is associated with increased risk of recurrent wheezing in childhood. Both acute and long-term alterations in airway functions are thought to be related to inefficient antiviral immune response. The airway epithelium, the first target of RSV, normally acts as an immunological barrier able to elicit an effective immune reaction but may also be programmed to directly promote a Th2 response, independently from Th2 lymphocyte involvement. Recognition of RSV transcripts and viral replication intermediates by bronchial epithelial cells brings about release of TSLP, IL-33, HMGB1, and IL-25, dubbed "alarmins." These epithelial cell-derived proteins are particularly effective in stimulating innate lymphoid cells 2 (ILC2) to release IL-4, IL-5, and IL-13. ILC2, reflect the innate counterparts of Th2 cells and, when activate, are potent promoters of airway inflammation and hyperresponsiveness in RSV bronchiolitis and childhood wheezing/asthma. Long-term epithelial progenitors or persistent epigenetic modifications of the airway epithelium following RSV infection may play a pathogenetic role in the short- and long-term increased susceptibility to obstructive lung diseases in response to RSV in the young. Additionally, ILC2 function may be further regulated by RSV-induced changes in gut microbiota community composition that can be associated with disease severity in infants. A better understanding of the alarmin-ILC interactions in childhood might provide insights into the mechanisms characterizing these immune-mediated diseases and indicate new targets for prevention and therapeutic interventions.
Collapse
Affiliation(s)
- Giovanni A Rossi
- Department of Pediatrics, Pediatric Pulmonology and Respiratory Endoscopy Unit, G. Gaslini institute and University Hospital, Genoa, Italy
| | - Stefania Ballarini
- Department of Medicine and Surgery, Section of Immunometabolism, Immunogenetics and Translational Immunology, University of Perugia, Perugia, Italy
| | - Pietro Salvati
- Department of Pediatrics, Pediatric Pulmonology and Respiratory Endoscopy Unit, G. Gaslini institute and University Hospital, Genoa, Italy
| | - Oliviero Sacco
- Department of Pediatrics, Pediatric Pulmonology and Respiratory Endoscopy Unit, G. Gaslini institute and University Hospital, Genoa, Italy
| | - Andrew A Colin
- Division of Pediatric Pulmonology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
68
|
PM2.5 Exposure and Asthma Development: The Key Role of Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3618806. [PMID: 35419163 PMCID: PMC9001082 DOI: 10.1155/2022/3618806] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/24/2022] [Indexed: 12/21/2022]
Abstract
Oxidative stress is defined as the imbalance between reactive oxygen species (ROS) production and the endogenous antioxidant defense system, leading to cellular damage. Asthma is a common chronic inflammatory airway disease. The presence of asthma tends to increase the production of reactive oxygen species (ROS), and the antioxidant system in the lungs is insufficient to mitigate it. Therefore, asthma can lead to an exacerbation of airway hyperresponsiveness and airway inflammation. PM2.5 exposure increases ROS levels. Meanwhile, the accumulation of ROS will further enhance the oxidative stress response, resulting in DNA, protein, lipid, and other cellular and molecular damage, leading to respiratory diseases. An in-depth study on the relationship between oxidative stress and PM2.5-related asthma is helpful to understand the pathogenesis and progression of the disease and provides a new direction for the treatment of the disease. This paper reviews the research progress of oxidative stress in PM2.5-induced asthma as well as highlights the therapeutic potentials of antioxidant approaches in treatment of asthma.
Collapse
|
69
|
Guilleminault L, Conde E, Reber LL. Pharmacological approaches to target type 2 cytokines in asthma. Pharmacol Ther 2022; 237:108167. [PMID: 35283171 DOI: 10.1016/j.pharmthera.2022.108167] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/17/2022] [Accepted: 03/07/2022] [Indexed: 02/06/2023]
Abstract
Asthma is the most common chronic lung disease, affecting more than 250 million people worldwide. The heterogeneity of asthma phenotypes represents a challenge for adequate assessment and treatment of the disease. However, approximately 50% of asthma patients present with chronic type 2 inflammation initiated by alarmins, such as IL-33 and thymic stromal lymphopoietin (TSLP), and driven by the TH2 interleukins IL-4, IL-5 and IL-13. These cytokines have therefore become important therapeutic targets in asthma. Here, we discuss current knowledge on the structure and functions of these cytokines in asthma. We review preclinical and clinical data obtained with monoclonal antibodies (mAbs) targeting these cytokines or their receptors, as well as novel strategies under development, including bispecific mAbs, designed ankyrin repeat proteins (DARPins), small molecule inhibitors and vaccines targeting type 2 cytokines.
Collapse
Affiliation(s)
- Laurent Guilleminault
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, 31024 Toulouse, France; Department of Respiratory Medicine, Toulouse University Hospital, Faculty of Medicine, Toulouse, France
| | - Eva Conde
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR 1222 INSERM, F-75015 Paris, France; Sorbonne University, ED394, F-75005 Paris, France
| | - Laurent L Reber
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, 31024 Toulouse, France.
| |
Collapse
|
70
|
Bamias G, Pizarro TT, Cominelli F. Immunological Regulation of Intestinal Fibrosis in Inflammatory Bowel Disease. Inflamm Bowel Dis 2022; 28:337-349. [PMID: 34904152 PMCID: PMC8919810 DOI: 10.1093/ibd/izab251] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Indexed: 02/06/2023]
Abstract
Intestinal fibrosis is a late-stage phenotype of inflammatory bowel disease (IBD), which underlies most of the long-term complications and surgical interventions in patients, particularly those with Crohn's disease. Despite these issues, antifibrotic therapies are still scarce, mainly due to the current lack of understanding concerning the pathogenetic mechanisms that mediate fibrogenesis in patients with chronic intestinal inflammation. In the current review, we summarize recent evidence regarding the cellular and molecular factors of innate and adaptive immunity that are considered critical for the initiation and amplification of extracellular matrix deposition and stricture formation. We focus on the role of cytokines by dissecting the pro- vs antifibrotic components of the immune response, while taking into consideration their temporal association to the progressive stages of the natural history of IBD. We critically present evidence from animal models of intestinal fibrosis and analyze inflammation-fibrosis interactions that occur under such experimental scenarios. In addition, we comment on recent findings from large-scale, single-cell profiling of fibrosis-relevant populations in IBD patients. Based on such evidence, we propose future potential targets for antifibrotic therapies to treat patients with IBD.
Collapse
Affiliation(s)
- Giorgos Bamias
- Gastrointestinal Unit, Third Academic Department of Internal Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, Athens, Greece
| | - Theresa T Pizarro
- Departments of Pathology and Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Fabio Cominelli
- Departments of Pathology and Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
71
|
He J, Du Y, Li G, Xiao P, Sun X, Song W, Lai L, Xia M, Zhang J, Wang Q. Myeloid Fbxw7 Prevents Pulmonary Fibrosis by Suppressing TGF-β Production. Front Immunol 2022; 12:760138. [PMID: 35069531 PMCID: PMC8767095 DOI: 10.3389/fimmu.2021.760138] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a group of chronic interstitial pulmonary diseases characterized by an inexorable decline in lung function with limited treatment options. The abnormal expression of transforming growth factor-β (TGF-β) in profibrotic macrophages is linked to severe pulmonary fibrosis, but the regulation mechanisms of TGF-β expression are incompletely understood. We found that decreased expression of E3 ubiquitin ligase Fbxw7 in peripheral blood mononuclear cells (PBMCs) was significantly related to the severity of pulmonary fibrosis in IPF patients. Fbxw7 is identified to be a crucial suppressing factor for pulmonary fibrosis development and progression in a mouse model induced by intratracheal bleomycin treatment. Myeloid cell-specific Fbxw7 deletion increases pulmonary monocyte-macrophages accumulation in lung tissue, and eventually promotes bleomycin-induced collagen deposition and progressive pulmonary fibrosis. Notably, the expression of TGF-β in profibrotic macrophages was significantly upregulated in myeloid cell-specific Fbxw7 deletion mice after bleomycin treatment. C-Jun has long been regarded as a critical transcription factor of Tgfb1, we clarified that Fbxw7 inhibits the expression of TGF-β in profibrotic macrophages by interacting with c-Jun and mediating its K48-linked ubiquitination and degradation. These findings provide insight into the role of Fbxw7 in the regulation of macrophages during the pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Jia He
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China
| | - Yue Du
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China
| | - Gaopeng Li
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China
| | - Peng Xiao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China
| | - Xingzheng Sun
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China
| | - Wenjun Song
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China
| | - Lihua Lai
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China
| | - Meng Xia
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China
| | - Jianhua Zhang
- Department of Medical Laboratory, School of Medicine, Shaoxing University, Shaoxing, China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.,The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
72
|
Hasan M, Paul NC, Paul SK, Saikat ASM, Akter H, Mandal M, Lee SS. Natural Product-Based Potential Therapeutic Interventions of Pulmonary Fibrosis. Molecules 2022; 27:1481. [PMID: 35268581 PMCID: PMC8911636 DOI: 10.3390/molecules27051481] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 11/16/2022] Open
Abstract
Pulmonary fibrosis (PF) is a disease-refractive lung condition with an increased rate of mortality. The potential factors causing PF include viral infections, radiation exposure, and toxic airborne chemicals. Idiopathic PF (IPF) is related to pneumonia affecting the elderly and is characterized by recurring scar formation in the lungs. An impaired wound healing process, defined by the dysregulated aggregation of extracellular matrix components, triggers fibrotic scar formation in the lungs. The potential pathogenesis includes oxidative stress, altered cell signaling, inflammation, etc. Nintedanib and pirfenidone have been approved with a conditional endorsement for the management of IPF. In addition, natural product-based treatment strategies have shown promising results in treating PF. In this study, we reviewed the recently published literature and discussed the potential uses of natural products, classified into three types-isolated active compounds, crude extracts of plants, and traditional medicine, consisting of mixtures of different plant products-in treating PF. These natural products are promising in the treatment of PF via inhibiting inflammation, oxidative stress, and endothelial mesenchymal transition, as well as affecting TGF-β-mediated cell signaling, etc. Based on the current review, we have revealed the signaling mechanisms of PF pathogenesis and the potential opportunities offered by natural product-based medicine in treating PF.
Collapse
Affiliation(s)
- Mahbub Hasan
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka 8100, Bangladesh; (N.C.P.); (S.K.P.); (A.S.M.S.); (M.M.)
