1
|
Lin L, Liao Z, Li Y, Pan S, Wu S, Sun QX, Li C. Transcriptomic analysis and validation study of key genes and the HIF‑1α/HO‑1 pathway associated with ferroptosis in neutrophilic asthma. Exp Ther Med 2024; 28:433. [PMID: 39347495 PMCID: PMC11425779 DOI: 10.3892/etm.2024.12722] [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: 02/04/2024] [Accepted: 06/19/2024] [Indexed: 10/01/2024] Open
Abstract
Ferroptosis, as a unique form of cell death caused by iron overload and lipid peroxidation, is involved in the pathogenesis of various inflammatory diseases of the airways. Inhibition of ferroptosis has become a novel strategy for reducing airway epithelial cell death and improving airway inflammation. The aim of the present study was to analyze and validate the key genes and signaling pathways associated with ferroptosis by bioinformatic methods combined with experimental analyzes in vitro and in vivo to aid the diagnosis and treatment of neutrophilic asthma. A total of 1,639 differentially expressed genes (DEGs) were identified in the transcriptome dataset. After overlapping with ferroptosis-related genes, 11 differentially expressed ferroptosis-related genes (DE-FRGs) were obtained. A new diagnostic model was constructed by these DE-FRGs from the transcriptome dataset with those from the GSE108417 dataset. The receiver operating characteristic curve analysis indicated that the area under the curve had good diagnostic performance (>0.8). As a result, four key DE-FRGs (CXCL2, HMOX1, IL-6 and SLC7A5) and biological pathway [hypoxia-inducible factor 1 (HIF-1) signaling pathway] associated with ferroptosis in neutrophilic asthma were identified by the bioinformatics analysis combined with experimental validation. The upstream regulatory network of key DE-FRGs and target drugs were predicted and the molecular docking results from screened 37 potential therapeutic drugs revealed that the 13 small-molecule drugs exhibited a higher stable binding to the primary proteins of key DE-FRGs. The results suggested that four key DE-FRGs and the HIF-1α/heme oxygenase 1 pathway associated with ferroptosis have potential as novel markers or targets for the diagnosis or treatment of neutrophilic asthma.
Collapse
Affiliation(s)
- Lu Lin
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Department of Pulmonary and Critical Care Medicine, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| | - Zenghua Liao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yinghua Li
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| | - Shitong Pan
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| | - Sihui Wu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qi-Xiang Sun
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chaoqian Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
2
|
Pasha MA, Hopp RJ, Habib N, Tang DD. Biomarkers in asthma, potential for therapeutic intervention. J Asthma 2024; 61:1376-1391. [PMID: 38805392 DOI: 10.1080/02770903.2024.2361783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/09/2024] [Accepted: 05/26/2024] [Indexed: 05/30/2024]
Abstract
Asthma is a heterogeneous disease characterized by multiple phenotypes with varying risk factors and therapeutic responses. This Commentary describes research on biomarkers for T2-"high" and T2-"low" inflammation, a hallmark of the disease. Patients with asthma who exhibit an increase in airway T2 inflammation are classified as having T2-high asthma. In this endotype, Type 2 cytokines interleukins (IL)-4, IL-5, and IL-13, plus other inflammatory mediators, lead to increased eosinophilic inflammation and elevated fractional exhaled nitric oxide (FeNO). In contrast, T2-low asthma has no clear definition. Biomarkers are considered valuable tools as they can help identify various phenotypes and endotypes, as well as treatment response to standard treatment or potential therapeutic targets, particularly for biologics. As our knowledge of phenotypes and endotypes expands, biologics are increasingly integrated into treatment strategies for severe asthma. These treatments block specific inflammatory pathways or single mediators. While single or composite biomarkers may help to identify subsets of patients who might benefit from these treatments, only a few inflammatory biomarkers have been validated for clinical application. One example is sputum eosinophilia, a particularly useful biomarker, as it may suggest corticosteroid responsiveness or reflect non-compliance to inhaled corticosteroids. As knowledge develops, a meaningful goal would be to provide individualized care to patients with asthma.
Collapse
Affiliation(s)
- M Asghar Pasha
- Department of Medicine, Division of Allergy and Immunology, Albany Medical College, Albany, NY, USA
| | - Russell J Hopp
- Department of Pediatrics, University of NE Medical Center and Children's Hospital and Medical Center, Omaha, NE, USA
| | - Nazia Habib
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Dale D Tang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| |
Collapse
|
3
|
Sey EA, Warris A. The gut-lung axis: the impact of the gut mycobiome on pulmonary diseases and infections. OXFORD OPEN IMMUNOLOGY 2024; 5:iqae008. [PMID: 39193472 PMCID: PMC11316619 DOI: 10.1093/oxfimm/iqae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 08/29/2024] Open
Abstract
The gastrointestinal tract contains a diverse microbiome consisting of bacteria, fungi, viruses and archaea. Although these microbes usually reside as commensal organisms, it is now well established that higher abundance of specific bacterial or fungal species, or loss of diversity in the microbiome can significantly affect development, progression and outcomes in disease. Studies have mainly focused on the effects of bacteria, however, the impact of other microbes, such as fungi, has received increased attention in the last few years. Fungi only represent around 0.1% of the total gut microbial population. However, key fungal taxa such as Candida, Aspergillus and Wallemia have been shown to significantly impact health and disease. The composition of the gut mycobiome has been shown to affect immunity at distal sites, such as the heart, lung, brain, pancreas, and liver. In the case of the lung this phenomenon is referred to as the 'gut-lung axis'. Recent studies have begun to explore and unveil the relationship between gut fungi and lung immunity in diseases such as asthma and lung cancer, and lung infections caused by viruses, bacteria and fungi. In this review we will summarize the current, rapidly growing, literature describing the impact of the gut mycobiome on respiratory disease and infection.
Collapse
Affiliation(s)
- Emily A Sey
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, EX4 4QD, UK
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, EX4 4QD, UK
| |
Collapse
|
4
|
Zhang K, Zhang M, Su H, Zhao F, Wang D, Zhang Y, Cao G, Zhang Y. Regulation of Inflammatory Responses of Cow Mammary Epithelial Cells through MAPK Signaling Pathways of IL-17A Cytokines. Animals (Basel) 2024; 14:1572. [PMID: 38891619 PMCID: PMC11171030 DOI: 10.3390/ani14111572] [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: 04/23/2024] [Revised: 05/19/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
The aim of this study is to explore the mechanism of IL-17A infection in the development of bacterial mastitis in dairy cows. In this study, RT-qPCR and ELISA were used to measure the promoting effect of IL-17A on the generation of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) and chemokine (IL-8). In addition, Western blot (WB) was applied to measure the influences of IL-17A on the inflammation-related ERK and p38 proteins in the MAPK pathways. The results show that under the stimulation of LPS on cow mammary epithelial cells (CMECs), cytokines IL-1β, IL-6, IL-8, TNF-α, and IL-17A will exhibit significantly increased expression levels (p < 0.05). With inhibited endogenous expression of IL-17A, cytokines IL-1β, IL-6, IL-8, and TNF-α will present reduced genetic expression (p < 0.01), with reduced phosphorylation levels of ERK and p38 proteins in the MAPK signaling pathways (p < 0.001). Upon the exogenous addition of the IL-17A cytokine, the genetic expression of cytokines IL-1β, IL-6, IL-8, and TNF-α will increase (p < 0.05), with increased phosphorylation levels of the ERK and p38 proteins in the MAPK signaling pathways (p < 0.001). These results indicate that under the stimulation of CMECs with LPS, IL-17A can be expressed together with relevant inflammatory cytokines. Meanwhile, the inflammatory responses of mammary epithelial cells are directly proportional to the expression levels of IL-17A inhibited alone or exogenously added. In summary, this study shows that IL-17A could be used as an important indicator for assessing the bacterial infections of mammary glands, indicating that IL-17A could be regarded as one potential therapeutic target for mastitis.
Collapse
Affiliation(s)
- Kai Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010011, China; (K.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.); (Y.Z.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Min Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010011, China; (K.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.); (Y.Z.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Hong Su
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010011, China; (K.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.); (Y.Z.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Feifei Zhao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010011, China; (K.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.); (Y.Z.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Daqing Wang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010011, China; (K.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.); (Y.Z.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Yuanyuan Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010011, China; (K.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.); (Y.Z.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Guifang Cao
- College of Life Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Yong Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010011, China; (K.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.); (Y.Z.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| |
Collapse
|
5
|
Zhang D, Liu H, Zhao F, Guo P, Li J, Lu T, Li Z, Li S. Exploring the relationship between Treg-mediated risk in COPD and lung cancer through Mendelian randomization analysis and scRNA-seq data integration. BMC Cancer 2024; 24:453. [PMID: 38605291 PMCID: PMC11010300 DOI: 10.1186/s12885-024-12076-1] [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: 12/08/2023] [Accepted: 03/03/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Evidence from observational studies suggests an association between chronic obstructive pulmonary disease (COPD) and lung cancer. The potential interactions between the immune system and the lungs may play a causative role in COPD and lung cancer and offer therapeutic prospects. However, the causal association and the immune-mediated mechanisms between COPD and lung cancer remain to be determined. METHODS We employed a two-sample Mendelian randomization (MR) approach to investigate the causal association between COPD and lung cancer. Additionally, we examined whether immune cell signals were causally related to lung cancer, as well as whether COPD was causally associated with immune cell signals. Furthermore, through two-step Mendelian randomization, we investigated the mediating effects of immune cell signals in the causal association between COPD and lung cancer. Leveraging publicly available genetic data, our analysis included 468,475 individuals of European ancestry with COPD, 492,803 individuals of European ancestry with lung cancer, and 731 immune cell signatures of European ancestry. Additionally, we conducted single-cell transcriptome sequencing analysis on COPD, lung cancer, and control samples to validate our findings. FINDINGS We found a causal association between COPD and lung cancer (odds ratio [OR] = 1.63, 95% confidence interval [CI] = 1.31-2.02, P-value < 0.001). We also observed a causal association between COPD and regulatory T cells (odds ratio [OR] = 1.19, 95% confidence interval [CI] = 1.01-1.40, P-value < 0.05), as well as a causal association between regulatory T cells and lung cancer (odds ratio [OR] = 1.02, 95% confidence interval [CI] = 1.002-1.045, P-value < 0.05). Furthermore, our two-step Mendelian randomization analysis demonstrated that COPD is associated with lung cancer through the mediation of regulatory T cells. These findings were further validated through single-cell sequencing analysis, confirming the mediating role of regulatory T cells in the association between COPD and lung cancer. INTERPRETATION As far as we are aware, we are the first to combine single-celled immune cell data with two-sample Mendelian randomization. Our analysis indicates a causal association between COPD and lung cancer, with regulatory T cells playing an intermediary role.
Collapse
Affiliation(s)
- Dengfeng Zhang
- Provincial Center for Clinical Laboratories,Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Haitao Liu
- College of Life Science, Inner Mongolia University, Hohhot, China
| | - Fangchao Zhao
- Provincial Center for Clinical Laboratories,Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Pengfei Guo
- Provincial Center for Clinical Laboratories,Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Li
- Provincial Center for Clinical Laboratories,Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tianxing Lu
- Provincial Center for Clinical Laboratories,Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhirong Li
- Provincial Center for Clinical Laboratories,Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Shujun Li
- Provincial Center for Clinical Laboratories,Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
6
|
Liu Y, Cheng K, Sun M, Ding C, Li T, Jia Y, Wang C, Zhu X, Song X, Jia R, Wang Q, Zhang Y, Sun X. UBD participates in neutrophilic asthma by promoting the activation of IL-17 signaling. Int J Biol Macromol 2024; 264:130581. [PMID: 38447828 DOI: 10.1016/j.ijbiomac.2024.130581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/24/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024]
Abstract
Neutrophilic asthma is a persistent and severe inflammatory lung disease characterized by neutrophil activation and the mechanisms of which are not completely elucidated. Ubiquitin D (UBD) is a ubiquitin-like modifier participating in infections, immune responses, and tumorigenesis, while whether UBD involves in neutrophilic asthma needs further study. In this study, we initially found that UBD expression was significantly elevated and interleukin 17 (IL-17) signaling was enriched in the endobronchial biopsies of severe asthma along with neutrophils increasing by bioinformatics analysis. We further confirmed that UBD was upregulated in the lung tissues of neutrophilic asthma mouse model. UBD overexpression promoted IL-17 signaling activation. Knockdown of UBD suppressed the activation of IL-17 signaling. UBD interacted with TRAF2 and reduced the total and the K48-linked ubiquitination of TRAF2. However, IL-17 A stimulation increased both the total and the K48-linked ubiquitination of TRAF2. Together, these findings indicated that UBD was upregulated and played a critical role in IL-17 signaling which contributed to a better understanding of the complex mechanisms in neutrophilic asthma.
Collapse
Affiliation(s)
- Yuchun Liu
- Henan International Joint Laboratory of Children's Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, 450000, China.
| | - Kang Cheng
- Institute of Biomedical Research, Henan Academy of Sciences, Zhengzhou 450000, China
| | - Meng Sun
- The Third People's Hospital of Zhengzhou, Cancer Hospital of Zhengzhou, The First Mercy Hospital of Henan Province, 450000, China
| | - Cong Ding
- Henan International Joint Laboratory of Children's Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, 450000, China
| | - Tao Li
- Henan International Joint Laboratory of Children's Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, 450000, China
| | - Yangyang Jia
- Henan International Joint Laboratory of Children's Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, 450000, China
| | - Chengbo Wang
- Henan International Joint Laboratory of Children's Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, 450000, China
| | - Xiangzhan Zhu
- Henan International Joint Laboratory of Children's Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, 450000, China
| | - Xiaorui Song
- Henan International Joint Laboratory of Children's Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, 450000, China
| | - Rui Jia
- Henan International Joint Laboratory of Children's Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, 450000, China
| | - Qionglin Wang
- Henan International Joint Laboratory of Children's Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, 450000, China
| | - Yaodong Zhang
- Henan International Joint Laboratory of Children's Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, 450000, China
| | - Xiaomin Sun
- Henan International Joint Laboratory of Children's Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, 450000, China.
| |
Collapse
|
7
|
Dill-McFarland KA, Altman MC, Esnault S, Jarjour NN, Busse WW, Rosenkranz MA. Molecular pathways underlying lung-brain axis signaling in asthma: Relevance for psychopathology and neuroinflammation. J Allergy Clin Immunol 2024; 153:111-121. [PMID: 37730134 PMCID: PMC10841090 DOI: 10.1016/j.jaci.2023.07.025] [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: 12/13/2022] [Revised: 07/12/2023] [Accepted: 07/20/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND Accumulating evidence indicates that asthma has systemic effects and affects brain function. Although airway inflammation is proposed to initiate afferent communications with the brain, the signaling pathways have not been established. OBJECTIVE We sought to identify the cellular and molecular pathways involved in afferent lung-brain communication during airway inflammation in asthma. METHODS In 23 adults with mild asthma, segmental bronchial provocation with allergen (SBP-Ag) was used to provoke airway inflammation and retrieve bronchoalveolar lavage fluid for targeted protein analysis and RNA sequencing to determine gene expression profiles. Neural responses to emotional cues in nodes of the salience network were assessed with functional magnetic resonance imaging at baseline and 48 hours after SBP-Ag. RESULTS Cell deconvolution and gene coexpression network analysis identified 11 cell-associated gene modules that changed in response to SBP-Ag. SBP-Ag increased bronchoalveolar lavage eosinophils and expression of an eosinophil-associated module enriched for genes related to TH17-type inflammation (eg, IL17A), as well as cell proliferation in lung and brain (eg, NOTCH1, VEGFA, and LIF). Increased expression of genes in this module, as well as several TH17-type inflammation-related proteins, was associated with an increase from baseline in salience network reactivity. CONCLUSIONS Our results identify a specific inflammatory pathway linking asthma-related airway inflammation and emotion-related neural function. Systemically, TH17-type inflammation has been implicated in both depression and neuroinflammation, with impacts on long-term brain health. Thus, our data emphasize that inflammation in the lung in asthma may have profound effects outside of the lung that may be targetable with novel therapeutic approaches.
