1
|
Boucher M, Henry C, Gélinas L, Packwood R, Rojas-Ruiz A, Fereydoonzad L, Graham P, Bossé Y. High throughput screening of airway constriction in mouse lung slices. Sci Rep 2024; 14:20133. [PMID: 39210022 PMCID: PMC11362152 DOI: 10.1038/s41598-024-71170-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The level of airway constriction in thin slices of lung tissue is highly variable. Owing to the labor-intensive nature of these experiments, determining the number of airways to be analyzed in order to allocate a reliable value of constriction in one mouse is challenging. Herein, a new automated device for physiology and image analysis was used to facilitate high throughput screening of airway constriction in lung slices. Airway constriction was first quantified in slices of lungs from male BALB/c mice with and without experimental asthma that were inflated with agarose through the trachea or trans-parenchymal injections. Random sampling simulations were then conducted to determine the number of airways required per mouse to quantify maximal constriction. The constriction of 45 ± 12 airways per mouse in 32 mice were analyzed. Mean maximal constriction was 37.4 ± 32.0%. The agarose inflating technique did not affect the methacholine response. However, the methacholine constriction was affected by experimental asthma (p = 0.003), shifting the methacholine concentration-response curve to the right, indicating a decreased sensitivity. Simulations then predicted that approximately 35, 16 and 29 airways per mouse are needed to quantify the maximal constriction mean, standard deviation and coefficient of variation, respectively; these numbers varying between mice and with experimental asthma.
Collapse
Affiliation(s)
- Magali Boucher
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ)-Université Laval, Pavillon M, room 2687, 2725, chemin Sainte-Foy, Quebec, Qc, G1V 4G5, Canada
| | - Cyndi Henry
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ)-Université Laval, Pavillon M, room 2687, 2725, chemin Sainte-Foy, Quebec, Qc, G1V 4G5, Canada
| | - Louis Gélinas
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ)-Université Laval, Pavillon M, room 2687, 2725, chemin Sainte-Foy, Quebec, Qc, G1V 4G5, Canada
| | - Rosalie Packwood
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ)-Université Laval, Pavillon M, room 2687, 2725, chemin Sainte-Foy, Quebec, Qc, G1V 4G5, Canada
| | - Andrés Rojas-Ruiz
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ)-Université Laval, Pavillon M, room 2687, 2725, chemin Sainte-Foy, Quebec, Qc, G1V 4G5, Canada
| | | | | | - Ynuk Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ)-Université Laval, Pavillon M, room 2687, 2725, chemin Sainte-Foy, Quebec, Qc, G1V 4G5, Canada.
| |
Collapse
|
2
|
Wu D, Zhang X, Zimmerly KM, Wang R, Livingston A, Iwawaki T, Kumar A, Wu X, Campen M, Mandell MA, Liu M, Yang XO. Unconventional Activation of IRE1 Enhances Th17 Responses and Promotes Airway Neutrophilia. Am J Respir Cell Mol Biol 2024; 71:169-181. [PMID: 38593442 PMCID: PMC11299091 DOI: 10.1165/rcmb.2023-0424oc] [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: 12/01/2023] [Accepted: 04/09/2024] [Indexed: 04/11/2024] Open
Abstract
Heightened unfolded protein responses (UPRs) are associated with the risk for asthma, including severe asthma. Treatment-refractory severe asthma manifests a neutrophilic phenotype with T helper (Th)17 responses. However, how UPRs participate in the deregulation of Th17 cells leading to neutrophilic asthma remains elusive. This study found that the UPR sensor IRE1 is induced in the murine lung with fungal asthma and is highly expressed in Th17 cells relative to naive CD4+ T cells. Cytokine (e.g., IL-23) signals induce the IRE1-XBP1s axis in a JAK2-dependent manner. This noncanonical activation of the IRE1-XBP1s pathway promotes UPRs and cytokine secretion by both human and mouse Th17 cells. Ern1 (encoding IRE1) deficiency decreases the expression of endoplasmic reticulum stress factors and impairs the differentiation and cytokine secretion of Th17 cells. Genetic ablation of Ern1 leads to alleviated Th17 responses and airway neutrophilia in a fungal airway inflammation model. Consistently, IL-23 activates the JAK2-IRE1-XBP1s pathway in vivo and enhances Th17 responses and neutrophilic infiltration into the airway. Taken together, our data indicate that IRE1, noncanonically activated by cytokine signals, promotes neutrophilic airway inflammation through the UPR-mediated secretory function of Th17 cells. The findings provide a novel insight into the fundamental understanding of IRE1 in Th17-biased TH2-low asthma.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Molecular Genetics and Microbiology and
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, and
| | | | - Ruoning Wang
- Department of Molecular Genetics and Microbiology and
| | | | - Takao Iwawaki
- Division of Cell Medicine, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, Texas; and
| | - Xiang Wu
- Department of Molecular Genetics and Microbiology and
- Department of Parasitology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha, China
| | - Matthew Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico
| | | | - Meilian Liu
- Department of Biochemistry and Molecular Biology, School of Medicine, and
| | | |
Collapse
|
3
|
Liu T, Woodruff PG, Zhou X. Advances in non-type 2 severe asthma: from molecular insights to novel treatment strategies. Eur Respir J 2024; 64:2300826. [PMID: 38697650 PMCID: PMC11325267 DOI: 10.1183/13993003.00826-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
Asthma is a prevalent pulmonary disease that affects more than 300 million people worldwide and imposes a substantial economic burden. While medication can effectively control symptoms in some patients, severe asthma attacks, driven by airway inflammation induced by environmental and infectious exposures, continue to be a major cause of asthma-related mortality. Heterogeneous phenotypes of asthma include type 2 (T2) and non-T2 asthma. Non-T2 asthma is often observed in patients with severe and/or steroid-resistant asthma. This review covers the molecular mechanisms, clinical phenotypes, causes and promising treatments of non-T2 severe asthma. Specifically, we discuss the signalling pathways for non-T2 asthma including the activation of inflammasomes, interferon responses and interleukin-17 pathways, and their contributions to the subtypes, progression and severity of non-T2 asthma. Understanding the molecular mechanisms and genetic determinants underlying non-T2 asthma could form the basis for precision medicine in severe asthma treatment.
Collapse
Affiliation(s)
- Tao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, China
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Leslie E, Miller M, Lafuze A, Svyatskaya S, Choi GS, Broide DH. PGAP3 regulates human bronchial epithelial cell mRNAs present in asthma and respiratory virus reference data sets. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.03.24309917. [PMID: 39006429 PMCID: PMC11245055 DOI: 10.1101/2024.07.03.24309917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
PGAP3 is a glycosylphosphatidylinositol (GPI) phospholipase gene localized within chromosome 17q12-21, a region highly linked to asthma. Although much is known about the function of other chromosome 17q12-21 genes expressed at increased levels in bronchial epithelium such as ORMDL3 and GSDMB, little is known about the function of increased PGAP3 expression in bronchial epithelium in the context of asthma. The aim of this study was therefore to determine whether increased PGAP3 expression in human bronchial epithelial cells regulated expression of mRNA pathways important to the pathogenesis of asthma by utilizing RNA-sequencing and bioinformatic analysis. We performed RNA-sequencing on normal human bronchial epithelial cells transfected with PGAP3 for 24 and 48 hours. PGAP3 regulated genes were compared to asthma and respiratory virus (influenza A, rhinovirus, respiratory syncytial virus) reference data sets to identify PGAP3 target genes and pathways. Approximately 9% of the upregulated PGAP3-induced genes were found in an asthma reference data set, 41% in a rhinovirus reference data set, 33% in an influenza A reference data set, and 3% in a respiratory syncytial virus reference data set. PGAP3 significantly upregulated the expression of several genes associated with the innate immune response and viral signatures of respiratory viruses associated with asthma exacerbations. Two of the highest expressed genes induced by PGAP3 are RSAD2, OASL, and IFN-λ, which are anti-viral genes associated with asthma. PGAP3 also upregulated the antiviral gene BST2, which like PGAP3 is a GPI-anchored protein. We conclude that PGAP3 expression in human bronchial epithelial cells regulates expression of genes known to be linked to asthma, and also regulates the bronchial epithelial expression of genes pertinent to the pathogenesis of respiratory viral triggered asthma exacerbations.
Collapse
Affiliation(s)
- Eric Leslie
- Department of Medicine, University of California, San Diego; La Jolla, California, USA
| | - Marina Miller
- Department of Medicine, University of California, San Diego; La Jolla, California, USA
| | - Allison Lafuze
- Department of Medicine, University of California, San Diego; La Jolla, California, USA
| | - Sofya Svyatskaya
- Department of Medicine, University of California, San Diego; La Jolla, California, USA
| | - Gil-Soon Choi
- Department of Medicine, University of California, San Diego; La Jolla, California, USA
- Department of Internal Medicine, Kosin University College of Medicine, Republic of Korea
| | - David H Broide
- Department of Medicine, University of California, San Diego; La Jolla, California, USA
| |
Collapse
|
5
|
Koziol-White C, Gebski E, Cao G, Panettieri RA. Precision cut lung slices: an integrated ex vivo model for studying lung physiology, pharmacology, disease pathogenesis and drug discovery. Respir Res 2024; 25:231. [PMID: 38824592 PMCID: PMC11144351 DOI: 10.1186/s12931-024-02855-6] [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: 04/08/2024] [Accepted: 05/18/2024] [Indexed: 06/03/2024] Open
Abstract
Precision Cut Lung Slices (PCLS) have emerged as a sophisticated and physiologically relevant ex vivo model for studying the intricacies of lung diseases, including fibrosis, injury, repair, and host defense mechanisms. This innovative methodology presents a unique opportunity to bridge the gap between traditional in vitro cell cultures and in vivo animal models, offering researchers a more accurate representation of the intricate microenvironment of the lung. PCLS require the precise sectioning of lung tissue to maintain its structural and functional integrity. These thin slices serve as invaluable tools for various research endeavors, particularly in the realm of airway diseases. By providing a controlled microenvironment, precision-cut lung slices empower researchers to dissect and comprehend the multifaceted interactions and responses within lung tissue, thereby advancing our understanding of pulmonary pathophysiology.
Collapse
Affiliation(s)
- Cynthia Koziol-White
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA.
| | - Eric Gebski
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| | - Gaoyaun Cao
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| |
Collapse
|
6
|
Song X, Liang J, Lin S, Xie Y, Ke C, Ao D, Lu J, Chen X, He Y, Liu X, Li W. Gut-lung axis and asthma: A historical review on mechanism and future perspective. Clin Transl Allergy 2024; 14:e12356. [PMID: 38687096 PMCID: PMC11060082 DOI: 10.1002/clt2.12356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 03/24/2024] [Accepted: 04/17/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Gut microbiota are closely related to the development and regulation of the host immune system by regulating the maturation of immune cells and the resistance to pathogens, which affects the host immunity. Early use of antibiotics disrupts the homeostasis of gut microbiota and increases the risk of asthma. Gut microbiota actively interact with the host immune system via the gut-lung axis, a bidirectional communication pathway between the gut and lung. The manipulation of gut microbiota through probiotics, helminth therapy, and fecal microbiota transplantation (FMT) to combat asthma has become a hot research topic. BODY: This review mainly describes the current immune pathogenesis of asthma, gut microbiota and the role of the gut-lung axis in asthma. Moreover, the potential of manipulating the gut microbiota and its metabolites as a treatment strategy for asthma has been discussed. CONCLUSION The gut-lung axis has a bidirectional effect on asthma. Gut microecology imbalance contributes to asthma through bacterial structural components and metabolites. Asthma, in turn, can also cause intestinal damage through inflammation throughout the body. The manipulation of gut microbiota through probiotics, helminth therapy, and FMT can inform the treatment strategies for asthma by regulating the maturation of immune cells and the resistance to pathogens.
Collapse
Affiliation(s)
- Xiu‐Ling Song
- Department of PediatricsAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Juan Liang
- Department of PediatricsAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Shao‐Zhu Lin
- Department of PediatricsAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Yu‐Wei Xie
- Department of PediatricsAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Chuang‐Hong Ke
- Department of PediatricsAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Dang Ao
- Department of PediatricsAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Jun Lu
- Department of PediatricsAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Xue‐Mei Chen
- Department of PediatricsAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Ying‐Zhi He
- Department of PediatricsAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Xiao‐Hua Liu
- Department of PediatricsAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| | - Wen Li
- Department of PediatricsAffiliated Hospital of Guangdong Medical UniversityZhanjiangChina
| |
Collapse
|
7
|
Jakwerth CA, Weckmann M, Illi S, Charles H, Zissler UM, Oelsner M, Guerth F, Omony J, Nemani SSP, Grychtol R, Dittrich AM, Skevaki C, Foth S, Weber S, Alejandre Alcazar MA, van Koningsbruggen-Rietschel S, Brock R, Blau S, Hansen G, Bahmer T, Rabe KF, Brinkmann F, Kopp MV, Chaker AM, Schaub B, von Mutius E, Schmidt-Weber CB. 17q21 Variants Disturb Mucosal Host Defense in Childhood Asthma. Am J Respir Crit Care Med 2024; 209:947-959. [PMID: 38064241 PMCID: PMC11531215 DOI: 10.1164/rccm.202305-0934oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 12/07/2023] [Indexed: 03/13/2024] Open
Abstract
Rationale: The strongest genetic risk factor for childhood-onset asthma, the 17q21 locus, is associated with increased viral susceptibility and disease-promoting processes.Objectives: To identify biological targets underlying the escalated viral susceptibility associated with the clinical phenotype mediated by the 17q21 locus.Methods: Genome-wide transcriptome analysis of nasal brush samples from 261 children (78 healthy, 79 with wheezing at preschool age, 104 asthmatic) within the ALLIANCE (All-Age-Asthma) cohort, with a median age of 10.0 (range, 1.0-20.0) years, was conducted to explore the impact of their 17q21 genotype (SNP rs72163891). Concurrently, nasal secretions from the same patients and visits were collected, and high-sensitivity mesoscale technology was employed to measure IFN protein levels.Measurements and Main Results: This study revealed that the 17q21 risk allele induces a genotype- and asthma/wheeze phenotype-dependent enhancement of mucosal GSDMB expression as the only relevant 17q21-encoded gene in children with preschool wheeze. Increased GSDMB expression correlated with the activation of a type-1 proinflammatory, cell-lytic immune, and natural killer signature, encompassing key genes linked to an IFN type-2-signature (IFNG, CXCL9, CXCL10, KLRC1, CD8A, GZMA). Conversely, there was a reduction in IFN type 1 and type 3 expression signatures at the mRNA and protein levels.Conclusions: This study demonstrates a novel disease-driving mechanism induced by the 17q21 risk allele. Increased mucosal GSDMB expression is associated with a cell-lytic immune response coupled with compromised airway immunocompetence. These findings suggest that GSDMB-related airway cell death and perturbations in the mucosal IFN signature account for the increased vulnerability of 17q21 risk allele carriers to respiratory viral infections during early life, opening new options for future biological interventions.The All-Age-Asthma (ALLIANCE) cohort is registered at www.clinicaltrials.gov (pediatric arm, NCT02496468).
