101
|
McDermott M, Cerullo AR, Parziale J, Achrak E, Sultana S, Ferd J, Samad S, Deng W, Braunschweig AB, Holford M. Advancing Discovery of Snail Mucins Function and Application. Front Bioeng Biotechnol 2021; 9:734023. [PMID: 34708024 PMCID: PMC8542881 DOI: 10.3389/fbioe.2021.734023] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/06/2021] [Indexed: 01/12/2023] Open
Abstract
Mucins are a highly glycosylated protein family that are secreted by animals for adhesion, hydration, lubrication, and other functions. Despite their ubiquity, animal mucins are largely uncharacterized. Snails produce mucin proteins in their mucous for a wide array of biological functions, including microbial protection, adhesion and lubrication. Recently, snail mucins have also become a lucrative source of innovation with wide ranging applications across chemistry, biology, biotechnology, and biomedicine. Specifically, snail mucuses have been applied as skin care products, wound healing agents, surgical glues, and to combat gastric ulcers. Recent advances in integrated omics (genomic, transcriptomic, proteomic, glycomic) technologies have improved the characterization of gastropod mucins, increasing the generation of novel biomaterials. This perspective describes the current research on secreted snail mucus, highlighting the potential of this biopolymer, and also outlines a research strategy to fulfill the unmet need of examining the hierarchical structures that lead to the enormous biological and chemical diversity of snail mucus genes.
Collapse
Affiliation(s)
- Maxwell McDermott
- Department of Chemistry and Biochemistry, Hunter College, New York, NY, United States
| | - Antonio R Cerullo
- Department of Chemistry and Biochemistry, Hunter College, New York, NY, United States
| | - James Parziale
- Department of Chemistry and Biochemistry, Hunter College, New York, NY, United States
| | - Eleonora Achrak
- Department of Chemistry and Biochemistry, Hunter College, New York, NY, United States
| | - Sharmin Sultana
- Department of Chemistry and Biochemistry, Hunter College, New York, NY, United States
| | - Jennifer Ferd
- Department of Chemistry and Biochemistry, Hunter College, New York, NY, United States
| | - Safiyah Samad
- Department of Chemistry and Biochemistry, Hunter College, New York, NY, United States
| | - William Deng
- Department of Chemistry and Biochemistry, Hunter College, New York, NY, United States
| | - Adam B Braunschweig
- Department of Chemistry and Biochemistry, Hunter College, New York, NY, United States.,Advanced Science Research Center, Graduate Center of New York, Graduate Department of Biochemistry, New York, NY, United States.,PhD Programs in Biochemistry and Chemistry Graduate Center of the City University of New York, New York, NY, United States
| | - Mandë Holford
- Department of Chemistry and Biochemistry, Hunter College, New York, NY, United States.,PhD Programs in Biochemistry and Chemistry Graduate Center of the City University of New York, New York, NY, United States.,PhD Program in Biology Graduate Center of the City University of New York, New York, NY, United States.,Department of Invertebrate Zoology, The American Museum of Natural History, New York, NY, United States
| |
Collapse
|
102
|
Dhasmana A, Dhasmana S, Kotnala S, A A, Kashyap VK, Shaji PD, Laskar P, Khan S, Pellicano R, Fagoonee S, Haque S, Yallapu MM, Chauhan SC, Jaggi M. A topography of immunotherapies against gastrointestinal malignancies. Panminerva Med 2021; 64:56-71. [PMID: 34664484 DOI: 10.23736/s0031-0808.21.04541-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Gastrointestinal (GI) cancers are one of the leading causes of death worldwide. Although various approaches are implemented to improve the health condition of GI patients, none of the treatment protocols promise for eradicating cancer. However, a treatment mechanism against any kind of disease condition is already existing executing inside the human body. The 'immune system' is highly efficient to detect and destroy the unfavourable events of the body including tumor cells. The immune system can restrict the growth and proliferation of cancer. Cancer cells behave much smarter and adopt new mechanisms for hiding from the immune cells. Thus, cancer immunotherapy might play a decisive role to train the immune system against cancer. In this review, we have discussed the immunotherapy permitted for the treatment of GI cancers. We have discussed various methods and mechanisms, periodic development of cancer immunotherapies, approved biologicals, completed and ongoing clinical trials, role of various biopharmaceuticals, and epigenetic factors involved in GI cancer immunotherapies (graphical abstract Figure 1).
Collapse
Affiliation(s)
- Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA.,Department of Biosciences and Cancer Research Institute, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, India
| | - Swati Dhasmana
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Sudhir Kotnala
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Anukriti A
- Department of Biosciences, School of Liberal Arts and Sciences, Mody University, Lakshamgarh, Rajasthan, India
| | - Vivek K Kashyap
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Poornima D Shaji
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Partha Laskar
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Sheema Khan
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | | | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging (CNR), Molecular Biotechnology Center, Turin, Italy
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia.,Bursa Uludağ University Faculty of Medicine, Görükle Campus, Nilüfer, Bursa, Turkey
| | - Murali M Yallapu
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX, USA - meena.jaggi @utrgv.edu.,South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| |
Collapse
|
103
|
Lee GH, Jin SW, Choi JH, Han EH, Hwang YP, Choi CY, Jeong HG. Influence of o,p'-DDT on MUC5AC expression via regulation of NF-κB/AP-1 activation in human lung epithelial cells. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2021; 84:836-845. [PMID: 34167450 DOI: 10.1080/15287394.2021.1944943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
o,p'-Dichlorodiphenyltrichloroethane (o,p'-DDT) is a representative endocrine disruptor, and exposure to o,p'-DDT may produce immune disorders and inflammation, leading to various diseases such as cancer. Chronic airway inflammation is characterized by excessive mucus secretion resulting in chronic obstructive pulmonary disease (COPD). Mucin 5AC (MUC5AC), one of the mucus genes, plays an important role in mucus secretion and inflammation in the airways. The aim of this study was to examine the effects of o,p'-DDT on the regulation of MUC5AC expression in human lung epithelial A549 cell line. o,p'-DDT increased mRNA levels and the promoter activity of MUC5AC. Transient transfection with mutation promoter constructs of MUC5AC demonstrated that nuclear factor kappa-b (NF-κB) and activator protein 1(AP-1) response elements were essential for the consequences of o,p'-DDT on MUC5AC expression. In addition, o,p'-DDT induced phosphorylation of ERK, JNK, p38, and Akt, which are involved in the regulation of MUC5AC expression. It is noteworthy that inhibitors of NF-κB, AP-1, Akt, and MAPKs blocked enhanced o,p'-DDT-induced MUC5AC mRNA expression. Data indicate that o,p'-DDT increase in NF-κB, and AP-1 transcriptional activation-dependent MUC5AC expression is associated with stimulation of Akt and MAPK signaling pathways in A549 cells.
Collapse
Affiliation(s)
- Gi Ho Lee
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Sun Woo Jin
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Jae Ho Choi
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
- Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju, Republic of Korea
| | - Eun Hee Han
- Drug & Disease Target Research Team, Division of Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Cheongju, Republic of Korea
| | - Yong Pil Hwang
- Fisheries Promotion Division Team, Mokpo City, Mokpo, Republic of Korea
| | - Chul Yung Choi
- Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju, Republic of Korea
| | - Hye Gwang Jeong
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
104
|
Tsujikawa Y, Suzuki M, Sakane I. Isolation, identification, and impact on intestinal barrier integrity of Lactiplantibacillus plantarum from fresh tea leaves (Camellia sinensis). BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2021; 40:186-195. [PMID: 34631330 PMCID: PMC8484006 DOI: 10.12938/bmfh.2020-083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 07/19/2021] [Indexed: 11/05/2022]
Abstract
Lactic acid bacteria (LAB) are safe microorganisms that have been used in the processing of fermented food for centuries. The aim of this study was to isolate Lactobacillus from fresh tea leaves and examine the impact of an isolated strain on intestinal barrier integrity. First, the presence of Lactobacillus strains was investigated in fresh tea leaves from Kagoshima, Japan. Strains were isolated by growing on De Man, Rogosa and Sharpe (MRS) agar medium containing sodium carbonate, followed by the identification of one strain by polymerase chain reaction (PCR) and pheS sequence analysis, with the strain identified as Lactiplantibacillus plantarum and named L. plantarum LOC1. Second, the impact of strain LOC1 in its heat-inactivated form on intestinal barrier integrity was investigated. Strain LOC1, but not L. plantarum ATCC 14917T or L. plantarum ATCC 8014, significantly suppressed dextran sulfate sodium (DSS)-induced decreases in transepithelial electrical resistance values of Caco-2:HT29-MTX 100:0 and 90:10 co-cultures. Moreover, in Caco-2:HT29-MTX co-cultures (90:10 and 75:25), levels of occludin mRNA were significantly increased by strain LOC1 compared with untreated co-cultures, and strain LOC1 had higher mRNA levels of MUC2 and MUC4 mucins than L. plantarum ATCC 14917T and L. plantarum YT9. These results indicate that L. plantarum LOC1 may be used as a safe probiotic with beneficial effects on the intestinal barrier, suggesting that fresh tea leaves could be utilized as a safe source for isolating probiotics.