Collapse
Affiliation(s)
| | - Matthew C Altman
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Wash; Systems Immunology Program, Benaroya Research Institute, Seattle, Wash
| | - Stephane Esnault
- Division of Allergy, Pulmonary and Critical Care Medicine, University of Wisconsin-Madison, Madison, Wis
| | - Nizar N Jarjour
- Division of Allergy, Pulmonary and Critical Care Medicine, University of Wisconsin-Madison, Madison, Wis
| | - William W Busse
- Division of Allergy, Pulmonary and Critical Care Medicine, University of Wisconsin-Madison, Madison, Wis
| | - Melissa A Rosenkranz
- Center for Healthy Minds, University of Wisconsin-Madison, Madison, Wis; Department of Psychiatry, University of Wisconsin-Madison, Madison, Wis.
| |
Collapse
|
8
|
Lu H, Cao P. Neural Mechanisms Underlying the Coughing Reflex. Neurosci Bull 2023; 39:1823-1839. [PMID: 37606821 PMCID: PMC10661548 DOI: 10.1007/s12264-023-01104-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/15/2023] [Indexed: 08/23/2023] Open
Abstract
Breathing is an intrinsic natural behavior and physiological process that maintains life. The rhythmic exchange of gases regulates the delicate balance of chemical constituents within an organism throughout its lifespan. However, chronic airway diseases, including asthma and chronic obstructive pulmonary disease, affect millions of people worldwide. Pathological airway conditions can disrupt respiration, causing asphyxia, cardiac arrest, and potential death. The innervation of the respiratory tract and the action of the immune system confer robust airway surveillance and protection against environmental irritants and pathogens. However, aberrant activation of the immune system or sensitization of the nervous system can contribute to the development of autoimmune airway disorders. Transient receptor potential ion channels and voltage-gated Na+ channels play critical roles in sensing noxious stimuli within the respiratory tract and interacting with the immune system to generate neurogenic inflammation and airway hypersensitivity. Although recent studies have revealed the involvement of nociceptor neurons in airway diseases, the further neural circuitry underlying airway protection remains elusive. Unraveling the mechanism underpinning neural circuit regulation in the airway may provide precise therapeutic strategies and valuable insights into the management of airway diseases.
Collapse
Affiliation(s)
- Haicheng Lu
- National Institute of Biological Sciences, Beijing, 102206, China.
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Peng Cao
- National Institute of Biological Sciences, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China
| |
Collapse
|
9
|
Barbosa JAS, da Silva LLS, João JMLG, de Campos EC, Fukuzaki S, Camargo LDN, dos Santos TM, dos Santos HT, Bezerra SKM, Saraiva-Romanholo BM, Lopes FDTQDS, Bonturi CR, Oliva MLV, Leick EA, Righetti RF, Tibério IDFLC. Investigating the Effects of a New Peptide, Derived from the Enterolobium contortisiliquum Proteinase Inhibitor (EcTI), on Inflammation, Remodeling, and Oxidative Stress in an Experimental Mouse Model of Asthma-Chronic Obstructive Pulmonary Disease Overlap (ACO). Int J Mol Sci 2023; 24:14710. [PMID: 37834157 PMCID: PMC10573003 DOI: 10.3390/ijms241914710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
The synthesized peptide derived from Enterolobium contortisiliquum (pep3-EcTI) has been associated with potent anti-inflammatory and antioxidant effects, and it may be a potential new treatment for asthma-COPD overlap-ACO). Purpose: To investigate the primary sequence effects of pep3-EcTI in an experimental ACO. BALB/c mice were divided into eight groups: SAL (saline), OVA (ovalbumin), ELA (elastase), ACO (ovalbumin + elastase), ACO-pep3-EcTI (treated with inhibitor), ACO-DX (treated with dexamethasone), ACO-DX-pep3-EcTI (treated with dexamethasone and inhibitor), and SAL-pep3-EcTI (saline group treated with inhibitor). We evaluated the hyperresponsiveness to methacholine, exhaled nitric oxide, bronchoalveolar lavage fluid (BALF), mean linear intercept (Lm), inflammatory markers, tumor necrosis factor (TNF-α), interferon (IFN)), matrix metalloproteinases (MMPs), growth factor (TGF-β), collagen fibers, the oxidative stress marker inducible nitric oxide synthase (iNOS), transcription factors, and the signaling pathway NF-κB in the airways (AW) and alveolar septa (AS). Statistical analysis was conducted using one-way ANOVA and t-tests, significant when p < 0.05. ACO caused alterations in the airways and alveolar septa. Compared with SAL, ACO-pep3-EcTI reversed the changes in the percentage of resistance of the respiratory system (%Rrs), the elastance of the respiratory system (%Ers), tissue resistance (%Gtis), tissue elastance (%Htis), airway resistance (%Raw), Lm, exhaled nitric oxide (ENO), lymphocytes, IL-4, IL-5, IL-6, IL-10, IL-13, IL-17, TNF-α, INF-γ, MMP-12, transforming growth factor (TGF)-β, collagen fibers, and iNOS. ACO-DX reversed the changes in %Rrs, %Ers, %Gtis, %Htis, %Raw, total cells, eosinophils, neutrophils, lymphocytes, macrophages, IL-1β, IL-6, IL-10, IL-13, IL-17, TNF-α, INF-γ, MMP-12, TGF-β, collagen fibers, and iNOS. ACO-DX-pep3-EcTI reversed the changes, as was also observed for the pep3-EcTI and the ACO-DX-pep3-EcTI. Significance: The pep3-EcTI was revealed to be a promising strategy for the treatment of ACO, asthma, and COPD.
Collapse
Affiliation(s)
- Jéssica Anastácia Silva Barbosa
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil; (J.A.S.B.); (L.L.S.d.S.); (J.M.L.G.J.); (E.C.d.C.); (S.F.); (L.d.N.C.); (T.M.d.S.); (H.T.d.S.); (S.K.M.B.); (B.M.S.-R.); (F.D.T.Q.d.S.L.); (E.A.L.); (R.F.R.)
- Hospital Sírio Libanês, São Paulo 01308-050, Brazil
| | - Luana Laura Sales da Silva
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil; (J.A.S.B.); (L.L.S.d.S.); (J.M.L.G.J.); (E.C.d.C.); (S.F.); (L.d.N.C.); (T.M.d.S.); (H.T.d.S.); (S.K.M.B.); (B.M.S.-R.); (F.D.T.Q.d.S.L.); (E.A.L.); (R.F.R.)
| | - Juliana Morelli Lopes Gonçalves João
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil; (J.A.S.B.); (L.L.S.d.S.); (J.M.L.G.J.); (E.C.d.C.); (S.F.); (L.d.N.C.); (T.M.d.S.); (H.T.d.S.); (S.K.M.B.); (B.M.S.-R.); (F.D.T.Q.d.S.L.); (E.A.L.); (R.F.R.)
- Hospital Sírio Libanês, São Paulo 01308-050, Brazil
| | - Elaine Cristina de Campos
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil; (J.A.S.B.); (L.L.S.d.S.); (J.M.L.G.J.); (E.C.d.C.); (S.F.); (L.d.N.C.); (T.M.d.S.); (H.T.d.S.); (S.K.M.B.); (B.M.S.-R.); (F.D.T.Q.d.S.L.); (E.A.L.); (R.F.R.)
- Hospital Sírio Libanês, São Paulo 01308-050, Brazil
| | - Silvia Fukuzaki
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil; (J.A.S.B.); (L.L.S.d.S.); (J.M.L.G.J.); (E.C.d.C.); (S.F.); (L.d.N.C.); (T.M.d.S.); (H.T.d.S.); (S.K.M.B.); (B.M.S.-R.); (F.D.T.Q.d.S.L.); (E.A.L.); (R.F.R.)
| | - Leandro do Nascimento Camargo
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil; (J.A.S.B.); (L.L.S.d.S.); (J.M.L.G.J.); (E.C.d.C.); (S.F.); (L.d.N.C.); (T.M.d.S.); (H.T.d.S.); (S.K.M.B.); (B.M.S.-R.); (F.D.T.Q.d.S.L.); (E.A.L.); (R.F.R.)
- Hospital Sírio Libanês, São Paulo 01308-050, Brazil
| | - Tabata Maruyama dos Santos
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil; (J.A.S.B.); (L.L.S.d.S.); (J.M.L.G.J.); (E.C.d.C.); (S.F.); (L.d.N.C.); (T.M.d.S.); (H.T.d.S.); (S.K.M.B.); (B.M.S.-R.); (F.D.T.Q.d.S.L.); (E.A.L.); (R.F.R.)
- Hospital Sírio Libanês, São Paulo 01308-050, Brazil
| | - Henrique Tibucheski dos Santos
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil; (J.A.S.B.); (L.L.S.d.S.); (J.M.L.G.J.); (E.C.d.C.); (S.F.); (L.d.N.C.); (T.M.d.S.); (H.T.d.S.); (S.K.M.B.); (B.M.S.-R.); (F.D.T.Q.d.S.L.); (E.A.L.); (R.F.R.)
| | - Suellen Karoline Moreira Bezerra
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil; (J.A.S.B.); (L.L.S.d.S.); (J.M.L.G.J.); (E.C.d.C.); (S.F.); (L.d.N.C.); (T.M.d.S.); (H.T.d.S.); (S.K.M.B.); (B.M.S.-R.); (F.D.T.Q.d.S.L.); (E.A.L.); (R.F.R.)
| | - Beatriz Mangueira Saraiva-Romanholo
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil; (J.A.S.B.); (L.L.S.d.S.); (J.M.L.G.J.); (E.C.d.C.); (S.F.); (L.d.N.C.); (T.M.d.S.); (H.T.d.S.); (S.K.M.B.); (B.M.S.-R.); (F.D.T.Q.d.S.L.); (E.A.L.); (R.F.R.)
- Department of Medicine, University City of São Paulo, São Paulo 03071-000, Brazil
| | - Fernanda Degobbi Tenório Quirino dos Santos Lopes
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil; (J.A.S.B.); (L.L.S.d.S.); (J.M.L.G.J.); (E.C.d.C.); (S.F.); (L.d.N.C.); (T.M.d.S.); (H.T.d.S.); (S.K.M.B.); (B.M.S.-R.); (F.D.T.Q.d.S.L.); (E.A.L.); (R.F.R.)
| | - Camila Ramalho Bonturi
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), São Paulo 04039-002, Brazil; (C.R.B.); (M.L.V.O.)
| | - Maria Luiza Vilela Oliva
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), São Paulo 04039-002, Brazil; (C.R.B.); (M.L.V.O.)
| | - Edna Aparecida Leick
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil; (J.A.S.B.); (L.L.S.d.S.); (J.M.L.G.J.); (E.C.d.C.); (S.F.); (L.d.N.C.); (T.M.d.S.); (H.T.d.S.); (S.K.M.B.); (B.M.S.-R.); (F.D.T.Q.d.S.L.); (E.A.L.); (R.F.R.)
| | - Renato Fraga Righetti
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil; (J.A.S.B.); (L.L.S.d.S.); (J.M.L.G.J.); (E.C.d.C.); (S.F.); (L.d.N.C.); (T.M.d.S.); (H.T.d.S.); (S.K.M.B.); (B.M.S.-R.); (F.D.T.Q.d.S.L.); (E.A.L.); (R.F.R.)
- Hospital Sírio Libanês, São Paulo 01308-050, Brazil
| | - Iolanda de Fátima Lopes Calvo Tibério
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil; (J.A.S.B.); (L.L.S.d.S.); (J.M.L.G.J.); (E.C.d.C.); (S.F.); (L.d.N.C.); (T.M.d.S.); (H.T.d.S.); (S.K.M.B.); (B.M.S.-R.); (F.D.T.Q.d.S.L.); (E.A.L.); (R.F.R.)
| |
Collapse
|
10
|
Ma R, Su H, Jiao K, Liu J. Association Between IL-17 and Chronic Obstructive Pulmonary Disease: A Systematic Review and Meta-Analysis. Int J Chron Obstruct Pulmon Dis 2023; 18:1681-1690. [PMID: 37551391 PMCID: PMC10404405 DOI: 10.2147/copd.s412626] [Citation(s) in RCA: 3] [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: 03/20/2023] [Accepted: 07/24/2023] [Indexed: 08/09/2023] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease characterized by neutrophils airway infiltration. It is currently known that Interleukin-17 (IL-17) is an important pro-inflammatory factor. It can promote the accumulation of neutrophils and participate in the chronic inflammatory process of COPD. However, the value of IL-17 levels in the diagnosis and assessment of COPD remains controversial. In view of this, we conducted a systematic review and meta-analysis to assess its relevance. Methods We searched databases such as PubMed, Web of Science, Cochrane Library and Embase to extract original research. Results A total of 10 studies with 2268 participants were included in this meta-analysis. The results showed that the level of serum IL-17 in patients with stable COPD was significantly higher than that in healthy controls (standard mean difference SMD, 1.59, 95% CI 0.84-2.34; p<0.001). Compared with the stable COPD group, the serum IL-17 level in acute exacerbation (AECOPD) was significantly higher (SMD, 1.78, 95% CI 1.22-2.33; p<0.001). The level of IL-17 in sputum of COPD patients was also higher than that of healthy controls (SMD, 2.03, 95% CI 0.74-3.31; p<0.001). Conclusion Our results showed that IL-17 levels were elevated in serum and sputum in COPD patients compared with healthy controls, and IL-17 levels increased with disease progression. IL-17 serves as a potential biomarker to indicate the persistence of neutrophilic inflammation and exacerbation of COPD.