Collapse
Affiliation(s)
- Constanze A. Jakwerth
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Germany
| | - Markus Weckmann
- Member of the German Center for Lung Research (DZL), Germany
- Division of Epigenetics in Chronic Lung Disease, Priority Area Chronic Lung Diseases, Research Center Borstel-Leibniz Lung Center, Borstel, Germany
- Department of Pediatric Pneumology and Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Airway Research Center North, Borstel, Lübeck, Kiel, Grosshansdorf, Germany
| | - Sabina Illi
- Member of the German Center for Lung Research (DZL), Germany
- Institute for Asthma and Allergy Prevention, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
- Comprehensive Pneumology Center–Munich, Munich, Germany
| | - Helen Charles
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Germany
| | - Ulrich M. Zissler
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Germany
| | - Madlen Oelsner
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Germany
| | - Ferdinand Guerth
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Germany
| | - Jimmy Omony
- Member of the German Center for Lung Research (DZL), Germany
- Institute for Asthma and Allergy Prevention, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
- Comprehensive Pneumology Center–Munich, Munich, Germany
| | - Sai Sneha Priya Nemani
- Member of the German Center for Lung Research (DZL), Germany
- Department of Pediatric Pneumology and Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Airway Research Center North, Borstel, Lübeck, Kiel, Grosshansdorf, Germany
| | - Ruth Grychtol
- Member of the German Center for Lung Research (DZL), Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Hanover, Germany
| | - Anna-Maria Dittrich
- Member of the German Center for Lung Research (DZL), Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Hanover, Germany
| | - Chrysanthi Skevaki
- Member of the German Center for Lung Research (DZL), Germany
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics and
| | - Svenja Foth
- Member of the German Center for Lung Research (DZL), Germany
- Universities of Giessen and Marburg Lung Center, Philipps University Marburg and University Children’s Hospital Marburg, University of Marburg, Marburg, Germany
| | - Stefanie Weber
- Member of the German Center for Lung Research (DZL), Germany
- Universities of Giessen and Marburg Lung Center, Philipps University Marburg and University Children’s Hospital Marburg, University of Marburg, Marburg, Germany
| | - Miguel A. Alejandre Alcazar
- Member of the German Center for Lung Research (DZL), Germany
- Institute for Lung Health and Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Giessen, Germany
- Translational Experimental Pediatrics, Experimental Pulmonology, Department of Pediatrics
- Center for Molecular Medicine Cologne and Cologne Excellence Cluster on Stress Responses in Aging-associated Diseases, and
- Pediatric Pulmonology and Allergology, Department of Pediatrics, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany; and
| | - Silke van Koningsbruggen-Rietschel
- Member of the German Center for Lung Research (DZL), Germany
- Pediatric Pulmonology and Allergology, Department of Pediatrics, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany; and
| | - Robert Brock
- Member of the German Center for Lung Research (DZL), Germany
- Pediatric Pulmonology and Allergology, Department of Pediatrics, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany; and
| | - Samira Blau
- Member of the German Center for Lung Research (DZL), Germany
- Pediatric Pulmonology and Allergology, Department of Pediatrics, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany; and
| | - Gesine Hansen
- Member of the German Center for Lung Research (DZL), Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Hanover, Germany
- Cluster of Excellence 2115 (RESIST), Hannover Medical School, Hanover, Germany
| | - Thomas Bahmer
- Member of the German Center for Lung Research (DZL), Germany
- Airway Research Center North, Borstel, Lübeck, Kiel, Grosshansdorf, Germany
- Internal Medicine Department I, University Hospital Schleswig-Holstein–Campus Kiel, Kiel, Germany
| | - Klaus F. Rabe
- Member of the German Center for Lung Research (DZL), Germany
- Airway Research Center North, Borstel, Lübeck, Kiel, Grosshansdorf, Germany
- LungenClinic Grosshansdorf GmbH and Medical Clinics, Christian Albrechts University, Kiel, Germany
| | - Folke Brinkmann
- Member of the German Center for Lung Research (DZL), Germany
- Division of Epigenetics in Chronic Lung Disease, Priority Area Chronic Lung Diseases, Research Center Borstel-Leibniz Lung Center, Borstel, Germany
- Department of Pediatric Pneumology and Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Airway Research Center North, Borstel, Lübeck, Kiel, Grosshansdorf, Germany
| | - Matthias Volkmar Kopp
- Department of Pediatric Pneumology and Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Pediatric Respiratory Medicine, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Airway Research Center North, Borstel, Lübeck, Kiel, Grosshansdorf, Germany
| | - Adam M. Chaker
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
| | - Bianca Schaub
- Member of the German Center for Lung Research (DZL), Germany
- Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, Munich, Germany
- Comprehensive Pneumology Center–Munich, Munich, Germany
| | - Erika von Mutius
- Member of the German Center for Lung Research (DZL), Germany
- Institute for Asthma and Allergy Prevention, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
- Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, Munich, Germany
| | - Carsten B. Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Germany
| | - the ALLIANCE Study Group as part of the German Center for Lung Research
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Germany
- Institute for Asthma and Allergy Prevention, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
- Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, Munich, Germany
- Comprehensive Pneumology Center–Munich, Munich, Germany
- Division of Epigenetics in Chronic Lung Disease, Priority Area Chronic Lung Diseases, Research Center Borstel-Leibniz Lung Center, Borstel, Germany
- Department of Pediatric Pneumology and Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
- Pediatric Respiratory Medicine, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Airway Research Center North, Borstel, Lübeck, Kiel, Grosshansdorf, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Hanover, Germany
- Internal Medicine Department I, University Hospital Schleswig-Holstein–Campus Kiel, Kiel, Germany
- LungenClinic Grosshansdorf GmbH and Medical Clinics, Christian Albrechts University, Kiel, Germany
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics and
- Institute for Lung Health and Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Giessen, Germany
- Universities of Giessen and Marburg Lung Center, Philipps University Marburg and University Children’s Hospital Marburg, University of Marburg, Marburg, Germany
- Translational Experimental Pediatrics, Experimental Pulmonology, Department of Pediatrics
- Center for Molecular Medicine Cologne and Cologne Excellence Cluster on Stress Responses in Aging-associated Diseases, and
- Pediatric Pulmonology and Allergology, Department of Pediatrics, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany; and
- Cluster of Excellence 2115 (RESIST), Hannover Medical School, Hanover, Germany
| |
Collapse
|
8
|
Listyoko AS, Okazaki R, Harada T, Inui G, Yamasaki A. Impact of obesity on airway remodeling in asthma: pathophysiological insights and clinical implications. FRONTIERS IN ALLERGY 2024; 5:1365801. [PMID: 38562155 PMCID: PMC10982419 DOI: 10.3389/falgy.2024.1365801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
The prevalence of obesity among asthma patients has surged in recent years, posing a significant risk factor for uncontrolled asthma. Beyond its impact on asthma severity and patients' quality of life, obesity is associated with reduced lung function, increased asthma exacerbations, hospitalizations, heightened airway hyperresponsiveness, and elevated asthma-related mortality. Obesity may lead to metabolic dysfunction and immune dysregulation, fostering chronic inflammation characterized by increased pro-inflammatory mediators and adipocytokines, elevated reactive oxygen species, and reduced antioxidant activity. This chronic inflammation holds the potential to induce airway remodeling in individuals with asthma and obesity. Airway remodeling encompasses structural and pathological changes, involving alterations in the airway's epithelial and subepithelial layers, hyperplasia and hypertrophy of airway smooth muscle, and changes in airway vascularity. In individuals with asthma and obesity, airway remodeling may underlie heightened airway hyperresponsiveness and increased asthma severity, ultimately contributing to the development of persistent airflow limitation, declining lung function, and a potential increase in asthma-related mortality. Despite efforts to address the impact of obesity on asthma outcomes, the intricate mechanisms linking obesity to asthma pathophysiology, particularly concerning airway remodeling, remain incompletely understood. This comprehensive review discusses current research investigating the influence of obesity on airway remodeling, to enhance our understanding of obesity's role in the context of asthma airway remodeling.
Collapse
Affiliation(s)
- Aditya Sri Listyoko
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
- Pulmonology and Respiratory Medicine Department, Faculty of Medicine, Brawijaya University-Dr. Saiful Anwar General Hospital, Malang, Indonesia
| | - Ryota Okazaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Tomoya Harada
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Genki Inui
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Akira Yamasaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
9
|
Antonisamy B, Shailesh H, Hani Y, Ahmed LHM, Noor S, Ahmed SY, Alfaki M, Muhayimana A, Jacob SS, Balayya SK, Soloviov O, Liu L, Mathew LS, Wang K, Tomei S, Al Massih A, Mathew R, Karim MY, Ramanjaneya M, Worgall S, Janahi IA. Sphingolipids in Childhood Asthma and Obesity (SOAP Study): A Protocol of a Cross-Sectional Study. Metabolites 2023; 13:1146. [PMID: 37999242 PMCID: PMC10673587 DOI: 10.3390/metabo13111146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/25/2023] Open
Abstract
Asthma and obesity are two of the most common chronic conditions in children and adolescents. There is increasing evidence that sphingolipid metabolism is altered in childhood asthma and is linked to airway hyperreactivity. Dysregulated sphingolipid metabolism is also reported in obesity. However, the functional link between sphingolipid metabolism, asthma, and obesity is not completely understood. This paper describes the protocol of an ongoing study on sphingolipids that aims to examine the pathophysiology of sphingolipids in childhood asthma and obesity. In addition, this study aims to explore the novel biomarkers through a comprehensive multi-omics approach including genomics, genome-wide DNA methylation, RNA-Seq, microRNA (miRNA) profiling, lipidomics, metabolomics, and cytokine profiling. This is a cross-sectional study aiming to recruit 440 children from different groups: children with asthma and normal weight (n = 100), asthma with overweight or obesity (n = 100), overweight or obesity (n = 100), normal weight (n = 70), and siblings of asthmatic children with normal weight, overweight, or obesity (n = 70). These participants will be recruited from the pediatric pulmonology, pediatric endocrinology, and general pediatric outpatient clinics at Sidra Medicine, Doha, Qatar. Information will be obtained from self-reported questionnaires on asthma, quality of life, food frequency (FFQ), and a 3-day food diary that are completed by the children and their parents. Clinical measurements will include anthropometry, blood pressure, biochemistry, bioelectrical impedance, and pulmonary function tests. Blood samples will be obtained for sphingolipid analysis, serine palmitoyltransferase (SPT) assay, whole-genome sequencing (WGS), genome-wide DNA methylation study, RNA-Seq, miRNA profiling, metabolomics, lipidomics, and cytokine analysis. Group comparisons of continuous outcome variables will be carried out by a one-way analysis of variance or the Kruskal-Wallis test using an appropriate pairwise multiple comparison test. The chi-squared test or a Fisher's exact test will be used to test the associations between categorical variables. Finally, multivariate analysis will be carried out to integrate the clinical data with multi-omics data. This study will help us to understand the role of dysregulated sphingolipid metabolism in obesity and asthma. In addition, the multi-omics data from the study will help to identify novel genetic and epigenetic signatures, inflammatory markers, and mechanistic pathways that link asthma and obesity in children. Furthermore, the integration of clinical and multi-omics data will help us to uncover the potential interactions between these diseases and to offer a new paradigm for the treatment of pediatric obesity-associated asthma.
Collapse
Affiliation(s)
- Belavendra Antonisamy
- Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar; (B.A.); (H.S.); (Y.H.); (L.H.M.A.); (S.N.); (S.Y.A.); (M.A.); (A.M.)
| | - Harshita Shailesh
- Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar; (B.A.); (H.S.); (Y.H.); (L.H.M.A.); (S.N.); (S.Y.A.); (M.A.); (A.M.)
| | - Yahya Hani
- Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar; (B.A.); (H.S.); (Y.H.); (L.H.M.A.); (S.N.); (S.Y.A.); (M.A.); (A.M.)
| | - Lina Hayati M. Ahmed
- Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar; (B.A.); (H.S.); (Y.H.); (L.H.M.A.); (S.N.); (S.Y.A.); (M.A.); (A.M.)
| | - Safa Noor
- Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar; (B.A.); (H.S.); (Y.H.); (L.H.M.A.); (S.N.); (S.Y.A.); (M.A.); (A.M.)
| | - Salma Yahya Ahmed
- Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar; (B.A.); (H.S.); (Y.H.); (L.H.M.A.); (S.N.); (S.Y.A.); (M.A.); (A.M.)
| | - Mohamed Alfaki
- Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar; (B.A.); (H.S.); (Y.H.); (L.H.M.A.); (S.N.); (S.Y.A.); (M.A.); (A.M.)
| | - Abidan Muhayimana
- Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar; (B.A.); (H.S.); (Y.H.); (L.H.M.A.); (S.N.); (S.Y.A.); (M.A.); (A.M.)
| | - Shana Sunny Jacob
- Analytical Chemistry Core, Advanced Diagnostic Core Facilities, Sidra Medicine, Doha P.O. Box 26999, Qatar; (S.S.J.); (S.K.B.)
| | - Saroja Kotegar Balayya
- Analytical Chemistry Core, Advanced Diagnostic Core Facilities, Sidra Medicine, Doha P.O. Box 26999, Qatar; (S.S.J.); (S.K.B.)
| | - Oleksandr Soloviov
- Clinical Genomics Laboratory, Integrated Genomics Services, Sidra Medicine, Doha P.O. Box 26999, Qatar; (O.S.); (L.L.); (L.S.M.); (K.W.)
| | - Li Liu
- Clinical Genomics Laboratory, Integrated Genomics Services, Sidra Medicine, Doha P.O. Box 26999, Qatar; (O.S.); (L.L.); (L.S.M.); (K.W.)
| | - Lisa Sara Mathew
- Clinical Genomics Laboratory, Integrated Genomics Services, Sidra Medicine, Doha P.O. Box 26999, Qatar; (O.S.); (L.L.); (L.S.M.); (K.W.)
| | - Kun Wang
- Clinical Genomics Laboratory, Integrated Genomics Services, Sidra Medicine, Doha P.O. Box 26999, Qatar; (O.S.); (L.L.); (L.S.M.); (K.W.)
| | - Sara Tomei
- Omics Core, Integrated Genomics Services, Sidra Medicine, Doha P.O. Box 26999, Qatar; (S.T.); (A.A.M.); (R.M.)
| | - Alia Al Massih
- Omics Core, Integrated Genomics Services, Sidra Medicine, Doha P.O. Box 26999, Qatar; (S.T.); (A.A.M.); (R.M.)
| | - Rebecca Mathew
- Omics Core, Integrated Genomics Services, Sidra Medicine, Doha P.O. Box 26999, Qatar; (S.T.); (A.A.M.); (R.M.)
| | - Mohammed Yousuf Karim
- Department of Pathology, Sidra Medicine, Doha P.O. Box 26999, Qatar;
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar
| | - Manjunath Ramanjaneya
- Qatar Metabolic Institute, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar;
- Translational Research Institute, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar
| | - Stefan Worgall
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10021, USA;
| | - Ibrahim A. Janahi
- Department of Pediatric Medicine, Sidra Medicine, Doha P.O. Box 26999, Qatar; (B.A.); (H.S.); (Y.H.); (L.H.M.A.); (S.N.); (S.Y.A.); (M.A.); (A.M.)