Collapse
Affiliation(s)
- Yuji Tsujikawa
- Central Research Institute, ITO EN, Ltd., 21 Mekami, Makinohara-shi, Shizuoka 421-0516, Japan
| | - Masahiko Suzuki
- Central Research Institute, ITO EN, Ltd., 21 Mekami, Makinohara-shi, Shizuoka 421-0516, Japan
| | - Iwao Sakane
- Central Research Institute, ITO EN, Ltd., 21 Mekami, Makinohara-shi, Shizuoka 421-0516, Japan
| |
Collapse
|
105
|
Payne DD, Renz A, Dunphy LJ, Lewis T, Dräger A, Papin JA. An updated genome-scale metabolic network reconstruction of Pseudomonas aeruginosa PA14 to characterize mucin-driven shifts in bacterial metabolism. NPJ Syst Biol Appl 2021; 7:37. [PMID: 34625561 PMCID: PMC8501023 DOI: 10.1038/s41540-021-00198-2] [Citation(s) in RCA: 12] [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: 03/10/2021] [Accepted: 09/17/2021] [Indexed: 12/30/2022] Open
Abstract
Mucins are present in mucosal membranes throughout the body and play a key role in the microbe clearance and infection prevention. Understanding the metabolic responses of pathogens to mucins will further enable the development of protective approaches against infections. We update the genome-scale metabolic network reconstruction (GENRE) of one such pathogen, Pseudomonas aeruginosa PA14, through metabolic coverage expansion, format update, extensive annotation addition, and literature-based curation to produce iPau21. We then validate iPau21 through MEMOTE, growth rate, carbon source utilization, and gene essentiality testing to demonstrate its improved quality and predictive capabilities. We then integrate the GENRE with transcriptomic data in order to generate context-specific models of P. aeruginosa metabolism. The contextualized models recapitulated known phenotypes of unaltered growth and a differential utilization of fumarate metabolism, while also revealing an increased utilization of propionate metabolism upon MUC5B exposure. This work serves to validate iPau21 and demonstrate its utility for providing biological insights.
Collapse
Affiliation(s)
- Dawson D Payne
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Alina Renz
- Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Tübingen, Germany
- Department of Computer Science, University of Tübingen, Tübingen, Germany
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen, Tübingen, Germany
| | - Laura J Dunphy
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Taylor Lewis
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Andreas Dräger
- Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Tübingen, Germany
- Department of Computer Science, University of Tübingen, Tübingen, Germany
- Cluster of Excellence 'Controlling Microbes to Fight Infections', University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF) partner site, Tübingen, Germany
| | - Jason A Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
106
|
Iwashita J, Murata J. Integrin β1 subunit regulates cellular and secreted MUC5AC and MUC5B production in NCI-H292 human lung epithelial cells. Biochem Biophys Rep 2021; 28:101124. [PMID: 34504957 PMCID: PMC8416645 DOI: 10.1016/j.bbrep.2021.101124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022] Open
Abstract
The surface of the human respiratory tract is covered with a mucus layer containing mucin 5AC (MUC5AC) and mucin 5B (MUC5B) as the main components. This layer contributes to biological defense by eliminating irritants, but excessive MUC5AC secretion by the airway epithelial cells exacerbates asthma. Therefore, regulating mucin production is important for asthma treatment. In this study, the effects of integrin β1 subunit on MUC5AC and MUC5B production were examined in NCI–H292 human lung cancer epithelial cells. When integrin β1 was overexpressed, cellular and secreted MUC5AC levels were decreased, whereas cellular MUC5B production was increased. Conversely, integrin β1 depletion using siRNA increased cellular and secreted MUC5AC production, but decreased cellular MUC5B production. Further, the activity of extracellular signal-regulated kinase (ERK), which promotes MUC5AC production, was decreased by integrin β1 overexpression and increased by its depletion. These results suggest that integrin β1 suppresses MUC5AC production and promotes MUC5B production by downregulating ERK. We studied the regulation of MUC5AC and MUC5B production by integrin β1 subunit. Integrin β1 overexpression reduced MUC5AC, but increased MUC5B levels. Integrin β1 depletion increased MUC5AC production and ROS level, but decreased MUC5B production. Integrin β1 overexpression decreased ERK activity in NCI–H292 airway cells. Integrin β1 downregulates ERK to suppress MUC5AC & promote MUC5B production.
Collapse
Affiliation(s)
- Jun Iwashita
- Faculty of Bioresource Sciences, Akita Prefectural University, 241-438 Kaidobata-Nishi, Shimoshinjo-Nakano, Akita, Akita, 010-0195, Japan
| | - Jun Murata
- Faculty of Bioresource Sciences, Akita Prefectural University, 241-438 Kaidobata-Nishi, Shimoshinjo-Nakano, Akita, Akita, 010-0195, Japan
| |
Collapse
|
107
|
Guo J, Xu S. Astragaloside IV suppresses histamine-induced inflammatory factors and mucin 5 subtype AC overproduction in nasal epithelial cells via regulation of inflammation-related genes. Bioengineered 2021; 12:6045-6056. [PMID: 34482800 PMCID: PMC8806810 DOI: 10.1080/21655979.2021.1965813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Allergic rhinitis (AR) is a symptomatic allergic disease that leads to severe inflammation. Astragaloside IV (AS-IV) is a primary active component of Astragalus membranaceus and exerts immune-regulation and anti-inflammatory effects. However, the pharmacological effect of AS-IV in the nasal epithelial cells (NECs) has not been reported. The present study aimed to assess the effect of AS-IV on inflammatory cytokines and mucin 5 subtype AC (MUC5AC) overproduction in histamine (His)-stimulated NECs and its underlying mechanism. NECs were stimulated with or without His for 24 h in the absence or presence of AS-IV. The levels of inflammatory cytokines including IL-6, IL-8, MCP-1, IL-1β, granulocyte-macrophage colony-stimulating factor (GM-CSF), eotaxin, and MUC5AC were assayed. Our findings indicated that AS-IV inhibited His-evoked release and expression of inflammatory cytokines and MUC5AC in NECs. RNA-seq analyses indicated the significant changes in expression levels involved in inflammation genes upon treatment of His-induced NECs with AS-IV. Our findings indicated that AS-IV inhibited His-evoked inflammatory cytokines secretion and MUC5AC overproduction in NECs, which were partly mediated by regulation of inflammation-related genes. Therefore, our findings provided a scientific basis for the development of AS-IV as an effective agent for clinical therapeutic strategy in the treatment of AR.