Collapse
Affiliation(s)
- Ru Ma
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Gansu Provincial People’s Hospital, Lanzhou, Gansu Province, People’s Republic of China
| | - Hongling Su
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Gansu Provincial People’s Hospital, Lanzhou, Gansu Province, People’s Republic of China
| | - Keping Jiao
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Gansu Provincial People’s Hospital, Lanzhou, Gansu Province, People’s Republic of China
| | - Jian Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| |
Collapse
|
11
|
Arora A, Singh A. Exploring the role of neutrophils in infectious and noninfectious pulmonary disorders. Int Rev Immunol 2023; 43:41-61. [PMID: 37353973 DOI: 10.1080/08830185.2023.2222769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/31/2023] [Indexed: 06/25/2023]
Abstract
With the change in global environment, respiratory disorders are becoming more threatening to the health of people all over the world. These diseases are closely linked to performance of immune system. Within the innate arm of immune system, Neutrophils are an important moiety to serve as an immune defense barrier. They are one of the first cells recruited to the site of infection and plays a critical role in pathogenesis of various pulmonary diseases. It is established that the migration and activation of neutrophils can lead to inflammation either directly or indirectly and this inflammation caused is very crucial for the clearance of pathogens and resolution of infection. However, the immunopathological mechanisms involved to carry out the same is very complex and not well understood. Despite there being studies concentrating on the role of neutrophils in multiple respiratory diseases, there is still a long way to go in order to completely understand the complexity of the participation of neutrophils and mechanisms involved in the development of these respiratory diseases. In the present article, we have reviewed the literature to comprehensively provide an insight in the current development and advancements about the role of neutrophils in infectious respiratory disorders including viral respiratory disorders such as Coronavirus disease (COVID-19) and bacterial pulmonary disorders with a focused review on pulmonary tuberculosis as well as in noninfectious disorders like Chronic obstructive pulmonary disease (COPD) and asthma. Also, future directions into research and therapeutic targets have been discussed for further exploration.
Collapse
Affiliation(s)
- Alisha Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Archana Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
12
|
Goulart A, Boko MMM, Martins NS, Gembre AF, de Oliveira RS, Palma-Albornoz SP, Bertolini T, Ribolla PEM, Ramalho LNZ, Fraga-Silva TFDC, Bonato VLD. IL-22 Is Deleterious along with IL-17 in Allergic Asthma but Is Not Detrimental in the Comorbidity Asthma and Acute Pneumonia. Int J Mol Sci 2023; 24:10418. [PMID: 37445595 PMCID: PMC10341917 DOI: 10.3390/ijms241310418] [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: 05/10/2023] [Revised: 06/06/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023] Open
Abstract
There is evidence that IL-22 and IL-17 participate in the pathogenesis of allergic asthma. To investigate the role of IL-22, we used IL-22 deficient mice (IL-22 KO) sensitized and challenged with ovalbumin (OVA) and compared with wild type (WT) animals exposed to OVA. IL-22 KO animals exposed to OVA showed a decreased number and frequency of eosinophils, IL-5 and IL-13 in the airways, reduced mucus production and pulmonary inflammation. In addition, IL-22 KO animals exhibited a decreased percentage and number of lung CD11c+CD11b+ cells and increased apoptosis of eosinophils. Th17 cell transfer generated from IL-22 KO to animals previously sensitized and challenged with OVA caused a reduction in eosinophil frequency and number in the airways compared to animals transferred with Th17 cells generated from WT mice. Therefore, IL-22 is deleterious with concomitant secretion of IL-17. Our findings show a pro-inflammatory role for IL-22, confirmed in a model of allergen-free and allergen-specific immunotherapy. Moreover, during the comorbidity asthma and pneumonia that induces neutrophil inflammation, IL-22 was not detrimental. Our results show that targeting IL-22 would negatively affect the survival of eosinophils, reduce the expansion or migration of CD11c+CD11b+ cells, and negatively regulate allergic asthma.
Collapse
Affiliation(s)
- Amanda Goulart
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Sao Paulo, Brazil; (A.G.); (M.M.M.B.); (N.S.M.); (R.S.d.O.); (S.P.P.-A.); (T.B.)
| | - Mèdéton Mahoussi Michaël Boko
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Sao Paulo, Brazil; (A.G.); (M.M.M.B.); (N.S.M.); (R.S.d.O.); (S.P.P.-A.); (T.B.)
| | - Nubia Sabrina Martins
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Sao Paulo, Brazil; (A.G.); (M.M.M.B.); (N.S.M.); (R.S.d.O.); (S.P.P.-A.); (T.B.)
| | - Ana Flávia Gembre
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Sao Paulo, Brazil; (A.F.G.); (T.F.d.C.F.-S.)
| | - Rômulo Silva de Oliveira
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Sao Paulo, Brazil; (A.G.); (M.M.M.B.); (N.S.M.); (R.S.d.O.); (S.P.P.-A.); (T.B.)
| | - Sandra Patrícia Palma-Albornoz
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Sao Paulo, Brazil; (A.G.); (M.M.M.B.); (N.S.M.); (R.S.d.O.); (S.P.P.-A.); (T.B.)
| | - Thais Bertolini
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Sao Paulo, Brazil; (A.G.); (M.M.M.B.); (N.S.M.); (R.S.d.O.); (S.P.P.-A.); (T.B.)
| | | | - Leandra Naira Zambelli Ramalho
- Department of Pathology and Legal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Sao Paulo, Brazil;
| | - Thais Fernanda de Campos Fraga-Silva
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Sao Paulo, Brazil; (A.F.G.); (T.F.d.C.F.-S.)
| | - Vânia Luiza Deperon Bonato
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Sao Paulo, Brazil; (A.G.); (M.M.M.B.); (N.S.M.); (R.S.d.O.); (S.P.P.-A.); (T.B.)
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Sao Paulo, Brazil; (A.F.G.); (T.F.d.C.F.-S.)
| |
Collapse
|
13
|
da Silva CO, de Souza Nogueira J, do Nascimento AP, Victoni T, Bártholo TP, da Costa CH, Costa AMA, Valença SDS, Schmidt M, Porto LC. COPD Patients Exhibit Distinct Gene Expression, Accelerated Cellular Aging, and Bias to M2 Macrophages. Int J Mol Sci 2023; 24:9913. [PMID: 37373058 DOI: 10.3390/ijms24129913] [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: 04/26/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
COPD, one of world's leading contributors to morbidity and mortality, is characterized by airflow limitation and heterogeneous clinical features. Three main phenotypes are proposed: overlapping asthma/COPD (ACO), exacerbator, and emphysema. Disease severity can be classified as mild, moderate, severe, and very severe. The molecular basis of inflammatory amplification, cellular aging, and immune response are critical to COPD pathogenesis. Our aim was to investigate EP300 (histone acetylase, HAT), HDAC 2 (histone deacetylase), HDAC3, and HDAC4 gene expression, telomere length, and differentiation ability to M1/M2 macrophages. For this investigation, 105 COPD patients, 42 smokers, and 73 non-smoker controls were evaluated. We identified a reduced HDAC2 expression in patients with mild, moderate, and severe severity; a reduced HDAC3 expression in patients with moderate and severe severity; an increased HDAC4 expression in patients with mild severity; and a reduced EP300 expression in patients with severe severity. Additionally, HDAC2 expression was reduced in patients with emphysema and exacerbator, along with a reduced HDAC3 expression in patients with emphysema. Surprisingly, smokers and all COPD patients showed telomere shortening. COPD patients showed a higher tendency toward M2 markers. Our data implicate genetic changes in COPD phenotypes and severity, in addition to M2 prevalence, that might influence future treatments and personalized therapies.
Collapse
Affiliation(s)
- Camila Oliveira da Silva
- Laboratory of Histocompatibility and Cryopreservation, University of the State of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil
| | - Jeane de Souza Nogueira
- Laboratory of Histocompatibility and Cryopreservation, University of the State of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil
| | | | - Tatiana Victoni
- VetAgro Sup, University of Lyon, APCSe, 69280 Marcy l'Étoile, France
| | - Thiago Prudente Bártholo
- Department of Thorax, University of the State of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil
| | | | - Andrea Monte Alto Costa
- Tissue Repair Laboratory, University of the State of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil
| | - Samuel Dos Santos Valença
- Laboratory of Redox Biology, ICB, Federal University of Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Luís Cristóvão Porto
- Laboratory of Histocompatibility and Cryopreservation, University of the State of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil
| |
Collapse
|
14
|
Tan DJ, Lodge CJ, Walters EH, Lowe AJ, Bui DS, Bowatte G, Kandane‐Rathnayake R, Aldakheel FM, Erbas B, Hamilton GS, Thomas PS, Hew M, Tang MLK, Abramson MJ, Perret JL, Dharmage SC. Biomarkers of asthma relapse and lung function decline in adults with spontaneous asthma remission: A population-based cohort study. Allergy 2023; 78:957-967. [PMID: 36301194 PMCID: PMC10953440 DOI: 10.1111/all.15566] [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: 03/21/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND The extent to which biomarkers of asthma activity persist in spontaneous asthma remission and whether such markers are associated with future respiratory outcomes remained unclear. We investigated the association between sub-clinical inflammation in adults with spontaneous asthma remission and future asthma relapse and lung function decline. METHODS The Tasmanian Longitudinal Health Study is a population-based cohort (n = 8583). Biomarkers of systemic inflammation were measured on participants at age 45, and latent profile analysis was used to identify cytokine profiles. Bronchial hyperresponsiveness (BHR) and nitric oxide products in exhaled breath condensate (EBC NOx) were measured at age 50. Participants with spontaneous asthma remission at ages 45 (n = 466) and 50 (n = 318) were re-evaluated at age 53, and associations between baseline inflammatory biomarkers and subsequent asthma relapse and lung function decline were assessed. RESULTS We identified three cytokine profiles in adults with spontaneous asthma remission: average (34%), Th2-high (42%) and Th2-low (24%). Compared to the average profile, a Th2-high profile was associated with accelerated decline in post-BD FEV1 /FVC (MD -0.18% predicted per-year; 95% CI -0.33, -0.02), while a Th2-low profile was associated with accelerated decline in both post-BD FEV1 (-0.41%; -0.75, -0.06) and post-BD FVC (-0.31%; -0.62, 0.01). BHR and high TNF-α during spontaneous remission were associated with an increased risk of asthma relapse. In contrast, we found no evidence of association between EBC NOx and either asthma relapse or lung function decline. CONCLUSION BHR and serum inflammatory cytokines have prognostic value in adults with spontaneous asthma remission. At-risk individuals with BHR, Th2-high or Th2-low cytokine profiles may benefit from closer monitoring and on-going follow-up.
Collapse
Affiliation(s)
- Daniel J. Tan
- Allergy and Lung Health Unit, Centre for Epidemiology and BiostatisticsSchool of Population and Global Health, University of MelbourneMelbourneVictoriaAustralia
| | - Caroline J. Lodge
- Allergy and Lung Health Unit, Centre for Epidemiology and BiostatisticsSchool of Population and Global Health, University of MelbourneMelbourneVictoriaAustralia
| | - Eugene Haydn Walters
- Allergy and Lung Health Unit, Centre for Epidemiology and BiostatisticsSchool of Population and Global Health, University of MelbourneMelbourneVictoriaAustralia
- School of Medicine, University of TasmaniaHobartTasmaniaAustralia
| | - Adrian J. Lowe
- Allergy and Lung Health Unit, Centre for Epidemiology and BiostatisticsSchool of Population and Global Health, University of MelbourneMelbourneVictoriaAustralia
| | - Dinh S. Bui
- Allergy and Lung Health Unit, Centre for Epidemiology and BiostatisticsSchool of Population and Global Health, University of MelbourneMelbourneVictoriaAustralia
| | - Gayan Bowatte
- Allergy and Lung Health Unit, Centre for Epidemiology and BiostatisticsSchool of Population and Global Health, University of MelbourneMelbourneVictoriaAustralia
- Department of Basic Sciences, Faculty of Allied Health SciencesUniversity of PeradeniyaPeradeniyaSri Lanka
| | | | - Fahad M. Aldakheel
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesKing Saud UniversityRiyadhSaudi Arabia
| | - Bircan Erbas
- School of Psychology and Public HealthLa Trobe UniversityMelbourneVictoriaAustralia
- Violet Vines Marshman Centre for Rural Health ResearchLa Trobe UniversityBendigoVictoriaAustralia
| | - Garun S. Hamilton
- School of Clinical SciencesMonash UniversityMelbourneVictoriaAustralia
- Monash Lung, Sleep, Allergy and ImmunologyMonash HealthMelbourneVictoriaAustralia
| | - Paul S. Thomas
- Prince of Wales' Clinical School, Faculty of MedicineUniversity of New South WalesSydneyNew South WalesAustralia
| | - Mark Hew
- School of Public Health & Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
- The Alfred HospitalMelbourneVictoriaAustralia
| | - Mimi L. K. Tang
- Murdoch Children's Research InstituteMelbourneVictoriaAustralia
- Department of PaediatricsUniversity of MelbourneMelbourneVictoriaAustralia
| | - Michael J. Abramson
- School of Public Health & Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Jennifer L. Perret
- Allergy and Lung Health Unit, Centre for Epidemiology and BiostatisticsSchool of Population and Global Health, University of MelbourneMelbourneVictoriaAustralia
- Institute for Breathing and SleepMelbourneVictoriaAustralia
| | - Shyamali C. Dharmage
- Allergy and Lung Health Unit, Centre for Epidemiology and BiostatisticsSchool of Population and Global Health, University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
15
|
Lambré C, Barat Baviera JM, Bolognesi C, Chesson A, Cocconcelli PS, Crebelli R, Gott DM, Grob K, Lampi E, Mengelers M, Mortensen A, Rivière G, Silano (until 21 December 2020†) V, Steffensen I, Tlustos C, Vernis L, Zorn H, Batke M, Bignami M, Corsini E, FitzGerald R, Gundert‐Remy U, Halldorsson T, Hart A, Ntzani E, Scanziani E, Schroeder H, Ulbrich B, Waalkens‐Berendsen D, Woelfle D, Al Harraq Z, Baert K, Carfì M, Castoldi AF, Croera C, Van Loveren H. Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs. EFSA J 2023; 21:e06857. [PMID: 37089179 PMCID: PMC10113887 DOI: 10.2903/j.efsa.2023.6857] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023] Open
Abstract
In 2015, EFSA established a temporary tolerable daily intake (t-TDI) for BPA of 4 μg/kg body weight (bw) per day. In 2016, the European Commission mandated EFSA to re-evaluate the risks to public health from the presence of BPA in foodstuffs and to establish a tolerable daily intake (TDI). For this re-evaluation, a pre-established protocol was used that had undergone public consultation. The CEP Panel concluded that it is Unlikely to Very Unlikely that BPA presents a genotoxic hazard through a direct mechanism. Taking into consideration the evidence from animal data and support from human observational studies, the immune system was identified as most sensitive to BPA exposure. An effect on Th17 cells in mice was identified as the critical effect; these cells are pivotal in cellular immune mechanisms and involved in the development of inflammatory conditions, including autoimmunity and lung inflammation. A reference point (RP) of 8.2 ng/kg bw per day, expressed as human equivalent dose, was identified for the critical effect. Uncertainty analysis assessed a probability of 57-73% that the lowest estimated Benchmark Dose (BMD) for other health effects was below the RP based on Th17 cells. In view of this, the CEP Panel judged that an additional uncertainty factor (UF) of 2 was needed for establishing the TDI. Applying an overall UF of 50 to the RP, a TDI of 0.2 ng BPA/kg bw per day was established. Comparison of this TDI with the dietary exposure estimates from the 2015 EFSA opinion showed that both the mean and the 95th percentile dietary exposures in all age groups exceeded the TDI by two to three orders of magnitude. Even considering the uncertainty in the exposure assessment, the exceedance being so large, the CEP Panel concluded that there is a health concern from dietary BPA exposure.