- Department of Pediatrics, Weill Cornell Medicine-Qatar, Doha P.O. Box 24144, Qatar
| |
Collapse
|
10
|
Zhang Y. From gene identifications to therapeutic targets for asthma: Focus on great potentials of TSLP, ORMDL3, and GSDMB. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:139-147. [PMID: 39171126 PMCID: PMC11332877 DOI: 10.1016/j.pccm.2023.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Indexed: 08/23/2024]
Abstract
Asthma is a chronic respiratory disease, and clinically, asthma exacerbations remain difficult to treat. The disease is caused by combinations of and interactions between genetic and environmental factors. Genomic and genetic approaches identified many novel genes to treat asthma and brought new insights into the disease. The products of the genes have functional roles in regulating physiological or pathophysiological processes in airway structural cells and immune system cells. Genetic factors also interact with environmental factors such as air pollutants, and bacterial and viral infections to trigger the disease. Thymic stromal lymphopoietin (TSLP), orosomucoid-like 3 (ORMDL3), and gasdermin B (GSDMB) are three genes identified by genetic studies to have a great potential as therapeutic targets of asthma. TSLP is an important driver of type 2 inflammation. ORMDL3 mediates cell stress, sphingolipid synthesis, and viral and bacterial infections. GSDMB regulates cell pyroptosis through its N and C terminals and can bind sulfatides to influence inflammatory response. Investigating inhibitors or modulators for these pathways would bring a new landscape for therapeutics of asthma in future.
Collapse
Affiliation(s)
- Youming Zhang
- National Heart and Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK
| |
Collapse
|
11
|
Wu D, Zhang X, Zimmerly KM, Wang R, Wang C, Hunter R, Wu X, Campen M, Liu M, Yang XO. Unfolded protein response factor ATF6 augments T helper cell responses and promotes mixed granulocytic airway inflammation. Mucosal Immunol 2023; 16:499-512. [PMID: 37209959 PMCID: PMC10530451 DOI: 10.1016/j.mucimm.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/11/2023] [Indexed: 05/22/2023]
Abstract
The unfolded protein response (UPR) is associated with the risk of asthma, including treatment-refractory severe asthma. Recent studies demonstrated a pathogenic role of activating transcription factor 6a (ATF6a or ATF6), an essential UPR sensor, in airway structural cells. However, its role in T helper (TH) cells has not been well examined. In this study, we found that ATF6 was selectively induced by signal transducer and activator of transcription6 (STAT6) and STAT3 in TH2 and TH17 cells, respectively. ATF6 upregulated UPR genes and promoted the differentiation and cytokine secretion of TH2 and TH17 cells. T cell-specific Atf6-deficiency impaired TH2 and TH17 responses in vitro and in vivo and attenuated mixed granulocytic experimental asthma. ATF6 inhibitor Ceapin A7 suppressed the expression of ATF6 downstream genes and TH cell cytokines by both murine and human memory clusters of differentiation 4 (CD4)+ T cells. At the chronic stage of asthma, administration of Ceapin A7 lessened TH2 and TH17 responses, leading to alleviation of both airway neutrophilia and eosinophilia. Thus, our results demonstrate a critical role of ATF6 in TH2 and TH17 cell-driven mixed granulocytic airway disease, suggesting a novel option to combat steroid-resistant mixed and even T2-low endotypes of asthma by targeting ATF6.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Kourtney M Zimmerly
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Ruoning Wang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Chunqing Wang
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Russell Hunter
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, USA
| | - Xiang Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA; Department of Parasitology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha, China
| | - Matthew Campen
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, USA
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, USA.
| | - Xuexian O Yang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA.
| |
Collapse
|
12
|
Wu D, Zhang X, Zimmerly KM, Wang R, Livingston A, Iwawaki T, Kumar A, Wu X, Mandell MA, Liu M, Yang XO. Unconventional Activation of IRE1 Enhances TH17 Responses and Promotes Neutrophilic Airway Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.30.547286. [PMID: 37461622 PMCID: PMC10349957 DOI: 10.1101/2023.06.30.547286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Treatment-refractory severe asthma manifests a neutrophilic phenotype associated with TH17 responses. Heightened unfolded protein responses (UPRs) are associated with the risk of asthma, including severe asthma. However, how UPRs participate in the deregulation of TH17 cells leading to this type of asthma remains elusive. In this study, we investigated the role of the UPR sensor IRE1 in TH17 cell function and neutrophilic airway inflammation. We found that IRE1 is induced in fungal asthma and is highly expressed in TH17 cells relative to naïve CD4+ T cells. Cytokine (e.g. IL-23) signals induce the IRE1-XBP1s axis in a JAK2-dependent manner. This noncanonical activation of the IRE1-XBP1s pathway promotes UPRs and cytokine secretion by TH17 cells. Ern1 (encoding IRE1)-deficiency decreases the expression of ER stress factors and impairs the differentiation and cytokine secretion of TH17 cells. Genetic ablation of Ern1 leads to alleviated TH17 responses and airway neutrophilia in a Candida albicans asthma model. Consistently, IL-23 activates the JAK2-IRE1-XBP1s pathway in vivo and enhances TH17 responses and neutrophilic infiltration into the airway. Taken together, our data indicate that IRE1, noncanonically activated by cytokine signals, promotes neutrophilic airway inflammation through the UPRmediated secretory function of TH17 cells. The findings provide a novel insight into the fundamental understanding of IRE1 in TH17-biased TH2-low asthma.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Kourtney M. Zimmerly
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Ruoning Wang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Amanda Livingston
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Takao Iwawaki
- Division of Cell Medicine, Medical Research Institute, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| | - Xiang Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
- Department of Parasitology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha, China
| | - Michael A. Mandell
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Xuexian O. Yang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| |
Collapse
|
13
|
Adrish M, Akuthota P. Approach to non-type 2 asthma. Respir Med 2023:107327. [PMID: 37307904 DOI: 10.1016/j.rmed.2023.107327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 05/21/2023] [Accepted: 06/08/2023] [Indexed: 06/14/2023]
Affiliation(s)
- Muhammad Adrish
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - Praveen Akuthota
- Division of Pulmonary, Critical Care, Sleep Medicine & Physiology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
14
|
Zeng Z, Cheng M, Li M, Wang T, Wen F, Sanderson MJ, Sneyd J, Shen Y, Chen J. Inherent differences of small airway contraction and Ca 2+ oscillations in airway smooth muscle cells between BALB/c and C57BL/6 mouse strains. Front Cell Dev Biol 2023; 11:1202573. [PMID: 37346175 PMCID: PMC10279852 DOI: 10.3389/fcell.2023.1202573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 05/26/2023] [Indexed: 06/23/2023] Open
Abstract
BALB/c and C57BL/6 mouse strains are widely used as animal model in studies of respiratory diseases, such as asthma. Asthma is characterized by airway hyperresponsiveness, which is eventually resulted from the excessive airway smooth muscle (ASM) contraction mediated by Ca2+ oscillations in ASM cells. It is reported that BALB/c mice have inherently higher airway responsiveness, but show no different contractive response of tracheal ring as compared to C57BL/6 mice. However, whether the different airway responsiveness is due to the different extents of small airway contraction, and what's underlying mechanism remains unknown. Here, we assess agonist-induced small airway contraction and Ca2+ oscillations in ASM cells between BALB/c and C57BL/6 mice by using precision-cut lung slices (PCLS). We found that BALB/c mice showed an intrinsically stronger extent of small airway narrowing and faster Ca2+ oscillations in ASM cells in response to agonists. These differences were associated with a higher magnitude of Ca2+ influx via store-operated Ca2+ entry (SOCE), as a result of increased expression of SOCE components (STIM1, Orai1) in the ASM cells of small airway of BALB/c mice. An established mathematical model and experimental results suggested that the increased SOC current could result in increased agonist-induced Ca2+ oscillations. Therefore, the inherently higher SOC underlies the increased Ca2+ oscillation frequency in ASM cells and stronger small airway contraction in BALB/c mice, thus higher airway responsiveness in BALB/c than C57BL/6 mouse strain.
Collapse
Affiliation(s)
- Zijian Zeng
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Chengdu, Sichuan, China
| | - Mengxin Cheng
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Chengdu, Sichuan, China
| | - Meng Li
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Chengdu, Sichuan, China
| | - Tao Wang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Chengdu, Sichuan, China
| | - Fuqiang Wen
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Chengdu, Sichuan, China
| | - Michael J. Sanderson
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - James Sneyd
- Department of Mathematics, The University of Auckland, Auckland, New Zealand
| | - Yongchun Shen
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Chengdu, Sichuan, China
| | - Jun Chen
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, Chengdu, Sichuan, China
| |
Collapse
|
15
|
Beri P, Plunkett C, Barbara J, Shih CC, Barnes SW, Ross O, Choconta P, Trinh T, Gomez D, Litvin B, Walker J, Qiu M, Hammack S, Toyama EQ. A high-throughput 3D cantilever array to model airway smooth muscle hypercontractility in asthma. APL Bioeng 2023; 7:026104. [PMID: 37206658 PMCID: PMC10191677 DOI: 10.1063/5.0132516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 04/11/2023] [Indexed: 05/21/2023] Open
Abstract
Asthma is often characterized by tissue-level mechanical phenotypes that include remodeling of the airway and an increase in airway tightening, driven by the underlying smooth muscle. Existing therapies only provide symptom relief and do not improve the baseline narrowing of the airway or halt progression of the disease. To investigate such targeted therapeutics, there is a need for models that can recapitulate the 3D environment present in this tissue, provide phenotypic readouts of contractility, and be easily integrated into existing assay plate designs and laboratory automation used in drug discovery campaigns. To address this, we have developed DEFLCT, a high-throughput plate insert that can be paired with standard labware to easily generate high quantities of microscale tissues in vitro for screening applications. Using this platform, we exposed primary human airway smooth muscle cell-derived microtissues to a panel of six inflammatory cytokines present in the asthmatic niche, identifying TGF-β1 and IL-13 as inducers of a hypercontractile phenotype. RNAseq analysis further demonstrated enrichment of contractile and remodeling-relevant pathways in TGF-β1 and IL-13 treated tissues as well as pathways generally associated with asthma. Screening of 78 kinase inhibitors on TGF-β1 treated tissues suggests that inhibition of protein kinase C and mTOR/Akt signaling can prevent this hypercontractile phenotype from emerging, while direct inhibition of myosin light chain kinase does not. Taken together, these data establish a disease-relevant 3D tissue model for the asthmatic airway, which combines niche specific inflammatory cues and complex mechanical readouts that can be utilized in drug discovery efforts.
Collapse
Affiliation(s)
- Pranjali Beri
- Novartis Institutes for Biomedical Research, San Diego, California 92121, USA
| | | | - Joshua Barbara
- Novartis Institutes for Biomedical Research, San Diego, California 92121, USA
| | - Chien-Cheng Shih
- Novartis Institutes for Biomedical Research, San Diego, California 92121, USA
| | - S. Whitney Barnes
- Novartis Institutes for Biomedical Research, San Diego, California 92121, USA
| | - Olivia Ross
- Novartis Institutes for Biomedical Research, San Diego, California 92121, USA
| | - Paula Choconta
- Novartis Institutes for Biomedical Research, San Diego, California 92121, USA
| | - Ton Trinh
- Novartis Institutes for Biomedical Research, San Diego, California 92121, USA
| | - Datzael Gomez
- Novartis Institutes for Biomedical Research, San Diego, California 92121, USA
| | - Bella Litvin
- Novartis Institutes for Biomedical Research, San Diego, California 92121, USA
| | - John Walker
- Novartis Institutes for Biomedical Research, San Diego, California 92121, USA
| | - Minhua Qiu
- Novartis Institutes for Biomedical Research, San Diego, California 92121, USA
| | - Scott Hammack
- Novartis Institutes for Biomedical Research, San Diego, California 92121, USA
| | - Erin Quan Toyama
- Novartis Institutes for Biomedical Research, San Diego, California 92121, USA
| |
Collapse
|
16
|
Feng Y, Liu X, Wang Y, Du R, Mao H. Delineating asthma according to inflammation phenotypes with a focus on paucigranulocytic asthma. Chin Med J (Engl) 2023:00029330-990000000-00572. [PMID: 37185590 DOI: 10.1097/cm9.0000000000002456] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Indexed: 05/17/2023] Open
Abstract
ABSTRACT Asthma is characterized by chronic airway inflammation and airway hyper-responsiveness. However, the differences in pathophysiology and phenotypic symptomology make a diagnosis of "asthma" too broad hindering individualized treatment. Four asthmatic inflammatory phenotypes have been identified based on inflammatory cell profiles in sputum: eosinophilic, neutrophilic, paucigranulocytic, and mixed-granulocytic. Paucigranulocytic asthma may be one of the most common phenotypes in stable asthmatic patients, yet it remains much less studied than the other inflammatory phenotypes. Understanding of paucigranulocytic asthma in terms of phenotypic discrimination, distribution, stability, surrogate biomarkers, underlying pathophysiology, clinical characteristics, and current therapies is fragmented, which impedes clinical management of patients. This review brings together existing knowledge and ongoing research about asthma phenotypes, with a focus on paucigranulocytic asthma, in order to present a comprehensive picture that may clarify specific inflammatory phenotypes and thus improve clinical diagnoses and disease management.
Collapse
Affiliation(s)
- Yinhe Feng
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaoyin Liu
- West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yubin Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Rao Du
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hui Mao
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
17
|
James BN, Weigel C, Green CD, Brown RDR, Palladino END, Tharakan A, Milstien S, Proia RL, Martin RK, Spiegel S. Neutrophilia in severe asthma is reduced in Ormdl3 overexpressing mice. FASEB J 2023; 37:e22799. [PMID: 36753412 PMCID: PMC9990076 DOI: 10.1096/fj.202201821r] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/11/2023] [Accepted: 01/20/2023] [Indexed: 02/09/2023]
Abstract
Genome-wide association studies have linked the ORM (yeast)-like protein isoform 3 (ORMDL3) to asthma severity. Although ORMDL3 is a member of a family that negatively regulates serine palmitoyltransferase (SPT) and thus biosynthesis of sphingolipids, it is still unclear whether ORMDL3 and altered sphingolipid synthesis are causally related to non-Th2 severe asthma associated with a predominant neutrophil inflammation and high interleukin-17 (IL-17) levels. Here, we examined the effects of ORMDL3 overexpression in a preclinical mouse model of allergic lung inflammation that is predominantly neutrophilic and recapitulates many of the clinical features of severe human asthma. ORMDL3 overexpression reduced lung and circulating levels of dihydrosphingosine, the product of SPT. However, the most prominent effect on sphingolipid levels was reduction of circulating S1P. The LPS/OVA challenge increased markers of Th17 inflammation with a predominant infiltration of neutrophils into the lung. A significant decrease of neutrophil infiltration was observed in the Ormdl3 transgenic mice challenged with LPS/OVA compared to the wild type and concomitant decrease in IL-17, that plays a key role in the pathogenesis of neutrophilic asthma. LPS decreased survival of murine neutrophils, which was prevented by co-treatment with S1P. Moreover, S1P potentiated LPS-induced chemotaxis of neutrophil, suggesting that S1P can regulate neutrophil survival and recruitment following LPS airway inflammation. Our findings reveal a novel connection between ORMDL3 overexpression, circulating levels of S1P, IL-17 and neutrophil recruitment into the lung, and questions the potential involvement of ORMDL3 in the pathology, leading to development of severe neutrophilic asthma.