Collapse
Affiliation(s)
- Jie Guo
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Luoyang Central Hospital of Zhengzhou University, Luoyang Henan, China
| | - Shuai Xu
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Luoyang Central Hospital of Zhengzhou University, Luoyang Henan, China
| |
Collapse
|
108
|
Maternal H-antigen secretor status is an early biomarker for potential preterm delivery. J Perinatol 2021; 41:2147-2155. [PMID: 33235282 DOI: 10.1038/s41372-020-00870-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 10/08/2020] [Accepted: 11/04/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Pre-pregnancy or first trimester biomarkers predicting preterm delivery are lacking. The purpose of this study was to determine whether maternal H-antigen (secretor status) is a potential biomarker for preterm delivery. METHODS This cohort study examined maternal saliva samples and birth data gathered by the National Children's Study Vanguard pilot phase (2009-2014) and included 300 women who were ≥18 years old and provided birth data and saliva samples. The maternal secretor status phenotype was determined by quantifying H-antigen in saliva using enzyme-linked immunoassay. Mothers were stratified by secretor status and multivariable analysis estimated adjusted associations with preterm delivery. RESULTS Maternal lack of H-antigen production was an independent risk factor for preterm delivery after adjusting for known confounders (aOR 4.53; 95% CI: 1.74, 11.81; P = 0.002). CONCLUSIONS Maternal H-antigen may be a biomarker identifying women at-risk for preterm delivery. Prospective cohort studies validating these findings are needed.
Collapse
|
109
|
Liu Z, Yan J, Tong L, Liu S, Zhang Y. The role of exosomes from BALF in lung disease. J Cell Physiol 2021; 237:161-168. [PMID: 34388259 PMCID: PMC9292261 DOI: 10.1002/jcp.30553] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 07/06/2021] [Accepted: 07/30/2021] [Indexed: 01/03/2023]
Abstract
Exosomes are released from a variety of immune cells and nonimmune cells, the phospholipid vesicle bilayer membrane structure actively secreted into tissues. Recently, exosomes were demonstrated to be effectively delivered proteins, cholesterol, lipids, and amounts of DNA, mRNA, and noncoding RNAs to a target cell or tissue from a host cell. These can be detected in blood, urine, exhaled breath condensates, bronchoalveolar lavage fluid (BALF), ascites, and cerebrospinal fluid. BALF is a clinical examination method for obtaining alveolar cells and biochemical components, reflecting changes in the lungs, so it is also called liquid biopsy. Exosomes from BALF become a new method for intercellular communication and well‐documented in various pulmonary diseases. In chronic obstructive pulmonary disease (COPD), BALF exosomes can predict the degree of COPD damage and serve as an effective monitoring indicator for airflow limitation and airway remodeling. It also mediates antigen presentation in the airways to the adaptive immune system as well as costimulatory effects. Furthermore, BALF exosomes from acute lung injury and infective diseases are closely related to various infections and lack of oxygen status. BALF exosomes play an important role in the diagnosis and prognosis of lung cancer. The effect of immunomodulatory role for BALF exosomes in adaptive and innate immune responses has been studied in sarcoidosis. The intercellular communication in the microenvironment of BALF exosomes in pulmonary fibrosis and lung remodeling have been studied. In this review, we summarize the novel findings of exosomes in BALF, executed function by protein, miRNA, DNA cytokine, and so on in several pulmonary diseases.
Collapse
Affiliation(s)
- Ziyu Liu
- Department of Pediatrics, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China.,School of Life Science, Jilin University, Changchun, Jilin, China
| | - Jiaqing Yan
- Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Lingling Tong
- Department of Pathology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Shouyue Liu
- Department of Neurosurgery, Second Hospital, Jilin University, Changchun, China
| | - Ying Zhang
- Department of Pediatrics, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| |
Collapse
|
110
|
Terlizzi V, Masi E, Francalanci M, Taccetti G, Innocenti D. Hypertonic saline in people with cystic fibrosis: review of comparative studies and clinical practice. Ital J Pediatr 2021; 47:168. [PMID: 34362426 PMCID: PMC8343926 DOI: 10.1186/s13052-021-01117-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/17/2021] [Indexed: 11/10/2022] Open
Abstract
Cystic fibrosis (CF) is a multisystem disorder, caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. These cause a reduced secretion of chloride, a marked absorption of sodium and, therefore, of water, through the epithelium, resulting in the formation of thickened secretions in organs such as lung or pancreas. These viscous secretions lead to airway obstruction, chronic infection and inflammation resulting in progressive lung damage, bronchiectasis and eventual respiratory failure. Although the average life expectancy has increased over the last 30 years, lung disease is the most common cause of death in people with CF. For these reasons, the improvement of sputum clearance is a major therapeutic aim in CF and early initiation of airway clearance is widely recommended and implemented. Symptomatic mucolytic therapy today is mainly based on inhalation of DNase, hypertonic saline or mannitol, in combination with physiotherapy. Mucolytic agents break down the gel structure of mucus and therefore decrease its elasticity and viscosity, reducing the pulmonary exacerbation frequency and to improve and stabilize lung function. Nevertheless, high quality studies comparing these mucolytic drugs are still few, and the individual experiences of patients and caregivers explain the high variability of their use globally. This review will summarize the current knowledge on hypertonic saline in the treatment of CF lung disease. Furthermore, we report the real-world prescription of inhaled mucolytic agents in CF.
Collapse
Affiliation(s)
- Vito Terlizzi
- Department of Paediatric Medicine, Cystic Fibrosis Regional Reference Center, Anna Meyer Children's Hospital, Viale Gaetano Pieraccini 24, 50139, Florence, Italy.
| | - Eleonora Masi
- Rehabilitation Unit, Anna Meyer Children's Hospital, Florence, Italy
| | - Michela Francalanci
- Department of Paediatric Medicine, Cystic Fibrosis Regional Reference Center, Anna Meyer Children's Hospital, Viale Gaetano Pieraccini 24, 50139, Florence, Italy
| | - Giovanni Taccetti
- Department of Paediatric Medicine, Cystic Fibrosis Regional Reference Center, Anna Meyer Children's Hospital, Viale Gaetano Pieraccini 24, 50139, Florence, Italy
| | - Diletta Innocenti
- Rehabilitation Unit, Anna Meyer Children's Hospital, Florence, Italy
| |
Collapse
|
111
|
Smyth T, Georas SN. Effects of ozone and particulate matter on airway epithelial barrier structure and function: a review of in vitro and in vivo studies. Inhal Toxicol 2021; 33:177-192. [PMID: 34346824 DOI: 10.1080/08958378.2021.1956021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The airway epithelium represents a crucial line of defense against the spread of inhaled pathogens. As the epithelium is the first part of the body to be exposed to the inhaled environment, it must act as both a barrier to and sentinel against any inhaled agents. Despite its vital role in limiting the spread of inhaled pathogens, the airway epithelium is also regularly exposed to air pollutants which disrupt its normal function. Here we review the current understanding of the structure and composition of the airway epithelial barrier, as well as the impact of inhaled pollutants, including the reactive gas ozone and particulate matter, on epithelial function. We discuss the current in vitro, rodent model, and human exposure findings surrounding the impact of various inhaled pollutants on epithelial barrier function, mucus production, and mucociliary clearance. Detailed information on how inhaled pollutants impact epithelial structure and function will further our understanding of the adverse health effects of air pollution exposure.
Collapse
Affiliation(s)
- Timothy Smyth
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Steve N Georas
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
112
|
Slot E, Boers R, Boers J, van IJcken WFJ, Tibboel D, Gribnau J, Rottier R, de Klein A. Genome wide DNA methylation analysis of alveolar capillary dysplasia lung tissue reveals aberrant methylation of genes involved in development including the FOXF1 locus. Clin Epigenetics 2021; 13:148. [PMID: 34325731 PMCID: PMC8323302 DOI: 10.1186/s13148-021-01134-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
Background Alveolar capillary dysplasia with or without misalignment of the pulmonary veins (ACD/MPV) is a lethal congenital lung disorder associated with a variety of heterozygous genomic alterations in the FOXF1 gene or its 60 kb enhancer. Cases without a genomic alteration in the FOXF1 locus have been described as well. The mechanisms responsible for FOXF1 haploinsufficiency and the cause of ACD/MPV in patients without a genomic FOXF1 variant are poorly understood, complicating the search for potential therapeutic targets for ACD/MPV. To investigate the contribution of aberrant DNA methylation, genome wide methylation patterns of ACD/MPV lung tissues were compared with methylation patterns of control lung tissues using the recently developed technique Methylated DNA sequencing (MeD-seq).