Collapse
|
16
|
Erjefält JS. Anatomical and histopathological approaches to asthma phenotyping. Respir Med 2023; 210:107168. [PMID: 36822489 DOI: 10.1016/j.rmed.2023.107168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023]
Abstract
Asthma is typically characterized by variable respiratory symptoms and airflow limitation. Along with the pathophysiology and symptoms are immunological and inflammatory processes. The last decades research has revealed that the immunology of asthma is highly heterogeneous. This has clinical consequences and identification of immunological phenotypes is currently used to guide biological treatment. The focus of this review is on another dimension of asthma diversity, namely anatomical heterogeneity. Immunopathological alterations may go beyond the central airways to also involve the distal airways, the alveolar parenchyma, and pulmonary vessels. Also, extrapulmonary tissues are affected. The anatomical distribution of inflammation in asthma has remained relatively poorly discussed despite its potential implication on both clinical presentation and response to treatment. There is today evidence that a significant proportion of the asthma patients has small airway disease with type 2 immunity, eosinophilia and smooth muscle infiltration of mast cells. The small airways in asthma are also subjected to remodelling, constriction, and luminal plugging, events that are likely to contribute to the elevated distal airway resistance seen in some patients. In cases when the inflammation extends into the alveolar parenchyma alveolar FCER1-high mast cells, eosinophilia, type 2 immunity and activated alveolar macrophages, together with modest interstitial remodelling, create a complex immunopathological picture. Importantly, the distal lung inflammation in asthma can be pharmacologically targeted by use of inhalers with more distal drug deposition. Biological treatments, which are readily distributed to the distal lung, may also be beneficial in eligible patients with more severe and anatomically widespread disease.
Collapse
Affiliation(s)
- Jonas S Erjefält
- Unit of Airway Inflammation, Department of Experimental Medical Research, Lund University, Lund, Sweden; Department of Allergology and Respiratory Medicine, Skane University Hospital, Lund, Sweden.
| |
Collapse
|
17
|
Neutrophil Extracellular Traps in Airway Diseases: Pathological Roles and Therapeutic Implications. Int J Mol Sci 2023; 24:ijms24055034. [PMID: 36902466 PMCID: PMC10003347 DOI: 10.3390/ijms24055034] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Neutrophils are important effector cells of the innate immune response that fight pathogens by phagocytosis and degranulation. Neutrophil extracellular traps (NETs) are released into the extracellular space to defend against invading pathogens. Although NETs play a defensive role against pathogens, excessive NETs can contribute to the pathogenesis of airway diseases. NETs are known to be directly cytotoxic to the lung epithelium and endothelium, highly involved in acute lung injury, and implicated in disease severity and exacerbation. This review describes the role of NET formation in airway diseases, including chronic rhinosinusitis, and suggests that targeting NETs could be a therapeutic strategy for airway diseases.
Collapse
|
18
|
Ganta S, Komaravalli PL, Ahmad S, Gaddam SL. Influence of genetic variants and mRNA expression of interleukin IL17A gene in asthma susceptibility. Gene 2023; 854:147119. [PMID: 36529350 DOI: 10.1016/j.gene.2022.147119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Asthma is a chronic respiratory disease characterized by coughing, wheezing, shortness of breath, chest tightness, overproduction of mucus, and expiratory airflow limitation, which affects >300 million people worldwide. It is triggered by the dynamic interplay of genetic factors and environmental exposure. Th17 cells are an emerging subset of CD4+ T cells, which secrete IL-17A. This proinflammatory cytokine has recently been associated with asthma, autoimmune diseases, and inflammatory disorders. The present case-control study was focused on identifying the involvement of the IL-17A gene in asthma pathogenesis among 150 clinically diagnosed asthma patients and 150 healthy controls (HCs) of South Indian origin. To carry out the study, we aimed to screen the genetic variants of rs2275913G/A and rs8193036C/T and also estimated the serum cytokine levels of the IL-17A cytokine of recruited subjects. Further, we evaluated mRNA expression in selected subjects to correlate with the genetic variants. The results revealed that the mean IL-17A serum levels (161.6 ± 380.1 pg/ml vs. 86.75 ± 90.01 pg/ml) and IgE levels (257.7 ± 133.3 pg/ml vs. 311.2 ± 160.5 pg/ml) in asthma patients were significantly high as compared to healthy controls (p < 0.05). The ROC curves were constructed to compare the cytokine levels of asthma patients and HC, and the area under the curve (AUC) for IL-17A cytokine was 0.64, indicating that the test was satisfactory and significant (95 % CI: 0.575-0.709; p < 0.001). Genotyping of rs2275913G/A polymorphism indicated a 1.6-fold risk (95 % CI-1.02-2.56; p = 0.04) for asthma patients compared to healthy controls, whereas no significant association was observed for rs8193036C/T polymorphism with asthma susceptibility. Under genetic models, GA and AA models showed a protective effect against the disease for rs2275913G/A. In contrast, no statistically significant result was observed among the models of rs8193036C/T when adjusted with age and sex. The mRNA expression levels of the gene were statistically high in patients compared to the HCs, with a 1.8-fold change (p < 0.0001). We conclude that the results indicate IL-17A rs2275913G/A is likely to contribute to protection against the disease, while IL-17A rs8193036C/T shows no association with the disease. However, no correlation was identified in serum cytokine levels concerning genotypes. This comprehensive information in the present study might contribute to developing novel therapeutic strategies for treating inflammatory diseases like asthma. Further studies are warranted to understand the diverse functions of IL-17A concerning its longitudinal stability and its response to clinical interventions with large sample sizes in various ethnicities.
Collapse
Affiliation(s)
- Soujanya Ganta
- Department of Genetics, Osmania University, Hyderabad, India
| | - Prasanna Latha Komaravalli
- Department of Genetics, Osmania University, Hyderabad, India; Global Medical Education and Research Foundation, Lakdi-ka-pool, Hyderabad, India
| | - Shazia Ahmad
- Bhagwan Mahavir Medical Research Centre, Hyderabad, India
| | - Suman Latha Gaddam
- Department of Genetics, Osmania University, Hyderabad, India; Bhagwan Mahavir Medical Research Centre, Hyderabad, India.
| |
Collapse
|
19
|
Zhang T, Shang F, Ma Y, Xu Y, Sun W, Song H. Caveolin-1 Promotes the Imbalance of Th17/Treg in Chronic Obstructive Pulmonary Disease by Regulating Hsp70 Expression. Int J Chron Obstruct Pulmon Dis 2023; 18:565-574. [PMID: 37077366 PMCID: PMC10106795 DOI: 10.2147/copd.s398780] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/27/2023] [Indexed: 04/21/2023] Open
Abstract
Objective To investigate whether the expression of Hsp70 is associated with Cav-1 in promoting the imbalance of Th17/Treg cells in COPD. Methods The plasma Cav-1, Hsp70 expression were quantified by enzyme-linked immunosorbent assay (ELISA). The frequencies of circulating Th17, Treg cells and Th17/Treg ratio were determined by flow cytometry. Peripheral blood mononuclear cells (PBMCs) from subjects were transfected with Cav-1 or control plasmids and Hsp70 plasmid. Results We found that Cav-1 expression was lower but the levels of Hsp70 and Th17 cells were higher in COPD than in healthy control (HC). Hsp70 expressions were positively correlated with Cav-1 levels, Th17 cells, and Th17/Treg ratio in COPD but not in HC. Cav-1 over-expression resulted in an increase in Hsp70 and Th17 levels. Suppressing Hsp70 expressing by small interfering RNA (siRNA), the decline of Th17 frequency was observed in Cav-1-overexpressed PBMCs. Conclusion Collectively, our results illuminate that Cav-1 contributes to the imbalance of Th17/Treg through potentially regulating Hsp70 expression.
Collapse
Affiliation(s)
- Tongsong Zhang
- Department of Oncology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong, 266042, People’s Republic of China
| | - Fangfang Shang
- Department of Pathology, No. 971 Hospital of People’s Liberation Army Navy, Qingdao, 266071, People’s Republic of China
| | - Yanhui Ma
- Department of Clinical Laboratory, Biotherapy Center, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, 266042, People’s Republic of China
| | - Yanxia Xu
- Department of Oncology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong, 266042, People’s Republic of China
| | - Weihong Sun
- Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong, 266042, People’s Republic of China
| | - Haiping Song
- Department of Oncology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong, 266042, People’s Republic of China
- Correspondence: Haiping Song; Yanxia Xu, Department of Oncology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, 127 Siliu South Road, Qingdao, 266042, People’s Republic of China, Tel +86 532 8496 2202; +86 532 84962203, Fax +86 532-84963506, Email ;
| |
Collapse
|
20
|
Iraji D, Oftedal BE, Wolff ASB. Th17 Cells: Orchestrators of Mucosal Inflammation and Potential Therapeutic Targets. Crit Rev Immunol 2023; 43:25-52. [PMID: 37831521 DOI: 10.1615/critrevimmunol.2023050360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
T helper 17 (Th17) cells represent a specialized subgroup of effector CD4+ T cells known for their role in provoking neutrophil-driven tissue inflammation, particularly within mucosal tissues. Although they are pivotal for defending the host against extracellular bacteria and fungi, they have also been associated with development of various T cell-mediated inflammatory conditions, autoimmune diseases, and even cancer. Notably, Th17 cells exhibit a dual nature, with different Th17 cell subtypes showcasing distinct effector functions and varying capacities to incite autoimmune tissue inflammation. Furthermore, Th17 cells exhibit significant plasticity, which carries important functional implications, both in terms of their expression of cytokines typically associated with other effector T cell subsets and in their interactions with regulatory CD4+ T cells. The intricate balance of Th17 cytokines can also be a double-edged sword in inflammation, autoimmunity, and cancer. Within this article, we delve into the mechanisms that govern the differentiation, function, and adaptability of Th17 cells. We culminate with an exploration of therapeutic potentials in harnessing the power of Th17 cells and their cytokines. Targeted interventions to modulate Th17 responses are emerging as promising strategies for autoimmunity, inflammation, and cancer treatment. By precisely fine-tuning Th17-related pathways, we may unlock new avenues for personalized therapeutic approaches, aiming to restore immune balance, alleviate the challenges of these disorders, and ultimately enhance the quality of life for individuals affected by them.
Collapse
Affiliation(s)
- Dorsa Iraji
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bergithe E Oftedal
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S B Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
21
|
Khalfaoui L, Pabelick CM. Airway smooth muscle in contractility and remodeling of asthma: potential drug target mechanisms. Expert Opin Ther Targets 2023; 27:19-29. [PMID: 36744401 DOI: 10.1080/14728222.2023.2177533] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Asthma is characterized by enhanced airway contractility and remodeling where airway smooth muscle (ASM) plays a key role, modulated by inflammation. Understanding the mechanisms by which ASM contributes to these features of asthma is essential for the development of novel asthma therapies. AREAS COVERED Inflammation in asthma contributes to a multitude of changes within ASM including enhanced airway contractility, proliferation, and fibrosis. Altered intracellular calcium ([Ca2+]i) regulation or Ca2+ sensitization contributes to airway hyperreactivity. Increased airway wall thickness from ASM proliferation and fibrosis contributes to structural changes seen with asthma. EXPERT OPINION ASM plays a significant role in multiple features of asthma. Increased ASM contractility contributes to hyperresponsiveness, while altered ASM proliferation and extracellular matrix production promote airway remodeling both influenced by inflammation of asthma and conversely even influencing the local inflammatory milieu. While standard therapies such as corticosteroids or biologics target inflammation, cytokines, or their receptors to alleviate asthma symptoms, these approaches do not address the underlying contribution of ASM to hyperresponsiveness and particularly remodeling. Therefore, novel therapies for asthma need to target abnormal contractility mechanisms in ASM and/or the contribution of ASM to remodeling, particularly in asthmatics resistant to current therapies.
Collapse
Affiliation(s)
- Latifa Khalfaoui
- Departments of Anesthesiology & Perioperative Medicine Mayo Clinic, Rochester, MN, USA
| | - Christina M Pabelick
- Departments of Anesthesiology & Perioperative Medicine Mayo Clinic, Rochester, MN, USA.,Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
22
|
Altieri A, Piyadasa H, Hemshekhar M, Osawa N, Recksiedler B, Spicer V, Hiemstra PS, Halayko AJ, Mookherjee N. Combination of IL-17A/F and TNF-α uniquely alters the bronchial epithelial cell proteome to enhance proteins that augment neutrophil migration. J Inflamm (Lond) 2022; 19:26. [PMCID: PMC9749191 DOI: 10.1186/s12950-022-00323-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 12/05/2022] [Indexed: 12/16/2022] Open
Abstract
Background The heterodimer interleukin (IL)-17A/F is elevated in the lungs in chronic respiratory disease such as severe asthma, along with the pro-inflammatory cytokine tumor necrosis factor-α (TNF-α). Although IL-17A/F and TNF-α are known to functionally cooperate to exacerbate airway inflammation, proteins altered by their interaction in the lungs are not fully elucidated. Results We used Slow Off-rate Modified Aptamer-based proteomic array to identify proteins that are uniquely and/or synergistically enhanced by concurrent stimulation with IL-17A/F and TNF-α in human bronchial epithelial cells (HBEC). The abundance of 38 proteins was significantly enhanced by the combination of IL-17A/F and TNF-α, compared to either cytokine alone. Four out of seven proteins that were increased > 2-fold were those that promote neutrophil migration; host defence peptides (HDP; Lipocalin-2 (LCN-2) and Elafin) and chemokines (IL-8, GROα). We independently confirmed the synergistic increase of these four proteins by western blots and ELISA. We also functionally confirmed that factors secreted by HBEC stimulated with the combination of IL-17A/F and TNF-α uniquely enhances neutrophil migration. We further showed that PI3K and PKC pathways selectively control IL-17A/F + TNF-α-mediated synergistic production of HDPs LCN-2 and Elafin, but not chemokines IL-8 and GROα. Using a murine model of airway inflammation, we demonstrated enhancement of IL-17A/F, TNF-α, LCN-2 and neutrophil chemokine KC in the lungs, thus corroborating our findings in-vivo. Conclusion This study identifies proteins and signaling mediated by concurrent IL-17A/F and TNF-α exposure in the lungs, relevant to respiratory diseases characterized by chronic inflammation, especially neutrophilic airway inflammation such as severe asthma. Supplementary Information The online version contains supplementary material available at 10.1186/s12950-022-00323-w.