Collapse
Affiliation(s)
- Briana N. James
- Department of Biochemistry and Molecular BiologyVirginia Commonwealth University School of MedicineRichmondVirginiaUSA
| | - Cynthia Weigel
- Department of Biochemistry and Molecular BiologyVirginia Commonwealth University School of MedicineRichmondVirginiaUSA
| | - Christopher D. Green
- Department of Biochemistry and Molecular BiologyVirginia Commonwealth University School of MedicineRichmondVirginiaUSA
| | - Ryan D. R. Brown
- Department of Biochemistry and Molecular BiologyVirginia Commonwealth University School of MedicineRichmondVirginiaUSA
| | - Elisa N. D. Palladino
- Department of Biochemistry and Molecular BiologyVirginia Commonwealth University School of MedicineRichmondVirginiaUSA
| | - Anuj Tharakan
- Department of Microbiology and ImmunologyVirginia Commonwealth University School of MedicineRichmondVirginiaUSA
| | - Sheldon Milstien
- Department of Biochemistry and Molecular BiologyVirginia Commonwealth University School of MedicineRichmondVirginiaUSA
| | - Richard L. Proia
- Genetics and Biochemistry BranchNational Institute of Diabetes and Digestive and Kidney Diseases, NIHBethesdaMarylandUSA
| | - Rebecca K. Martin
- Department of Microbiology and ImmunologyVirginia Commonwealth University School of MedicineRichmondVirginiaUSA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular BiologyVirginia Commonwealth University School of MedicineRichmondVirginiaUSA
| |
Collapse
|
18
|
Nowakowska J, Olechnowicz A, Langwiński W, Koteluk O, Lemańska Ż, Jóźwiak K, Kamiński K, Łosiewski W, Stegmayr J, Wagner D, Alsafadi HN, Lindstedt S, Dziuba M, Bielicka A, Graczyk Z, Szczepankiewicz A. Increased expression of ORMDL3 in allergic asthma: a case control and in vitro study. J Asthma 2023; 60:458-467. [PMID: 35321632 DOI: 10.1080/02770903.2022.2056896] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Asthma is the most frequent chronic disease in children. One of the most replicated genetic findings in childhood asthma is the ORMDL3 gene confirmed in several GWA studies in several pediatric populations. OBJECTIVES The purpose of this study was to analyze ORMDL3 variants and expression in childhood asthma in the Polish population. METHODS In the study we included 416 subject, 223 asthmatic children and 193 healthy control subjects. The analysis of two SNPs (rs3744246 and rs8076131) was performed using genotyping with TaqMan probes. The methylation of the ORMDL3 promoter was examined with Methylation Sensitive HRM (MS-HRM), covering 9 CpG sites. The expression of ORMDL3 was analyzed in PBMCs from pediatric patients diagnosed with allergic asthma and primary human bronchial epithelial cells derived from healthy subjects treated with IL-13, IL-4, or co-treatment with both cytokines to model allergic airway inflammation. RESULTS We found that ORMDL3 expression was increased in allergic asthma both in PBMCs from asthmatic patients as well as in human bronchial epithelial cells stimulated with the current cytokines. We did not observe significant differences between cases and controls either in the genotype distribution of analyzed SNPs (rs3744246 and rs8076131) nor in the level of promoter methylation. CONCLUSIONS Increased ORMDL3 expression is associated with pediatric allergic asthma and upregulated in the airways upon Th2-cytokines stimulation, but further functional studies are required to fully understand its role in this disease.
Collapse
Affiliation(s)
- Joanna Nowakowska
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Anna Olechnowicz
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Wojciech Langwiński
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Oliwia Koteluk
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Żaneta Lemańska
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Kacper Jóźwiak
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Kacper Kamiński
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Wojciech Łosiewski
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - John Stegmayr
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Darcy Wagner
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Hani N Alsafadi
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Sandra Lindstedt
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Maria Dziuba
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Antonina Bielicka
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Zuzanna Graczyk
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Aleksandra Szczepankiewicz
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
19
|
Gu W, Lei J, Zhu H, Xiao Y, Zhang Z, Zhao L. Effect of the BMPR-II-SMAD3/MRTF pathway on proliferation and migration of ASMCs and the mechanism in asthma. Mol Biol Rep 2022; 49:9283-9296. [PMID: 36008606 PMCID: PMC9515032 DOI: 10.1007/s11033-022-07764-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/30/2022] [Indexed: 11/30/2022]
Abstract
Background A variety of smooth muscle-specific genes and proteins, including SMAD3, BMPR-II, and MRTF, are involved in airway remodeling in asthma. As a receptor of bone morphogenetic protein (BMP) signaling, BMPR-II has important roles in airway remodeling in asthma. However, the underlying mechanism of BMPR-II in airway smooth muscle cells (ASMCs) in asthma remains incomplete. Methods Wistar rats were intraperitoneally injected with ovalbumin antigen suspension and aluminium hydroxide and, stimulated with ovalbumin nebulized inhalation to constructed asthma model. Primary ASMCs were isolated with collagenase I and identified by testing the α-SMA expression. Quantitative polymerase chain reaction (qPCR) and western blot assay were employed to detect the gene expression. CCK8, Transwell and Fluo-4 A assays were introduced to measure the cell viability, migration and intracellular Ca2+. Co-Immunoprecipitation (Co-IP) assay was applied to test the interaction among proteins. Results First, we observed significant increases in BMPR-II in asthmatic rat model and ASMCs at both the mRNA and protein levels. Second, we observed that silencing of siBMPR-II inhibited proliferation, migratory capacity and intracellular Ca2+ concentration in ASMCs. Furthermore, our study demonstrated that siBMPR-II inhibited the Smad3 expression and overexpression promoted the bioactivity of ASMCs. In addition, this study showed that p-Smad3 could interacted with MRTF and siMRTF inhibits the bioactivity of ASMCs. Finally, our results revealed BMPR-II-SMAD3/MRTF pathway affected the bioactivity of ASMCs. Conclusions This study indicates that the BMPR-II-SMAD3/MRTF signaling pathway is involved in the process of ASMCs remodeling, providing novel avenues for the identification of new therapeutic modalities.
Collapse
Affiliation(s)
- Wenbo Gu
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Jiahui Lei
- Zhengzhou University People's Hospital, Zhengzhou, China
| | - He Zhu
- Zhengzhou University People's Hospital, Zhengzhou, China
| | - Yali Xiao
- Zhengzhou University People's Hospital, Zhengzhou, China
| | - Zhenping Zhang
- People's Hospital of Zhongmu, Zhengzhou, China.,Henan Provincial People's Hospital, Zhengzhou, China
| | - Limin Zhao
- Zhengzhou University People's Hospital, Zhengzhou, China. .,Henan Provincial People's Hospital, Zhengzhou, China. .,Henan Univerity People's Hospital, Zhengzhou, China. .,Department of Respiratory Medicine, Henan Provincial People's Hospital (Zhengzhou University People's Hospital), No. 7 Weiwu Road, Zhengzhou, Henan, China.
| |
Collapse
|
20
|
Guo F, Hao Y, Zhang L, Croteau-Chonka DC, Thibault D, Kothari P, Li L, Levy BD, Zhou X, Raby BA. Asthma Susceptibility Gene ORMDL3 Promotes Autophagy in Human Bronchial Epithelium. Am J Respir Cell Mol Biol 2022; 66:661-670. [PMID: 35353673 PMCID: PMC9163638 DOI: 10.1165/rcmb.2021-0305oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 01/03/2022] [Indexed: 02/05/2023] Open
Abstract
The genome-wide association study (GWAS)-identified asthma susceptibility risk alleles on chromosome 17q21 increase the expression of ORMDL3 (ORMDL sphingolipid biosynthesis regulator 3) in lung tissue. Given the importance of epithelial integrity in asthma, we hypothesized that ORMDL3 directly impacted bronchial epithelial function. To determine whether and how ORMDL3 expression impacts the bronchial epithelium, in studies using both primary human bronchial epithelial cells and human bronchial epithelial cell line, 16HBE (16HBE14o-), we assessed the impact of ORMDL3 on autophagy. Studies included: autophagosome detection by electron microscopy, RFP-GFP-LC3B to assess autophagic activity, and Western blot analysis of autophagy-related proteins. Mechanistic assessments included immunoprecipitation assays, intracellular calcium mobilization assessments, and cell viability assays. Coexpression of ORMDL3 and autophagy-related genes was measured in primary human bronchial epithelial cells derived from 44 subjects. Overexpressing ORMDL3 demonstrated increased numbers of autophagosomes and increased levels of autophagy-related proteins LC3B, ATG3, ATG7, and ATG16L1. ORMDL3 overexpression promotes autophagy and subsequent cell death by impairing intracellular calcium mobilization through interacting with SERCA2. Strong correlation was observed between expression of ORMDL3 and autophagy-related genes in patient-derived bronchial epithelial cells. Increased ORMDL3 expression induces autophagy, possibly through interacting with SERCA2, thereby inhibiting intracellular calcium influx, and induces cell death, impairing bronchial epithelial function in asthma.
Collapse
Affiliation(s)
- Feng Guo
- Channing Division of Network Medicine and
| | - Yuan Hao
- Channing Division of Network Medicine and
- Division of Pulmonary Medicine, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts; and
| | - Li Zhang
- Channing Division of Network Medicine and
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | | | | | | | - Lijia Li
- Channing Division of Network Medicine and
| | - Bruce D. Levy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Xiaobo Zhou
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Benjamin A. Raby
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Division of Pulmonary Medicine, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts; and
| |
Collapse
|
21
|
Weng N, Miller M, Pham AK, Komor AC, Broide DH. Single-base editing of rs12603332 on chromosome 17q21 with a cytosine base editor regulates ORMDL3 and ATF6α expression. Allergy 2022; 77:1139-1149. [PMID: 34525218 DOI: 10.1111/all.15092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/08/2021] [Accepted: 08/09/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Genetic association studies have demonstrated that the SNP rs12603332 located on chromosome 17q21 is highly associated with the risk of the development of asthma. METHODS To determine whether SNP rs1260332 is functional in regulating levels of ORMDL3 expression, we used a Cytosine Base Editor (CBE) plasmid DNA or a CBE mRNA to edit the rs12603332 C risk allele to the T non-risk allele in a human lymphocyte cell line (i.e., Jurkat cells) and in primary human CD4 T cells that carry the C risk alleles. RESULTS Jurkat cells with the rs12603332 C risk allele expressed significantly higher levels of ORMDL3 mRNA, as well as the ORMDL3 regulated gene ATF6α as assessed by qPCR compared to Jurkat clones with the T non-risk allele. In primary human CD4 T cells, we edited 90 ± 3% of the rs12603332-C risk allele to the T non-risk allele and observed a reduction in ORMDL3 and ATF6α expression. Bioinformatic analysis predicted that the non-risk allele rs12603332-T could be the central element of the E-box binding motif (CANNTG) recognized by the E47 transcription factor. An EMSA assay confirmed the bioinformatics prediction demonstrating that a rs12603332-T containing probe bound to the transcription factor E47 in vitro. CONCLUSIONS SNP rs12603332 is functional in regulating the expression of ORMDL3 as well as ORMDL3 regulated gene ATF6α expression. In addition, we demonstrate the use of CBE technology in functionally interrogating asthma-associated SNPs using studies of primary human CD4 cells.
Collapse
Affiliation(s)
- Ning Weng
- Department of Medicine University of California San Diego La Jolla California USA
| | - Marina Miller
- Department of Medicine University of California San Diego La Jolla California USA
| | - Alexa K. Pham
- Department of Medicine University of California San Diego La Jolla California USA
| | - Alexis C. Komor
- Department of Chemistry and Biochemistry University of California San Diego La Jolla California USA
| | - David H. Broide
- Department of Medicine University of California San Diego La Jolla California USA
| |
Collapse
|
22
|
Chen R, Michaeloudes C, Liang Y, Bhavsar PK, Chung KF, Ip MSM, Mak JCW. ORMDL3 regulates cigarette smoke-induced endoplasmic reticulum stress in airway smooth muscle cells. J Allergy Clin Immunol 2022; 149:1445-1457.e5. [PMID: 34624393 DOI: 10.1016/j.jaci.2021.09.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/19/2021] [Accepted: 09/29/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND Orosomucoid 1-like protein 3 (ORMDL3), a transmembrane protein localized in the endoplasmic reticulum (ER), has been genetically associated with chronic obstructive pulmonary disease (COPD), in addition to childhood-onset asthma. However, the functional role of ORMDL3 in the pathogenesis of COPD is still unknown. OBJECTIVE Because cigarette smoke is the major risk factor for COPD, we aimed to investigate the role of ORMDL3 in cigarette smoke-induced human airway smooth muscle cell (HASMC) injury. METHODS The mRNA and protein expression of ORMDL3 was examined in HASMCs from nonsmokers and smokers without or with COPD. Knockdown of ORMDL3 in primary healthy HASMCs was performed using small interfering RNA before exposure to cigarette smoke medium (CSM) for 24 hours. Inflammatory, proliferative/apoptotic, ER stress, and mitochondrial markers were evaluated. RESULTS Elevation of ORMDL3 mRNA and protein expression was observed in HASMCs of smokers without or with COPD. CSM caused significant upregulation of ORMDL3 expression in healthy nonsmokers. ORMDL3 knockdown regulated CSM-induced inflammation, cell proliferation, and apoptosis. Silencing ORMDL3 led to reduction of CSM-induced ER stress via inhibition of unfolded protein response pathways such as activating transcription factor 6 and protein kinase RNA-like ER kinase. ORMDL3 was also involved in CSM-induced mitochondrial dysfunction via the mitochondrial fission process. CONCLUSIONS We report the induction of ORMDL3 in HASMCs after cigarette smoke exposure. ORMDL3 may mediate cigarette smoke-induced activation of unfolded protein response pathways during airway smooth muscle cell injury.
Collapse
Affiliation(s)
- Rui Chen
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Charalambos Michaeloudes
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Yingmin Liang
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Pankaj K Bhavsar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Mary S M Ip
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Judith C W Mak
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China; Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
23
|
Komlósi ZI, van de Veen W, Kovács N, Szűcs G, Sokolowska M, O'Mahony L, Akdis M, Akdis CA. Cellular and molecular mechanisms of allergic asthma. Mol Aspects Med 2021; 85:100995. [PMID: 34364680 DOI: 10.1016/j.mam.2021.100995] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 12/21/2022]
Abstract
Asthma is a chronic disease of the airways, which affects more than 350 million people worldwide. It is the most common chronic disease in children, affecting at least 30 million children and young adults in Europe. Asthma is a complex, partially heritable disease with a marked heterogeneity. Its development is influenced both by genetic and environmental factors. The most common, as well as the most well characterized subtype of asthma is allergic eosinophilic asthma, which is characterized by a type 2 airway inflammation. The prevalence of asthma has substantially increased in industrialized countries during the last 60 years. The mechanisms underpinning this phenomenon are incompletely understood, however increased exposure to various environmental pollutants probably plays a role. Disease inception is thought to be enabled by a disadvantageous shift in the balance between protective and harmful lifestyle and environmental factors, including exposure to protective commensal microbes versus infection with pathogens, collectively leading to airway epithelial cell damage and disrupted barrier integrity. Epithelial cell-derived cytokines are one of the main drivers of the type 2 immune response against innocuous allergens, ultimately leading to infiltration of lung tissue with type 2 T helper (TH2) cells, type 2 innate lymphoid cells (ILC2s), M2 macrophages and eosinophils. This review outlines the mechanisms responsible for the orchestration of type 2 inflammation and summarizes the novel findings, including but not limited to dysregulated epithelial barrier integrity, alarmin release and innate lymphoid cell stimulation.
Collapse
Affiliation(s)
- Zsolt I Komlósi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad Sqr. 4, 1089, Budapest, Hungary.