Results Eight ACD/MPV lung tissue samples and three control samples were sequenced and their mutual comparison resulted in identification of 319 differentially methylated regions (DMRs) genome wide, involving 115 protein coding genes. The potentially upregulated genes were significantly enriched in developmental signalling pathways, whereas potentially downregulated genes were mainly enriched in O-linked glycosylation. In patients with a large maternal deletion encompassing the 60 kb FOXF1 enhancer, DNA methylation patterns in this FOXF1 enhancer were not significantly different compared to controls. However, two hypermethylated regions were detected in the 60 kb FOXF1 enhancer of patients harbouring a FOXF1 point mutation. Lastly, a large hypermethylated region overlapping the first FOXF1 exon was found in one of the ACD/MPV patients without a known pathogenic FOXF1 variation.
Conclusion This is the first study providing genome wide methylation data on lung tissue of ACD/MPV patients. DNA methylation analyses in the FOXF1 locus excludes maternal imprinting of the 60 kb FOXF1 enhancer. Hypermethylation at the 60 kb FOXF1 enhancer might contribute to FOXF1 haploinsufficiency caused by heterozygous mutations in the FOXF1 coding region. Interestingly, DNA methylation analyses of patients without a genomic FOXF1 variant suggest that abnormal hypermethylation of exon 1 might play a role in some ACD/MPV in patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01134-1.
Collapse
Affiliation(s)
- Evelien Slot
- Department of Paediatric Surgery, Erasmus MC - Sophia Children's Hospital Rotterdam, Rotterdam, Netherlands.,Department of Clinical Genetics, Rm Ee2089, Erasmus MC Rotterdam, Wytemaweg 80, 3015 CN, Rotterdam, Netherlands
| | - Ruben Boers
- Department of Developmental Biology, Oncode Institute, Erasmus MC Rotterdam, Rotterdam, Netherlands
| | - Joachim Boers
- Department of Developmental Biology, Oncode Institute, Erasmus MC Rotterdam, Rotterdam, Netherlands
| | - Wilfred F J van IJcken
- Center for Biomics, Erasmus University Medical Center, Erasmus MC, Rotterdam, Netherlands.,Department of Cell Biology, Erasmus University Medical Center, Erasmus MC, Rotterdam, Netherlands
| | - Dick Tibboel
- Department of Paediatric Surgery, Erasmus MC - Sophia Children's Hospital Rotterdam, Rotterdam, Netherlands
| | - Joost Gribnau
- Department of Developmental Biology, Oncode Institute, Erasmus MC Rotterdam, Rotterdam, Netherlands
| | - Robbert Rottier
- Department of Paediatric Surgery, Erasmus MC - Sophia Children's Hospital Rotterdam, Rotterdam, Netherlands.,Department of Cell Biology, Erasmus University Medical Center, Erasmus MC, Rotterdam, Netherlands
| | - Annelies de Klein
- Department of Clinical Genetics, Rm Ee2089, Erasmus MC Rotterdam, Wytemaweg 80, 3015 CN, Rotterdam, Netherlands.
| |
Collapse
|
113
|
Voynow JA, Shinbashi M. Neutrophil Elastase and Chronic Lung Disease. Biomolecules 2021; 11:biom11081065. [PMID: 34439732 PMCID: PMC8394930 DOI: 10.3390/biom11081065] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 12/24/2022] Open
Abstract
Neutrophil elastase (NE) is a major inflammatory protease released by neutrophils and is present in the airways of patients with cystic fibrosis (CF), chronic obstructive pulmonary disease, non-CF bronchiectasis, and bronchopulmonary dysplasia. Although NE facilitates leukocyte transmigration to the site of infection and is required for clearance of Gram-negative bacteria, it also activates inflammation when released into the airway milieu in chronic inflammatory airway diseases. NE exposure induces airway remodeling with increased mucin expression and secretion and impaired ciliary motility. NE interrupts epithelial repair by promoting cellular apoptosis and senescence and it activates inflammation directly by increasing cytokine expression and release, and indirectly by triggering extracellular trap release and exosome release, which magnify protease activity and inflammation in the airway. NE inhibits innate immune function by digesting opsonins and opsonin receptors, degrading innate immune proteins such as lactoferrin, and inhibiting macrophage phagocytosis. Importantly, NE-directed therapies have not yet been effective in preventing the pathologic sequelae of NE exposure, but new therapies are being developed that offer both direct antiprotease activity and multifunctional anti-inflammatory properties.
Collapse
Affiliation(s)
- Judith A. Voynow
- Division of Pediatric Pulmonology, Children’s Hospital of Richmond at Virginia Commonwealth University, Richmond, VA 23298, USA
- Correspondence:
| | - Meagan Shinbashi
- School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA;
| |
Collapse
|
114
|
Groiss S, Somvilla I, Daxböck C, Fuchs J, Lang-Olip I, Stiegler P, Leber B, Liegl-Atzwanger B, Brislinger D. Quantification of increased MUC5AC expression in airway mucus of smoker using an automated image-based approach. Microsc Res Tech 2021; 85:5-18. [PMID: 34288207 DOI: 10.1002/jemt.23879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/06/2021] [Indexed: 12/19/2022]
Abstract
Microscopic analysis of mucus quantity and composition is crucial in research and diagnostics on muco-obstructive diseases. Currently used image-based methods are unable to extract concrete numeric values of mucosal proteins, especially on the expression of the key mucosal proteins MUC5AC and MUC5B. Since their levels increase under pathologic conditions such as extensive exposure to cigarette smoke, it is imperative to quantify them to improve treatment strategies of pulmonary diseases. This study presents a simple, image-based, and high-processing computational method that allows determining the ratio of MUC5AC and MUC5B within the overall airway mucus while providing information on their spatial distribution. The presented pipeline was optimized for automated downstream analysis using a combination of bright field and immunofluorescence imaging suitable for tracheal and bronchial tissue samples, and air-liquid interface (ALI) cell cultures. To validate our approach, we compared tracheal tissue and ALI cell cultures of isolated primary normal human bronchial epithelial cells derived from smokers and nonsmokers. Our data indicated 18-fold higher levels of MUC5AC in submucosal glands of smokers covering about 8% of mucosal areas compared to <1% in nonsmoking individuals, confirming results of previous studies. We further identified a subpopulation of nonsmokers with slightly elevated glandular MUC5AC levels suggesting moderate exposure to second-hand smoke or fine particulate air pollution. Overall, this study demonstrates a novel, user-friendly and freely available tool for digital pathology and the analysis of therapeutic interventions tested in ALI cell cultures.
Collapse
Affiliation(s)
- Silvia Groiss
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Ina Somvilla
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Christine Daxböck
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Julia Fuchs
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Ingrid Lang-Olip
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Philipp Stiegler
- Division of Transplantation Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Bettina Leber
- Division of Transplantation Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Bernadette Liegl-Atzwanger
- Diagnostic and Research Institute of Pathology, Diagnostic and Research Center for Molecular for Molecular Biomedicine, Medical University Graz, Graz, Austria
| | - Dagmar Brislinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
115
|
Kuang Z, Bennett RC, Lin J, Hao Y, Zhu L, Akinbi HT, Lau GW. Surfactant phospholipids act as molecular switches for premature induction of quorum sensing-dependent virulence in Pseudomonas aeruginosa. Virulence 2021; 11:1090-1107. [PMID: 32842850 PMCID: PMC7549932 DOI: 10.1080/21505594.2020.1809327] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The virulence behaviors of many Gram-negative bacterial pathogens are governed by quorum-sensing (QS), a hierarchical system of gene regulation that relies on population density by producing and detecting extracellular signaling molecules. Although extensively studied under in vitro conditions, adaptation of QS system to physiologically relevant host environment is not fully understood. In this study, we investigated the influence of lung environment on the regulation of Pseudomonas aeruginosa virulence factors by QS in a mouse model of acute pneumonia. When cultured under laboratory conditions in lysogeny broth, wild-type P. aeruginosa strain PAO1 began to express QS-regulated virulence factors elastase B (LasB) and rhamnolipids (RhlA) during transition from late-exponential into stationary growth phase. In contrast, during acute pneumonia as well as when cultured in mouse bronchial alveolar lavage fluids (BALF), exponential phase PAO1 bacteria at low population density prematurely expressed QS regulatory genes lasI-lasR and rhlI-rhlR and their downstream virulence genes lasB and rhlA. Further analysis indicated that surfactant phospholipids were the primary components within BALF that induced the synthesis of N-(3-oxododecanoyl)-L-homoserine lactone (C12-HSL), which triggered premature expression of LasB and RhlA. Both phenol extraction and phospholipase A2 digestion abolished the ability of mouse BALF to promote LasB and RhlA expression. In contrast, provision of the major surfactant phospholipid dipalmitoylphosphatidylcholine (DPPC) restored the expression of both virulence factors. Collectively, our study demonstrates P. aeruginosa modulates its QS to coordinate the expression of virulence factors during acute pneumonia by recognizing pulmonary surfactant phospholipids.