- Department of Oriental Biomedical Engineering, College of Health Sciences, Sangji University, Wonju 26339, Korea
| | - Nidhan Chandra Paul
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka 8100, Bangladesh; (N.C.P.); (S.K.P.); (A.S.M.S.); (M.M.)
| | - Shamrat Kumar Paul
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka 8100, Bangladesh; (N.C.P.); (S.K.P.); (A.S.M.S.); (M.M.)
| | - Abu Saim Mohammad Saikat
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka 8100, Bangladesh; (N.C.P.); (S.K.P.); (A.S.M.S.); (M.M.)
| | - Hafeza Akter
- Pharmacology and Toxicology Research Division, Health Medical Science Research Foundation, Dhaka 1207, Bangladesh;
| | - Manoj Mandal
- Department of Biochemistry and Molecular Biology, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka 8100, Bangladesh; (N.C.P.); (S.K.P.); (A.S.M.S.); (M.M.)
| | - Sang-Suk Lee
- Department of Oriental Biomedical Engineering, College of Health Sciences, Sangji University, Wonju 26339, Korea
| |
Collapse
|
73
|
Fu HT, Zhang Y, Zhang P, Wu H, Sun XQ, Shen SY, Dou DB. Tumor necrosis factor-α promotes airway mucus hypersecretion by repressing miR-146a-5p and miR-134-5p levels in human airway epithelial cells. Transl Cancer Res 2022; 10:4047-4056. [PMID: 35116702 PMCID: PMC8797934 DOI: 10.21037/tcr-20-3375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 07/14/2021] [Indexed: 12/12/2022]
Abstract
Background Airway mucus acts as an indispensable protective component of innate immune response against invading pathogens. However, airway mucus hypersecretion, largely consisting of mucin 5AC (MUC5AC), is the leading cause of airflow obstruction and airway hyperresponsiveness that contributes to chronic obstructive pulmonary disease (COPD). MicroRNAs (miRNAs) are frequently dysregulated in the pathogenesis of COPD, but the definite role of miRNAs in airway mucus hypersecretion is not well understood. Methods A cell model of mucus hypersecretion was established in 16HBE cells by treatment with TNF-α. Cell viability and apoptosis were assessed using cell counting kit-8 (CCK-8) and flow cytometry, respectively. The aberrant expression of miR-146a-5p and miR-134-5p was assayed in TNF-α-treated 16HBE cells, and the effect of miR-146a-5p and miR-134-5p on regulating MUC5AC expression was evaluated using quantitative real-time PCR (qPCR) and Western blot analysis. Results TNF-α treatment resulted in a significant decrease of cell viability, and increase of cell apoptosis and MUC5AC expression in 16HBE cells. Additionally, the expression of miR-134-5p and miR-146a-5p was markedly decreased in the cell model. Importantly, forced expression of miR-134-5p and miR-146a-5p significantly repressed TNF-α-induced upregulation of MUC5AC. Mechanistically, although miR-134-5p did not affect 16HBE cells viability and apoptosis, miR-134-5p partially blocked TNF-α-induced MUC5AC expression by inhibiting the activation of NF-κB signaling. On the other hand, miR-146a-5p enhanced cell viability and reduced cell apoptosis. miR-146a-5p also repressed TNF-α-induced MUC5AC expression by inhibiting p38 MAPK (mitogen-activated protein kinase) signaling activation. Conclusions The current data demonstrated that both miR-134-5p and miR-146a-5p conferred protection against TNF-α-induced mucus hypersecretion through repressing NF-κB and p38 MAPK signaling, indicating that miR-134-5p and miR-146a-5p may serve as the biomarker for COPD.
Collapse
Affiliation(s)
- Hui-Ting Fu
- Department of Traditional Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Zhang
- Department of Ultrasound, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Zhang
- Department of Traditional Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huan Wu
- Department of Traditional Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuan-Qiu Sun
- Department of Traditional Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shu-Yang Shen
- Department of Traditional Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan-Bo Dou
- Department of Traditional Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
74
|
Mazeraud A, Jamme M, Mancusi RL, Latroche C, Megarbane B, Siami S, Zarka J, Moneger G, Santoli F, Argaud L, Chillet P, Muller G, Bruel C, Asfar P, Beloncle F, Reignier J, Vinsonneau C, Schimpf C, Amour J, Goulenok C, Lemaitre C, Rohaut B, Mateu P, De Rudnicki S, Mourvillier B, Declercq PL, Schwebel C, Stoclin A, Garnier M, Madeux B, Gaudry S, Bailly K, Lamer C, Aegerter P, Rieu C, Sylla K, Lucas B, Sharshar T. Intravenous immunoglobulins in patients with COVID-19-associated moderate-to-severe acute respiratory distress syndrome (ICAR): multicentre, double-blind, placebo-controlled, phase 3 trial. THE LANCET. RESPIRATORY MEDICINE 2022; 10:158-166. [PMID: 34774185 PMCID: PMC8585489 DOI: 10.1016/s2213-2600(21)00440-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/26/2021] [Accepted: 09/13/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a major complication of COVID-19 and is associated with high mortality and morbidity. We aimed to assess whether intravenous immunoglobulins (IVIG) could improve outcomes by reducing inflammation-mediated lung injury. METHODS In this multicentre, double-blind, placebo-controlled trial, done at 43 centres in France, we randomly assigned patients (1:1) receiving invasive mechanical ventilation for up to 72 h with PCR confirmed COVID-19 and associated moderate-to-severe ARDS to receive either IVIG (2 g/kg over 4 days) or placebo. Random assignment was done with a web-based system and was stratified according to the participating centre and the duration of invasive mechanical ventilation before inclusion in the trial (<12 h, 12-24 h, and >24-72 h), and treatment was administered within the first 96 h of invasive mechanical ventilation. To minimise the risk of adverse events, the IVIG administration was divided into four perfusions of 0·5 g/kg each administered over at least 8 hours. Patients in the placebo group received an equivalent volume of sodium chloride 0·9% (10 mL/kg) over the same period. The primary outcome was the number of ventilation-free days by day 28, assessed according to the intention-to-treat principle. This trial was registered on ClinicalTrials.gov, NCT04350580. FINDINGS Between April 3, and October 20, 2020, 146 patients (43 [29%] women) were eligible for inclusion and randomly assigned: 69 (47%) patients to the IVIG group and 77 (53%) to the placebo group. The intention-to-treat analysis showed no statistical difference in the median number of ventilation-free days at day 28 between the IVIG group (0·0 [IQR 0·0-8·0]) and the placebo group (0·0 [0·0-6·0]; difference estimate 0·0 [0·0-0·0]; p=0·21). Serious adverse events were more frequent in the IVIG group (78 events in 22 [32%] patients) than in the placebo group (47 events in 15 [20%] patients; p=0·089). INTERPRETATION In patients with COVID-19 who received invasive mechanical ventilation for moderate-to-severe ARDS, IVIG did not improve clinical outcomes at day 28 and tended to be associated with an increased frequency of serious adverse events, although not significant. The effect of IVIGs on earlier disease stages of COVID-19 should be assessed in future trials. FUNDING Programme Hospitalier de Recherche Clinique.
Collapse
Affiliation(s)
- Aurélien Mazeraud
- Service d'Anesthésie-Réanimation, Groupe Hospitalier Université Paris Psychiatrie et Neurosciences, Pôle Neuro, Paris, France; Société Française d'Anesthésie-Réanimation Research Network, France; Department of Neurosiences, Université de Paris, Paris, France.
| | - Matthieu Jamme
- Service de Réanimation Polyvalente, Centre Hospitalier Intercommunal de Poissy Saint Germain en Laye, Poissy, France; INSERM U1018, CESP, Équipe Epidémiologie Clinique, Université Paris Saclay, Villejuif, France
| | - Rossella Letizia Mancusi
- Direction de la recherche clinique et de l'innovation, Groupe Hospitalier Universitaire Paris Psychiatrie et Neurosciences, Paris, France
| | - Claire Latroche
- Service d'Anesthésie-Réanimation, Groupe Hospitalier Université Paris Psychiatrie et Neurosciences, Pôle Neuro, Paris, France; Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016, Paris, France
| | - Bruno Megarbane
- Department of Neurosiences, Université de Paris, Paris, France; Service de Médecine Intensive et Réanimation, Centre Hospitalo-universitaire Lariboisière Paris, France
| | - Shidasp Siami
- Service de Réanimation Polyvalente, Centre Hospitalier Sud-Essonnes, Etampes, France
| | - Jonathan Zarka
- Service de Médecine Intensive et Réanimation, Grand hôpital de l'Est francilien site Marne-la-Vallée, Marne-la-Vallée, France
| | - Guy Moneger
- Service de Réanimation polyvalente, Hôpital Nord Franche Comté, Trevenans, France
| | - Francesco Santoli
- Service de Réanimation Médicale, Centre Hospitalo-universitaire Robert Ballanger, Aulnay, France
| | - Laurent Argaud
- Service de Médecine Intensive-Réanimation, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Patrick Chillet
- Service de Réanimation Polyvalente, Centre Hospitalier Chalons en Champagne, Chalons en Champagne, France
| | - Gregoire Muller
- Service de Médecine Intensive et Réanimation, Centre hospitalier, Orléans, France; Clinical Research in Intensive Care and Sepsis - TRIal Group for global Evaluation and Research in Sepsis research network, Tours, France
| | - Cedric Bruel
- Service de Réanimation Médico-chirurgicale, Groupe Hospitalier Paris Saint-Joseph, Paris, France
| | - Pierre Asfar
- Service de Médecine Intensive Réanimation, Centre Hospitalo-Universitaire d'Angers, Angers, France
| | - Francois Beloncle
- Service de Médecine Intensive Réanimation, Centre Hospitalo-Universitaire d'Angers, Angers, France
| | - Jean Reignier
- Service de Médecine Intensive Réanimation, Centre Hospitalier Universitaire de Nantes, France
| | - Christophe Vinsonneau
- Service de Médecine Intensive Réanimation, Centre Hospitalier de Béthune, Béthune, France
| | - Caroline Schimpf
- Service d'Anesthésie-Réanimation, Groupe Hospitalier Université Paris Psychiatrie et Neurosciences, Pôle Neuro, Paris, France
| | - Julien Amour
- Institute of Perfusion, Critical Care Medicine and Anesthesiology in Cardiac Surgery, Ramsay Health Care, Hôpital Privé Jacques Cartier, Massy, France
| | - Cyril Goulenok
- Intensive Care Unit, Ramsay Générale de Santé, Hôpital Privé Jacques Cartier, Massy, France
| | - Caroline Lemaitre
- Département de Gastroentérologie et Hépatologie, Hôpital Jacques Monod, Montivilliers, France; Département de Médecine Intensive et Réanimation, Hôpital Jacques Monod, Avenue Pierre Mendès France, Montivilliers, France
| | - Benjamin Rohaut
- Département de Neurologie, Neurointensive care unit, Assistance Publique -Hopitaux de Paris-Pitié Salpêtrière, Paris, France; Department of Neurology, Sorbonne Université, Paris, France; Institut du Cerveau-Paris Brain Institute-Institut du Cerveau et de la Moelle, Paris, France; Pinic Lab, INSERM, Paris, France; Centre National de la Recherche Scientifique, Paris, France
| | - Philippe Mateu