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| | - Nóra Kovács
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad Sqr. 4, 1089, Budapest, Hungary; Lung Health Hospital, Munkácsy Mihály Str. 70, 2045, Törökbálint, Hungary
| | - Gergő Szűcs
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad Sqr. 4, 1089, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Tömő Str. 25-29, 1083, Budapest, Hungary
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| | - Liam O'Mahony
- Department of Medicine and School of Microbiology, APC Microbiome Ireland, University College Cork, Ireland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| |
Collapse
|
24
|
Gao YY, Gao ZY. Extracellular Adenosine Diphosphate Stimulates CXCL10-Mediated Mast Cell Infiltration Through P2Y1 Receptor to Aggravate Airway Inflammation in Asthmatic Mice. Front Mol Biosci 2021; 8:621963. [PMID: 34291079 PMCID: PMC8287885 DOI: 10.3389/fmolb.2021.621963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/28/2021] [Indexed: 12/02/2022] Open
Abstract
Asthma is an inflammatory disease associated with variable airflow obstruction and airway inflammation. This study aimed to explore the role and mechanism of extracellular adenosine diphosphate (ADP) in the occurrence of airway inflammation in asthma. The expression of ADP in broncho-alveolar lavage fluid (BALF) of asthmatic patients was determined by enzyme linked immunosorbent assay (ELISA) and the expression of P2Y1 receptor in lung tissues was determined by reverse transcription-quantitative polymerase chain reaction. Asthmatic mouse model was induced using ovalbumin and the mice were treated with ADP to assess its effects on the airway inflammation and infiltration of mast cells (MCs). Additionally, alveolar epithelial cells were stimulated with ADP, and the levels of interleukin-13 (IL-13) and C-X-C motif chemokine ligand 10 (CXCL10) were measured by ELISA. We finally analyzed involvement of NF-κB signaling pathway in the release of CXCL10 in ADP-stimulated alveolar epithelial cells. The extracellular ADP was enriched in BALF of asthmatic patients, and P2Y1 receptor is highly expressed in lung tissues of asthmatic patients. In the OVA-induced asthma model, extracellular ADP aggravated airway inflammation and induced MC infiltration. Furthermore, ADP stimulated alveolar epithelial cells to secrete chemokine CXCL10 by activating P2Y1 receptor, whereby promoting asthma airway inflammation. Additionally, ADP activated the NF-κB signaling pathway to promote CXCL10 release. As a “danger signal” extracellular ADP could trigger and maintain airway inflammation in asthma by activating P2Y1 receptor. This study highlights the extracellular ADP as a promising anti-inflammatory target for the treatment of asthma.
Collapse
Affiliation(s)
- Yan-Yan Gao
- Department of Respiratory Medicine, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Zeng-Yan Gao
- Department of Respiratory Medicine, The Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Allergic asthma reflects the interplay between inflammatory mediators and immune, airway epithelial, and other cells. This review summarizes key insights in these areas over the past year. RECENT FINDINGS Key findings over the past year demonstrate that epithelial cells mediate tight junction breakdown to facilitate the development of asthma-like disease in mice. Innate lymph lymphoid cells (ILC), while previously shown to promote allergic airway disease, have now been shown to inhibit the development of severe allergic disease in mice. Fibrinogen cleavage products (previously shown to mediate allergic airway disease and macrophage fungistatic immunity by signaling through Toll-like receptor 4) have now been shown to first bind to the integrin Mac-1 (CD11c/CD18). Therapeutically, recent discoveries include the development of the antiasthma drug PM-43I that inhibits the allergy-related transcription factors STAT5 and STAT6 in mice, and confirmatory evidence of the efficacy of the antifungal agent voriconazole in human asthma. SUMMARY Studies over the past year provide critical new insight into the mechanisms by which epithelial cells, ILC, and coagulation factors contribute to the expression of asthma-like disease and further support the development antiasthma drugs that block STAT factors and inhibit fungal growth in the airways.
Collapse
|
26
|
Pham AK, Miller M, Rosenthal P, Das S, Weng N, Jang S, Kurten RC, Badrani J, Doherty TA, Oliver B, Broide DH. ORMDL3 expression in ASM regulates hypertrophy, hyperplasia via TPM1 and TPM4, and contractility. JCI Insight 2021; 6:136911. [PMID: 33661765 PMCID: PMC8119187 DOI: 10.1172/jci.insight.136911] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/26/2021] [Indexed: 12/21/2022] Open
Abstract
ORM1-like 3 (ORMDL3) has strong genetic linkage to childhood onset asthma. To determine whether ORMDL3 selective expression in airway smooth muscle (ASM) influences ASM function, we used Cre-loxP techniques to generate transgenic mice (hORMDL3Myh11eGFP-cre), which express human ORMDL3 selectively in smooth muscle cells. In vitro studies of ASM cells isolated from the bronchi of hORMDL3Myh11eGFP-cre mice demonstrated that they developed hypertrophy (quantitated by FACS and image analysis), developed hyperplasia (assessed by BrdU incorporation), and expressed increased levels of tropomysin proteins TPM1 and TPM4. siRNA knockdown of TPM1 or TPM4 demonstrated their importance to ORMDL3-mediated ASM proliferation but not hypertrophy. In addition, ASM derived from hORMDL3Myh11eGFP-cre mice had increased contractility to histamine in vitro, which was associated with increased levels of intracellular Ca2+; increased cell surface membrane Orai1 Ca2+ channels, which mediate influx of Ca2+ into the cytoplasm; and increased expression of ASM contractile genes sarco/endoplasmic reticulum Ca2+ ATPase 2b and smooth muscle 22. In vivo studies of hORMDL3Myh11eGFP-cre mice demonstrated that they had a spontaneous increase in ASM and airway hyperreactivity (AHR). ORMDL3 expression in ASM thus induces changes in ASM (hypertrophy, hyperplasia, increased contractility), which may explain the contribution of ORMDL3 to the development of AHR in childhood onset asthma, which is highly linked to ORMDL3 on chromosome 17q12-21.
Collapse
Affiliation(s)
- Alexa K. Pham
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Marina Miller
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Peter Rosenthal
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Sudipta Das
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Ning Weng
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Sunghoon Jang
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Richard C. Kurten
- Department of Pediatrics, Arkansas Children’s Research Institute, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jana Badrani
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Taylor A. Doherty
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Veterans Affairs San Diego Health Care System, La Jolla, California, USA
| | - Brian Oliver
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - David H. Broide
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| |
Collapse
|
27
|
Zhang Y, Hua L, Liu QH, Chu SY, Gan YX, Wu M, Bao YX, Chen Q, Zhang J. Household mold exposure interacts with inflammation-related genetic variants on childhood asthma: a case-control study. BMC Pulm Med 2021; 21:114. [PMID: 33810791 PMCID: PMC8019181 DOI: 10.1186/s12890-021-01484-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/24/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A number of studies have examined the association between mold exposure and childhood asthma. However, the conclusions were inconsistent, which might be partly attributable to the lack of consideration of gene function, especially the key genes affecting the pathogenesis of childhood asthma. Research on the interactions between genes and mold exposure on childhood asthma is still very limited. We therefore examined whether there is an interaction between inflammation-related genes and mold exposure on childhood asthma. METHODS A case-control study with 645 asthmatic children and 910 non-asthmatic children aged 3-12 years old was conducted. Eight single nucleotide polymorphisms (SNPs) in inflammation-related genes were genotyped using MassARRAY assay. Mold exposure was defined as self-reported visible mold on the walls. Associations between visible mold exposure, SNPs and childhood asthma were evaluated using logistic regression models. In addition, crossover analyses were used to estimate the gene-environment interactions on childhood asthma on an additive scale. RESULTS After excluding children without information on visible mold exposure or SNPs, 608 asthmatic and 839 non-asthmatic children were included in the analyses. Visible mold exposure was reported in 151 asthmatic (24.8%) and 119 non-asthmatic children (14.2%) (aOR 2.19, 95% CI 1.62-2.97). The rs7216389 SNP in gasdermin B gene (GSDMB) increased the risk of childhood asthma with each C to T substitution in a dose-dependent pattern (additive model, aOR 1.32, 95% CI 1.11-1.57). Children carrying the rs7216389 T allele and exposed to visible mold dramatically increased the risk of childhood asthma (aOR 3.21; 95% CI 1.77-5.99). The attributable proportion due to the interaction (AP: 0.47, 95% CI 0.03-0.90) and the relative excess risk due to the interaction (RERI: 1.49, 95% CI 0-2.99) were statistically significant. CONCLUSIONS In the present study, there was a significant additive interaction between visible mold exposure and rs7216389 SNP on childhood asthma. Future studies need to consider the gene-environment interactions when exploring the risk factors of childhood asthma.
Collapse
Affiliation(s)
- Yu Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Li Hua
- Department of Pediatric Pulmonology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Quan-Hua Liu
- Department of Pediatric Pulmonology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Shu-Yuan Chu
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Yue-Xin Gan
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Min Wu
- Department of Chinese Traditional Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yi-Xiao Bao
- Department of Pediatric Pulmonology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Qian Chen
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Jun Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
28
|
Li J, He Q, Wang L, Chen D, Qiu C, Xu P, Lu Y, Zeng Y, Chen R. SET knockdown attenuated phenotype modulation and calcium channel associated markers of airway smooth muscle cells in asthmatic mice. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:657. [PMID: 33987355 PMCID: PMC8106076 DOI: 10.21037/atm-21-573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Dysfunctional phenotype modulation and calcium channels in airway smooth muscle cells (ASMCs) are important characteristics of airway remodeling in chronic asthma. However, the mechanisms underlying these pathological processes remain unclear. SET (I2PP2A, inhibitor-2 of protein phosphatase 2A) has many significant functions and is involved in various physiological and pathological processes. This study aimed to determine the function of SET in chronic asthma. Methods BALB/c mice were sensitized by ovalbumin injection and repeated inhalation of ovalbumin. The Penh value was measured using the Buxco whole body plethysmography system. A short hairpin RNA of the SET gene was designed and transfected into ASMCs derived from asthmatic mice. Flow cytometry of Annexin-V/propidium iodide staining was used for evaluating cell apoptosis. Western blot was adopted to measure the expression levels of ASMCs phenotype modulation markers and calcium channel-associated proteins. Results The results showed that shRNA targeting SET significantly decreased the expression of SET, and enhanced the apoptosis of ASMCs. SET knockdown promoted the expression of contractile phenotype markers such as α-SMA (alpha smooth muscle Actin), SM-MHC (smooth muscle Myosin heavy chain), and calponin, and inhibited the expression of synthetic phenotype markers including vimentin and CD44. The expression of the calcium channel-related proteins STIM1 (Stromal interaction molecule 1) and Orai1 were also inhibited after SET knockdown. Conclusions These data demonstrated that SET participated in the development of airway dysfunction in asthma, suggesting that the silencing of SET may be a new therapeutic target for the treatment of asthma patients.
Collapse
Affiliation(s)
- Jie Li
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Qi He
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Lingwei Wang
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Dandan Chen
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Chen Qiu
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Peng Xu
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Yongzhen Lu
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Yuwei Zeng
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Rongchang Chen
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| |
Collapse
|
29
|
Li J, Ullah MA, Jin H, Liang Y, Lin L, Wang J, Peng X, Liao H, Li Y, Ge Y, Li L. ORMDL3 Functions as a Negative Regulator of Antigen-Mediated Mast Cell Activation via an ATF6-UPR-Autophagy-Dependent Pathway. Front Immunol 2021; 12:604974. [PMID: 33679742 PMCID: PMC7933793 DOI: 10.3389/fimmu.2021.604974] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
Antigen (Ag)-mediated mast cell activation plays a critical role in the immunopathology of IgE-dependent allergic diseases. Restraining the signaling cascade that regulates the release of mast cell-derived inflammatory mediators is an attractive therapeutic strategy to treat allergic diseases. Orosomucoid-like-3 (ORMDL3) regulates the endoplasmic reticulum stress (ERS)-induced unfolded protein response (UPR) and autophagy. Although ERS/UPR/autophagy pathway is crucial in Ag-induced mast cell activation, it is unknown whether ORMDL3 regulates the ERS/UPR/autophagy pathway during mast cell activation. In this study, we found that ORMDL3 expression was downregulated in Ag-activated MC/9 cells. Overexpression of ORMDL3 significantly inhibited degranulation, and cytokine/chemokine production, while the opposite effect was observed with ORMDL3 knockdown in MC/9 cells. Importantly, ORMDL3 overexpression upregulated mediators of ERS-UPR (SERCA2b, ATF6) and autophagy (Beclin 1 and LC3BII). Knockdown of ATF6 and/or inhibition of autophagy reversed the decreased degranulation and cytokine/chemokine expression caused by ORMDL3 overexpression. Moreover, in vivo knockdown of ORMDL3 and/or ATF6 enhanced passive cutaneous anaphylaxis (PCA) reactions in mouse ears. These data indicate that ORMDL3 suppresses Ag-mediated mast cell activation via an ATF6 UPR-autophagy dependent pathway and thus, attenuates anaphylactic reaction. This highlights a potential mechanism to intervene in mast cell mediated diseases.
Collapse
Affiliation(s)
- Jia Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Md Ashik Ullah
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Hongping Jin
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Yuting Liang
- Center of Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lihui Lin
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan Wang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Peng
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huanjin Liao
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanning Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiqin Ge
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
Gui H, Levin AM, Hu D, Sleiman P, Xiao S, Mak ACY, Yang M, Barczak AJ, Huntsman S, Eng C, Hochstadt S, Zhang E, Whitehouse K, Simons S, Cabral W, Takriti S, Abecasis G, Blackwell TW, Kang HM, Nickerson DA, Germer S, Lanfear DE, Gilliland F, Gauderman WJ, Kumar R, Erle DJ, Martinez FD, Hakonarson H, Burchard EG, Williams LK. Mapping the 17q12-21.1 Locus for Variants Associated with Early-Onset Asthma in African Americans. Am J Respir Crit Care Med 2021; 203:424-436. [PMID: 32966749 PMCID: PMC7885840 DOI: 10.1164/rccm.202006-2623oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/21/2020] [Indexed: 01/12/2023] Open
Abstract
Rationale: The 17q12-21.1 locus is one of the most highly replicated genetic associations with asthma. Individuals of African descent have lower linkage disequilibrium in this region, which could facilitate identifying causal variants.Objectives: To identify functional variants at 17q12-21.1 associated with early-onset asthma among African American individuals.Methods: We evaluated African American participants from SAPPHIRE (Study of Asthma Phenotypes and Pharmacogenomic Interactions by Race-Ethnicity) (n = 1,940), SAGE II (Study of African Americans, Asthma, Genes and Environment) (n = 885), and GCPD-A (Study of the Genetic Causes of Complex Pediatric Disorders-Asthma) (n = 2,805). Associations with asthma onset at ages under 5 years were meta-analyzed across cohorts. The lead signal was reevaluated considering haplotypes informed by genetic ancestry (i.e., African vs. European). Both an expression-quantitative trait locus analysis and a phenome-wide association study were performed on the lead variant.Measurements and Main Results: The meta-analyzed results from SAPPHIRE, SAGE II, and the GCPD-A identified rs11078928 as the top association for early-onset asthma. A haplotype analysis suggested that the asthma association partitioned most closely with the rs11078928 genotype. Genetic ancestry did not appear to influence the effect of this variant. In the expression-quantitative trait locus analysis, rs11078928 was related to alternative splicing of GSDMB (gasdermin-B) transcripts. The phenome-wide association study of rs11078928 suggested that this variant was predominantly associated with asthma and asthma-associated symptoms.Conclusions: A splice-acceptor polymorphism appears to be a causal variant for asthma at the 17q12-21.1 locus. This variant appears to have the same magnitude of effect in individuals of African and European descent.