Collapse
Affiliation(s)
- Zhizhou Kuang
- Department of Pathobiology, University of Illinois at Urbana-Champaign , Urbana, IL, USA
| | - Richard C Bennett
- Department of Pathobiology, University of Illinois at Urbana-Champaign , Urbana, IL, USA
| | - Jingjun Lin
- Department of Pathobiology, University of Illinois at Urbana-Champaign , Urbana, IL, USA
| | - Yonghua Hao
- Department of Pathobiology, University of Illinois at Urbana-Champaign , Urbana, IL, USA
| | - Luchang Zhu
- Department of Pathobiology, University of Illinois at Urbana-Champaign , Urbana, IL, USA
| | - Henry T Akinbi
- Division of Pulmonary Medicine, Cincinnati Children Hospital , Cincinnati, OH, USA
| | - Gee W Lau
- Department of Pathobiology, University of Illinois at Urbana-Champaign , Urbana, IL, USA
| |
Collapse
|
116
|
Hicks-Berthet J, Ning B, Federico A, Tilston-Lunel A, Matschulat A, Ai X, Lenburg ME, Beane J, Monti S, Varelas X. Yap/Taz inhibit goblet cell fate to maintain lung epithelial homeostasis. Cell Rep 2021; 36:109347. [PMID: 34260916 PMCID: PMC8346236 DOI: 10.1016/j.celrep.2021.109347] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/22/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022] Open
Abstract
Proper lung function relies on the precise balance of specialized epithelial cells that coordinate to maintain homeostasis. Herein, we describe essential roles for the transcriptional regulators YAP/TAZ in maintaining lung epithelial homeostasis, reporting that conditional deletion of Yap and Wwtr1/Taz in the lung epithelium of adult mice results in severe defects, including alveolar disorganization and the development of airway mucin hypersecretion. Through in vivo lineage tracing and in vitro molecular experiments, we reveal that reduced YAP/TAZ activity promotes intrinsic goblet transdifferentiation of secretory airway epithelial cells. Global gene expression and chromatin immunoprecipitation sequencing (ChIP-seq) analyses suggest that YAP/TAZ act cooperatively with TEA domain (TEAD) transcription factors and the NuRD complex to suppress the goblet cell fate program, directly repressing the SPDEF gene. Collectively, our study identifies YAP/TAZ as critical factors in lung epithelial homeostasis and offers molecular insight into the mechanisms promoting goblet cell differentiation, which is a hallmark of many lung diseases.
Collapse
Affiliation(s)
- Julia Hicks-Berthet
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Boting Ning
- Department of Medicine, Computational Biomedicine Section, Boston University School of Medicine, Boston, MA 02118, USA
| | - Anthony Federico
- Department of Medicine, Computational Biomedicine Section, Boston University School of Medicine, Boston, MA 02118, USA; Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Andrew Tilston-Lunel
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Adeline Matschulat
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Xingbin Ai
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Marc E Lenburg
- Department of Medicine, Computational Biomedicine Section, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jennifer Beane
- Department of Medicine, Computational Biomedicine Section, Boston University School of Medicine, Boston, MA 02118, USA
| | - Stefano Monti
- Department of Medicine, Computational Biomedicine Section, Boston University School of Medicine, Boston, MA 02118, USA; Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
117
|
Harris A. Human molecular genetics and the long road to treating cystic fibrosis. Hum Mol Genet 2021; 30:R264-R273. [PMID: 34245257 DOI: 10.1093/hmg/ddab191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/04/2021] [Accepted: 07/06/2021] [Indexed: 11/13/2022] Open
Abstract
The causative gene in cystic fibrosis was identified in 1989, three years before the publication of the first issue of Human Molecular Genetics. CFTR was among the first genes underlying a common inherited disorder to be cloned, and hence its subsequent utilization towards a cure for CF provides a roadmap for other monogenic diseases. Over the past 30 years the advances that built upon knowledge of the gene and the CFTR protein to develop effective therapeutics have been remarkable, and yet the setbacks have also been challenging. Technological progress in other fields has often circumvented the barriers. This review focuses on key aspects of CF diagnostics and current approaches to develop new therapies for all CFTR mutations. It also highlights the major research advances that underpinned progress towards treatments, and considers the remaining obstacles.
Collapse
Affiliation(s)
- Ann Harris
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| |
Collapse
|
118
|
Hara S, Tojima I, Shimizu S, Kouzaki H, Shimizu T. 17,18-Epoxyeicosatetraenoic Acid Inhibits TNF-α-Induced Inflammation in Cultured Human Airway Epithelium and LPS-Induced Murine Airway Inflammation. Am J Rhinol Allergy 2021; 36:106-114. [PMID: 34236247 DOI: 10.1177/19458924211027682] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND 17,18-Epoxyeicosatetraenoic acid (17,18-EpETE), an eicosapentaenoic acid metabolite, is generated from dietary oil in the gut, and antiinflammatory activity of 17,18-EpETE was recently reported. OBJECTIVE To evaluate the inhibitory effects of 17,18-EpETE in airway inflammation, we examined in vitro and in vivo effects on mucus production, neutrophil infiltration, and cytokine/chemokine production in airway epithelium. METHODS Nasal tissue localization of G protein-coupled receptor 40 (GPR40), a receptor of 17,18-EpETE, was determined by immunohistochemical staining. Expression of GPR40 mRNA in nasal mucosa of chronic rhinosinusitis (CRS) patients and control subjects was determined by reverse transcription-polymerase chain reaction (RT-PCR). The in vitro effects on airway epithelial cells were examined using normal human bronchial epithelial cells and NCI-H292 cells. To examine the in vivo effects of 17,18-EpETE on airway inflammation, we induced goblet cell metaplasia, mucus production, and neutrophil infiltration in mouse nasal epithelium by intranasal lipopolysaccharide (LPS) instillation. RESULTS GPR40 is mainly expressed in human nasal epithelial cells and submucosal gland cells. RT-PCR analysis revealed that the expression of GPR40 mRNA was increased in nasal tissues from CRS patients compared with those from control subjects. 17,18-EpETE significantly inhibited tumor necrosis factor (TNF)-α-induced production of interleukin (IL)-6 , IL-8, and mucin from cultured human airway epithelial cells dose dependently, and these antiinflammatory effects on cytokine production were abolished by GW1100, a selective GPR40 antagonist. Intraperitoneal injection or intranasal instillation of 17,18-EpETE significantly attenuated LPS-induced mucus production and neutrophil infiltration in mouse nasal epithelium. Inflammatory cytokine/chemokine production in lung tissues and bronchoalveolar lavage fluids was also inhibited. CONCLUSION These results indicate that 17,18-EpETE plays a regulatory role in mucus hypersecretion and neutrophil infiltration in nasal inflammation. Local or systemic administration may provide a new therapeutic approach for the treatment of intractable airway disease such as CRS.
Collapse
Affiliation(s)
- Shiori Hara
- 13051Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Ichiro Tojima
- 13051Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Shino Shimizu
- 13051Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Hideaki Kouzaki
- 13051Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Takeshi Shimizu
- 13051Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|
119
|
Morin CD, Déziel E, Gauthier J, Levesque RC, Lau GW. An Organ System-Based Synopsis of Pseudomonas aeruginosa Virulence. Virulence 2021; 12:1469-1507. [PMID: 34180343 PMCID: PMC8237970 DOI: 10.1080/21505594.2021.1926408] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Driven in part by its metabolic versatility, high intrinsic antibiotic resistance, and a large repertoire of virulence factors, Pseudomonas aeruginosa is expertly adapted to thrive in a wide variety of environments, and in the process, making it a notorious opportunistic pathogen. Apart from the extensively studied chronic infection in the lungs of people with cystic fibrosis (CF), P. aeruginosa also causes multiple serious infections encompassing essentially all organs of the human body, among others, lung infection in patients with chronic obstructive pulmonary disease, primary ciliary dyskinesia and ventilator-associated pneumonia; bacteremia and sepsis; soft tissue infection in burns, open wounds and postsurgery patients; urinary tract infection; diabetic foot ulcers; chronic suppurative otitis media and otitis externa; and keratitis associated with extended contact lens use. Although well characterized in the context of CF, pathogenic processes mediated by various P. aeruginosa virulence factors in other organ systems remain poorly understood. In this review, we use an organ system-based approach to provide a synopsis of disease mechanisms exerted by P. aeruginosa virulence determinants that contribute to its success as a versatile pathogen.