- Service de Réanimation polyvalente, Centre Hospitalier Interrégional Nord Ardennes, Charleville-Mézières, France
| | - Stephane De Rudnicki
- Service d'Anesthésie Réanimation, Hôpital d'instruction des Armées de Percy, Clamart, France
| | - Bruno Mourvillier
- Service de Médecine Intensive et Réanimation Polyvalente, Centre Hospitalo-Universitaire Robert Debré, Reims, France
| | | | - Carole Schwebel
- Service de Médecine Intensive et Réanimation, Centre Hospitalo-Universitaire de Grenoble Alpe, Grenoble, France
| | | | - Marc Garnier
- Service d'Anesthésie-Réanimation, Centre Hospitalo-Universitaire Saint-Antoine, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Benjamin Madeux
- Service de Réanimation Polyvalente, Centre Hospitalier Intercommunal de Poissy Saint Germain en Laye, Poissy, France; Service de Médecine Intensive et Réanimation, Centre Hospitalier De Tarbes, Tarbes, France
| | - Stéphane Gaudry
- Service de Médecine Intensive et Réanimation Centre Hospitalo-Universitaire Avicenne, Bobigny, France
| | - Karine Bailly
- Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016, Paris, France
| | - Christian Lamer
- Service de Réanimation Polyvalente, Institut mutualiste Montsouris, Paris, France
| | - Philippe Aegerter
- Groupement inter-régional de recherche clinique et d'innovation - Île de France, Cellule Méthodologie, Paris, France; Équipe d'Épidémiologie respiratoire intégrative, Centre de recherche en Epidémiologie et Santé des Populations, U1018 INSERM Université Paris Saclay - Université Versailles Saint Quentin en Yveline, Villejuif, France
| | - Christine Rieu
- Service d'Anesthésie-Réanimation, Groupe Hospitalier Université Paris Psychiatrie et Neurosciences, Pôle Neuro, Paris, France
| | - Khaoussou Sylla
- Direction de la recherche clinique et de l'innovation, Groupe Hospitalier Universitaire Paris Psychiatrie et Neurosciences, Paris, France
| | - Bruno Lucas
- Department of Neurosiences, Université de Paris, Paris, France; Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016, Paris, France
| | - Tarek Sharshar
- Service d'Anesthésie-Réanimation, Groupe Hospitalier Université Paris Psychiatrie et Neurosciences, Pôle Neuro, Paris, France; Department of Neurosiences, Université de Paris, Paris, France; INSERM UMR S894, Sorbonne Université, Paris, France
| |
Collapse
|
75
|
Frøssing L, Hvidtfeldt M, Silberbrandt A, Sverrild A, Porsbjerg C. Missing sputum samples are common in asthma intervention studies and successful collection at follow-up is related to improvement in clinical outcomes. ERJ Open Res 2022; 8:00612-2021. [PMID: 35141327 PMCID: PMC8819258 DOI: 10.1183/23120541.00612-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/21/2021] [Indexed: 11/23/2022] Open
Abstract
With only modest agreement between airway and systemic eosinophilia, biomarkers directly assessing the level and type of airway inflammation are becoming increasingly important, both for targeting treatment to the individual patient and for assessing effect [1]. Several factors significantly impact ability to produce a sputum sample after an anti-inflammatory intervention and these authors argue that the widely used complete-case analysis is inappropriate for paired sputum-based outcome measureshttps://bit.ly/3qN2pk5
Collapse
|
76
|
Yue L, Dong J, Wang J, Yin X, Ren X, Xu O, Shan C. High mobility group box 1 inhibition by BoxA attenuates ovalbumin-induced allergic rhinitis in mice. Autoimmunity 2022; 55:118-126. [PMID: 35007167 DOI: 10.1080/08916934.2021.2021512] [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: 11/02/2022]
Abstract
This study was designed to evaluate the effects of BoxA on allergic rhinitis (AR). Ovalbumin (OVA)-induced AR mice model was employed and BoxA was administered to AR mice. AR symptoms, levels of cytokines and chemokines, and the expression of high mobility group box 1 (HMGB1), TLR2, and TLR4 were measured. BoxA treatment significantly ameliorated AR symptoms, decreased level of histamine, OVA-specific antibodies, suppressed the infiltration of immune cells in nasal tissues, inhibited the expression of IL-4, IL-6, IL-5, TNF-α, IL-13, IL-17, IL-2 while promoting the expression of IL-10, suppressed the expression of HMGB1, TLR2, and TLR4 in AR mice. BoxA ameliorated allergic rhinitis in mice by inhibiting HMGB1.
Collapse
Affiliation(s)
- Liyan Yue
- ENT Department 1, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jinhui Dong
- ENT Department 1, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jianxing Wang
- ENT Department 1, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoyan Yin
- ENT Department 1, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiumin Ren
- ENT Department 1, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ou Xu
- ENT Department 1, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chunguang Shan
- ENT Department 1, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
77
|
Mo Y, Kim Y, Bang JY, Jung J, Lee CG, Elias JA, Kang HR. Mesenchymal Stem Cells Attenuate Asthmatic Inflammation and Airway Remodeling by Modulating Macrophages/Monocytes in the IL-13-Overexpressing Mouse Model. Immune Netw 2022; 22:e40. [DOI: 10.4110/in.2022.22.e40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yosep Mo
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yujin Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Ji-Young Bang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jiung Jung
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Chun-Geun Lee
- Brown University, Molecular Microbiology and Immunology, Providence, Rhode Island, United States
| | - Jack A. Elias
- Brown University, Molecular Microbiology and Immunology, Providence, Rhode Island, United States
| | - Hye-Ryun Kang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
78
|
Jaiswal AK, Yadav J, Makhija S, Mazumder S, Mitra AK, Suryawanshi A, Sandey M, Mishra A. Irg1/itaconate metabolic pathway is a crucial determinant of dendritic cells immune-priming function and contributes to resolute allergen-induced airway inflammation. Mucosal Immunol 2022; 15:301-313. [PMID: 34671116 PMCID: PMC8866123 DOI: 10.1038/s41385-021-00462-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 02/04/2023]
Abstract
Itaconate is produced from the mitochondrial TCA cycle enzyme aconitase decarboxylase (encoded by immune responsive gene1; Irg1) that exerts immunomodulatory function in myeloid cells. However, the role of the Irg1/itaconate pathway in dendritic cells (DC)-mediated airway inflammation and adaptive immunity to inhaled allergens, which are the primary antigen-presenting cells in allergic asthma, remains largely unknown. House dust mite (HDM)-challenged Irg1-/- mice displayed increases in eosinophilic airway inflammation, mucous cell metaplasia, and Th2 cytokine production with a mechanism involving impaired mite antigen presentations by DC. Adoptive transfer of HDM-pulsed DC from Irg1-deficient mice into naïve WT mice induced a similar phenotype of elevated type 2 airway inflammation and allergic sensitization. Untargeted metabolite analysis of HDM-pulsed DC revealed itaconate as one of the most abundant polar metabolites that potentially suppress mitochondrial oxidative damage. Furthermore, the immunomodulatory effect of itaconate was translated in vivo, where intranasal administration of 4-octyl itaconate 4-OI following antigen priming attenuated the manifestations of HDM-induced airway disease and Th2 immune response. Taken together, these data demonstrated for the first time a direct regulatory role of the Irg1/itaconate pathway in DC for the development of type 2 airway inflammation and suggest a possible therapeutic target in modulating allergic asthma.
Collapse
Affiliation(s)
- Anil Kumar Jaiswal
- grid.252546.20000 0001 2297 8753From the Laboratory of Lung Inflammation, Auburn University, Auburn, AL USA ,grid.252546.20000 0001 2297 8753Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL USA
| | - Jyoti Yadav
- grid.252546.20000 0001 2297 8753From the Laboratory of Lung Inflammation, Auburn University, Auburn, AL USA ,grid.252546.20000 0001 2297 8753Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL USA
| | - Sangeet Makhija
- grid.252546.20000 0001 2297 8753From the Laboratory of Lung Inflammation, Auburn University, Auburn, AL USA ,grid.252546.20000 0001 2297 8753Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL USA
| | - Suman Mazumder
- grid.252546.20000 0001 2297 8753Department of Drug Discovery and Development, Auburn University, Auburn, AL USA ,grid.252546.20000 0001 2297 8753Center for Pharmacogenomics and Single-Cell Omics, Harrison School of Pharmacy, Auburn University, Auburn, AL USA
| | - Amit Kumar Mitra
- grid.252546.20000 0001 2297 8753Department of Drug Discovery and Development, Auburn University, Auburn, AL USA ,grid.252546.20000 0001 2297 8753Center for Pharmacogenomics and Single-Cell Omics, Harrison School of Pharmacy, Auburn University, Auburn, AL USA
| | - Amol Suryawanshi
- grid.252546.20000 0001 2297 8753Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL USA
| | - Maninder Sandey
- grid.252546.20000 0001 2297 8753Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL USA
| | - Amarjit Mishra
- grid.252546.20000 0001 2297 8753From the Laboratory of Lung Inflammation, Auburn University, Auburn, AL USA ,grid.252546.20000 0001 2297 8753Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL USA
| |
Collapse
|
79
|
Kim HJ, Song JY, Park TI, Choi WS, Kim JH, Kwon OS, Lee JY. The effects of BRL-50481 on ovalbumin-induced asthmatic lung inflammation exacerbated by co-exposure to Asian sand dust in the murine model. Arch Pharm Res 2022; 45:51-62. [PMID: 34984603 PMCID: PMC8726530 DOI: 10.1007/s12272-021-01367-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 12/25/2021] [Indexed: 12/16/2022]
Abstract
Asian sand dust (ASD), which mainly originates in China and Mongolia in the spring and blows into Korea, can exacerbate respiratory and immunological diseases. This study aims to observe effects of co-exposure to ASD on ovalbumin (OVA)-induced asthmatic lung inflammation and of treatment with a phosphodiesterase 7 (PDE7) inhibitor in a mouse model. The challenge with OVA increased airway hyperresponsiveness (AHR) and inflammatory cell infiltration into the lung tissue. Interleukin (IL)-13, tumor necrosis factor-alpha, monocyte-protein-1, mucin, and antigen-specific IgE and IgG1 production increased in mouse serum. The co-exposure of ASD significantly exacerbated these effects in this asthma model. Notably, the administration of a PDE7 inhibitor, BRL-50481 (BRL), significantly reduced AHR, infiltration of inflammatory cells into the lungs, and the levels of type 2 T helper cell-related cytokines, antigen-specific immunoglobulins, and mucin. Thus, the administration of BRL ameliorated OVA-induced allergic asthmatic responses exacerbated by co-exposure to ASD. This study suggests that PDE7 inhibition can be a therapeutic strategy for inflammatory lung diseases and asthma via the regulation of T lymphocytes and reduction of IL-13, and, consequently, mucin production.