Collapse
Affiliation(s)
- Hongsheng Gui
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research and
| | - Albert M. Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Michigan
| | | | - Patrick Sleiman
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shujie Xiao
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research and
| | | | - Mao Yang
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research and
| | | | | | | | - Samantha Hochstadt
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research and
| | - Ellen Zhang
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research and
| | - Kyle Whitehouse
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research and
| | - Samantha Simons
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research and
| | - Whitney Cabral
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research and
| | - Sami Takriti
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research and
| | - Gonçalo Abecasis
- Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan
| | - Thomas W. Blackwell
- Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan
| | - Hyun Min Kang
- Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan
| | - Deborah A. Nickerson
- Department of Genome Sciences, University of Washington, Seattle, Washington
- Northwest Genomics Center, Seattle, Washington
- Brotman Baty Institute, Seattle, Washington
| | | | - David E. Lanfear
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research and
| | - Frank Gilliland
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - W. James Gauderman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Rajesh Kumar
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; and
| | - David J. Erle
- Department of Medicine
- Lung Biology Center
- CoLabs, and
| | - Fernando D. Martinez
- Arizona Respiratory Center and
- Department of Pediatrics, University of Arizona, Tucson, Arizona
| | - Hakon Hakonarson
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Esteban G. Burchard
- Department of Medicine
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California
| | - L. Keoki Williams
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research and
| |
Collapse
|
31
|
Ding Z, Yu F, Sun Y, Jiao N, Shi L, Wan J, Liu Q. ORMDL3 Promotes Angiogenesis in Chronic Asthma Through the ERK1/2/VEGF/MMP-9 Pathway. Front Pediatr 2021; 9:708555. [PMID: 35252072 PMCID: PMC8888883 DOI: 10.3389/fped.2021.708555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
AIM Angiogenesis plays a vital role in airway remodeling in chronic asthma. ORMDL3 has been identified to be closely associated with the development of asthma remodeling. This study was to investigate the mechanism of ORMDL3 in angiogenesis of chronic asthma. METHODS BALB/c mice were divided into three groups, including an asthmatic group (group A), a budesonide-treated group (group B), and a normal control group (group C). Hematoxylin and eosin and Masson staining were used to evaluate the pathological changes. Angiogenesis in lung tissue was examined by CD31 staining. The changes of ORMDL3, ERK1/2, and angiogenesis-associated MMP-9 and Vascular endothelial growth factor (VEGF) expression were examined. Furthermore, ORMDL3, MMP-9, and VEGF mRNA and protein levels were examined after transfection in BEAS-2B cells with the ORMDL3-overexpressed lentiviral vector. RESULTS Compared with the control group, asthmatic mice indicated more severe airway angiogenesis with increased ORMDL3, ERK1/2, MMP-9, and VEGF expression. Budesonide alleviated airway angiogenesis, and CD31 expression was positive with the levels of ORMDL3, MMP-9, and VEGF (P < 0.01). After successful transfection in BEAS-2B cells with the ORMDL3-overexpressing lentiviral vector, VEGF, and MMP-9 expression were activated in vitro (P < 0.01). CONCLUSION In conclusion, our study provides novel evidence that ORMDL3 promotes angiogenesis through upregulating VEGF and MMP-9 in chronic asthma.
Collapse
Affiliation(s)
- Zhen Ding
- Department of Pediatrics, Zibo Hospital of Traditional Chinese Medicine, Zibo, China
| | - Fei Yu
- Department of Pediatrics, Zibo Hospital of Traditional Chinese Medicine, Zibo, China
| | - Yan Sun
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ning Jiao
- Department of Pediatrics, Zibo Hospital of Traditional Chinese Medicine, Zibo, China
| | - Lina Shi
- Department of Pediatrics, Zibo Hospital of Traditional Chinese Medicine, Zibo, China
| | - Jinghong Wan
- Department of Pediatrics, Zibo Hospital of Traditional Chinese Medicine, Zibo, China
| | - Qinghua Liu
- Department of Pediatrics, Zibo Hospital of Traditional Chinese Medicine, Zibo, China
| |
Collapse
|
32
|
Heras AF, Veerappan A, Silver RB, Emala CW, Worgall TS, Perez-Zoghbi J, Worgall S. Increasing Sphingolipid Synthesis Alleviates Airway Hyperreactivity. Am J Respir Cell Mol Biol 2020; 63:690-698. [PMID: 32706610 DOI: 10.1165/rcmb.2020-0194oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Impaired sphingolipid synthesis is linked genetically to childhood asthma and functionally to airway hyperreactivity (AHR). The objective was to investigate whether sphingolipid synthesis could be a target for asthma therapeutics. The effects of GlyH-101 and fenretinide via modulation of de novo sphingolipid synthesis on AHR was evaluated in mice deficient in SPT (serine palmitoyl-CoA transferase), the rate-limiting enzyme of sphingolipid synthesis. The drugs were also used directly in human airway smooth-muscle and epithelial cells to evaluate changes in de novo sphingolipid metabolites and calcium release. GlyH-101 and fenretinide increased sphinganine and dihydroceramides (de novo sphingolipid metabolites) in lung epithelial and airway smooth-muscle cells, decreased the intracellular calcium concentration in airway smooth-muscle cells, and decreased agonist-induced contraction in proximal and peripheral airways. GlyH-101 also decreased AHR in SPT-deficient mice in vivo. This study identifies the manipulation of sphingolipid synthesis as a novel metabolic therapeutic strategy to alleviate AHR.
Collapse
Affiliation(s)
| | | | | | | | - Tilla S Worgall
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | | | - Stefan Worgall
- Department of Pediatrics.,Department of Genetic Medicine, and.,Drukier Institute for Children's Health, Weill Cornell Medicine, New York, New York; and
| |
Collapse
|
33
|
Luthers CR, Dunn TM, Snow AL. ORMDL3 and Asthma: Linking Sphingolipid Regulation to Altered T Cell Function. Front Immunol 2020; 11:597945. [PMID: 33424845 PMCID: PMC7793773 DOI: 10.3389/fimmu.2020.597945] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/02/2020] [Indexed: 12/21/2022] Open
Abstract
Orosomucoid like 3 (ORMDL3) encodes an ER-resident transmembrane protein that regulates the activity of serine palmitoyltransferase (SPT), the first and rate-limiting enzyme for sphingolipid biosynthesis in cells. A decade ago, several genome wide association studies revealed single nucleotide polymorphisms associated with increased ORMDL3 protein expression and susceptibility to allergic asthma. Since that time, numerous studies have investigated how altered ORMDL3 expression might predispose to asthma and other autoimmune/inflammatory diseases. In this brief review, we focus on growing evidence suggesting that heightened ORMDL3 expression specifically in CD4+ T lymphocytes, the central orchestrators of adaptive immunity, constitutes a major underlying mechanism of asthma pathogenesis by skewing their differentiation and function. Furthermore, we explore how sphingolipid modulation in T cells might be responsible for these effects, and how further studies may interrogate this intriguing hypothesis.
Collapse
Affiliation(s)
- Christopher R Luthers
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Teresa M Dunn
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Department of Biochemistry, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Andrew L Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
34
|
Dang H, Polineni D, Pace RG, Stonebraker JR, Corvol H, Cutting GR, Drumm ML, Strug LJ, O’Neal WK, Knowles MR. Mining GWAS and eQTL data for CF lung disease modifiers by gene expression imputation. PLoS One 2020; 15:e0239189. [PMID: 33253230 PMCID: PMC7703903 DOI: 10.1371/journal.pone.0239189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 09/02/2020] [Indexed: 12/18/2022] Open
Abstract
Genome wide association studies (GWAS) have identified several genomic loci with candidate modifiers of cystic fibrosis (CF) lung disease, but only a small proportion of the expected genetic contribution is accounted for at these loci. We leveraged expression data from CF cohorts, and Genotype-Tissue Expression (GTEx) reference data sets from multiple human tissues to generate predictive models, which were used to impute transcriptional regulation from genetic variance in our GWAS population. The imputed gene expression was tested for association with CF lung disease severity. By comparing and combining results from alternative approaches, we identified 379 candidate modifier genes. We delved into 52 modifier candidates that showed consensus between approaches, and 28 of them were near known GWAS loci. A number of these genes are implicated in the pathophysiology of CF lung disease (e.g., immunity, infection, inflammation, HLA pathways, glycosylation, and mucociliary clearance) and the CFTR protein biology (e.g., cytoskeleton, microtubule, mitochondrial function, lipid metabolism, endoplasmic reticulum/Golgi, and ubiquitination). Gene set enrichment results are consistent with current knowledge of CF lung disease pathogenesis. HLA Class II genes on chr6, and CEP72, EXOC3, and TPPP near the GWAS peak on chr5 are most consistently associated with CF lung disease severity across the tissues tested. The results help to prioritize genes in the GWAS regions, predict direction of gene expression regulation, and identify new candidate modifiers throughout the genome for potential therapeutic development.
Collapse
Affiliation(s)
- Hong Dang
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| | - Deepika Polineni
- University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Rhonda G. Pace
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| | - Jaclyn R. Stonebraker
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| | - Harriet Corvol
- Pediatric Pulmonary Department, Assistance Publique-Hôpitaux sde Paris (AP-HP), Hôpital Trousseau, Institut National de la Santé et la Recherche Médicale (INSERM) U938, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Paris 6, Paris, France
| | - Garry R. Cutting
- McKusick-Nathans Institute of Genetic Medicine, Baltimore, Maryland, United States of America
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Mitchell L. Drumm
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Lisa J. Strug
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Wanda K. O’Neal
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| | - Michael R. Knowles
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
35
|
Shi X, Chai X, Yang Y, Cheng Q, Jiao Y, Chen H, Huang J, Yang C, Liu J. A tissue-specific collaborative mixed model for jointly analyzing multiple tissues in transcriptome-wide association studies. Nucleic Acids Res 2020; 48:e109. [PMID: 32978944 PMCID: PMC7641735 DOI: 10.1093/nar/gkaa767] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/14/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022] Open
Abstract
Transcriptome-wide association studies (TWASs) integrate expression quantitative trait loci (eQTLs) studies with genome-wide association studies (GWASs) to prioritize candidate target genes for complex traits. Several statistical methods have been recently proposed to improve the performance of TWASs in gene prioritization by integrating the expression regulatory information imputed from multiple tissues, and made significant achievements in improving the ability to detect gene-trait associations. Unfortunately, most existing multi-tissue methods focus on prioritization of candidate genes, and cannot directly infer the specific functional effects of candidate genes across different tissues. Here, we propose a tissue-specific collaborative mixed model (TisCoMM) for TWASs, leveraging the co-regulation of genetic variations across different tissues explicitly via a unified probabilistic model. TisCoMM not only performs hypothesis testing to prioritize gene-trait associations, but also detects the tissue-specific role of candidate target genes in complex traits. To make full use of widely available GWASs summary statistics, we extend TisCoMM to use summary-level data, namely, TisCoMM-S2. Using extensive simulation studies, we show that type I error is controlled at the nominal level, the statistical power of identifying associated genes is greatly improved, and the false-positive rate (FPR) for non-causal tissues is well controlled at decent levels. We further illustrate the benefits of our methods in applications to summary-level GWASs data of 33 complex traits. Notably, apart from better identifying potential trait-associated genes, we can elucidate the tissue-specific role of candidate target genes. The follow-up pathway analysis from tissue-specific genes for asthma shows that the immune system plays an essential function for asthma development in both thyroid and lung tissues.
Collapse
Affiliation(s)
- Xingjie Shi
- Department of Statistics, Nanjing University of Finance and Economics, Nanjing, China
- Centre for Quantitative Medicine, Health Services & Systems Research, Duke-NUS Medical School, Singapore
| | - Xiaoran Chai
- Beijing Advanced Innovation Center for Genomics (ICG) & Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China
- School of Medicine, National University of Singapore, Singapore
| | - Yi Yang
- Centre for Quantitative Medicine, Health Services & Systems Research, Duke-NUS Medical School, Singapore
| | - Qing Cheng
- Centre for Quantitative Medicine, Health Services & Systems Research, Duke-NUS Medical School, Singapore
| | - Yuling Jiao
- School of Mathematics and Statistics, and Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, China
| | - Haoyue Chen
- School of International Studies, Zhejiang University, Hangzhou, China
| | - Jian Huang
- Department of Statistics and Actuarial Science, University of Iowa, USA
| | - Can Yang
- Department of Mathematics, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Jin Liu
- Centre for Quantitative Medicine, Health Services & Systems Research, Duke-NUS Medical School, Singapore
| |
Collapse
|
36
|
Mechanisms of non-type 2 asthma. Curr Opin Immunol 2020; 66:123-128. [PMID: 33160187 DOI: 10.1016/j.coi.2020.10.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/04/2020] [Indexed: 12/17/2022]
Abstract
Non-type 2 inflammation (Non-T2)-mediated asthma is difficult to define due to lack of signature biomarkers. It exists in the absence of T2-high or eosinophilic inflammation and includes neutrophilic and paucigranulocytic subtypes. Several cell types and cytokines, including Th1, Th17, IL-6, and IL-17, contribute to mechanisms of non-T2 asthma. Neutrophil extracellular traps (NETs) and inflammasome activation likely play a role in severe neutrophilic asthma. Several mechanisms lead to uncoupling of airway hyperresponsiveness and remodeling from airway inflammation in paucigranulocytic asthma. Recent research on transcriptomics and proteomics in non-T2 asthma is discussed in this review. Investigations of specific drug therapies for non-T2 asthma have been disappointing, and remain an important area for future clinical studies.