Collapse
Affiliation(s)
- Charles D Morin
- Centre Armand-Frappier Santé Biotechnologie, Institut National De La Recherche Scientifique (INRS), Laval, Quebec, Canada
| | - Eric Déziel
- Centre Armand-Frappier Santé Biotechnologie, Institut National De La Recherche Scientifique (INRS), Laval, Quebec, Canada
| | - Jeff Gauthier
- Département De Microbiologie-infectiologie Et Immunologie, Institut De Biologie Intégrative Et Des Systèmes (IBIS), Université Laval, Québec City, Quebec, Canada
| | - Roger C Levesque
- Département De Microbiologie-infectiologie Et Immunologie, Institut De Biologie Intégrative Et Des Systèmes (IBIS), Université Laval, Québec City, Quebec, Canada
| | - Gee W Lau
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, US
| |
Collapse
|
120
|
Understanding the Clinical Impact of MUC5AC Expression on Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13123059. [PMID: 34205412 PMCID: PMC8235261 DOI: 10.3390/cancers13123059] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Management of pancreatic cancer is challenging as there are limited treatment options, and most cases are diagnosed at advanced stages. In addition, there are no dependable tests available to predict bad outcomes or treatment responses in current clinical practice. Here, we shed light on the available evidence on mucin, MUC5AC in predicting the outcome of pancreatic cancers. We also discuss variants of MUC5AC believed to have a role in the malignant transformation of pancreatic tissues. Abstract Mucin-5AC (MUC5AC) is a heavily glycosylated gel-forming secreted mucin with a reliable prognostic value when detected in multiple malignancies. It is highly prevalent (70%) in PDA and is nonexistent in normal pancreatic tissues. Retrospective studies on PDA tumor tissue (detected by immunohistochemistry or IHC)) have investigated the prognostic value of MUC5AC expression but were equivocal. Some studies associated it with poor outcomes (survival or pathological features such as lymph node disease, vascular/neural invasion in resected tumors), while others have concluded that it is a good prognostic marker. The examination of expression level threshold (5%, 10%, or 25%) and the detected region (apical vs. cytoplasmic) were variable among the studies. The maturation stage and glycoform of MUC5AC detected also differed with the Monoclonal antibody (Mab) employed for IHC. CLH2 detects less mature/less glycosylated versions while 45M1 or 21-1 detect mature/more glycosylated forms. Interestingly, aberrantly glycosylated variants of MUC5AC were detected using lectin assays (Wheat Germ Agglutinin-MUC5AC), and Mabs such as NPC-1C and PAM4 have are more specific to malignant pancreatic tissues. NPC-1C and PAM4 antibody reactive epitopes on MUC5AC are immunogenic and could represent specific changes on the native MUC5AC glycoprotein linked to carcinogenesis. It was never studied to predict treatment response.
Collapse
|
121
|
Xu-Chen X, Weinstock J, Rastogi D, Koumbourlis A, Nino G. The airway epithelium during infancy and childhood: A complex multicellular immune barrier. Basic review for clinicians. Paediatr Respir Rev 2021; 38:9-15. [PMID: 34030977 PMCID: PMC8859843 DOI: 10.1016/j.prrv.2021.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/13/2021] [Indexed: 12/13/2022]
Abstract
The airway epithelium is a complex multicellular layer that extends from the nasopharynx to the small airways. It functions as an immune respiratory barrier during early life that develops, matures, and regenerates to adapt to the changes in the environment. While airway epithelial abnormalities have been identified in several clinical disorders, there is increasing interest in understanding its basic regulation and structure in humans. Indeed, recent advances in technology (e.g. single-cell analysis and new human airway epithelial cell models) have allowed us to identify additional cellular subtypes and functions that overall have greatly improved our understanding of the airway epithelium during health and disease. In this review we summarize key features of the airway epithelium including: 1) multilayer structure and cell heterogeneity; 2) adaptability to different environmental and developmental stimuli; 3) innate recognition; and 4) orchestration of immune responses. We discuss these features with a translational and clinical prospective focusing on the development of human respiratory immunity, particularly during early life.
Collapse
Affiliation(s)
| | | | | | | | - Gustavo Nino
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, George Washington University, Washington, D.C, USA.
| |
Collapse
|
122
|
Groth R, Cravigan LT, Niazi S, Ristovski Z, Johnson GR. In situ measurements of human cough aerosol hygroscopicity. J R Soc Interface 2021; 18:20210209. [PMID: 33947221 PMCID: PMC8097516 DOI: 10.1098/rsif.2021.0209] [Citation(s) in RCA: 12] [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: 03/13/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022] Open
Abstract
The airborne dynamics of respiratory droplets, and the transmission routes of pathogens embedded within them, are governed primarily by the diameter of the particles. These particles are composed of the fluid which lines the respiratory tract, and is primarily mucins and salts, which will interact with the atmosphere and evaporate to reach an equilibrium diameter. Measuring organic volume fraction (OVF) of cough aerosol has proved challenging due to large variability and low material volume produced after coughing. Here, the diametric hygroscopic growth factors (GF) of the cough aerosol produced by healthy participants were measured in situ using a rotating aerosol suspension chamber and a humidification tandem differential mobility analyser. Using hygroscopicity models, it was estimated that the average OVF in the evaporated cough aerosol was 0.88 ± 0.07 and the average GF at 90% relative humidity (RH) was 1.31 ± 0.03. To reach equilibrium in dry air the droplets will reduce in diameter by a factor of approximately 2.8 with an evaporation factor of 0.36 ± 0.05. Hysteresis was observed in cough aerosol at RH = ∼35% and RH = ∼65% for efflorescence and deliquescence, respectively, and may depend on the OVF. The same behaviour and GF were observed in nebulized bovine bronchoalveolar lavage fluid.
Collapse
Affiliation(s)
- Robert Groth
- International Laboratory for Air Quality and Health (ILAQH), School of Earth and Atmospheric Sciences, Queensland University of Technology, Brisbane, Australia
| | - Luke T. Cravigan
- International Laboratory for Air Quality and Health (ILAQH), School of Earth and Atmospheric Sciences, Queensland University of Technology, Brisbane, Australia
| | - Sadegh Niazi
- International Laboratory for Air Quality and Health (ILAQH), School of Earth and Atmospheric Sciences, Queensland University of Technology, Brisbane, Australia
| | - Zoran Ristovski
- International Laboratory for Air Quality and Health (ILAQH), School of Earth and Atmospheric Sciences, Queensland University of Technology, Brisbane, Australia
| | - Graham R. Johnson
- International Laboratory for Air Quality and Health (ILAQH), School of Earth and Atmospheric Sciences, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
123
|
Choi TY, Kim JH, Jo S, Lee S, Na HG, Choi YS, Song SY, Kim YD, Bae CH. Ginsenoside Rb1 Attenuates TGF-β1-Induced MUC4/5AC Expression and Epithelial-Mesenchymal Transition in Human Airway Epithelial Cells. KOREAN JOURNAL OF OTORHINOLARYNGOLOGY-HEAD AND NECK SURGERY 2021; 64:232-239. [DOI: 10.3342/kjorl-hns.2020.00150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/01/2020] [Indexed: 07/25/2023]
Abstract
Background and Objectives Ginsenoside Rb1 is the main metabolite of Panax ginseng. It is known to have many beneficial properties including anti-inflammatory, antitumoral and antioxidant effects. However, the therapeutic effects of ginenoside Rb1 on inflammatory airway diseases have not been elucidated. Therefore, we investigated the effects of ginsenoside Rb1 on the TGF-β1-induced mucin gene expression and epithelial-mesenchymal transition (EMT) in human airway epithelial cells.Materials and Method We evaluated the effects of ginsenoside Rb1 on the changes of MUC4, MUC5AC, occludin, claudin 4, claudin 18, neural (N)-cadherin, and epithelial (E)-cadherin expression by TGF-β1 in NCI-H292 cells using reverse, real-time polymerase chain reaction, enzyme-linked immunosorbent assay, and western blot.Results TGF-β1 significantly increased MUC4/5AC expression. Rb1 inhibited TGF-β1- induced MUC4/5AC expression. In addition, TGF-β1 significantly attenuated occludin, claudin 18, and E-cadherin expressions but induced claudin 4 and N-cadherin expressions. On the other hand, Rb1 reversed changes in the TGF-β1- mediated expressions of cell junction molecules.Conclusion These results suggest that ginsenoside Rb1 attenuates TGF-β1-induced MUC4/5AC expressions and EMT in the human airway epithelial cells. These findings are important data demonstrating the potential of ginsenoside Rb1 as a therapeutic agent for inflammatory airway diseases.