Collapse
Affiliation(s)
- Hong Jo Kim
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jin Yong Song
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Tae Il Park
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Won Seok Choi
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jong Heon Kim
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Oh Seong Kwon
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ji-Yun Lee
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea.
- Pathophysiology, College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
80
|
Pera T, Loblundo C, Penn RB. Pharmacological Management of Asthma and COPD. COMPREHENSIVE PHARMACOLOGY 2022:762-802. [DOI: 10.1016/b978-0-12-820472-6.00095-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
81
|
Klimek L, Hagemann J, Welkoborsky HJ, Cuevas M, Casper I, Förster-Rurmann U, Klimek F, Hintschich CA, Huppertz T, Bergmann KC, Tomazic PV, Bergmann C, Becker S. T2-Inflammation bei entzündlichen Atemwegserkrankungen: Grundlage neuer Behandlungsoptionen. Laryngorhinootologie 2021; 101:96-108. [PMID: 34937094 DOI: 10.1055/a-1709-7899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- L Klimek
- Zentrum für Rhinologie und Allergologie, Wiesbaden.,Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, Universitätsmedizin Mainz
| | - J Hagemann
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, Universitätsmedizin Mainz
| | | | - M Cuevas
- Klinik und Poliklinik für HNO-Heilkunde, Universitätsklinikum Carl Gustav Carus, TU Dresden
| | - I Casper
- Zentrum für Rhinologie und Allergologie, Wiesbaden
| | | | - F Klimek
- Zentrum für Rhinologie und Allergologie, Wiesbaden
| | - C A Hintschich
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, Universitätsklinikum Regensburg
| | - T Huppertz
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde, Universitätsmedizin Mainz
| | - K-Ch Bergmann
- Klinik für Dermatologie, Venerologie und Allergie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health
| | - P V Tomazic
- HNO-Universitätsklinik Graz, Medizinische Universität Graz
| | | | | |
Collapse
|
82
|
Dy ABC, Langlais PR, Barker NK, Addison KJ, Tanyaratsrisakul S, Boitano S, Christenson SA, Kraft M, Meyers D, Bleecker ER, Li X, Ledford JG. Myeloid-associated differentiation marker is a novel SP-A-associated transmembrane protein whose expression on airway epithelial cells correlates with asthma severity. Sci Rep 2021; 11:23392. [PMID: 34862427 PMCID: PMC8642528 DOI: 10.1038/s41598-021-02869-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022] Open
Abstract
Surfactant protein A (SP-A) is well-known for its protective role in pulmonary immunity. Previous studies from our group have shown that SP-A mediates eosinophil activities, including degranulation and apoptosis. In order to identify potential binding partners on eosinophils for SP-A, eosinophil lysates were subjected to SP-A pull-down and tandem mass spectrometry (MS/MS) analysis. We identified one membrane-bound protein, myeloid-associated differentiation marker (MYADM), as a candidate SP-A binding partner. Blocking MYADM on mouse and human eosinophils ex vivo prevented SP-A from inducing apoptosis; blocking MYADM in vivo led to increased persistence of eosinophilia and airway hyper-responsiveness in an ovalbumin (OVA) allergy model and increased airways resistance and mucus production in a house dust mite (HDM) asthma model. Examination of a subset of participants in the Severe Asthma Research Program (SARP) cohort revealed a significant association between epithelial expression of MYADM in asthma patients and parameters of airway inflammation, including: peripheral blood eosinophilia, exhaled nitric oxide (FeNO) and the number of exacerbations in the past 12 months. Taken together, our studies provide the first evidence of MYADM as a novel SP-A-associated protein that is necessary for SP-A to induce eosinophil apoptosis and we bring to light the potential importance of this previously unrecognized transmembrane protein in patients with asthma.
Collapse
Affiliation(s)
- Alane Blythe C Dy
- Clinical Translational Sciences, University of Arizona Health Sciences, Tucson, AZ, 85721, USA
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA
| | - Paul R Langlais
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Natalie K Barker
- Division of Endocrinology, Department of Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Kenneth J Addison
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA
| | | | - Scott Boitano
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Physiology, University of Arizona, Tucson, AZ, 85724, USA
| | - Stephanie A Christenson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, 94117, USA
| | - Monica Kraft
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA
- Department of Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Deborah Meyers
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Eugene R Bleecker
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Xingnan Li
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Julie G Ledford
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, 85724, USA.
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA.
- , 1230 N Cherry Avenue, BSRL Building, Tucson, AZ, 85719, USA.
| |
Collapse
|
83
|
Lewis BW, Jackson D, Amici SA, Walum J, Guessas M, Guessas S, Coneglio E, Boda AV, Guerau-de-Arellano M, Grayson MH, Britt RD. Corticosteroid insensitivity persists in the absence of STAT1 signaling in severe allergic airway inflammation. Am J Physiol Lung Cell Mol Physiol 2021; 321:L1194-L1205. [PMID: 34755542 PMCID: PMC8715027 DOI: 10.1152/ajplung.00244.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Corticosteroid insensitivity in asthma limits the ability to effectively manage severe asthma, which is characterized by persistent airway inflammation, airway hyperresponsiveness (AHR), and airflow obstruction despite corticosteroid treatment. Recent reports indicate that corticosteroid insensitivity is associated with increased interferon-γ (IFN-γ) levels and T-helper (Th) 1 lymphocyte infiltration in severe asthma. Signal transducer and activator of transcription 1 (STAT1) activation by IFN-γ is a key signaling pathway in Th1 inflammation; however, its role in the context of severe allergic airway inflammation and corticosteroid sensitivity remains unclear. In this study, we challenged wild-type (WT) and Stat1-/- mice with mixed allergens (MA) augmented with c-di-GMP [bis-(3'-5')-cyclic dimeric guanosine monophosphate], an inducer of Th1 cell infiltration with increased eosinophils, neutrophils, Th1, Th2, and Th17 cells. Compared with WT mice, Stat1-/- had reduced neutrophils, Th1, and Th17 cell infiltration. To evaluate corticosteroid sensitivity, mice were treated with either vehicle, 1 or 3 mg/kg fluticasone propionate (FP). Corticosteroids significantly reduced eosinophil infiltration and cytokine levels in both c-di-GMP + MA-challenged WT and Stat1-/- mice. However, histological and functional analyses show that corticosteroids did not reduce airway inflammation, epithelial mucous cell abundance, airway smooth muscle mass, and AHR in c-di-GMP + MA-challenged WT or Stat1-/- mice. Collectively, our data suggest that increased Th1 inflammation is associated with a decrease in corticosteroid sensitivity. However, increased airway pathology and AHR persist in the absence of STAT1 indicate corticosteroid insensitivity in structural airway cells is a STAT1 independent process.
Collapse
Affiliation(s)
- Brandon W. Lewis
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Devine Jackson
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Stephanie A. Amici
- 5Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, Ohio
| | - Joshua Walum
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Manel Guessas
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Sonia Guessas
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Elise Coneglio
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Akhila V. Boda
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Mireia Guerau-de-Arellano
- 5Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, Ohio,6Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio,7Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio,8Department of Neuroscience, The Ohio State University, Columbus, Ohio
| | - Mitchell H. Grayson
- 2Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio,3Division of Allergy and Immunology, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio,4Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Rodney D. Britt
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio,4Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
84
|
Rubbino F, Greco L, di Cristofaro A, Gaiani F, Vetrano S, Laghi L, Bonovas S, Piovani D. Journey through Crohn's Disease Complication: From Fistula Formation to Future Therapies. J Clin Med 2021; 10:jcm10235548. [PMID: 34884247 PMCID: PMC8658128 DOI: 10.3390/jcm10235548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/19/2022] Open
Abstract
Crohn’s Disease (CD) is a chronic inflammatory disorder in which up to 50% of patients develop fistula within 20 years after the initial diagnosis, and half of these patients suffer perianal fistulizing disease. The etiopathogenesis of CD-related perianal fistula is still unclear, and its phenotypical and molecular characteristics are even more indefinite. A better understanding would be crucial to develop targeted and more effective therapeutic strategies. At present, the most accredited theory for the formation of CD-related fistula identifies the epithelial-to-mesenchymal transition (EMT) as the driving force. It has been well recognized that CD carries an increased risk of malignancy, particularly mucinous adenocarcinoma is often associated with long-standing fistula in CD patients. Despite the availability of multiple treatment options, perianal fistulizing CD represents a therapeutic challenge and is associated with an important impact on patients’ quality of life. To date, the most effective management is multidisciplinary with the cooperation of gastroenterologists, surgeons, radiologists, and nutritionists and the best recommended treatment is a combination of medical and surgical approaches.
Collapse
Affiliation(s)
- Federica Rubbino
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy; (L.G.); (A.d.C.); (L.L.)
- Correspondence: (F.R.); (S.B.)
| | - Luana Greco
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy; (L.G.); (A.d.C.); (L.L.)
| | - Alessio di Cristofaro
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy; (L.G.); (A.d.C.); (L.L.)
| | - Federica Gaiani
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy;
- Gastroenterology and Endoscopy Unit, University-Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Stefania Vetrano
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (S.V.); (D.P.)
| | - Luigi Laghi
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy; (L.G.); (A.d.C.); (L.L.)
- Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43126 Parma, Italy;
| | - Stefanos Bonovas
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (S.V.); (D.P.)
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
- Correspondence: (F.R.); (S.B.)
| | - Daniele Piovani
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy; (S.V.); (D.P.)