Collapse
Affiliation(s)
- Anthony Altieri
- grid.21613.370000 0004 1936 9609Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB Canada ,grid.21613.370000 0004 1936 9609Department of Immunology, University of Manitoba, Winnipeg, MB Canada
| | - Hadeesha Piyadasa
- grid.21613.370000 0004 1936 9609Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB Canada ,grid.21613.370000 0004 1936 9609Department of Immunology, University of Manitoba, Winnipeg, MB Canada ,grid.168010.e0000000419368956Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA USA
| | - Mahadevappa Hemshekhar
- grid.21613.370000 0004 1936 9609Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB Canada
| | - Natasha Osawa
- grid.21613.370000 0004 1936 9609Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB Canada
| | - Breann Recksiedler
- grid.21613.370000 0004 1936 9609Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB Canada
| | - Victor Spicer
- grid.21613.370000 0004 1936 9609Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB Canada
| | - Pieter S Hiemstra
- grid.10419.3d0000000089452978Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrew J Halayko
- grid.21613.370000 0004 1936 9609Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB Canada ,grid.460198.20000 0004 4685 0561Biology of Breathing Group, The Children’s Hospital Research Institute of Manitoba, Winnipeg, MB Canada
| | - Neeloffer Mookherjee
- grid.21613.370000 0004 1936 9609Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB Canada ,grid.21613.370000 0004 1936 9609Department of Immunology, University of Manitoba, Winnipeg, MB Canada ,grid.460198.20000 0004 4685 0561Biology of Breathing Group, The Children’s Hospital Research Institute of Manitoba, Winnipeg, MB Canada
| |
Collapse
|
23
|
Current Limitations and Recent Advances in the Management of Asthma. Dis Mon 2022:101483. [DOI: 10.1016/j.disamonth.2022.101483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
24
|
Investigational Treatments in Phase I and II Clinical Trials: A Systematic Review in Asthma. Biomedicines 2022; 10:biomedicines10092330. [PMID: 36140430 PMCID: PMC9496184 DOI: 10.3390/biomedicines10092330] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Inhaled corticosteroids (ICS) remain the mainstay of asthma treatment, along with bronchodilators serving as control agents in combination with ICS or reliever therapy. Although current pharmacological treatments improve symptom control, health status, and the frequency and severity of exacerbations, they do not really change the natural course of asthma, including disease remission. Considering the highly heterogeneous nature of asthma, there is a strong need for innovative medications that selectively target components of the inflammatory cascade. The aim of this review was to systematically assess current investigational agents in Phase I and II randomised controlled trials (RCTs) over the last five years. Sixteen classes of novel therapeutic options were identified from 19 RCTs. Drugs belonging to different classes, such as the anti-interleukin (IL)-4Rα inhibitors, anti-IL-5 monoclonal antibodies (mAbs), anti-IL-17A mAbs, anti-thymic stromal lymphopoietin (TSLP) mAbs, epithelial sodium channel (ENaC) inhibitors, bifunctional M3 receptor muscarinic antagonists/β2-adrenoceptor agonists (MABAs), and anti-Fel d 1 mAbs, were found to be effective in the treatment of asthma, with lung function being the main assessed outcome across the RCTs. Several novel investigational molecules, particularly biologics, seem promising as future disease-modifying agents; nevertheless, further larger studies are required to confirm positive results from Phase I and II RCTs.
Collapse
|
25
|
Androgen Plays a Potential Novel Hormonal Therapeutic Role in Th17 Cells Predominant Neutrophilic Severe Asthma by Attenuating BECs Regulated Th17 Cells Differentiation via MBD2 Expression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3096528. [PMID: 36062195 PMCID: PMC9436621 DOI: 10.1155/2022/3096528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/02/2022] [Indexed: 12/05/2022]
Abstract
T helper 17 (Th17) cells subtype of non-T2 asthma is less responsive (resistant) to inhaled corticosteroids (ICS), so also called severe asthma. Methyl-CpG-binding domain protein 2 (MBD2) regulates the differentiation of the Th17 cells, showing the possibility of a therapeutic target in severe asthma. Androgen tends to show beneficial therapeutic effects and is a “hot research topic,” but its role in the differentiation and expression of Th17 cells via MBD2 is still unknown. The aim of this study was to evaluate how sex hormone interacts with MBD2 and affects the differentiation and expression of Th17 cells in severe asthma. Here, first, we measured the concentration of androgen, estrogen, and androgen estrogen ratio from subjects and correlated it with severe asthma status. Then, we established an animal model and bronchial epithelial cells (BECs) model of severe asthma to evaluate the role of MBD2 in the differentiation and expression of Th17 cells (IL-17), the therapeutic potential of sex hormones in severe asthma, and the effect of sex hormones in BECs regulated Th17 cells differentiation via MBD2 at the cellular level. Increased MBD2 expression and Th17 cells differentiation were noted in the animal and the BECs severe asthma models. Th17 cell differentiation and expression were MBD2 dependent. Androgen attenuated the differentiation of BECs regulated Th17 cells via MBD2 showing BECs as a therapeutic target of androgen, and these findings postulate the novel role of androgen in Th17 cells predominant neutrophilic severe asthma therapy through targeting MBD2.
Collapse
|
26
|
Ritzmann F, Lunding LP, Bals R, Wegmann M, Beisswenger C. IL-17 Cytokines and Chronic Lung Diseases. Cells 2022; 11:2132. [PMID: 35883573 PMCID: PMC9318387 DOI: 10.3390/cells11142132] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/27/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
IL-17 cytokines are expressed by numerous cells (e.g., gamma delta (γδ) T, innate lymphoid (ILC), Th17, epithelial cells). They contribute to the elimination of bacteria through the induction of cytokines and chemokines which mediate the recruitment of inflammatory cells to the site of infection. However, IL-17-driven inflammation also likely promotes the progression of chronic lung diseases, such as chronic obstructive pulmonary disease (COPD), lung cancer, cystic fibrosis, and asthma. In this review, we highlight the role of IL-17 cytokines in chronic lung diseases.
Collapse
Affiliation(s)
- Felix Ritzmann
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
- Helmholtz Institute for Pharmaceutical Research, 66123 Saarbrücken, Germany
| | - Lars Peter Lunding
- Division of Lung Immunology, Priority Area Asthma and Allergy, Research Center Borstel—Leibniz Lung Center, 23845 Borstel, Germany; (L.P.L.); (M.W.)
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Robert Bals
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
- Helmholtz Institute for Pharmaceutical Research, 66123 Saarbrücken, Germany
| | - Michael Wegmann
- Division of Lung Immunology, Priority Area Asthma and Allergy, Research Center Borstel—Leibniz Lung Center, 23845 Borstel, Germany; (L.P.L.); (M.W.)
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
| |
Collapse
|
27
|
Liu M, Wang J, Sun X. A Meta-Analysis on Vitamin D Supplementation and Asthma Treatment. Front Nutr 2022; 9:860628. [PMID: 35873428 PMCID: PMC9300755 DOI: 10.3389/fnut.2022.860628] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 05/25/2022] [Indexed: 12/23/2022] Open
Abstract
Background Vitamin D, as an immunomodulator, may be related to the therapeutic effect of asthma patients, but the research in this area is still controversial. The aim of this meta-analysis was to analyze the role of vitamin D supplementation in the treatment of asthma patients. Materials and Methods Randomized Controlled Trials (RCTs) of vitamin D supplementation in asthma were searched in PubMed, EMBASE, and the Cochrane library. Primary outcomes were forced expiratory volume in one second (FEV1), asthma exacerbations, Asthma Control Test scores (ACT scores), and fractional exhaled nitric oxide (FENO). Results A total of 10 RCTs were included, including 1,349 patients. Vitamin D supplementation didn't affect the ACT scores (SMD = 0.04, 95% CI = -0.13 to 0.21, P = 0.87), FEV1 (SMD = 0.04, 95% CI = -0.35 to 0.43, P < 0.01) and FENO (SMD = -0.01, 95% CI = -0.22 to 0.20, P = 0.27), but reduced the rate of asthma exacerbations (RR = 0.69, 95% CI = 0.41 to 0.88, P < 0.01), especially in subgroups of children (RR = 0.46, 95% CI = 0.30 to 0.70, P = 0.83) and follow up time less than 6 months (RR = 0.45, 95% CI = 0.32 to 0.63, P = 0.95). Additionally, though there was only one study included in the subgroup, it significantly enhanced FEV1 at the last visit for patients whose FEV1 baseline value was less than 70% (SMD = 0.94, 95% CI = 0.47 to 1.41). Conclusion Vitamin D supplementation can reduce asthma exacerbations, especially in children, and within 6 months of follow up time. In addition, vitamin D has a positive effect on improving FEV1 of patients whose FEV1 baseline value is less than 70%, but more RCTs are still needed to support this conclusion. Systematic Review Registration [https://inplasy.com], identifier [10.37766/inplasy20 22.6.0049].
Collapse
Affiliation(s)
- Meiqi Liu
- Department of Respiratory Medicine, Xi’an Children’s Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Jun Wang
- Department and Institute of Infectious Disease, Xi’an Children’s Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Xinrong Sun
- Department of Respiratory Medicine, Xi’an Children’s Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
28
|
The role of Th17 cells: explanation of relationship between periodontitis and COPD? Inflamm Res 2022; 71:1011-1024. [PMID: 35781342 DOI: 10.1007/s00011-022-01602-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/13/2022] [Indexed: 11/05/2022] Open
Abstract
Periodontitis and chronic obstructive pulmonary disease (COPD) are chronic inflammatory diseases with common risk factors, such as long-term smoking, age, and social deprivation. Many observational studies have shown that periodontitis and COPD are correlated. Moreover, they share a common pathophysiological process involving local accumulation of inflammatory cells and cytokines and damage of soft tissues. The T helper 17 (Th17) cells and the related cytokines, interleukin (IL)-17, IL-22, IL-1β, IL-6, IL-23, and transforming growth factor (TGF)-β, play a crucial regulatory role during the pathophysiological process. This paper reviewed the essential roles of Th17 lineage in the occurrence of periodontitis and COPD. The gaps in the study of their common pathological mechanism were also evaluated to explore future research directions. Therefore, this review can provide study direction for the association between periodontitis and COPD and new ideas for the clinical diagnosis and treatment of the two diseases.
Collapse
|
29
|
Rahmawati SF, Vos R, Bos IST, Kerstjens HAM, Kistemaker LEM, Gosens R. Function-specific IL-17A and dexamethasone interactions in primary human airway epithelial cells. Sci Rep 2022; 12:11110. [PMID: 35773318 PMCID: PMC9247091 DOI: 10.1038/s41598-022-15393-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/23/2022] [Indexed: 11/09/2022] Open
Abstract
Asthmatics have elevated levels of IL-17A compared to healthy controls. IL-17A is likely to contribute to reduced corticosteroid sensitivity of human airway epithelium. Here, we aimed to investigate the mechanistic underpinnings of this reduced sensitivity in more detail. Differentiated primary human airway epithelial cells (hAECs) were exposed to IL-17A in the absence or presence of dexamethasone. Cells were then collected for RNA sequencing analysis or used for barrier function experiments. Mucus was collected for volume measurement and basal medium for cytokine analysis. 2861 genes were differentially expressed by IL-17A (Padj < 0.05), of which the majority was not sensitive to dexamethasone (< 50% inhibition). IL-17A did inhibit canonical corticosteroid genes, such as HSD11B2 and FKBP5 (p < 0.05). Inflammatory and goblet cell metaplasia markers, cytokine secretion and mucus production were all induced by IL-17A, and these effects were not prevented by dexamethasone. Dexamethasone did reverse IL-17A-stimulated epithelial barrier disruption, and this was associated with gene expression changes related to cilia function and development. We conclude that IL-17A induces function-specific corticosteroid-insensitivity. Whereas inflammatory response genes and mucus production in primary hAECs in response to IL-17A were corticosteroid-insensitive, corticosteroids were able to reverse IL-17A-induced epithelial barrier disruption.
Collapse
Affiliation(s)
- Siti Farah Rahmawati
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
- Department of Pharmacology and Clinical Pharmacy, Institut Teknologi Bandung, Bandung, Indonesia
- University of Groningen, University Medical Center Groningen (UMCG), Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Rémon Vos
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen (UMCG), Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - I Sophie T Bos
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen (UMCG), Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Huib A M Kerstjens
- Department of Pulmonary Medicine, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen (UMCG), Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Loes E M Kistemaker
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
- Aquilo Contract Research, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen (UMCG), Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
- University of Groningen, University Medical Center Groningen (UMCG), Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.
| |
Collapse
|
30
|
An inhibitor of RORγ for chronic pulmonary obstructive disease treatment. Sci Rep 2022; 12:8744. [PMID: 35610240 PMCID: PMC9130233 DOI: 10.1038/s41598-022-12251-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 05/03/2022] [Indexed: 11/23/2022] Open
Abstract
The role of RORγ as a transcription factor for Th17 cell differentiation and thereby regulation of IL-17 levels is well known. Increased RORγ expression along with IL-17A levels was observed in animal models, immune cells and BAL fluid of COPD patients. Increased IL-17A levels in severe COPD patients are positively correlated with decreased lung functions and increased severity symptoms and emphysema, supporting an urgency to develop novel therapies modulating IL-17 or RORγ for COPD treatment. We identified a potent RORγ inhibitor, PCCR-1 using hit to lead identification followed by extensive lead optimization by structure–activity relationship. PCCR-1 resulted in RORγ inhibition with a high degree of specificity in a biochemical assay, with > 300-fold selectivity over other isoforms of ROR. Our data suggest promising potency for IL-17A inhibition in human and canine PBMCs and mouse splenocytes with no significant impact on Th1 and Th2 cytokines. In vivo, PCCR-1 exhibited significant efficacy in the acute CS model with dose-dependent inhibition of the PD biomarkers that correlated well with the drug concentration in lung and BAL fluid, demonstrating an acceptable safety profile. This inhibitor effectively inhibited IL-17A release in whole blood and BALf samples from COPD patients. Overall, we identified a selective inhibitor of RORγ to pursue further development of novel scaffolds for COPD treatment.