Collapse
|
124
|
Borneol in cardio-cerebrovascular diseases: Pharmacological actions, mechanisms, and therapeutics. Pharmacol Res 2021; 169:105627. [PMID: 33892091 DOI: 10.1016/j.phrs.2021.105627] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 12/21/2022]
Abstract
With the coming acceleration of global population aging, the incidence rate of cardio-cerebrovascular diseases (CVDs) is increasing. It has become the leading cause of human mortality. As a natural drug, borneol (BO) not only has anti-inflammatory, anti-oxidant, anti-apoptotic, anti-coagulant activities and improves energy metabolism but can also promote drugs to enter the target organs or tissues through various physiological barriers, such as the blood-brain barrier (BBB), mucous membrane, skin. Thus, it has a significant therapeutic effect on various CVDs, which has been confirmed in a large number of studies. However, the pharmacological actions and mechanisms of BO on CVDs have not been fully investigated. Hence, this review summarizes the pharmacological actions and possible mechanisms of BO, which provides novel ideas for the treatment of CVDs.
Collapse
|
125
|
Mucin expression, epigenetic regulation and patient survival: A toolkit of prognostic biomarkers in epithelial cancers. Biochim Biophys Acta Rev Cancer 2021; 1876:188538. [PMID: 33862149 DOI: 10.1016/j.bbcan.2021.188538] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022]
Abstract
Twenty mucin genes have been identified and classified in two groups (encoding secreted and membrane-bound proteins). Secreted mucins participate in mucus formation by assembling a 3-dimensional network via oligomerization, whereas membrane-bound mucins are anchored to the outer membrane mediating extracellular interactions and cell signaling. Both groups have been associated with carcinogenesis progression in epithelial cancers, and are therefore considered as potential therapeutic targets. In the present review, we discuss the link between mucin expression patterns and patient survival and propose mucins as prognosis biomarkers of epithelial cancers (esophagus, gastric, pancreatic, colorectal, lung, breast or ovarian cancers). We also investigate the relationship between mucin expression and overall survival in the TCGA dataset. In particular, epigenetic mechanisms regulating mucin gene expression, such as aberrant DNA methylation and histone modification, are interesting as they are also associated with diagnosis or prognosis significance. Indeed, mucin hypomethylation has been shown to be associated with carcinogenesis progression and was linked to prognosis in colon cancer or pancreatic cancer patients. Finally we describe the relationship between mucin expression and non-coding RNAs that also may serve as biomarkers. Altogether the concomitant knowledge of specific mucin-pattern expression and epigenetic regulation could be translated as biomarkers with a better specificity/sensitivity performance in several epithelial cancers.
Collapse
|
126
|
Shaheen S, Maqbool K, Beg OA, Gul F. Thermal analysis of airway mucus clearance by ciliary activity in the presence of inertial forces. SN APPLIED SCIENCES 2021. [DOI: 10.1007/s42452-021-04439-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
AbstractIn this study heat transfer effects on cilia induced mucus flow in human airways is presented. The elliptic wave pattern of cilia tips produces metachronal wave which enables the transportation of highly viscous mucus with nonzero inertial forces. Upper Convective Maxwell model is considered as mucus. The governing partial differential equations are transformed from the fixed frame to the wave frame by using Galilean transformation and viscous dissipation is also incorporated in the energy equation. The non-linear governing equations are evaluated by the perturbation technique by using software “MATHEMATICA” and pressure rise is computed by numerical integration. The impact of interested parameters on temperature profile, velocity, pressure rise and pressure gradient are plotted by the graphs. The comparison of velocities due to symplectic and antiplectic metachronal wave are also achieved graphically.
Collapse
|
127
|
The Application of Bicarbonate Recovers the Chemical-Physical Properties of Airway Surface Liquid in Cystic Fibrosis Epithelia Models. BIOLOGY 2021; 10:biology10040278. [PMID: 33805545 PMCID: PMC8065534 DOI: 10.3390/biology10040278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 01/24/2023]
Abstract
Cystic fibrosis (CF) is a genetic disease associated with the defective function of the cystic fibrosis transmembrane conductance regulator (CFTR) protein that causes obstructive disease and chronic bacterial infections in airway epithelia. Deletion of phenylalanine at position 508, p.F508del, the most frequent mutation among CF patients, causes a folding and traffic defect, resulting in a dramatic reduction in the CFTR expression. To investigate whether the direct application of bicarbonate could modify the properties of the airway surface liquid (ASL), we measured the micro-viscosity, fluid transport and pH of human bronchial epithelial cells monolayers. We have demonstrated that the treatment of a CF-epithelia with an iso-osmotic solution containing bicarbonate is capable of reducing both, the ASL viscosity and the apical fluid re-absorption. We suggest the possibility of design a supportive treatment based on topical application of bicarbonate, or any other alkaline buffer.
Collapse
|
128
|
Zajac M, Dreano E, Edwards A, Planelles G, Sermet-Gaudelus I. Airway Surface Liquid pH Regulation in Airway Epithelium Current Understandings and Gaps in Knowledge. Int J Mol Sci 2021; 22:3384. [PMID: 33806154 PMCID: PMC8037888 DOI: 10.3390/ijms22073384] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 12/22/2022] Open
Abstract
Knowledge on the mechanisms of acid and base secretion in airways has progressed recently. The aim of this review is to summarize the known mechanisms of airway surface liquid (ASL) pH regulation and their implication in lung diseases. Normal ASL is slightly acidic relative to the interstitium, and defects in ASL pH regulation are associated with various respiratory diseases, such as cystic fibrosis. Basolateral bicarbonate (HCO3-) entry occurs via the electrogenic, coupled transport of sodium (Na+) and HCO3-, and, together with carbonic anhydrase enzymatic activity, provides HCO3- for apical secretion. The latter mainly involves CFTR, the apical chloride/bicarbonate exchanger pendrin and paracellular transport. Proton (H+) secretion into ASL is crucial to maintain its relative acidity compared to the blood. This is enabled by H+ apical secretion, mainly involving H+/K+ ATPase and vacuolar H+-ATPase that carry H+ against the electrochemical potential gradient. Paracellular HCO3- transport, the direction of which depends on the ASL pH value, acts as an ASL protective buffering mechanism. How the transepithelial transport of H+ and HCO3- is coordinated to tightly regulate ASL pH remains poorly understood, and should be the focus of new studies.