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| |
Collapse
|
85
|
Zhang M, Duffen JL, Nocka KH, Kasaian MT. IL-13 Controls IL-33 Activity through Modulation of ST2. THE JOURNAL OF IMMUNOLOGY 2021; 207:3070-3080. [PMID: 34789557 DOI: 10.4049/jimmunol.2100655] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/13/2021] [Indexed: 12/12/2022]
Abstract
IL-33 is a multifunctional cytokine that mediates local inflammation upon tissue damage. IL-33 is known to act on multiple cell types including group 2 innate lymphoid cells (ILC2s), Th2 cells, and mast cells to drive production of Th2 cytokines including IL-5 and IL-13. IL-33 signaling activity through transmembrane ST2L can be inhibited by soluble ST2 (sST2), which acts as a decoy receptor. Previous findings suggested that modulation of IL-13 levels in mice lacking decoy IL-13Rα2, or mice lacking IL-13, impacted responsiveness to IL-33. In this study, we used Il13 -/- mice to investigate whether IL-13 regulates IL-33 activity by modulating the transmembrane and soluble forms of ST2. In Il13 -/- mice, the effects of IL-33 administration were exacerbated relative to wild type (WT). Il13 -/- mice administered IL-33 i.p. had heightened splenomegaly, more immune cells in the peritoneum including an expanded ST2L+ ILC2 population, increased eosinophilia in the spleen and peritoneum, and reduced sST2 in the circulation and peritoneum. In the spleen, lung, and liver of mice given IL-33, gene expression of both isoforms of ST2 was increased in Il13 -/- mice relative to WT. We confirmed fibroblasts to be an IL-13-responsive cell type that can regulate IL-33 activity through production of sST2. This study elucidates the important regulatory activity that IL-13 exerts on IL-33 through induction of IL-33 decoy receptor sST2 and through modulation of ST2L+ ILC2s.
Collapse
Affiliation(s)
- Melvin Zhang
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| | - Jennifer L Duffen
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| | - Karl H Nocka
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| | - Marion T Kasaian
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| |
Collapse
|
86
|
Liu Y, Wei L, He C, Chen R, Meng L. Lipoxin A4 inhibits ovalbumin-induced airway inflammation and airway remodeling in a mouse model of asthma. Chem Biol Interact 2021; 349:109660. [PMID: 34537180 DOI: 10.1016/j.cbi.2021.109660] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/24/2021] [Accepted: 09/14/2021] [Indexed: 12/19/2022]
Abstract
Asthma is a chronic respiratory disease, which is characterized by airway inflammation, remodeling and airway hyperresponsiveness. Airway remodeling is caused by long-term inflammation of the airways. Lipoxin A4 (LXA4) is a natural eicosanoid with powerful anti-inflammatory properties, and has been shown to serve a critical role in orchestrating pulmonary inflammation and airway hyper-responsiveness in asthmatic mice. However, its effect on airway remodeling is unknown. Female BALB/c mice were used to establish a mouse model of asthma which were sensitized and challenged by ovalbumin (OVA). LXA4 was intranasally administrated prior to the challenge. The results of our study indicated that LXA4 suppressed the OVA-induced inflammatory cell infiltration and T helper type 2 (Th2) cytokines secretion in the mouse model of asthma. Characteristics of airway remodeling, such as thickening of the bronchial wall and smooth muscle, overdeposition of collagen, and overexpression of α-smooth muscle actin (α-SMA) and collagen-I were reversed by LXA4. Furthermore, LXA4 suppressed the aberrant activation of the signal transducer and activator of transcription 3 (STAT3) pathway in the lung tissues of asthmatic mice. In conclusion, these findings demonstrated that LXA4 alleviated allergic airway inflammation and remodeling in asthmatic mice, which may be related to the inhibition of STAT3 pathway.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, 271000, China
| | - Li Wei
- Department of Respiratory Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, 271000, China
| | - Chao He
- Department of Gastrointestinal Surgery, Taian City Central Hospital, Taian, Shandong, China
| | - Ran Chen
- Department of Respiratory Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, 271000, China
| | - Ling Meng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, 271000, China.
| |
Collapse
|
87
|
Corcoran TE, Huber AS, Hill SL, Locke LW, Weber L, Muthukrishnan A, Heidrich EM, Wenzel S, Myerburg MM. Mucociliary Clearance Differs in Mild Asthma by Levels of Type 2 Inflammation. Chest 2021; 160:1604-1613. [PMID: 34029561 PMCID: PMC8628176 DOI: 10.1016/j.chest.2021.05.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Although mucus plugging is a well-reported feature of asthma, whether asthma and type 2 inflammation affect mucociliary clearance (MCC) is unknown. RESEARCH QUESTION Does type 2 inflammation influence mucus clearance rates in patients with mild asthma who are not receiving corticosteroids? STUDY DESIGN AND METHODS The clearance rates of inhaled radiolabeled particles were compared between patients with mild asthma with low (n = 17) and high (n = 18) levels of T2 inflammation. Fraction exhaled nitric oxide (Feno) was used to prospectively segregate subjects into T2 Lo (Feno < 25 ppb) and T2 Hi (Feno > 35 ppb) cohorts. Bronchial brush samples were collected with fiber-optic bronchoscopy, and quantitative polymerase chain reaction was performed to measure expression of genes associated with T2 asthma. MCC rate comparisons were also made with a historical group of healthy control subjects (HCs, n = 12). RESULTS The T2 Lo cohort demonstrated increased MCC when compared with both T2 Hi and historic HCs. MCC within the T2 Hi group varied significantly, with some subjects having low or zero clearance. MCC decreased with increasing expression of several markers of T2 airway inflammation (CCL26, NOS2, and POSTN) and with Feno. MUC5AC and FOXJ1 expression was similar between the T2Lo and T2Hi cohorts. INTERPRETATION Increasing T2 inflammation was associated with decreasing MCC. High rates of MCC in T2 Lo subjects may indicate a compensatory mechanism present in mild disease but lost with high levels of inflammation. Future studies are required to better understand mechanisms and whether impairments in MCC in more severe asthma drive worse clinical outcomes.
Collapse
Affiliation(s)
- Timothy E Corcoran
- Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, PA; Department of Bioengineering, University of Pittsburgh, PA; Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA.
| | - Alex S Huber
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, PA
| | - Sherri L Hill
- Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, PA
| | - Landon W Locke
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH
| | - Lawrence Weber
- Nuclear Medicine Department, University of Pittsburgh Medical Center, PA
| | | | - Elisa M Heidrich
- Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, PA
| | - Sally Wenzel
- Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, PA; Department of Environmental & Occupational Health, University of Pittsburgh, PA
| | - Mike M Myerburg
- Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, PA
| |
Collapse
|
88
|
Nicholas BD, Kiprovski A, Perez D, Mehta R, Murphy MK, Li Z, Tampio A. Changes in Eustachian Tube Mucosa in Mice After Short-Term Tobacco and E-cigarette Smoke Exposure. Laryngoscope 2021; 132:648-654. [PMID: 34599608 DOI: 10.1002/lary.29887] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/05/2021] [Accepted: 08/30/2021] [Indexed: 11/09/2022]
Abstract
OBJECTIVES To evaluate histologic changes in middle ear and eustachian tube (ET) mucosa of mice after exposure to tobacco or electronic cigarette (e-cigarette) smoke. To determine whether there were any mitigating effects of middle ear application of anti-IL-13 or the epidermal growth factor receptor antagonist AG1478 on noted changes within ET mucosa. STUDY DESIGN Controlled animal study. METHODS Fifty BALB/cJ mice were randomly assigned to one of five groups: A control group with no smoke exposure, two groups exposed to tobacco smoke, and two groups exposed to e-cigarette vapor. Within the exposed groups after 4 weeks of exposure, one ear was infiltrated with a saline hydrogel and the other ear with hydrogel of either Anti-IL-13 or AG1478. After four more weeks of exposure, the animals were euthanized and the ETs were evaluated for mucosal changes. RESULTS Compared to control animals with no smoke exposure, there were significant decreases in the numbers of goblet cells within the ET mucosa of mice exposed to tobacco smoke and e-cigarette vapor. No significant differences in cilia, mucin, or squamous metaplasia were noted. Neither anti-IL-13 nor AG178 significantly altered goblet cell count in the ET mucosa of mice exposed to tobacco smoke; however, both agents significantly increased goblet cells within the ET mucosa of mice exposed to e-cigarette vapor. CONCLUSION Short-term tobacco smoke and e-cigarette vapor significantly decrease goblet cell count in mouse ET mucosa. Middle ear application of both anti-IL-13 and AG1478 resulted in an increase in goblet cell count among mice exposed to e-cigarette vapor, but not to tobacco smoke. LEVEL OF EVIDENCE NA Laryngoscope, 2021.
Collapse
Affiliation(s)
- Brian D Nicholas
- Department of Otolaryngology-Head and Neck Surgery, Upstate Medical University, Syracuse, New York, U.S.A
| | | | - Diandra Perez
- Department of Pathology, Upstate Medical University, Syracuse, New York, U.S.A
| | - Rohin Mehta
- Department of Pathology, Upstate Medical University, Syracuse, New York, U.S.A
| | - Michael K Murphy
- Department of Otolaryngology-Head and Neck Surgery, Upstate Medical University, Syracuse, New York, U.S.A
| | - Zhenfeng Li
- Department of Applied Statistics, Syracuse University, Syracuse, New York, U.S.A
| | - Alex Tampio
- Department of Otolaryngology-Head and Neck Surgery, Upstate Medical University, Syracuse, New York, U.S.A
| |
Collapse
|
89
|
Noto CN, Hoft SG, Bockerstett KA, Jackson NM, Ford EL, Vest LS, DiPaolo RJ. IL13 Acts Directly on Gastric Epithelial Cells to Promote Metaplasia Development During Chronic Gastritis. Cell Mol Gastroenterol Hepatol 2021; 13:623-642. [PMID: 34587523 PMCID: PMC8715193 DOI: 10.1016/j.jcmgh.2021.09.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS It is well established that chronic inflammation promotes gastric cancer-associated metaplasia, but little is known regarding the mechanisms by which immune cells and cytokines regulate metaplastic cellular changes. The goals of this study were to identify interleukin 13 (IL13)-producing immune cells, determine the gastric epithelial cell response(s) to IL13, and establish the role(s) of IL13 in metaplasia development. METHODS Experiments used an established mouse model of autoimmune gastritis (TxA23), TxA23×Il4ra-/- mice, which develop gastritis but do not express the IL4/IL13-receptor subunit IL4Rα, and TxA23×Il13-Yfp mice, which express yellow fluorescent protein in IL13-producing cells. Flow cytometry was used to measure IL13 secretion and identify IL13-producing immune cells. Mouse and human gastric organoids were cultured with IL13 to determine epithelial cell response(s) to IL13. Single-cell RNA sequencing was performed on gastric epithelial cells from healthy and inflamed mouse stomachs. Mice with gastritis were administered IL13-neutralizing antibodies and stomachs were analyzed by histopathology and immunofluorescence. RESULTS We identified 6 unique subsets of IL13-producing immune cells in the inflamed stomach. Organoid cultures showed that IL13 acts directly on gastric epithelium to induce a metaplastic phenotype. IL4Rα-deficient mice did not progress to metaplasia. Single-cell RNA sequencing determined that gastric epithelial cells from IL4Rα-deficient mice up-regulated inflammatory genes but failed to up-regulate metaplasia-associated transcripts. Neutralization of IL13 significantly reduced and reversed metaplasia development in mice with gastritis. CONCLUSIONS IL13 is made by a variety of immune cell subsets during chronic gastritis and promotes gastric cancer-associated metaplastic epithelial cell changes. Neutralization of IL13 reduces metaplasia severity during chronic gastritis.