Collapse
|
37
|
Liu Y, Bochkov YA, Eickhoff JC, Hu T, Zumwalde NA, Tan JW, Lopez C, Fichtinger PS, Reddy TR, Overmyer KA, Gumperz JE, Coon J, Mathur SK, Gern JE, Smith JA. Orosomucoid-like 3 Supports Rhinovirus Replication in Human Epithelial Cells. Am J Respir Cell Mol Biol 2020; 62:783-792. [PMID: 32078788 DOI: 10.1165/rcmb.2019-0237oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Polymorphism at the 17q21 gene locus and wheezing responses to rhinovirus (RV) early in childhood conspire to increase the risk of developing asthma. However, the mechanisms mediating this gene-environment interaction remain unclear. In this study, we investigated the impact of one of the 17q21-encoded genes, ORMDL3 (orosomucoid-like 3), on RV replication in human epithelial cells. ORMDL3 knockdown inhibited RV-A16 replication in HeLa, BEAS-2B, A549, and NCI-H358 epithelial cell lines and primary nasal and bronchial epithelial cells. Inhibition varied by RV species, as both minor and major group RV-A subtypes RV-B52 and RV-C2 were inhibited but not RV-C15 or RV-C41. ORMDL3 siRNA did not affect expression of the major group RV-A receptor ICAM-1 or initial internalization of RV-A16. The two major outcomes of ORMDL3 activity, SPT (serine palmitoyl-CoA transferase) inhibition and endoplasmic reticulum (ER) stress induction, were further examined: silencing ORMDL3 decreased RV-induced ER stress and IFN-β mRNA expression. However, pharmacologic induction of ER stress and concomitant increased IFN-β inhibited RV-A16 replication. Conversely, blockade of ER stress with tauroursodeoxycholic acid augmented replication, pointing to an alternative mechanism for the effect of ORMDL3 knockdown on RV replication. In comparison, the SPT inhibitor myriocin increased RV-A16 but not RV-C15 replication and negated the inhibitory effect of ORMDL3 knockdown. Furthermore, lipidomics analysis revealed opposing regulation of specific sphingolipid species (downstream of SPT) by myriocin and ORMDL3 siRNA, correlating with the effect of these treatments on RV replication. Together, these data revealed a requirement for ORMDL3 in supporting RV replication in epithelial cells via SPT inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Paul S Fichtinger
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | | | - Katherine A Overmyer
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin; and.,Morgridge Institute for Research, Madison, Wisconsin
| | | | - Joshua Coon
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin; and.,Morgridge Institute for Research, Madison, Wisconsin
| | - Sameer K Mathur
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | | | - Judith A Smith
- Department of Pediatrics.,Department of Medical Microbiology and Immunology, and
| |
Collapse
|
38
|
Wills-Karp M. At last - linking ORMDL3 polymorphisms, decreased sphingolipid synthesis, and asthma susceptibility. J Clin Invest 2020; 130:604-607. [PMID: 31929192 DOI: 10.1172/jci134333] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Asthma is a common chronic respiratory disease that has a heritable component. Polymorphisms in the endoplasmic reticular protein orosomucoid-like protein 3 (ORMDL3), which regulates sphingolipid homeostasis, have been strongly linked with childhood-onset asthma. Despite extensive investigation, a link between ORMDL3 asthma-risk genotypes and altered sphingolipid synthesis has been lacking. In this issue of the JCI, Ono et al. establish a clear association between nonallergic childhood asthma, lower whole-blood sphingolipids, and asthma-risk 17q21 genotypes. These results demonstrate that genetic variants in ORMDL3 may confer a risk of developing childhood asthma through dysregulation of sphingolipid synthesis. As such, modulation of sphingolipids may represent a promising avenue of therapeutic development for childhood asthma.
Collapse
|
39
|
Miller M, Rosenthal P, Weng N, Pham A, Hur GY, Elliot J, Green FHY, James A, Broide DH. Chromosome 17q21 SNP rs8076131 risk allele associates with airway smooth muscle hypertrophy in fatal asthma. Clin Exp Allergy 2020; 50:1270-1273. [PMID: 32672387 DOI: 10.1111/cea.13708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/03/2020] [Accepted: 06/25/2020] [Indexed: 01/11/2023]
Affiliation(s)
- Marina Miller
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Peter Rosenthal
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ning Weng
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Alex Pham
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Gyu-Young Hur
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - John Elliot
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Francis H Y Green
- Department of Pathology & Laboratory Medicine, University of Calgary, Alberta, Canada
| | - Alan James
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - David H Broide
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
40
|
Dileepan M, Ha SG, Rastle-Simpson S, Ge XN, Greenberg YG, Wijesinghe DS, Contaifer D, Rao SP, Sriramarao P. Pulmonary delivery of ORMDL3 short hairpin RNA - a potential tool to regulate allergen-induced airway inflammation. Exp Lung Res 2020; 46:243-257. [PMID: 32578458 DOI: 10.1080/01902148.2020.1781297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Aim/Purpose: Exposure to various allergens has been shown to increase expression of ORMDL3 in the lung in models of allergic asthma. Studies using genetically modified (transgenic or knock out) mice have revealed some of the functions of ORMDL3 in asthma pathogenesis, although amid debate. The goal of this study was to use targeted post-transcriptional downregulation of ORMDL3 in allergen-challenged wild-type (WT) mice by RNA interference to further elucidate the functional role of ORMDL3 in asthma pathogenesis and evaluate a potential therapeutic option.Methods: Allergen (ovalbumin [OVA])-challenged WT mice were administered intranasally (i.n) with a single dose of five short hairpin RNA (shRNA) constructs with different target sequence for murine ORMDL3 cloned in a lentiviral vector or with the empty vector (control). Mice were evaluated for allergen-induced airway hyperresponsiveness (AHR) and various features of airway inflammation after 72 hours.Results: I.n administration of a single dose of ORMDL3 shRNAs to OVA-challenged mice resulted in reduction of ORMDL3 gene expression in the lungs associated with a significant reduction in AHR to inhaled methacholine and in the number of inflammatory cells recruited in the airways, specifically eosinophils, as well as in airway mucus secretion compared to OVA-challenged mice that received the empty vector. Administration of ORMDL3 shRNAs also significantly inhibited levels of IL-13, eotaxin-2 and sphingosine in the lungs. Additionally, ORMDL3 shRNAs significantly inhibited the allergen-mediated increase in monohexyl ceramides C22:0 and C24:0.Conclusions: Post-transcriptional down regulation of ORMDL3 in allergic lungs using i.n-delivered ORMDL3 shRNA (akin to inhaled therapy) attenuates development of key features of airway allergic disease, confirming the involvement of ORMDL3 in allergic asthma pathogenesis and serving as a model for a potential therapeutic strategy.
Collapse
Affiliation(s)
- Mythili Dileepan
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, USA
| | - Sung Gil Ha
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, USA
| | | | - Xiao Na Ge
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, USA.,Merck & Co., Inc, Palo Alto, CA, USA
| | - Yana G Greenberg
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, USA
| | - Dayanjan S Wijesinghe
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Daniel Contaifer
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Savita P Rao
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, USA
| | - P Sriramarao
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, USA
| |
Collapse
|
41
|
Böll S, Ziemann S, Ohl K, Klemm P, Rieg AD, Gulbins E, Becker KA, Kamler M, Wagner N, Uhlig S, Martin C, Tenbrock K, Verjans E. Acid sphingomyelinase regulates T H 2 cytokine release and bronchial asthma. Allergy 2020; 75:603-615. [PMID: 31494944 DOI: 10.1111/all.14039] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 08/01/2019] [Accepted: 08/19/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Allergic diseases and especially allergic asthma are widespread diseases with high prevalence in childhood, but also in adults. Acid sphingomyelinase (ASM) is a key regulator of the sphingolipid pathway. Previous studies defined the association of ASM with the pathogenesis of TH 1-directed lung diseases like cystic fibrosis and acute lung injury. Here, we define the role of ASM in TH 2-regulated allergic bronchial asthma. METHODS To determine the role of Asm under baseline conditions, wild-type (WT) and Asm-/- mice were ventilated with a flexiVent setup and bronchial hyperresponsiveness was determined using acetylcholine. Flow cytometry and cytokine measurements in bronchoalveolar lavage fluid and lung tissue were followed by in vitro TH 2 differentiations with cells from WT and Asm-/- mice and blockade of Asm with amitriptyline. As proof of principle, we conducted an ovalbumin-induced model of asthma in WT- and Asm-/- mice. RESULTS At baseline, Asm-/- mice showed better lung mechanics, but unaltered bronchial hyperresponsiveness. Higher numbers of Asm-/- T cells in bronchoalveolar lavage fluid released lower levels of IL-4 and IL-5, and these results were paralleled by decreased production of typical TH 2 cytokines in Asm-/- T lymphocytes in vitro. This phenotype could be imitated by incubation of T cells with amitriptyline. In the ovalbumin asthma model, Asm-/- animals were protected from high disease activity and showed better lung functions and lower levels of eosinophils and TH 2 cytokines. CONCLUSION Asm deficiency could induce higher numbers of TH 2 cells in the lung, but those cells release decreased TH 2 cytokine levels. Hereby, Asm-/- animals are protected from bronchial asthma, which possibly offers novel therapeutic strategies, for example, with ASM blockade.
Collapse
Affiliation(s)
- Svenja Böll
- Department of Pediatrics Medical Faculty RWTH Aachen University University Hospital Aachen Aachen Germany
- Institute of Pharmacology and Toxicology RWTH Aachen University University Hospital Aachen Aachen Germany
| | - Sebastian Ziemann
- Institute of Pharmacology and Toxicology RWTH Aachen University University Hospital Aachen Aachen Germany
- Department of Anaesthesiology Medical Faculty RWTH Aachen University University Hospital Aachen Aachen Germany
| | - Kim Ohl
- Department of Pediatrics Medical Faculty RWTH Aachen University University Hospital Aachen Aachen Germany
| | - Patricia Klemm
- Department of Pediatrics Medical Faculty RWTH Aachen University University Hospital Aachen Aachen Germany
| | - Annette D. Rieg
- Institute of Pharmacology and Toxicology RWTH Aachen University University Hospital Aachen Aachen Germany
- Department of Anaesthesiology Medical Faculty RWTH Aachen University University Hospital Aachen Aachen Germany
| | - Erich Gulbins
- Department of Molecular Biology University Hospital Essen University of Duisburg‐Essen Essen Germany
- Department of Surgery University of Cincinnati Cincinnati OH USA
| | - Katrin Anne Becker
- Department of Molecular Biology University Hospital Essen University of Duisburg‐Essen Essen Germany
| | - Markus Kamler
- Thoracic Transplantation Thoracic and Cardiovascular Surgery University Hospital Essen University of Duisburg‐Essen Essen Germany
| | - Norbert Wagner
- Department of Pediatrics Medical Faculty RWTH Aachen University University Hospital Aachen Aachen Germany
| | - Stefan Uhlig
- Institute of Pharmacology and Toxicology RWTH Aachen University University Hospital Aachen Aachen Germany
| | - Christian Martin
- Institute of Pharmacology and Toxicology RWTH Aachen University University Hospital Aachen Aachen Germany
| | - Klaus Tenbrock
- Department of Pediatrics Medical Faculty RWTH Aachen University University Hospital Aachen Aachen Germany
| | - Eva Verjans
- Department of Pediatrics Medical Faculty RWTH Aachen University University Hospital Aachen Aachen Germany
- Institute of Pharmacology and Toxicology RWTH Aachen University University Hospital Aachen Aachen Germany
| |
Collapse
|
42
|
Liu C, Zhong G, Zhou Y, Yang Y, Tan Y, Li Y, Gao X, Sun W, Li J, Jin X, Cao D, Yuan X, Liu Z, Liang S, Li Y, Du R, Zhao Y, Xue J, Zhao D, Song J, Ling S, Li Y. Alteration of calcium signalling in cardiomyocyte induced by simulated microgravity and hypergravity. Cell Prolif 2020; 53:e12783. [PMID: 32101357 PMCID: PMC7106961 DOI: 10.1111/cpr.12783] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/21/2020] [Accepted: 02/02/2020] [Indexed: 12/14/2022] Open
Abstract
Objectives Cardiac Ca2+ signalling plays an essential role in regulating excitation‐contraction coupling and cardiac remodelling. However, the response of cardiomyocytes to simulated microgravity and hypergravity and the effects on Ca2+ signalling remain unknown. Here, we elucidate the mechanisms underlying the proliferation and remodelling of HL‐1 cardiomyocytes subjected to rotation‐simulated microgravity and 4G hypergravity. Materials and Methods The cardiomyocyte cell line HL‐1 was used in this study. A clinostat and centrifuge were used to study the effects of microgravity and hypergravity, respectively, on cells. Calcium signalling was detected with laser scanning confocal microscopy. Protein and mRNA levels were detected by Western blotting and real‐time PCR, respectively. Wheat germ agglutinin (WGA) staining was used to analyse cell size. Results Our data showed that spontaneous calcium oscillations and cytosolic calcium concentration are both increased in HL‐1 cells after simulated microgravity and 4G hypergravity. Increased cytosolic calcium leads to activation of calmodulin‐dependent protein kinase II/histone deacetylase 4 (CaMKII/HDAC4) signalling and upregulation of the foetal genes ANP and BNP, indicating cardiac remodelling. WGA staining indicated that cell size was decreased following rotation‐simulated microgravity and increased following 4G hypergravity. Moreover, HL‐1 cell proliferation was increased significantly under hypergravity but not rotation‐simulated microgravity. Conclusions Our study demonstrates for the first time that Ca2+/CaMKII/HDAC4 signalling plays a pivotal role in myocardial remodelling under rotation‐simulated microgravity and hypergravity.
Collapse
Affiliation(s)
- Caizhi Liu
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Guohui Zhong
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | | | | | - Yingjun Tan
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yuheng Li
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Xingcheng Gao
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Weijia Sun
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Jianwei Li
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Xiaoyan Jin
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Dengchao Cao
- State Key Laboratory of Agrobiotechnology, College of Life Sciences, China Agricultural University, Beijing, China
| | - Xinxin Yuan
- State Key Laboratory of Agrobiotechnology, College of Life Sciences, China Agricultural University, Beijing, China
| | - Zizhong Liu
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Shuai Liang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Youyou Li
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Ruikai Du
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yinlong Zhao
- Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Jianqi Xue
- Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Dingsheng Zhao
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Jinping Song
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Shukuan Ling
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yingxian Li
- State Key Lab of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
43
|
Cheng Q, Shang Y, Huang W, Zhang Q, Li X, Zhou Q. p300 mediates the histone acetylation of ORMDL3 to affect airway inflammation and remodeling in asthma. Int Immunopharmacol 2019; 76:105885. [PMID: 31536903 DOI: 10.1016/j.intimp.2019.105885] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/20/2019] [Accepted: 09/05/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Bronchial asthma is affected by both environmental and genetic factors. The orosomucoid 1-like protein 3 (ORMDL3) gene is related to childhood asthma and is involved in airway inflammation and airway remodeling. The ORMDL3 promoter contains binding sites for the histone acetylase p300. Gene expression can be affected by epigenetic modifications. This study aimed to investigate whether the p300-mediated histone acetylation (HAT) of ORMDL3 gene affects airway inflammation and remodeling in asthma. METHODS 16HBE14o- cells were transfected with various concentrations of a wild-type p300 plasmid or p300HAT-deletion plasmids. A dual luciferase reporter assay was used to examine the effect of p300-mediated HAT on the ORMDL3 promoter. Thirty BALB/c mice were randomly divided into a control group, an ovalbumin (OVA)-induced asthma group and an asthma + C646 (a selective inhibitor of p300) group. Noninvasive lung function tests were conducted to examine airway hyperreactivity (AHR) in the different groups. HE and Masson's trichrome staining was performed to examine airway remodeling and inflammation. Immunohistochemistry, western blotting and real-time PCR were used to analyze ORMDL3 expression in lung tissues. ELISA and western blotting were used to evaluate the HAT status in lung tissue. The ChIP assay was used to determine the relationship of the ORMDL3 promoter to p300 or acetylated histone H3 (aceH3). RESULTS p300 activated transcription from the ORMDL3 promoter, resulting in an increase in endogenous ORMDL3 mRNA levels. ORMDL3 promoter activity was reduced when the HAT activity of p300 was lost. ORMDL3 expression was elevated, and HAT activity was high in the lung tissues of asthmatic mice. p300 and aceH3 bound to the promoter region of ORMDL3. In the asthma group, the amounts of p300 and aceH3 recruited to the ORMDL3 promoter were increased. C646 inhibited p300 expression and reduced HAT activity and aceH3 levels in asthmatic mice, thereby reducing ORMDL3 expression and relieving AHR and airway remodeling. CONCLUSION p300-mediated HAT modulates the expression of the asthma susceptibility gene ORMDL3, thereby improving the process of airway inflammation and remodeling in asthma.