Collapse
|
31
|
Abstract
ABSTRACT Severe asthma is "asthma which requires treatment with high dose inhaled corticosteroids (ICS) plus a second controller (and/or systemic corticosteroids) to prevent it from becoming 'uncontrolled' or which remains 'uncontrolled' despite this therapy." The state of control was defined by symptoms, exacerbations and the degree of airflow obstruction. Therefore, for the diagnosis of severe asthma, it is important to have evidence for a diagnosis of asthma with an assessment of its severity, followed by a review of comorbidities, risk factors, triggers and an assessment of whether treatment is commensurate with severity, whether the prescribed treatments have been adhered to and whether inhaled therapy has been properly administered. Phenotyping of severe asthma has been introduced with the definition of a severe eosinophilic asthma phenotype characterized by recurrent exacerbations despite being on high dose ICS and sometimes oral corticosteroids, with a high blood eosinophil count and a raised level of nitric oxide in exhaled breath. This phenotype has been associated with a Type-2 (T2) inflammatory profile with expression of interleukin (IL)-4, IL-5, and IL-13. Molecular phenotyping has also revealed non-T2 inflammatory phenotypes such as Type-1 or Type-17 driven phenotypes. Antibody treatments targeted at the T2 targets such as anti-IL5, anti-IL5Rα, and anti-IL4Rα antibodies are now available for treating severe eosinophilic asthma, in addition to anti-immunoglobulin E antibody for severe allergic asthma. No targeted treatments are currently available for non-T2 inflammatory phenotypes. Long-term azithromycin and bronchial thermoplasty may be considered. The future lies with molecular phenotyping of the airway inflammatory process to refine asthma endotypes for precision medicine.
Collapse
|
32
|
Aslani MR, Sharghi A, Boskabady MH, Ghobadi H, Keyhanmanesh R, Alipour MR, Ahmadi M, Saadat S, Naghizadeh P. Altered gene expression levels of IL-17/TRAF6/MAPK/USP25 axis and pro-inflammatory cytokine levels in lung tissue of obese ovalbumin-sensitized rats. Life Sci 2022; 296:120425. [PMID: 35202642 DOI: 10.1016/j.lfs.2022.120425] [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: 11/04/2021] [Revised: 01/31/2022] [Accepted: 02/16/2022] [Indexed: 11/28/2022]
Abstract
AIMS The association between asthma and obesity has been shown but its accurate mechanism is unknown. In the current study, we sought to investigate the gene expression levels of IL-17/TRAF6/MAPK/USP25 axis and pro-inflammatory cytokine level (IL-6, IL-1β, and TNF-α) in obese Ovalbumin (OVA)-sensitized female and male Wistar rats lung tissue. MAIN METHODS Animals in both males and females were divided into eight groups (four groups in each sex) based on diet and OVA-sensitization: normal diet, a normal diet with OVA-sensitization, high-fat diet (HFD), and OVA-sensitization with an HFD. KEY FINDINGS In both sexes, obese OVA-sensitized rats, the methacholine concentration-response curve shifted to the left and EC50 methacholine decreased. Increased pro-inflammatory cytokines as well as elevated IL-17/TRAF6/MAPK axis genes and decreased USP25 gene expression were identified in obese OVA-sensitized groups. SIGNIFICANCE The results indicate that in obese OVA-sensitized rats, the IL-17 axis were involved in the pathogenesis of the disease and can be considered as a therapeutic target in subjects with obesity-related asthma.
Collapse
Affiliation(s)
- Mohammad Reza Aslani
- Department of Physiology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Afshan Sharghi
- Department of Community Medicine, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mohammad Hossein Boskabady
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hassan Ghobadi
- Internal Medicine Department, Pulmonary Division, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Rana Keyhanmanesh
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mahdi Ahmadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeideh Saadat
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Physiology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Parya Naghizadeh
- Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
33
|
Huang J, Zhang J, Wang X, Jin Z, Zhang P, Su H, Sun X. Effect of Probiotics on Respiratory Tract Allergic Disease and Gut Microbiota. Front Nutr 2022; 9:821900. [PMID: 35295917 PMCID: PMC8920559 DOI: 10.3389/fnut.2022.821900] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Allergy is a hypersensitivity reaction triggered by specific cell or antibody-mediated immune mechanisms. Allergies have increased in industrialized countries in recent decades. The rise in allergic respiratory diseases such as allergic rhinitis (AR) and allergic asthma (AA) is a potential threat to public health. Searches were conducted using PubMed, Google Scholar and Medline using the following key terms: allergic rhinitis OR asthma AND probiotics, allergic airway inflammation AND immune disorders, probiotics OR gut microbiota AND allergic disease, probiotics AND inflammatory. Studies from all years were included, specifically those published within the last 10 years. Some review articles and their reference lists were searched to identify related articles. The role of microbiota in respiratory allergic diseases has attracted more and more attention. Pieces of evidence suggested that the development of allergic diseases causes a possible imbalance in the composition of the gut microbiota. Compared to colonized mice, germ-free mice exhibit exaggerated allergic airway responses, suggesting that microbial host interactions play an important role in the development of allergic diseases. Probiotics modulate both the innate and adaptive inflammatory immune responses, often used as dietary supplements to provide health benefits in gastrointestinal disorders. Probiotics may serve as immunomodulators and activators of host defense pathways. Besides, oral probiotics can modulate the immune response in the respiratory system. Recently, studies in humans and animals have demonstrated the role of probiotic in RA and AA. To understand the characterization, microbiota, and the potential role of probiotics intervention of AA/AR, this review provides an overview of clinical features of AA and AR, probiotics for the prevention and treatment of AR, AA, changes in gut microbiota, and their mechanisms of action.
Collapse
Affiliation(s)
- Jinli Huang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Juan Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xingzhi Wang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zenghui Jin
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Panpan Zhang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hui Su
- Department of Geratology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xin Sun
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
34
|
Margelidon-Cozzolino V, Tsicopoulos A, Chenivesse C, de Nadai P. Role of Th17 Cytokines in Airway Remodeling in Asthma and Therapy Perspectives. FRONTIERS IN ALLERGY 2022; 3:806391. [PMID: 35386663 PMCID: PMC8974749 DOI: 10.3389/falgy.2022.806391] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/10/2022] [Indexed: 12/07/2022] Open
Abstract
Airway remodeling is a frequent pathological feature of severe asthma leading to permanent airway obstruction in up to 50% of cases and to respiratory disability. Although structural changes related to airway remodeling are well-characterized, immunological processes triggering and maintaining this phenomenon are still poorly understood. As a consequence, no biotherapy targeting cytokines are currently efficient to treat airway remodeling and only bronchial thermoplasty may have an effect on bronchial nerves and smooth muscles with uncertain clinical relevance. Th17 cytokines, including interleukin (IL)-17 and IL-22, play a role in neutrophilic inflammation in severe asthma and may be involved in airway remodeling. Indeed, IL-17 is increased in sputum from severe asthmatic patients, induces the expression of "profibrotic" cytokines by epithelial, endothelial cells and fibroblasts, and provokes human airway smooth muscle cell migration in in vitro studies. IL-22 is also increased in asthmatic samples, promotes myofibroblast differentiation, epithelial-mesenchymal transition and proliferation and migration of smooth muscle cells in vitro. Accordingly, we also found high levels of IL-17 and IL-22 in a mouse model of dog-allergen induced asthma characterized by a strong airway remodeling. Clinical trials found no effect of therapy targeting IL-17 in an unselected population of asthmatic patients but showed a potential benefit in a sub-population of patients exhibiting a high level of airway reversibility, suggesting a potential role on airway remodeling. Anti-IL-22 therapies have not been evaluated in asthma yet but were demonstrated efficient in severe atopic dermatitis including an effect on skin remodeling. In this review, we will address the role of Th17 cytokines in airway remodeling through data from in vitro, in vivo and translational studies, and examine the potential place of Th17-targeting therapies in the treatment of asthma with airway remodeling.
Collapse
Affiliation(s)
- Victor Margelidon-Cozzolino
- Univ. Lille, CNRS, INSERM, CHU de Lille, Institut Pasteur de Lille, Unité INSERM U1019-UMR9017-CIIL-Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Anne Tsicopoulos
- Univ. Lille, CNRS, INSERM, CHU de Lille, Institut Pasteur de Lille, Unité INSERM U1019-UMR9017-CIIL-Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Cécile Chenivesse
- Univ. Lille, CNRS, INSERM, CHU de Lille, Institut Pasteur de Lille, Unité INSERM U1019-UMR9017-CIIL-Centre d'Infection et d'Immunité de Lille, Lille, France
- CRISALIS (Clinical Research Initiative in Severe Asthma: a Lever for Innovation & Science), F-CRIN Network, INSERM US015, Toulouse, France
| | - Patricia de Nadai
- Univ. Lille, CNRS, INSERM, CHU de Lille, Institut Pasteur de Lille, Unité INSERM U1019-UMR9017-CIIL-Centre d'Infection et d'Immunité de Lille, Lille, France
| |
Collapse
|
35
|
Xie Y, Abel PW, Casale TB, Tu Y. T H17 cells and corticosteroid insensitivity in severe asthma. J Allergy Clin Immunol 2022; 149:467-479. [PMID: 34953791 PMCID: PMC8821175 DOI: 10.1016/j.jaci.2021.12.769] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/30/2021] [Accepted: 12/15/2021] [Indexed: 02/03/2023]
Abstract
Asthma is classically described as having either a type 2 (T2) eosinophilic phenotype or a non-T2 neutrophilic phenotype. T2 asthma usually responds to classical bronchodilation therapy and corticosteroid treatment. Non-T2 neutrophilic asthma is often more severe. Patients with non-T2 asthma or late-onset T2 asthma show poor response to the currently available anti-inflammatory therapies. These therapeutic failures result in increased morbidity and cost associated with asthma and pose a major health care problem. Recent evidence suggests that some non-T2 asthma is associated with elevated TH17 cell immune responses. TH17 cells producing Il-17A and IL-17F are involved in the neutrophilic inflammation and airway remodeling processes in severe asthma and have been suggested to contribute to the development of subsets of corticosteroid-insensitive asthma. This review explores the pathologic role of TH17 cells in corticosteroid insensitivity of severe asthma and potential targets to treat this endotype of asthma.
Collapse
Affiliation(s)
- Yan Xie
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, USA
| | - Peter W. Abel
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, USA
| | - Thomas B. Casale
- Department of Internal Medicine, University of South Florida School of Medicine, Tampa, FL, USA
| | - Yaping Tu
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, USA
| |
Collapse
|
36
|
Kim Y, Kim J, Mo Y, Park DE, Lee HS, Jung JW, Kang HR. Cigarette smoke extract contributes to the inception and aggravation of asthmatic inflammation by stimulating innate immunity. ALLERGY ASTHMA & RESPIRATORY DISEASE 2022. [DOI: 10.4168/aard.2022.10.3.145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Yujin Kim
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Jeonghyeon Kim
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Yosep Mo
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Da Eun Park
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Hyun-Seung Lee
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Jae-Woo Jung
- Department of Internal Medicine, Chung-Ang University Hospital, Seoul, Korea
| | - Hye-Ryun Kang
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
37
|
Brightling CE, Nair P, Cousins DJ, Louis R, Singh D. Risankizumab in Severe Asthma - A Phase 2a, Placebo-Controlled Trial. N Engl J Med 2021; 385:1669-1679. [PMID: 34706172 DOI: 10.1056/nejmoa2030880] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Interleukin-23 has been implicated in airway inflammation that is mediated by type 2 and type 17 cytokines. Whether targeting interleukin-23 in the treatment of asthma improves disease control and reduces airway inflammation is unclear. METHODS We conducted a phase 2a, multicenter, randomized, double-blind, placebo-controlled, 24-week, parallel-group trial to assess the efficacy and safety of risankizumab, an anti-interleukin-23p19 monoclonal antibody, in adults with severe asthma. Patients were assigned to receive 90 mg of risankizumab or placebo, administered subcutaneously once every 4 weeks. The primary end point was the time to the first asthma worsening. Asthma worsening was defined as deterioration from baseline on 2 or more consecutive days; deterioration was considered to be a decrease of at least 30% in the morning peak expiratory flow or an increase from baseline of at least 50% in the number of puffs of rescue medication in a 24-hour period (equating to at least four additional puffs), a severe asthma exacerbation, or an increase of 0.75 or more points on the 5-item Asthma Control Questionnaire (ACQ-5; scores range from 0 to 6, with higher scores indicating less control). Secondary end points were the annualized rate of asthma worsening, the annualized rate of severe exacerbations, the ACQ-5 score, and the forced expiratory volume in 1 second. Exploratory end points were assessed with the use of sputum cytologic analysis and gene expression analysis, and safety was assessed. RESULTS A total of 105 patients received risankizumab and 109 received placebo. The clinical characteristics of the patients were similar in the two groups. The time to the first asthma worsening was shorter with risankizumab than with placebo (median, 40 days vs. 86 days; hazard ratio, 1.46; 95% confidence interval [CI], 1.05 to 2.04; P = 0.03). The rate ratio for annualized asthma worsening with risankizumab as compared with placebo was 1.49 (95% CI, 1.12 to 1.99), and the rate ratio for severe exacerbations was 1.13 (95% CI, 0.75 to 1.70). Sputum transcriptomic pathway analysis showed that genes involved in the activation of natural killer cells and cytotoxic T cells and the activation of the type 1 helper T and type 17 helper T transcription factors were down-regulated by risankizumab. No safety concerns were associated with risankizumab therapy. CONCLUSIONS Risankizumab treatment was not beneficial in severe asthma. The time to the first asthma worsening was shorter and the annualized rate of asthma worsening was higher with risankizumab than with placebo. (Funded by AbbVie and Boehringer Ingelheim; ClinicalTrials.gov number, NCT02443298.).