Collapse
Affiliation(s)
- Christine N Noto
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Stella G Hoft
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Kevin A Bockerstett
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Nicholas M Jackson
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Eric L Ford
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Luke S Vest
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Richard J DiPaolo
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
90
|
Balkrishna A, Solleti SK, Singh H, Singh R, Sharma N, Varshney A. Biotite-Calx Based Traditional Indian Medicine Sahastraputi-Abhrak-Bhasma Prophylactically Mitigates Allergic Airway Inflammation in a Mouse Model of Asthma by Amending Cytokine Responses. J Inflamm Res 2021; 14:4743-4760. [PMID: 34557016 PMCID: PMC8455516 DOI: 10.2147/jir.s313955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/25/2021] [Indexed: 12/15/2022] Open
Abstract
Purpose Asthma is a heterogeneous airway inflammatory disease with limited therapeutic options. Traditional medicine is extensively used for treating various ailments including asthma. Sahastraputi-Abhrak-Bhasma (SPAB) is a biotite-calx based Indian medicine. Methods We have tested for the anti-inflammatory and anti-asthmatic properties of SPAB, using a mouse model of ovalbumin-induced allergic asthma in-vivo and cell-based assays in-vitro. Histological analysis, qPCR and ELISA were performed to assess the pathology. SEM, EDX and XRD-analysis were performed to characterize the SPAB particles. Results SEM, EDX and XRD-analysis identified the presence of SPAB particle of 100 nm–~1µm diameter and contains annite-1M, aluminium silicate, kyanite, aluminium oxide, magnesium silicate, and maghemite in the samples. Ova-challenge resulted in severe inflammatory responses, airway remodelling and increased oxidative burden in lungs. Importantly, prophylactic treatment with SPAB significantly attenuated allergen induced leukocyte infiltration specifically eosinophils, lymphocytes, macrophages and neutrophils in BALF. Ova-induced mucus hypersecretion, peri-bronchial collagen deposition, inflammatory cell infiltration and bronchial epithelial thickening were significantly abrogated upon SPAB treatment. qPCR and ELISA analysis identified that allergen induced increases in IL-5, IL-13, IL-33, IFN-γ and IL-1β cytokines mRNA in whole lungs and the levels of IL-6, IL-1β and TNF-α proteins in BALF were significantly attenuated upon oral SPAB treatment. SPAB restored allergen induced decreases in anti-oxidant markers in lungs. In-vitro, SPAB attenuated the secretion of IL-6, and TNF-α from human bronchial epithelial cells and modestly inhibited NF-kB/AP-1 pathway in HEK cells. Conclusion Taken together, our results experimentally validated the prophylactic ameliorative potential of the Indian classical medicine Sahastraputi-Abhrak-Bhasma against asthma associated airway inflammation.
Collapse
Affiliation(s)
- Acharya Balkrishna
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, Uttarakhand, India.,Department of Allied and Applied Sciences, University of Patanjali, Patanjali Yog Peeth, Haridwar, Uttarakhand, India.,Patanjali UK Trust, Glasgow, UK
| | - Siva Kumar Solleti
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, Uttarakhand, India
| | - Hoshiyar Singh
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, Uttarakhand, India
| | - Rani Singh
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, Uttarakhand, India
| | - Niti Sharma
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, Uttarakhand, India
| | - Anurag Varshney
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, Uttarakhand, India.,Department of Allied and Applied Sciences, University of Patanjali, Patanjali Yog Peeth, Haridwar, Uttarakhand, India.,Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
91
|
Luo Y, Li L, Chen X, Gou H, Yan K, Xu Y. Effects of lactate in immunosuppression and inflammation: Progress and prospects. Int Rev Immunol 2021; 41:19-29. [PMID: 34486916 DOI: 10.1080/08830185.2021.1974856] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Lactate used to be considered as a waste product of glucose metabolism. However, accumulating evidence has revealed its crucial role in regulating various biological and pathological processes. Hypoxia, inflammation, viral infection, and tumor promote the production of lactate. Then lactate activates G protein-coupled receptor 81 (GPR81) or shuttles across membranes by monocarboxylate-transporters (MCTs) to execute its intricate effects. Many studies highlighted the function of lactate in regulating dendritic cells, monocytes, natural killer cells, mast cells, T cells, tumor cells, fibroblasts, macrophages polarization, and the differentiation of Th1, Th17, MDSCs, Tregs; all of which play a role in maintaining the immune homeostasis of the host when challenged with the noxious stimuli. In this review, we summarized the influence of lactate in diverse tissue-specific cells, and discuss their effects on viral infection, acute inflammation, chronic inflammation, sepsis, and tumor immunosuppression. The goal of this review is to expose that lactate has a double-edged effect on host immunity and accompanying inflammatory reactions, which could be a potentially effective target for treating the tumor and multiple infectious diseases.
Collapse
Affiliation(s)
- Ying Luo
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Lu Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Xu Chen
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Huiqing Gou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Ke Yan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Yan Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
92
|
Rupani H, Fong WCG, Kyyaly A, Kurukulaaratchy RJ. Recent Insights into the Management of Inflammation in Asthma. J Inflamm Res 2021; 14:4371-4397. [PMID: 34511973 PMCID: PMC8421249 DOI: 10.2147/jir.s295038] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/17/2021] [Indexed: 12/11/2022] Open
Abstract
The present prevailing inflammatory paradigm in asthma is of T2-high inflammation orchestrated by key inflammatory cells like Type 2 helper lymphocytes, innate lymphoid cells group 2 and associated cytokines. Eosinophils are key components of this T2 inflammatory pathway and have become key therapeutic targets. Real-world evidence on the predominant T2-high nature of severe asthma is emerging. Various inflammatory biomarkers have been adopted in clinical practice to aid asthma characterization including airway measures such as bronchoscopic biopsy and lavage, induced sputum analysis, and fractional exhaled nitric oxide. Blood measures like eosinophil counts have also gained widespread usage and multicomponent algorithms combining different parameters are now appearing. There is also growing interest in potential future biomarkers including exhaled volatile organic compounds, micro RNAs and urinary biomarkers. Additionally, there is a growing realisation that asthma is a heterogeneous state with numerous phenotypes and associated treatable traits. These may show particular inflammatory patterns and merit-specific management approaches that could improve asthma patient outcomes. Inhaled corticosteroids (ICS) remain the mainstay of asthma management but their use earlier in the course of disease is being advocated. Recent evidence suggests potential roles for ICS in combination with long-acting beta-agonists (LABA) for as needed use in mild asthma whilst maintenance and reliever therapy regimes have gained widespread acceptance. Other anti-inflammatory strategies including ultra-fine particle ICS, leukotriene receptor antagonists and macrolide antibiotics may show efficacy in particular phenotypes too. Monoclonal antibody biologic therapies have recently entered clinical practice with significant impacts on asthma outcomes. Understanding of the efficacy and use of those agents is becoming clearer with a growing body of real-world evidence as is their potential applicability to other treatable comorbid traits. In conclusion, the evolving understanding of T2 driven inflammation alongside a treatable traits disease model is enhancing therapeutic approaches to address inflammation in asthma.
Collapse
Affiliation(s)
- Hitasha Rupani
- Department of Respiratory Medicine, University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| | - Wei Chern Gavin Fong
- Clinical and Experimental Sciences, University of Southampton, Southampton, UK
- David Hide Asthma and Allergy Research Centre, Isle of Wight NHS Trust, Isle of Wight, UK
| | - Aref Kyyaly
- Clinical and Experimental Sciences, University of Southampton, Southampton, UK
- David Hide Asthma and Allergy Research Centre, Isle of Wight NHS Trust, Isle of Wight, UK
| | - Ramesh J Kurukulaaratchy
- Department of Respiratory Medicine, University Hospitals Southampton NHS Foundation Trust, Southampton, UK
- Clinical and Experimental Sciences, University of Southampton, Southampton, UK
- David Hide Asthma and Allergy Research Centre, Isle of Wight NHS Trust, Isle of Wight, UK
- NIHR Biomedical Research Centre, University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
93
|
Majumder N, Goldsmith WT, Kodali VK, Velayutham M, Friend SA, Khramtsov VV, Nurkiewicz TR, Erdely A, Zeidler-Erdely PC, Castranova V, Harkema JR, Kelley EE, Hussain S. Oxidant-induced epithelial alarmin pathway mediates lung inflammation and functional decline following ultrafine carbon and ozone inhalation co-exposure. Redox Biol 2021; 46:102092. [PMID: 34418598 PMCID: PMC8385153 DOI: 10.1016/j.redox.2021.102092] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 11/29/2022] Open
Abstract
Environmental inhalation exposures are inherently mixed (gases and particles), yet regulations are still based on single toxicant exposures. While the impacts of individual components of environmental pollution have received substantial attention, the impact of inhalation co-exposures is poorly understood. Here, we mechanistically investigated pulmonary inflammation and lung function decline after inhalation co-exposure and individual exposures to ozone (O3) and ultrafine carbon black (CB). Environmentally/occupationally relevant lung deposition levels in mice were achieved after inhalation of stable aerosols with similar aerodynamic and mass median distributions. X-ray photoemission spectroscopy detected increased surface oxygen contents on particles in co-exposure aerosols. Compared with individual exposures, co-exposure aerosols produced greater acellular and cellular oxidants detected by electron paramagnetic resonance (EPR) spectroscopy, and in vivo immune-spin trapping (IST), as well as synergistically increased lavage neutrophils, lavage proteins and inflammation related gene/protein expression. Co-exposure induced a significantly greater respiratory function decline compared to individual exposure. A synthetic catalase-superoxide dismutase mimetic (EUK-134) significantly blunted lung inflammation and respiratory function decline confirming the role of oxidant imbalance. We identified a significant induction of epithelial alarmin (thymic stromal lymphopoietin-TSLP)-dependent interleukin-13 pathway after co-exposure, associated with increased mucin and interferon gene expression. We provided evidence of interactive outcomes after air pollution constituent co-exposure and identified a key mechanistic pathway that can potentially explain epidemiological observation of lung function decline after an acute peak of air pollution. Developing and studying the co-exposure scenario in a standardized and controlled fashion will enable a better mechanistic understanding of how environmental exposures result in adverse outcomes. Interaction with O3 mediates free radical production on the surface of carbon black (CB) particles. Oxidants mediate co-exposure (CB + O3)-induced lung function decline. EUK-134 (a synthetic superoxide-catalase mimetic) abrogates CB + O3-induced lung inflammation. CB + O3 co-exposure induces greater lung inflammation than individual exposures. Epithelial alarmin (TSLP) contributes significantly to the CB + O3 toxicity.