Collapse
Affiliation(s)
- Qi Cheng
- Pediatric Pulmonology Department, Shengjing Hospital of China Medical University, 36th Sanhao Street, Heping District, Shenyang 110004, PR China.
| | - Yunxiao Shang
- Pediatric Pulmonology Department, Shengjing Hospital of China Medical University, 36th Sanhao Street, Heping District, Shenyang 110004, PR China.
| | - Wanjie Huang
- Pediatric Pulmonology Department, Shengjing Hospital of China Medical University, 36th Sanhao Street, Heping District, Shenyang 110004, PR China
| | - Qinzhen Zhang
- Pediatric Pulmonology Department, Shengjing Hospital of China Medical University, 36th Sanhao Street, Heping District, Shenyang 110004, PR China
| | - Xiang Li
- Pediatric Pulmonology Department, Shengjing Hospital of China Medical University, 36th Sanhao Street, Heping District, Shenyang 110004, PR China
| | - Qianlan Zhou
- Pediatric Pulmonology Department, Shengjing Hospital of China Medical University, 36th Sanhao Street, Heping District, Shenyang 110004, PR China
| |
Collapse
|
44
|
Bacharier LB, Mori A, Kita H. Advances in asthma, asthma-COPD overlap, and related biologics in 2018. J Allergy Clin Immunol 2019; 144:906-919. [PMID: 31476323 DOI: 10.1016/j.jaci.2019.08.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 01/14/2023]
Abstract
Over the past year, numerous important advances in our understanding of multiple aspects of asthma, ranging from disease pathogenesis to epidemiology to therapeutics, have been reported. This review is a compilation of highlights from articles published largely in the Journal of Allergy and Clinical Immunology and supplemented by articles published elsewhere that have substantially advanced the fields of asthma, chronic obstructive pulmonary disease (COPD), and asthma-COPD overlap and biologic therapies for these disorders. The intention of this article is not to provide a comprehensive review but rather to focus on several areas that have developed quickly and/or received extensive attention from our readers.
Collapse
Affiliation(s)
- Leonard B Bacharier
- Division of Pediatric Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine and St Louis Children's Hospital, St Louis, Mo.
| | - Akio Mori
- Department of Advanced Medicine, Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan
| | - Hirohito Kita
- Division of Allergic Diseases, Department of Medicine and Department of Immunology, Mayo Clinic, Rochester, Minn; Division of Allergic Diseases, Department of Medicine and Department of Immunology, Mayo Clinic, Scottsdale
| |
Collapse
|
45
|
Abstract
Current management of severe asthma relying either on guidelines (bulk approach) or on disease phenotypes (stratified approach) did not improve the burden of the disease. Several severe phenotypes are described: clinical, functional, morphological, inflammatory, molecular and microbiome-related. However, phenotypes do not necessarily relate to or give insights into the underlying pathogenetic mechanisms which are described by the disease endotypes. Based on the major immune-inflammatory pathway involved type-2 high, type-2 low and mixed endotypes are described for severe asthma, with several shared pathogenetic pathways such as genetic and epigenetic, metabolic, neurogenic and remodelling subtypes. The concept of multidimensional endotyping as un unbiased approach to severe asthma is discussed, together with new tools and targets facilitating the shift from the stratified to the precision medicine approach.
Collapse
|
46
|
James B, Milstien S, Spiegel S. ORMDL3 and allergic asthma: From physiology to pathology. J Allergy Clin Immunol 2019; 144:634-640. [PMID: 31376405 DOI: 10.1016/j.jaci.2019.07.023] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/18/2019] [Accepted: 07/26/2019] [Indexed: 01/10/2023]
Abstract
There is a strong genetic component to asthma, and numerous genome-wide association studies have identified ORM1 (yeast)-like protein 3 (ORMDL3) as a gene associated with asthma susceptibility. However, how ORMDL3 contributes to asthma pathogenesis and its physiologic functions is not well understood and a matter of great debate. This rostrum describes recent advances and new insights in understanding of the multifaceted functions of ORMDL3 in patients with allergic asthma. We also suggest a potential unifying paradigm and discuss molecular mechanisms for the pathologic functions of ORMDL3 in asthma related to its evolutionarily conserved role in regulation of sphingolipid homeostasis. Finally, we briefly survey the utility of sphingolipid metabolites as potential biomarkers for allergic asthma.
Collapse
Affiliation(s)
- Briana James
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Va
| | - Sheldon Milstien
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Va
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Va.
| |
Collapse
|
47
|
Miller M, Broide DH. Why Is ORMDL3 on Chromosome 17q21 Highly Linked to Asthma? Am J Respir Crit Care Med 2019; 199:404-406. [PMID: 30365391 DOI: 10.1164/rccm.201810-1941ed] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Marina Miller
- 1 Department of Medicine University of California San Diego La Jolla, California
| | - David H Broide
- 1 Department of Medicine University of California San Diego La Jolla, California
| |
Collapse
|
48
|
Willis-Owen SAG, Cookson WOC, Moffatt MF. The Genetics and Genomics of Asthma. Annu Rev Genomics Hum Genet 2019; 19:223-246. [PMID: 30169121 DOI: 10.1146/annurev-genom-083117-021651] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Asthma is a common, clinically heterogeneous disease with strong evidence of heritability. Progress in defining the genetic underpinnings of asthma, however, has been slow and hampered by issues of inconsistency. Recent advances in the tools available for analysis-assaying transcription, sequence variation, and epigenetic marks on a genome-wide scale-have substantially altered this landscape. Applications of such approaches are consistent with heterogeneity at the level of causation and specify patterns of commonality with a wide range of alternative disease traits. Looking beyond the individual as the unit of study, advances in technology have also fostered comprehensive analysis of the human microbiome and its varied roles in health and disease. In this article, we consider the implications of these technological advances for our current understanding of the genetics and genomics of asthma.
Collapse
Affiliation(s)
- Saffron A G Willis-Owen
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, United Kingdom; , ,
| | - William O C Cookson
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, United Kingdom; , ,
| | - Miriam F Moffatt
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, United Kingdom; , ,
| |
Collapse
|
49
|
Shrine N, Portelli MA, John C, Soler Artigas M, Bennett N, Hall R, Lewis J, Henry AP, Billington CK, Ahmad A, Packer RJ, Shaw D, Pogson ZEK, Fogarty A, McKeever TM, Singapuri A, Heaney LG, Mansur AH, Chaudhuri R, Thomson NC, Holloway JW, Lockett GA, Howarth PH, Djukanovic R, Hankinson J, Niven R, Simpson A, Chung KF, Sterk PJ, Blakey JD, Adcock IM, Hu S, Guo Y, Obeidat M, Sin DD, van den Berge M, Nickle DC, Bossé Y, Tobin MD, Hall IP, Brightling CE, Wain LV, Sayers I. Moderate-to-severe asthma in individuals of European ancestry: a genome-wide association study. THE LANCET. RESPIRATORY MEDICINE 2019; 7:20-34. [PMID: 30552067 PMCID: PMC6314966 DOI: 10.1016/s2213-2600(18)30389-8] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/12/2018] [Accepted: 09/13/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Few genetic studies that focus on moderate-to-severe asthma exist. We aimed to identity novel genetic variants associated with moderate-to-severe asthma, see whether previously identified genetic variants for all types of asthma contribute to moderate-to-severe asthma, and provide novel mechanistic insights using expression analyses in patients with asthma. METHODS In this genome-wide association study, we used a two-stage case-control design. In stage 1, we genotyped patient-level data from two UK cohorts (the Genetics of Asthma Severity and Phenotypes [GASP] initiative and the Unbiased BIOmarkers in PREDiction of respiratory disease outcomes [U-BIOPRED] project) and used data from the UK Biobank to collect patient-level genomic data for cases and controls of European ancestry in a 1:5 ratio. Cases were defined as having moderate-to-severe asthma if they were taking appropriate medication or had been diagnosed by a doctor. Controls were defined as not having asthma, rhinitis, eczema, allergy, emphysema, or chronic bronchitis as diagnosed by a doctor. For stage 2, an independent cohort of cases and controls (1:5) was selected from the UK Biobank only, with no overlap with stage 1 samples. In stage 1 we undertook a genome-wide association study of moderate-to-severe asthma, and in stage 2 we followed up independent variants that reached the significance threshold of p less than 1 × 10-6 in stage 1. We set genome-wide significance at p less than 5 × 10-8. For novel signals, we investigated their effect on all types of asthma (mild, moderate, and severe). For all signals meeting genome-wide significance, we investigated their effect on gene expression in patients with asthma and controls. FINDINGS We included 5135 cases and 25 675 controls for stage 1, and 5414 cases and 21 471 controls for stage 2. We identified 24 genome-wide significant signals of association with moderate-to-severe asthma, including several signals in innate or adaptive immune-response genes. Three novel signals were identified: rs10905284 in GATA3 (coded allele A, odds ratio [OR] 0·90, 95% CI 0·88-0·93; p=1·76 × 10-10), rs11603634 in the MUC5AC region (coded allele G, OR 1·09, 1·06-1·12; p=2·32 × 10-8), and rs560026225 near KIAA1109 (coded allele GATT, OR 1·12, 1·08-1·16; p=3·06 × 10-9). The MUC5AC signal was not associated with asthma when analyses included mild asthma. The rs11603634 G allele was associated with increased expression of MUC5AC mRNA in bronchial epithelial brush samples via proxy SNP rs11602802; (p=2·50 × 10-5) and MUC5AC mRNA was increased in bronchial epithelial samples from patients with severe asthma (in two independent analyses, p=0·039 and p=0·022). INTERPRETATION We found substantial shared genetic architecture between mild and moderate-to-severe asthma. We also report for the first time genetic variants associated with the risk of developing moderate-to-severe asthma that regulate mucin production. Finally, we identify candidate causal genes in these loci and provide increased insight into this difficult to treat population. FUNDING Asthma UK, AirPROM, U-BIOPRED, UK Medical Research Council, and Rosetrees Trust.
Collapse
Affiliation(s)
- Nick Shrine
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Michael A Portelli
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Catherine John
- Department of Health Sciences, University of Leicester, Leicester, UK
| | | | - Neil Bennett
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Robert Hall
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Jon Lewis
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Amanda P Henry
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Charlotte K Billington
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Azaz Ahmad
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Richard J Packer
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Dominick Shaw
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Zara E K Pogson
- Division of Epidemiology and Public Health, University of Nottingham, Nottingham, UK
| | - Andrew Fogarty
- Division of Epidemiology and Public Health, University of Nottingham, Nottingham, UK
| | - Tricia M McKeever
- Division of Epidemiology and Public Health, University of Nottingham, Nottingham, UK
| | - Amisha Singapuri
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK; Glenfield Hospital, Leicester, UK
| | - Liam G Heaney
- Centre for Infection and Immunity, Queen's University of Belfast, Belfast, UK
| | - Adel H Mansur
- Respiratory Medicine, Birmingham Heartlands Hospital and University of Birmingham, Birmingham, UK
| | - Rekha Chaudhuri
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Neil C Thomson
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - John W Holloway
- Human Development and Health, Clinical and Experimental Sciences, Faculty of Medicine and National Institute of Health Biomedical Research Centre, Southampton, University of Southampton, Southampton, UK
| | - Gabrielle A Lockett
- Human Development and Health, Clinical and Experimental Sciences, Faculty of Medicine and National Institute of Health Biomedical Research Centre, Southampton, University of Southampton, Southampton, UK
| | - Peter H Howarth
- Human Development and Health, Clinical and Experimental Sciences, Faculty of Medicine and National Institute of Health Biomedical Research Centre, Southampton, University of Southampton, Southampton, UK
| | - Ratko Djukanovic
- Human Development and Health, Clinical and Experimental Sciences, Faculty of Medicine and National Institute of Health Biomedical Research Centre, Southampton, University of Southampton, Southampton, UK
| | - Jenny Hankinson
- Division of Infection Immunity and Respiratory Medicine, The University of Manchester, Manchester Academic Health Science Centre, and Manchester University NHS Foundation Trust, Manchester, UK
| | - Robert Niven
- Division of Infection Immunity and Respiratory Medicine, The University of Manchester, Manchester Academic Health Science Centre, and Manchester University NHS Foundation Trust, Manchester, UK
| | - Angela Simpson
- Division of Infection Immunity and Respiratory Medicine, The University of Manchester, Manchester Academic Health Science Centre, and Manchester University NHS Foundation Trust, Manchester, UK
| | - Kian Fan Chung
- The National Heart and Lung Institute, Imperial College, London, UK
| | - Peter J Sterk
- Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - John D Blakey
- Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Ian M Adcock
- The National Heart and Lung Institute, Imperial College, London, UK
| | - Sile Hu
- Data Science Institute, Imperial College, London, UK
| | - Yike Guo
- Data Science Institute, Imperial College, London, UK
| | - Maen Obeidat
- The University of British Columbia Center for Heart Lung Innovation, St Paul's Hospital Vancouver, Vancouver, BC, Canada
| | - Don D Sin
- The University of British Columbia Center for Heart Lung Innovation, St Paul's Hospital Vancouver, Vancouver, BC, Canada; Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Maarten van den Berge
- University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen Research Institute for Asthma and COPD Research Institute, Groningen, Netherlands
| | | | - Yohan Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Molecular Medicine, Laval University, Quebec City, QC, Canada
| | - Martin D Tobin
- Department of Health Sciences, University of Leicester, Leicester, UK; National Institute for Health Research, Leicester Respiratory Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Ian P Hall
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Christopher E Brightling
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK; National Institute for Health Research, Leicester Respiratory Biomedical Research Centre, University of Leicester, Leicester, UK; Glenfield Hospital, Leicester, UK
| | - Louise V Wain
- Department of Health Sciences, University of Leicester, Leicester, UK; National Institute for Health Research, Leicester Respiratory Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Ian Sayers
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK.
| |
Collapse
|
50
|
Ma X, Long F, Yun Y, Dang J, Wei S, Zhang Q, Li J, Zhang H, Zhang W, Wang Z, Liu Q, Zou C. ORMDL3 and its implication in inflammatory disorders. Int J Rheum Dis 2018; 21:1154-1162. [PMID: 29879314 DOI: 10.1111/1756-185x.13324] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A growing body of evidence has suggested the genetic association of ORMDL3 gene (ORMDL Sphingolipid Biosynthesis Regulator 3) polymorphisms with a diverse set of inflammatory disorders that include bronchial asthma, inflammatory bowel disease, ankylosing spondylitis and atherosclerosis. Gene functional investigations have revealed the particular relevance of ORMDL3 in endoplasmic reticulum stress, lipid metabolism and inflammatory reactions. Additionally, several reports have recently added a new dimension to our understanding of the modulation of ORMDL3 gene expression in inflammation. This mini-review summarizes the pertinent publications regarding the genetic association studies and mechanistic exploration of ORMDL3 in common inflammatory disorders.
Collapse
Affiliation(s)
- Xiaochun Ma
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China.,Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, China
| | - Feng Long
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, China
| | - Yan Yun
- Brain Research Institute, Qilu Hospital of Shandong University, Jinan, China
| | - Jie Dang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, China.,Department of Medical Genetics and Cell Biology, Ningxia Medical University, Yinchuan, China
| | - Shijun Wei
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, China
| | - Qian Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Jinzhang Li
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Haizhou Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Wenlong Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Zhengjun Wang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Qiji Liu
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, Shandong University School of Medicine, Jinan, China
| | - Chengwei Zou
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| |
Collapse
|