Collapse
Affiliation(s)
- Christopher E Brightling
- From the Institute for Lung Health, Leicester NIHR Biomedical Research Centre, Department of Respiratory Sciences, University of Leicester, Leicester (C.E.B., D.J.C.), and the University of Manchester, Manchester University NHS Foundation Trust, Manchester (D.S.) - both in the United Kingdom; the Firestone Institute for Respiratory Health, McMaster University and St. Joseph's Healthcare, Hamilton, ON, Canada (P.N.); and the GIGA-I3 Research Unit, University of Liege, Department of Pneumology, Centre Hospitalier Universitaire Liège, Liege, Belgium (R.L.)
| | - Parameswaran Nair
- From the Institute for Lung Health, Leicester NIHR Biomedical Research Centre, Department of Respiratory Sciences, University of Leicester, Leicester (C.E.B., D.J.C.), and the University of Manchester, Manchester University NHS Foundation Trust, Manchester (D.S.) - both in the United Kingdom; the Firestone Institute for Respiratory Health, McMaster University and St. Joseph's Healthcare, Hamilton, ON, Canada (P.N.); and the GIGA-I3 Research Unit, University of Liege, Department of Pneumology, Centre Hospitalier Universitaire Liège, Liege, Belgium (R.L.)
| | - David J Cousins
- From the Institute for Lung Health, Leicester NIHR Biomedical Research Centre, Department of Respiratory Sciences, University of Leicester, Leicester (C.E.B., D.J.C.), and the University of Manchester, Manchester University NHS Foundation Trust, Manchester (D.S.) - both in the United Kingdom; the Firestone Institute for Respiratory Health, McMaster University and St. Joseph's Healthcare, Hamilton, ON, Canada (P.N.); and the GIGA-I3 Research Unit, University of Liege, Department of Pneumology, Centre Hospitalier Universitaire Liège, Liege, Belgium (R.L.)
| | - Renaud Louis
- From the Institute for Lung Health, Leicester NIHR Biomedical Research Centre, Department of Respiratory Sciences, University of Leicester, Leicester (C.E.B., D.J.C.), and the University of Manchester, Manchester University NHS Foundation Trust, Manchester (D.S.) - both in the United Kingdom; the Firestone Institute for Respiratory Health, McMaster University and St. Joseph's Healthcare, Hamilton, ON, Canada (P.N.); and the GIGA-I3 Research Unit, University of Liege, Department of Pneumology, Centre Hospitalier Universitaire Liège, Liege, Belgium (R.L.)
| | - Dave Singh
- From the Institute for Lung Health, Leicester NIHR Biomedical Research Centre, Department of Respiratory Sciences, University of Leicester, Leicester (C.E.B., D.J.C.), and the University of Manchester, Manchester University NHS Foundation Trust, Manchester (D.S.) - both in the United Kingdom; the Firestone Institute for Respiratory Health, McMaster University and St. Joseph's Healthcare, Hamilton, ON, Canada (P.N.); and the GIGA-I3 Research Unit, University of Liege, Department of Pneumology, Centre Hospitalier Universitaire Liège, Liege, Belgium (R.L.)
| |
Collapse
|
38
|
Fung KY, Louis C, Metcalfe RD, Kosasih CC, Wicks IP, Griffin MDW, Putoczki TL. Emerging roles for IL-11 in inflammatory diseases. Cytokine 2021; 149:155750. [PMID: 34689057 DOI: 10.1016/j.cyto.2021.155750] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/16/2022]
Abstract
Interleukin-11 (IL-11) is a cytokine that has been strongly implicated in the pathogenesis of fibrotic diseases and solid malignancies. Elevated IL-11 expression is also associated with several non-malignant inflammatory diseases where its function remains less well-characterized. Here, we summarize current literature surrounding the contribution of IL-11 to the pathogenesis of autoimmune inflammatory diseases, including rheumatoid arthritis, multiple sclerosis, diabetes and systemic sclerosis, as well as other chronic inflammatory conditions such as periodontitis, asthma, chronic obstructive pulmonary disease, psoriasis and colitis.
Collapse
Affiliation(s)
- Ka Yee Fung
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Victoria 3053, Australia.
| | - Cynthia Louis
- Department of Medical Biology, University of Melbourne, Victoria 3053, Australia; Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Victoria 3052, Australia
| | - Riley D Metcalfe
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Technology Institute, University of Melbourne, Victoria 3010, Australia
| | - Clara C Kosasih
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Technology Institute, University of Melbourne, Victoria 3010, Australia
| | - Ian P Wicks
- Department of Medical Biology, University of Melbourne, Victoria 3053, Australia; Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Victoria 3052, Australia; Rheumatology Unit, The Royal Melbourne Hospital, Victoria 3050, Australia
| | - Michael D W Griffin
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Technology Institute, University of Melbourne, Victoria 3010, Australia
| | - Tracy L Putoczki
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Victoria 3053, Australia.
| |
Collapse
|
39
|
Yuksel H, Ocalan M, Yilmaz O. E-Cadherin: An Important Functional Molecule at Respiratory Barrier Between Defence and Dysfunction. Front Physiol 2021; 12:720227. [PMID: 34671272 PMCID: PMC8521047 DOI: 10.3389/fphys.2021.720227] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022] Open
Abstract
While breathing, many microorganisms, harmful environmental particles, allergens, and environmental pollutants enter the human airways. The human respiratory tract is lined with epithelial cells that act as a functional barrier to these harmful factors and provide homeostasis between external and internal environment. Intercellular epithelial junctional proteins play a role in the formation of the barrier. E-cadherin is a calcium-dependent adhesion molecule and one of the most important molecules involved in intercellular epithelial barier formation. E-cadherin is not only physical barrier element but also regulates cell proliferation, differentiation and the immune response to environmental noxious agents through various transcription factors. In this study, we aimed to review the role of E-cadherin in the formation of airway epithelial barier, its status as a result of exposure to various environmental triggers, and respiratory diseases associated with its dysfunction. Moreover, the situations in which its abnormal activation can be noxious would be discussed.
Collapse
Affiliation(s)
- Hasan Yuksel
- Department of Pediatric Allergy and Pulmonology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| | - Merve Ocalan
- Department of Pediatric Allergy and Immunology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| | - Ozge Yilmaz
- Department of Pediatric Allergy and Pulmonology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| |
Collapse
|
40
|
Hussain MS, Sharma P, Dhanjal DS, Khurana N, Vyas M, Sharma N, Mehta M, Tambuwala MM, Satija S, Sohal SS, Oliver BGG, Sharma HS. Nanotechnology based advanced therapeutic strategies for targeting interleukins in chronic respiratory diseases. Chem Biol Interact 2021; 348:109637. [PMID: 34506765 DOI: 10.1016/j.cbi.2021.109637] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/22/2021] [Accepted: 09/06/2021] [Indexed: 02/07/2023]
Abstract
Both communicable and non-communicable chronic respiratory conditions have accorded for suffering of millions of people of all ages and stated to be leading cause of death, morbidity, economic and social pressures, and disability-adjusted life-years (DALYs) worldwide. These illnesses impair patient's health and negatively impacts families and society, particularly in low and middle-income countries. Chronic respiratory diseases (CRDs) affect different organs of respiratory system, involving airways, parenchyma, and pulmonary vasculature. As the number of respiratory diseases are exponentially escalating but still the stakeholders are not paying attention towards its serious complications. Currently, the treatment being used primarily focusses only on alleviating symptoms of these illness rather delivering the therapeutic agent at target site for optimal care and/or prevention. Lately, extensive research is being conducted on airways and systemic inflammation, oxidative stress, airway, or parenchymal rehabilitation. From which macrophages, neutrophils, and T cells, as well as structural cells as fibroblasts, epithelial, endothelial, and smooth muscle cells have been found to be active participants that are involved in these chronic respiratory diseases. The pathogenesis of all these chronic respiratory diseases gets caused differently via mediators and proteins, including cytokines, chemokines, growth factors and oxidants. Presently, the target of prescription therapies is to reduce the inflammation of airways and relieve the airway contraction. In all studies, cytokines have been found to play an imperative role in fostering chronic airway inflammation and remodelling. Owing to the limitations of conventional treatments, the current review aims to summarize the current knowledge about the chronic respiratory disease and discuss further about the various conventional methods that can be used for treating this ailment. Additionally, it also highlights and discusses about the advanced drug delivery system that are being used for targeting the interleukins for the treatment of CRDs.
Collapse
Affiliation(s)
- Md Sadique Hussain
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Parvarish Sharma
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Daljeet Singh Dhanjal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Navneet Khurana
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Manish Vyas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Neha Sharma
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Meenu Mehta
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, Londonderry, BT52 1SA, United Kingdom
| | - Saurabh Satija
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, 7248, Australia
| | - Brian G G Oliver
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia; Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Hari S Sharma
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands.
| |
Collapse
|
41
|
Rahmawati SF, te Velde M, Kerstjens HAM, Dömling ASS, Groves MR, Gosens R. Pharmacological Rationale for Targeting IL-17 in Asthma. FRONTIERS IN ALLERGY 2021; 2:694514. [PMID: 35387016 PMCID: PMC8974835 DOI: 10.3389/falgy.2021.694514] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/07/2021] [Indexed: 01/09/2023] Open
Abstract
Asthma is a respiratory disease that currently affects around 300 million people worldwide and is defined by coughing, shortness of breath, wheezing, mucus overproduction, chest tightness, and expiratory airflow limitation. Increased levels of interleukin 17 (IL-17) have been observed in sputum, nasal and bronchial biopsies, and serum of patients with asthma compared to healthy controls. Patients with higher levels of IL-17 have a more severe asthma phenotype. Biologics are available for T helper 2 (Th2)-high asthmatics, but the Th17-high subpopulation has a relatively low response to these treatments, rendering it a rather severe asthma phenotype to treat. Several experimental models suggest that targeting the IL-17 pathway may be beneficial in asthma. Moreover, as increased activation of the Th17/IL-17 axis is correlated with reduced inhaled corticosteroids (ICS) sensitivity, targeting the IL-17 pathway might reverse ICS unresponsiveness. In this review, we present and discuss the current knowledge on the role of IL-17 in asthma and its interaction with the Th2 pathway, focusing on the rationale for therapeutic targeting of the IL-17 pathway.
Collapse
Affiliation(s)
- Siti Farah Rahmawati
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Department of Pharmacology and Clinical Pharmacy, Institut Teknologi Bandung, Bandung, Indonesia
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Centre Groningen (UMCG), Groningen, Netherlands
| | - Maurice te Velde
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Centre Groningen (UMCG), Groningen, Netherlands
| | - Huib A. M. Kerstjens
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Centre Groningen (UMCG), Groningen, Netherlands
- Department of Pulmonary Medicine, University of Groningen and University Medical Center Groningen (UMCG), Groningen, Netherlands
| | | | | | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Centre Groningen (UMCG), Groningen, Netherlands
- *Correspondence: Reinoud Gosens
| |
Collapse
|
42
|
Zhang S, Fan Y, Qin L, Fang X, Zhang C, Yue J, Bai W, Wang G, Chen Z, Renz H, Skevaki C, Liu X, Xie M. IL-1β augments TGF-β inducing epithelial-mesenchymal transition of epithelial cells and associates with poor pulmonary function improvement in neutrophilic asthmatics. Respir Res 2021; 22:216. [PMID: 34344357 PMCID: PMC8336269 DOI: 10.1186/s12931-021-01808-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/23/2021] [Indexed: 02/08/2023] Open
Abstract
Background Neutrophilic asthmatics (NA) have less response to inhaled corticosteroids. We aimed to find out the predictor of treatment response in NA. Methods Asthmatics (n = 115) and healthy controls (n = 28) underwent clinical assessment during 6-month follow-up with standardized therapy. Asthmatics were categorized by sputum differential cell count. The mRNA expressions were measured by RT-qPCR for sputum cytokines (IFN-γ, IL-1β, IL-27, FOXP3, IL-17A, and IL-5). The protein of IL-1β in sputum supernatant was detected by ELISA. Reticular basement membranes (RBM) were measured in the biopsy samples. The role and signaling pathways of IL-1β mediating the epithelial-mesenchymal transition (EMT) process were explored through A549 cells. Results NA had increased baseline sputum cell IL-1β expression compared to eosinophilic asthmatics (EA). After follow-up, NA had less improvement in FEV1 compared to EA. For all asthmatics, sputum IL-1β mRNA was positively correlated with protein expression. Sputum IL-1β mRNA and protein levels were negatively correlated to FEV1 improvement. After subgrouping, the correlation between IL-1β mRNA and FEV1 improvement was significant in NA but not in EA. Thickness of RBM in asthmatics was greater than that of healthy controls and positively correlated with neutrophil percentage in bronchoalveolar lavage fluid. In vitro experiments, the process of IL-1β augmenting TGF-β1-induced EMT cannot be abrogated by glucocorticoid or montelukast sodium, but can be reversed by MAPK inhibitors. Conclusions IL-1β level in baseline sputum predicts the poor lung function improvement in NA. The potential mechanism may be related to IL-1β augmenting TGF-β1-induced steroid-resistant EMT through MAPK signaling pathways. Trial registration: This study was approved by the Ethics Committee of Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (IRB ID: 20150406). Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01808-7.
Collapse
Affiliation(s)
- Shengding Zhang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Yu Fan
- Department of Respiratory and Critical Care Medicine, Qiandongnanzhou People's Hospital, Kaili, China
| | - Lu Qin
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Xiaoyu Fang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Cong Zhang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Junqing Yue
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Wenxue Bai
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China
| | - Gang Wang
- Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihong Chen
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Harld Renz
- Institute of Laboratory Medicine, Philipps Universität Marburg, Marburg, Germany.,Universities of Giessen and Marburg Lung Center (UGMLC), and the German Center for Lung Research (DZL), Marburg, Germany
| | - Chrysanthi Skevaki
- Institute of Laboratory Medicine, Philipps Universität Marburg, Marburg, Germany.,Universities of Giessen and Marburg Lung Center (UGMLC), and the German Center for Lung Research (DZL), Marburg, Germany
| | - Xiansheng Liu
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China. .,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China.
| | - Min Xie
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China. .,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Key Laboratory of Respiratory Diseases, National Ministry of Health of the People's Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan, China.
| |
Collapse
|
43
|
Murphy RC, Pavord ID, Alam R, Altman MC. Management Strategies to Reduce Exacerbations in non-T2 Asthma. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:2588-2597. [PMID: 34246435 DOI: 10.1016/j.jaip.2021.04.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 11/25/2022]
Abstract
There have been considerable advances in our understanding of asthmatic airway inflammation, resulting in a paradigm shift of classifying individuals on the basis of either the presence or the absence of type 2 (T2) inflammatory markers. Several novel monoclonal antibody therapies targeting T2 cytokines have demonstrated significant clinical effects including reductions in acute exacerbations and improvements in asthma-related quality of life and lung function for individuals with T2-high asthma. However, there have been fewer advancements in developing therapies for those without evidence of T2 airway inflammation (so-called non-T2 asthma). Here, we review the heterogeneity of molecular mechanisms responsible for initiation and regulation of non-T2 inflammation and discuss both current and potential future therapeutic options for individuals with non-T2 asthma.
Collapse
Affiliation(s)
- Ryan C Murphy
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Wash; Center for Lung Biology, Department of Medicine, University of Washington, Seattle, Wash.
| | - Ian D Pavord
- Respiratory Medicine Unit and Oxford Respiratory NIHR Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Rafeul Alam
- Division of Allergy and Immunology, Department of Medicine, National Jewish Health and University of Colorado, Denver, Colo
| | - Matthew C Altman
- Center for Lung Biology, Department of Medicine, University of Washington, Seattle, Wash; Division of Allergy and Immunology, University of Washington, Seattle, Wash
| |
Collapse
|
44
|
Li L, Fang Z, Lee YK, Zhao J, Zhang H, Lu W, Chen W. Prophylactic effects of oral administration of Lactobacillus casei on house dust mite-induced asthma in mice. Food Funct 2021; 11:9272-9284. [PMID: 33047743 DOI: 10.1039/d0fo01363c] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This study investigated the prophylactic effects of five Lactobacillus casei strains on house dust mite (HDM)-induced asthma in mice. BALB/c mice were orally administered with L. casei strains for one week before HDM treatment. Ketotifen and Lactobacillus rhamnosus GG were used as positive controls. All L. casei strains decreased the number of granulocytes and the levels of Th2 and Th17 inflammatory cytokines in the lungs, L. casei3 significantly decreased the airway inflammation score. Further studies showed that L. casei3, L. casei4, and L. casei5 decreased the chemokine levels, L. casei2, L. casei4, and L. casei5 promoted the secretion of secretory immunoglobulin A (sIgA), L. casei2 upregulated the interleukin (IL)-10 levels, and L. casei1 had no effect on these immune indices. L. casei1 and L. casei4 decreased the serum levels of total IgE and HDM-specific IgG1, respectively. L. casei3 and L. casei5 decreased both HDM-specific IgG1 and total IgE levels. L. casei2 did not affect the levels of these immunoglobulins. The gut microbiota analysis revealed that all five L. casei strains enhanced the richness of the gut microbiota mainly by increasing the abundance of Firmicutes, while there were differences at the genus level.Thus, the prophylactic effects of L. casei on HDM-induced mixed chronic airway inflammatory asthma exerted as they differentially affected the immune responses and gut microbiota composition. L. casei3, which exhibited the highest prophylactic effect, increased the acetate and propionate contents in a strain-dependent manner.