Collapse
Affiliation(s)
- Nairrita Majumder
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, USA; Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, USA
| | - William T Goldsmith
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, USA; Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, USA
| | - Vamsi K Kodali
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, USA; Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, USA; National Institute for Occupational Safety and Health, USA
| | | | - Sherri A Friend
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, USA; National Institute for Occupational Safety and Health, USA
| | - Valery V Khramtsov
- Department of Biochemistry, School of Medicine, West Virginia University, USA
| | - Timothy R Nurkiewicz
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, USA; Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, USA
| | - Aaron Erdely
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, USA; Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, USA; National Institute for Occupational Safety and Health, USA
| | - Patti C Zeidler-Erdely
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, USA; Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, USA; National Institute for Occupational Safety and Health, USA
| | - Vince Castranova
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, USA
| | - Jack R Harkema
- Department of Pathobiology and Diagnostic Investigation, School of Veterinary Medicine, Michigan State University, USA
| | - Eric E Kelley
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, USA; Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, USA
| | - Salik Hussain
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, USA; Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, USA.
| |
Collapse
|
94
|
Deshpande NP, Riordan SM, Gorman CJ, Nielsen S, Russell TL, Correa-Ospina C, Fernando BSM, Waters SA, Castaño-Rodríguez N, Man SM, Tedla N, Wilkins MR, Kaakoush NO. Multi-omics of the esophageal microenvironment identifies signatures associated with progression of Barrett's esophagus. Genome Med 2021; 13:133. [PMID: 34412659 PMCID: PMC8375061 DOI: 10.1186/s13073-021-00951-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 08/11/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The enrichment of Gram-negative bacteria of oral origin in the esophageal microbiome has been associated with the development of metaplasia. However, to date, no study has comprehensively assessed the relationships between the esophageal microbiome and the host. METHODS Here, we examine the esophageal microenvironment in gastro-esophageal reflux disease and metaplasia using multi-omics strategies targeting the microbiome and host transcriptome, followed by targeted culture, comparative genomics, and host-microbial interaction studies of bacterial signatures of interest. RESULTS Profiling of the host transcriptome from esophageal mucosal biopsies revealed profound changes during metaplasia. Importantly, five biomarkers showed consistent longitudinal changes with disease progression from reflux disease to metaplasia. We showed for the first time that the esophageal microbiome is distinct from the salivary microbiome and the enrichment of Campylobacter species as a consistent signature in disease across two independent cohorts. Shape fitting and matrix correlation identified associations between the microbiome and host transcriptome profiles, with a novel co-exclusion relationship found between Campylobacter and napsin B aspartic peptidase. Targeted culture of Campylobacter species from the same cohort revealed a subset of isolates to have a higher capacity to survive within primary human macrophages. Comparative genomic analyses showed these isolates could be differentiated by specific genomic features, one of which was validated to be associated with intracellular fitness. Screening for these Campylobacter strain-specific signatures in shotgun metagenomics data from another cohort showed an increase in prevalence with disease progression. Comparative transcriptomic analyses of primary esophageal epithelial cells exposed to the Campylobacter isolates revealed expression changes within those infected with strains with high intracellular fitness that could explain the increased likelihood of disease progression. CONCLUSIONS We provide a comprehensive assessment of the esophageal microenvironment, identifying bacterial strain-specific signatures with high relevance to progression of metaplasia.
Collapse
Affiliation(s)
- Nandan P Deshpande
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Stephen M Riordan
- Gastrointestinal and Liver Unit, The Prince of Wales Hospital, Randwick, NSW, 2031, Australia
| | - Claire J Gorman
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Shaun Nielsen
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Tonia L Russell
- Ramaciotti Centre for Genomics, UNSW Sydney, Sydney, NSW, 2052, Australia
| | | | - Bentotage S M Fernando
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Shafagh A Waters
- School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, 2052, Australia
| | | | - Si Ming Man
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, 2601, Australia
| | - Nicodemus Tedla
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Marc R Wilkins
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
- Ramaciotti Centre for Genomics, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Nadeem O Kaakoush
- School of Medical Sciences, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia.
| |
Collapse
|
95
|
Akkoc T, O'Mahony L, Ferstl R, Akdis C, Akkoc T. Mouse Models of Asthma: Characteristics, Limitations and Future Perspectives on Clinical Translation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1376:119-133. [PMID: 34398449 DOI: 10.1007/5584_2021_654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Asthma is a complex and heterogeneous inflammatory airway disease primarily characterized by airway obstruction, which affects up to 15% of the population in Westernized countries with an increasing prevalence. Descriptive laboratory and clinical studies reveal that allergic asthma is due to an immunological inflammatory response and is significantly influenced by an individual's genetic background and environmental factors. Due to the limitations associated with human experiments and tissue isolation, direct mouse models of asthma provide important insights into the disease pathogenesis and in the discovery of novel therapeutics. A wide range of asthma models are currently available, and the correct model system for a given experimental question needs to be carefully chosen. Despite recent advances in the complexity of murine asthma models, for example humanized murine models and the use of clinically relevant allergens, the limitations of the murine system should always be acknowledged, and it remains to be seen if any single murine model can accurately replicate all the clinical features associated with human asthmatic disease.
Collapse
Affiliation(s)
- Tolga Akkoc
- Genetic Engineering and Biotechnology Institute, Tubitak Marmara Research Center, Kocaeli, Turkey.
| | - Liam O'Mahony
- Department of Medicine and Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ruth Ferstl
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Cezmi Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Davos, Switzerland
| | - Tunc Akkoc
- Department of Pediatric Allergy-Immunology, School of Medicine, Marmara University, Istanbul, Turkey
| |
Collapse
|
96
|
Yang R, Weisshaar M, Mele F, Benhsaien I, Dorgham K, Han J, Croft CA, Notarbartolo S, Rosain J, Bastard P, Puel A, Fleckenstein B, Glimcher LH, Di Santo JP, Ma CS, Gorochov G, Bousfiha A, Abel L, Tangye SG, Casanova JL, Bustamante J, Sallusto F. High Th2 cytokine levels and upper airway inflammation in human inherited T-bet deficiency. J Exp Med 2021; 218:e20202726. [PMID: 34160550 PMCID: PMC8225679 DOI: 10.1084/jem.20202726] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/16/2021] [Accepted: 05/27/2021] [Indexed: 12/20/2022] Open
Abstract
We have described a child suffering from Mendelian susceptibility to mycobacterial disease (MSMD) due to autosomal recessive, complete T-bet deficiency, which impairs IFN-γ production by innate and innate-like adaptive, but not mycobacterial-reactive purely adaptive, lymphocytes. Here, we explore the persistent upper airway inflammation (UAI) and blood eosinophilia of this patient. Unlike wild-type (WT) T-bet, the mutant form of T-bet from this patient did not inhibit the production of Th2 cytokines, including IL-4, IL-5, IL-9, and IL-13, when overexpressed in T helper 2 (Th2) cells. Moreover, Herpesvirus saimiri-immortalized T cells from the patient produced abnormally large amounts of Th2 cytokines, and the patient had markedly high plasma IL-5 and IL-13 concentrations. Finally, the patient's CD4+ αβ T cells produced most of the Th2 cytokines in response to chronic stimulation, regardless of their antigen specificities, a phenotype reversed by the expression of WT T-bet. T-bet deficiency thus underlies the excessive production of Th2 cytokines, particularly IL-5 and IL-13, by CD4+ αβ T cells, causing blood eosinophilia and UAI. The MSMD of this patient results from defective IFN-γ production by innate and innate-like adaptive lymphocytes, whereas the UAI and eosinophilia result from excessive Th2 cytokine production by adaptive CD4+ αβ T lymphocytes.
Collapse
Affiliation(s)
- Rui Yang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
| | - Marc Weisshaar
- Institute of Microbiology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Federico Mele
- Center of Medical Immunology, Institute for Research in Biomedicine, Faculty of Biomedical Sciences, University of Italian Switzerland, Bellinzona, Switzerland
| | - Ibtihal Benhsaien
- Laboratory of Clinical Immunology, Inflammation, and Allergy, Faculty of Medicine and Pharmacy of Casablanca, King Hassan II University, Casablanca, Morocco
- Clinical Immunology Unit, Department of Pediatric Infectious Diseases, Children's Hospital, Centre Hospitalo-Universitaire Averroes, Casablanca, Morocco
| | - Karim Dorgham
- Sorbonne University, Institut national de la santé et de la recherche médicale, Center for Immunology and Microbial Infections-Paris, Paris, France
| | - Jing Han
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
| | - Carys A. Croft
- Innate Immunity Unit, Institut Pasteur, Paris, France
- Institut national de la santé et de la recherche médicale U1223, Paris, France
- University of Paris, Paris, France
| | - Samuele Notarbartolo
- Center of Medical Immunology, Institute for Research in Biomedicine, Faculty of Biomedical Sciences, University of Italian Switzerland, Bellinzona, Switzerland
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale Unité Mixte de Recherches 1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Paul Bastard
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale Unité Mixte de Recherches 1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Anne Puel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale Unité Mixte de Recherches 1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Bernhard Fleckenstein
- Institute for Clinical and Molecular Virology, University Erlangen-Nuremberg, Erlangen, Germany
| | - Laurie H. Glimcher
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Department of Immunology, Harvard Medical School, Boston, MA
| | - James P. Di Santo
- Innate Immunity Unit, Institut Pasteur, Paris, France
- Institut national de la santé et de la recherche médicale U1223, Paris, France
| | - Cindy S. Ma
- Garvan Institute of Medical Research, Darlinghurst, Australia
- St. Vincent’s Clinical School, Faculty of Medicine and Health, University of New South Wales, Sydney, Darlinghurst, Australia
| | - Guy Gorochov
- Sorbonne University, Institut national de la santé et de la recherche médicale, Center for Immunology and Microbial Infections-Paris, Paris, France
- Assistance Publique–Hôpitaux de Paris, Department of Immunology, Paris, France
| | - Aziz Bousfiha
- Laboratory of Clinical Immunology, Inflammation, and Allergy, Faculty of Medicine and Pharmacy of Casablanca, King Hassan II University, Casablanca, Morocco
- Clinical Immunology Unit, Department of Pediatric Infectious Diseases, Children's Hospital, Centre Hospitalo-Universitaire Averroes, Casablanca, Morocco
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale Unité Mixte de Recherches 1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Stuart G. Tangye
- Garvan Institute of Medical Research, Darlinghurst, Australia
- St. Vincent’s Clinical School, Faculty of Medicine and Health, University of New South Wales, Sydney, Darlinghurst, Australia
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale Unité Mixte de Recherches 1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
- Howard Hughes Medical Institute, New York, NY
| | - Jacinta Bustamante
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale Unité Mixte de Recherches 1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Federica Sallusto
- Institute of Microbiology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
- Center of Medical Immunology, Institute for Research in Biomedicine, Faculty of Biomedical Sciences, University of Italian Switzerland, Bellinzona, Switzerland
| |
Collapse
|
97
|
Ono J, Takai M, Kamei A, Azuma Y, Izuhara K. Pathological Roles and Clinical Usefulness of Periostin in Type 2 Inflammation and Pulmonary Fibrosis. Biomolecules 2021; 11:1084. [PMID: 34439751 PMCID: PMC8391913 DOI: 10.3390/biom11081084] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
Periostin is known to be a useful biomarker for various diseases. In this article, we focus on allergic diseases and pulmonary fibrosis, for which we and others are now developing detection systems for periostin as a biomarker. Biomarker-based precision medicine in the management of type 2 inflammation and fibrotic diseases since heterogeneity is of utmost importance. Periostin expression is induced by type 2 cytokines (interleukin-4/-13) or transforming growth factor-β, and plays a vital role in the pathogenesis of allergic inflammation or interstitial lung disease, respectively, andits serum levels are correlated disease severity, prognosis and responsiveness to the treatment. We first summarise the importance of type 2 biomarker and then describe the pathological role of periostin in the development and progression of type 2 allergic inflammation and pulmonary fibrosis. In addition, then, we summarise the recent development of assay methods for periostin detection, and analyse the diseases in which periostin concentration is elevated in serum and local biological fluids and its usefulness as a biomarker. Furthermore, we describe recent findings of periostin as a biomarker in the use of biologics or anti-fibrotic therapy. Finally, we describe the factors that influence the change in periostin concentration under the healthy conditions.
Collapse
Affiliation(s)
- Junya Ono
- Shino-Test Corporation, 2-29-14 Oonodai Minami-ku, Sagamihara, Kanagawa 252-0331, Japan; (M.T.); (A.K.); (Y.A.)
| | - Masayuki Takai
- Shino-Test Corporation, 2-29-14 Oonodai Minami-ku, Sagamihara, Kanagawa 252-0331, Japan; (M.T.); (A.K.); (Y.A.)
- Division of Medical Biochemistry, Department of Biomolecular Science, Saga Medical School, 5-1-1 Nabeshima, Saga 849-8501, Japan;
| | - Ayami Kamei
- Shino-Test Corporation, 2-29-14 Oonodai Minami-ku, Sagamihara, Kanagawa 252-0331, Japan; (M.T.); (A.K.); (Y.A.)
| | - Yoshinori Azuma
- Shino-Test Corporation, 2-29-14 Oonodai Minami-ku, Sagamihara, Kanagawa 252-0331, Japan; (M.T.); (A.K.); (Y.A.)
| | - Kenji Izuhara
- Division of Medical Biochemistry, Department of Biomolecular Science, Saga Medical School, 5-1-1 Nabeshima, Saga 849-8501, Japan;
| |
Collapse
|
98
|
Perkins TN, Oury TD. The perplexing role of RAGE in pulmonary fibrosis: causality or casualty? Ther Adv Respir Dis 2021; 15:17534666211016071. [PMID: 34275342 PMCID: PMC8293846 DOI: 10.1177/17534666211016071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease in which most patients die within 3 years of diagnosis. With an unknown etiology, IPF results in progressive fibrosis of the lung parenchyma, diminishing normal lung function, which results in respiratory failure, and eventually, death. While few therapies are available to reduce disease progression, patients continue to advance toward respiratory failure, leaving lung transplantation the only viable option for survival. As incidence and mortality rates steadily increase, the need for novel therapeutics is imperative. The receptor for advanced glycation endproducts (RAGE) is most highly expressed in the lungs and plays a significant role in a number of chronic lung diseases. RAGE has long been linked to IPF; however, confounding data from both human and experimental studies have left an incomplete and perplexing story. This review examines the present understanding of the role of RAGE in human and experimental models of IPF, drawing parallels to recent advances in RAGE biology. Moreover, this review discusses the role of RAGE in lung injury response, type 2 immunity, and cellular senescence, and how such mechanisms may relate to RAGE as both a biomarker of disease progression and potential therapeutic target in IPF.The reviews of this paper are available via the supplemental material section.
Collapse
Affiliation(s)
- Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, 3550 Terrace Street, S-784 Scaife Hall, Pittsburgh, PA 15261, USA
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
99
|
Sun YB, Liu M, Fan XS, Zhou LP, Li MW, Hu FY, Yue QF, Zhang YM. Effects of cigarette smoke on the aggravation of ovalbumin-induced asthma and the expressions of TRPA1 and tight junctions in mice. Mol Immunol 2021; 135:62-72. [PMID: 33873095 DOI: 10.1016/j.molimm.2021.04.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 03/29/2021] [Accepted: 04/06/2021] [Indexed: 10/21/2022]
Abstract
The occurrence of asthma is closely related to environmental factors such as cigarette smoke (CS), one of the common risk factors. Environmental stimuli have the potential to activate transient receptor potential ankyrin 1 (TRPA1) and cause or aggravate asthma. The destruction of tight junctions (TJs) between airway epithelial cells by environmental stimuli in asthma has been researched. It is worth exploring whether CS can injury TJs and aggravate asthma by activating TRPA1. The objective of this study was to investigate the aggravation of CS on ovalbumin (OVA)-induced asthma related phenotypes and TJs expression in mice, and to explore the relationship between TRPA1 and the expression of TJs protein. Female wild type (WT) C57BL/6 mice, induced by OVA, CS and OVA plus CS (OVA + CS) respectively, were used to establish a 42-day asthma model, and mice with TRPA1 knockout (TRPA1-/-) were treated in the same way. This study detected the number of inflammatory cells in peripheral blood and bronchoalveolar lavage fluid (BALF), the levels of IL-4, IL-5, IL-13 in BALF, enhanced pause (Penh) of lung function, pathological changes and the gene and protein expressions of TRPA1 and TJs (including ZO-1, Occludin and Claudin-2) in lung tissues. Compared with normal saline (NS) group, WT mice in the OVA group and OVA + CS group were significantly higher in asthma related phenotypes. The WT-OVA + CS group also showed higher Penh value, levels of IL-5 and IL-13 in BALF and lung tissue injury scores when compared with the WT-OVA group and WT-CS group. However, WT-OVA + CS group mice had significantly larger number of neutrophils in BALF than the WT-OVA group, and had larger number of eosinophils in peripheral blood and higher levels of IL-4 in BALF than the WT-CS group. Meanwhile, compared with the WT-NS group, the expressions of TRPA1 and Claudin-2 in lung tissues increased in other three groups while their expressions of ZO-1 and Occludin decreased, among which, the WT-OVA + CS group showed more remarkable changes. Compared with the WT-OVA + CS group, mice in the TRPA1-/--OVA + CS showed a significant decrease in the number of inflammatory cells, levels of IL-4, IL-5 and IL-13 in BALF, Penh value and lung tissue injury score, and a downregulation of Claudin-2 expression while an upregulation of ZO-1 and Occludin expressions. In addition, the airway inflammation and injury, and the expressions of ZO-1, Occluding and Claudin-2 expressions were found with no statistic differences between TRPA1-/--OVA group and TRPA1-/--OVA + CS group. These results suggest that CS has aggravated the airway inflammation, pathological damage and destruction of TJs in airway epithelium of OVA-induced asthmatic mice, the processes of which are related to the increase of TRPA1 expression.
Collapse
Affiliation(s)
- Yu-Bo Sun
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Mo Liu
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Xin-Sheng Fan
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China; Jiangsu Key Laboratory for High Technology Research of Traditional Chinese Medicine Formulae, Nanjing, 210023, Jiangsu Province, China.
| | - Li-Ping Zhou
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Meng-Wen Li
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Fang-Yuan Hu
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Qin-Fei Yue
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Yi-Ming Zhang
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| |
Collapse
|
100
|
Xie M, Park D, Sica GL, Deng X. Bcl2-induced DNA replication stress promotes lung carcinogenesis in response to space radiation. Carcinogenesis 2021; 41:1565-1575. [PMID: 32157295 DOI: 10.1093/carcin/bgaa021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 02/18/2020] [Accepted: 03/05/2020] [Indexed: 11/12/2022] Open
Abstract
Space radiation is characterized by high-linear energy transfer (LET) ionizing radiation. The relationships between the early biological effects of space radiation and the probability of cancer in humans are poorly understood. Bcl2 not only functions as a potent antiapoptotic molecule but also as an oncogenic protein that induces DNA replication stress. To test the role and mechanism of Bcl2 in high-LET space radiation-induced lung carcinogenesis, we created lung-targeting Bcl2 transgenic C57BL/6 mice using the CC10 promoter to drive Bcl2 expression selectively in lung tissues. Intriguingly, lung-targeting transgenic Bcl2 inhibits ribonucleotide reductase activity, reduces dNTP pool size and retards DNA replication fork progression in mouse bronchial epithelial cells. After exposure of mice to space radiation derived from 56iron, 28silicon or protons, the incidence of lung cancer was significantly higher in lung-targeting Bcl2 transgenic mice than in wild-type mice, indicating that Bcl2-induced DNA replication stress promotes lung carcinogenesis in response to space radiation. The findings provide some evidence for the relative effectiveness of space radiation and Bcl-2 at inducing lung cancer in mice.
Collapse
Affiliation(s)
- Maohua Xie
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Dongkyoo Park
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Gabriel L Sica
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Xingming Deng
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|