Collapse
Affiliation(s)
- Lingzhi Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhifeng Fang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yuan-Kun Lee
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China. and School of Food Science and Technology, Jiangnan University, Wuxi 214122, China and National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China and Beijing Innovation Centre of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
| |
Collapse
|
45
|
Santos RVC, Cunha EGC, de Mello GSV, Rizzo JÂ, de Oliveira JF, do Carmo Alves de Lima M, da Rocha Pitta I, da Rocha Pitta MG, de Melo Rêgo MJB. New Oxazolidines Inhibit the Secretion of IFN-γ and IL-17 by PBMCS from Moderate to Severe Asthmatic Patients. Med Chem 2021; 17:289-297. [PMID: 32914717 DOI: 10.2174/1573406416666200910151950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 07/01/2020] [Accepted: 07/20/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Moderate to severe asthma could be induced by diverse proinflammatory cytokines, as IL-17 and IFN-γ, which are also related to treatment resistance and airway hyperresponsiveness. Oxazolidines emerged as a novel approach for asthma treatment, since some chemical peculiarities were suggested by previous studies. OBJECTIVE The present study aimed to evaluate the IL-17A and IFN-γ modulatory effect of two new oxazolidine derivatives (LPSF/NB-12 and -13) on mononucleated cells of patients with moderate and severe asthma. METHODS The study first looked at potential targets for oxazolidine derivatives using SWISS-ADME. After the synthesis of the compounds, cytotoxicity and cytokine levels were analyzed. RESULTS We demonstrated that LPSF/NB-12 and -13 reduced IFN-γ and IL-17 production in peripheral blood mononucleated cells from asthmatic patients in a concentrated manner. Our in silico analysis showed the neurokinin-1 receptor as a common target for both compounds, which is responsible for diverse proinflammatory effects of moderate and severe asthma. CONCLUSION The work demonstrated a novel approach against asthma, which deserves further studies of its mechanisms of action.
Collapse
Affiliation(s)
- Renata Virgínia Cavalcanti Santos
- Laboratorio de Imunomodulacao e Novas Abordagens Terapeuticas (LINAT), Nucleo de Pesquisa em Inovacao Terapeutica Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Eudes Gustavo Constantino Cunha
- Laboratorio de Imunomodulacao e Novas Abordagens Terapeuticas (LINAT), Nucleo de Pesquisa em Inovacao Terapeutica Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Gabriela Souto Vieira de Mello
- Laboratorio de Imunomodulacao e Novas Abordagens Terapeuticas (LINAT), Nucleo de Pesquisa em Inovacao Terapeutica Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - José Ângelo Rizzo
- Servico de Pneumologia, Hospital das Clinicas, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Jamerson Ferreira de Oliveira
- Laboratorio de Quimica e Inovacao Terapeutica (LQIT), Departamento de Antibioticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Maria do Carmo Alves de Lima
- Laboratorio de Quimica e Inovacao Terapeutica (LQIT), Departamento de Antibioticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Ivan da Rocha Pitta
- Laboratorio de Imunomodulacao e Novas Abordagens Terapeuticas (LINAT), Nucleo de Pesquisa em Inovacao Terapeutica Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Maira Galdino da Rocha Pitta
- Laboratorio de Imunomodulacao e Novas Abordagens Terapeuticas (LINAT), Nucleo de Pesquisa em Inovacao Terapeutica Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Moacyr Jesus Barreto de Melo Rêgo
- Laboratorio de Imunomodulacao e Novas Abordagens Terapeuticas (LINAT), Nucleo de Pesquisa em Inovacao Terapeutica Suely Galdino (NUPIT-SG), Universidade Federal de Pernambuco, Recife, PE, Brazil
| |
Collapse
|
46
|
Mammen MJ, Ali J, Aurora A, Sharma UC, Aalinkeel R, Mahajan SD, Sands M, Schwartz SA. IL-17 Is a Key Regulator of Mucin-Galectin-3 Interactions in Asthma. Int J Cell Biol 2021; 2021:9997625. [PMID: 34221020 PMCID: PMC8211528 DOI: 10.1155/2021/9997625] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/24/2021] [Indexed: 12/28/2022] Open
Abstract
Mucus hypersecretion and chronic airway inflammation are standard characteristics of several airway diseases, such as chronic obstructive pulmonary disease and asthma. Increased mucus secretion from increased mucin gene expression in the airway epithelium is associated with poor prognosis and mortality. We previously showed that the absence of tissue inhibitor of metalloproteinase 1 (TIMP-1) enhances lung inflammation, airway hyperreactivity, and lung remodeling in asthma in an ovalbumin (OVA) asthma model of TIMP-1 knockout (TIMPKO) mice as compared to wild-type (WT) controls and mediated by increased galectin-3 (Gal-3) levels. Additionally, we have shown that in the lung epithelial cell line A549, Gal-3 inhibition increases interleukin-17 (IL-17) levels, leading to increased mucin expression in the airway epithelium. Therefore, in the current study, we further examined the relationship between Gal-3 and the production of IL-17-axis cytokines and critical members of the mucin family in the murine TIMPKO asthma model and the lung epithelium cell line A549. While Gal-3 may regulate a Th1/Th2 response, IL-17 could stimulate the mucin genes, MUC5B and MUC5AC. Gal-3 and IL-17 interactions induce mucus expression in OVA-sensitized mice. We conclude that Gal-3 may play an essential role in the pathogenesis of asthma, and modulation of Gal-3 may prove helpful in the treatment of this disease.
Collapse
Affiliation(s)
- Manoj J. Mammen
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, State University of New York at Buffalo, 875 Ellicott Street, Buffalo, NY 14203, USA
| | - Jamil Ali
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, State University of New York at Buffalo, 875 Ellicott Street, Buffalo, NY 14203, USA
- Division of Allergy, Immunology & Rheumatology, Department of Medicine, State University of New York at Buffalo, 875 Ellicott Street, Buffalo, NY 14203, USA
| | - Amita Aurora
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, State University of New York at Buffalo, 875 Ellicott Street, Buffalo, NY 14203, USA
- Division of Allergy, Immunology & Rheumatology, Department of Medicine, State University of New York at Buffalo, 875 Ellicott Street, Buffalo, NY 14203, USA
| | - Umesh C. Sharma
- Division of Cardiology, Department of Medicine, 875 Ellicott Street, Buffalo, NY 14203, USA
| | - Ravikumar Aalinkeel
- Division of Allergy, Immunology & Rheumatology, Department of Medicine, State University of New York at Buffalo, 875 Ellicott Street, Buffalo, NY 14203, USA
| | - Supriya D. Mahajan
- Division of Allergy, Immunology & Rheumatology, Department of Medicine, State University of New York at Buffalo, 875 Ellicott Street, Buffalo, NY 14203, USA
| | - Mark Sands
- Division of Allergy, Immunology & Rheumatology, Department of Medicine, State University of New York at Buffalo, 875 Ellicott Street, Buffalo, NY 14203, USA
- WNY VA Healthcare System, Buffalo, NY 14215, USA
| | - Stanley A. Schwartz
- Division of Allergy, Immunology & Rheumatology, Department of Medicine, State University of New York at Buffalo, 875 Ellicott Street, Buffalo, NY 14203, USA
| |
Collapse
|
47
|
Hayes KS, Grencis RK. Trichuris muris and comorbidities - within a mouse model context. Parasitology 2021; 148:1-9. [PMID: 34078488 PMCID: PMC8660644 DOI: 10.1017/s0031182021000883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/22/2021] [Accepted: 05/26/2021] [Indexed: 01/15/2023]
Abstract
Trichuris muris is a mouse intestinal parasitic nematode that inhabits the large intestine of its host and induces a strong immune response. The effects of this strong anti-parasite response can be found locally within the intestinal niche and also systemically, having effects on multiple organs. Additionally, the anti-parasite response can have multiple effects on infectious organisms and on microbiota that the host is harbouring. It has been shown that Th1 responses induced by T. muris can affect progression of bowel inflammation, cause colitic-like intestinal inflammation, reduce barrier function and intestinal mucosal responses. In the brain, T. muris can exacerbate stroke outcome and other neurological conditions. In the lung, T. muris can suppress airway inflammation and alter immune responses to other parasites. Additionally, T. muris induced responses can inhibit anti-tumour immunity. Although this parasite maintains a localized niche in the large intestine, its effects can be far-reaching and substantially impact other infections through modulation of bystander immune responses.
Collapse
Affiliation(s)
- Kelly S. Hayes
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Richard K. Grencis
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Trust Centre for Cell Matrix Research and Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
48
|
Dual interleukin-17A/F deficiency protects against acute and chronic response to cigarette smoke exposure in mice. Sci Rep 2021; 11:11508. [PMID: 34075087 PMCID: PMC8169846 DOI: 10.1038/s41598-021-90853-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 05/18/2021] [Indexed: 12/19/2022] Open
Abstract
IL-17A and IL-17F are both involved in the pathogenesis of neutrophilic inflammation observed in COPD and severe asthma. To explore this, mice deficient in both Il17a and Il17f and wild type (WT) mice were exposed to cigarette smoke or environmental air for 5 to 28 days and changes in inflammatory cells in bronchoalveolar lavage (BAL) fluid were determined. We also measured the mRNA expression of keratinocyte derived chemokine (Kc), macrophage inflammatory protein-2 (Mip2), granulocyte–macrophage colony stimulating factor (Gmcsf) and matrix metalloproteinase-9 (Mmp9 ) in lung tissue after 8 days, and lung morphometric changes after 24 weeks of exposure to cigarette smoke compared to air-exposed control animals. Macrophage counts in BAL fluid initially peaked at day 8 and again on day 28, while neutrophil counts peaked between day 8 and 12 in WT mice. Mice dual deficient with Il17a and 1l17f showed similar kinetics with macrophages and neutrophils, but cell numbers at day 8 and mRNA expression of Kc, Gmcsf and Mmp9 were significantly reduced. Furthermore, airspaces in WT mice became larger after cigarette smoke exposure for 24 weeks, whereas this was not seen dual Il17a and 1l17f deficient mice. Combined Il17a and Il17f deficiency resulted in significant attenuation of neutrophilic inflammatory response and protection against structural lung changes after long term cigarette smoke exposure compared with WT mice. Dual IL-17A/F signalling plays an important role in pro-inflammatory responses associated with histological changes induced by cigarette smoke exposure.
Collapse
|
49
|
Fitzpatrick AM, Chipps BE, Holguin F, Woodruff PG. T2-"Low" Asthma: Overview and Management Strategies. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 8:452-463. [PMID: 32037109 DOI: 10.1016/j.jaip.2019.11.006] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/11/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023]
Abstract
Although the term "asthma" has been applied to all patients with airway lability and variable chest symptoms for centuries, phenotypes of asthma with distinct clinical and molecular features that may warrant different treatment approaches are well recognized. Patients with type 2 (T2)-"high" asthma are characterized by upregulation of T2 immune pathways (ie, IL-4 and IL-13 gene sets) and eosinophilic airway inflammation, whereas these features are absent in patients with T2-"low" asthma and may contribute to poor responsiveness to corticosteroid treatment. This review details definitions and clinical features of T2-"low" asthma, potential mechanisms and metabolic aspects, pediatric considerations, and potential treatment approaches. Priority research questions for T2-"low" asthma are also discussed.
Collapse
Affiliation(s)
| | - Bradley E Chipps
- Capital Allergy and Respiratory Disease Center, Sacramento, Calif
| | - Fernando Holguin
- University of Colorado, Pulmonary Sciences and Critical Care Medicine, Denver, Colo
| | - Prescott G Woodruff
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, and the Cardiovascular Research Institute, University of California, San Francisco, Calif
| |
Collapse
|
50
|
He Z, Wu J, Zeng X, Bao H, Liu X. Role of the Notch ligands Jagged1 and Delta4 in Th17/Treg immune imbalance in a mouse model of chronic asthma. Exp Lung Res 2021; 47:289-299. [PMID: 34096812 DOI: 10.1080/01902148.2021.1933653] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/30/2021] [Accepted: 05/19/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Asthma is associated with a T helper (Th)17/regulatory T (Treg) cells immune imbalance where the Notch signaling pathway contributes vitally. This study aimed to explore the role of Notch ligands Jagged1 and Delta4 in the Th17/Treg immune imbalance of chronic asthmatic mice. METHODS The experimental animals were randomly assigned to the Saline, ovalbumin (OVA), and OVA + γ-secretase inhibitor (GSI) groups. A mouse model of chronic asthma was induced by OVA sensitization and challenge. GSI was injected intraperitoneally before the OVA challenge in the OVA + GSI group. Lung function, lung histopathology and immunohistochemistry to assess airway inflammation, enzyme-linked immunosorbent assay to measure cytokines levels, flow cytometry to measure the proportions of Th17 (Th17%) and Treg% in CD4+T cells, quantitative real-time polymerase chain reaction and western blot to measure mRNA and protein levels of Jagged1 and Delta4 in lung tissue, and correlation analysis were performed. RESULTS Lung function and histopathology and IL-4, IL-13, and IFN-γ levels in the bronchoalveolar lavage fluid (BALF) of chronic asthmatic mice showed characteristic changes of asthma. The Th17%, Th17/Treg ratio, BALF and serum IL-17 levels, and IL-17/IL-10 ratio increased significantly in the OVA group, while the Treg% and IL-10 level significantly decreased. mRNA and protein expression levels of Jagged1 and Delta4 increased significantly. GSI could reduce the Th17%, Th17/Treg ratio, IL-17, IL-17/IL-10 ratio, and Jagged1 expression in chronic asthmatic mice. The mRNA and protein levels of Jagged1 and Delta4 were positively correlated with the Th17/Treg ratio in the OVA group, while only those of Jagged1 were positively correlated with the Th17/Treg ratio in the OVA + GSI group. CONCLUSIONS In chronic asthmatic mice, the Th17/Treg ratio increased, and the Notch ligands Jagged1 and Delta4 were overactive and positively regulated the Th17/Treg imbalance. GSI partially inhibited Jagged1 and relieved the Th17/Treg imbalance.
Collapse
Affiliation(s)
- Zhen He
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Jirong Wu
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Xiaoli Zeng
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Hairong Bao
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xiaoju Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|