Collapse
Affiliation(s)
- Miroslaw Zajac
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-776 Warsaw, Poland;
| | - Elise Dreano
- Institut Necker Enfants Malades, INSERM U1151, 75015 Paris, France;
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, 75006 Paris, France;
| | - Aurelie Edwards
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA;
| | - Gabrielle Planelles
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, 75006 Paris, France;
- Laboratoire de Physiologie rénale et Tubulopathies, CNRS ERL 8228, 75006 Paris, France
| | - Isabelle Sermet-Gaudelus
- Institut Necker Enfants Malades, INSERM U1151, 75015 Paris, France;
- Centre de Recherche des Cordeliers, Sorbonne Université, INSERM, Université de Paris, 75006 Paris, France;
- Centre de Référence Maladies Rares, Mucoviscidose et Maladies de CFTR, Hôpital Necker Enfants Malades, 75015 Paris, France
- Clinical Trial Network, European Cystic Fibrosis Society, BT2 Belfast, Ireland
- European Respiratory Network Lung, 75006 Paris, France
| |
Collapse
|
129
|
Sanchez-Guzman D, Boland S, Brookes O, Mc Cord C, Lai Kuen R, Sirri V, Baeza Squiban A, Devineau S. Long-term evolution of the epithelial cell secretome in preclinical 3D models of the human bronchial epithelium. Sci Rep 2021; 11:6621. [PMID: 33758289 PMCID: PMC7988136 DOI: 10.1038/s41598-021-86037-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/10/2021] [Indexed: 01/31/2023] Open
Abstract
The human bronchial epithelium is the first line of defense against atmospheric particles, pollutants, and respiratory pathogens such as the novel SARS-CoV-2. The epithelial cells form a tight barrier and secrete proteins that are major components of the mucosal immune response. Functional in vitro models of the human lung are essential for screening the epithelial response and assessing the toxicity and barrier crossing of drugs, inhaled particles, and pollutants. However, there is a lack of models to investigate the effect of chronic exposure without resorting to animal testing. Here, we developed a 3D model of the human bronchial epithelium using Calu-3 cell line and demonstrated its viability and functionality for 21 days without subculturing. We investigated the effect of reduced Fetal Bovine Serum supplementation in the basal medium and defined the minimal supplementation needed to maintain a functional epithelium, so that the amount of exogenous serum proteins could be reduced during drug testing. The long-term evolution of the epithelial cell secretome was fully characterized by quantitative mass spectrometry in two preclinical models using Calu-3 or primary NHBE cells. 408 common secreted proteins were identified while significant differences in protein abundance were observed with time, suggesting that 7-10 days are necessary to establish a mature secretome in the Calu-3 model. The associated Reactome pathways highlight the role of the secreted proteins in the immune response of the bronchial epithelium. We suggest this preclinical 3D model can be used to evaluate the long-term toxicity of drugs or particles on the human bronchial epithelium, and subsequently to investigate their effect on the epithelial cell secretions.
Collapse
Affiliation(s)
| | - Sonja Boland
- Université de Paris, BFA, UMR 8251, CNRS, 75013, Paris, France
| | - Oliver Brookes
- Université de Paris, BFA, UMR 8251, CNRS, 75013, Paris, France
| | - Claire Mc Cord
- Université de Paris, BFA, UMR 8251, CNRS, 75013, Paris, France
| | - René Lai Kuen
- Cellular and Molecular Imaging Facility, US25 Inserm-3612 CNRS, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | - Valentina Sirri
- Université de Paris, BFA, UMR 8251, CNRS, 75013, Paris, France
| | | | | |
Collapse
|
130
|
Sun Y, Shi Z, Lin Y, Zhang M, Liu J, Zhu L, Chen Q, Bi J, Li S, Ni Z, Wang X. Benzo(a)pyrene induces MUC5AC expression through the AhR/mitochondrial ROS/ERK pathway in airway epithelial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 210:111857. [PMID: 33421718 DOI: 10.1016/j.ecoenv.2020.111857] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 06/12/2023]
Abstract
OBJECTIVES Benzo(a)pyrene (BaP) is a ubiquitous air pollutants, and BaP exposure leads to a risk of respiratory diseases. The oversecretion of airway mucus and high expression of mucin 5AC (MUC5AC) are associated with common respiratory disorders caused by air pollution. We aimed to investigate the effect of BaP on MUC5AC expression, especially the mechanisms by which BaP induces MUC5AC gene expression. METHODS The human airway epithelial cell NCI-H292 was used to test the effects of BaP on the expression of MUC5AC in vitro. MUC5AC mRNA and protein expression were assessed with real-time quantitative PCR, immunochemistry, and western blotting. A luciferase assay was conducted to detect the activity of the promoter. The total cellular ROS and mitochondrial ROS were measured by corresponding probes. Small-interfering RNAs were used for gene silencing. AhR-overexpressing cell lines were constructed by transfection with AhR overexpression lentivirus. RESULTS We found that BaP stimulation upregulated the MUC5AC mRNA and protein levels and activated the ERK pathway. Suppressing ERK with U0126 (an ERK inhibitor) or knocking down ERK with siRNA decreased BaP-induced MUC5AC expression. The luciferase activity transfected with the MUC5AC promoter and cAMP-response element (CRE) was increased after BaP treatment, whereas CREB siRNA suppressed the BaP-induced overexpression of MUC5AC. In addition, BaP increased mitochondrial ROS production, and Mito-TEMP, a mitochondrial ROS inhibitor, inhibited BaP-induced MUC5AC expression and ERK activation. BaP increased the mRNA levels of CYP1A1 and CYP1B1, while Alizarin, a CYP1s inhibitor, suppressed the effects of BaP, including the MUC5AC overexpression, ERK activation and mitochondrial ROS generation. BaP induced the translocation of aryl hydrocarbon receptor (AhR) from the cytoplasm to the nucleus. SiRNA-mediated knockdown or chemical inhibition of AhR decreased the BaP-induced expression of MUC5AC, while the overexpression of AhR significantly enhanced the BaP-induced expression of MUC5AC. ITE, an endogenous AhR ligand, also upregulated the mRNA and protein expression of MUC5AC. Furthermore, resveratrol treatment inhibited the BaP-induced MUC5AC overexpression, AhR translocation, mitochondrial ROS production and ERK pathway activation. CONCLUSION Here, we highlighted the crucial role of AhR/mitochondrial ROS/ERK pathway activation in BaP-induced MUC5AC overexpression and identified resveratrol as a promising drug to reduce BaP-induced MUC5AC overexpression.
Collapse
Affiliation(s)
- Yipeng Sun
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China; Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zhaowen Shi
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Yuhua Lin
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Mengzhe Zhang
- Department of Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Jinjin Liu
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Linyun Zhu
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Qingge Chen
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Junjie Bi
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Shanqun Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zhenhua Ni
- Central lab, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China.
| | - Xiongbiao Wang
- Department of Respiratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China.
| |
Collapse
|
131
|
Zangari T, Ortigoza MB, Lokken-Toyli KL, Weiser JN. Type I Interferon Signaling Is a Common Factor Driving Streptococcus pneumoniae and Influenza A Virus Shedding and Transmission. mBio 2021; 12:e03589-20. [PMID: 33593970 PMCID: PMC8545127 DOI: 10.1128/mbio.03589-20] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 01/27/2023] Open
Abstract
The dynamics underlying respiratory contagion (the transmission of infectious agents from the airways) are poorly understood. We investigated host factors involved in the transmission of the leading respiratory pathogen Streptococcus pneumoniae Using an infant mouse model, we examined whether S. pneumoniae triggers inflammatory pathways shared by influenza A virus (IAV) to promote nasal secretions and shedding from the upper respiratory tract to facilitate transit to new hosts. Here, we show that amplification of the type I interferon (IFN-I) response is a critical host factor in this process, as shedding and transmission by both IAV and S. pneumoniae were decreased in pups lacking the common IFN-I receptor (Ifnar1-/- mice). Additionally, providing exogenous recombinant IFN-I to S. pneumoniae-infected pups was sufficient to increase bacterial shedding. The expression of IFN-stimulated genes (ISGs) was upregulated in S. pneumoniae-infected wild-type (WT) but not Ifnar1-/- mice, including genes involved in mucin type O-glycan biosynthesis; this correlated with an increase in secretions in S. pneumoniae- and IAV-infected WT compared to Ifnar1-/- pups. S. pneumoniae stimulation of ISGs was largely dependent on its pore-forming toxin, pneumolysin, and coinfection with IAV and S. pneumoniae resulted in synergistic increases in ISG expression. We conclude that the induction of IFN-I signaling appears to be a common factor driving viral and bacterial respiratory contagion.IMPORTANCE Respiratory tract infections are a leading cause of childhood mortality and, globally, Streptococcus pneumoniae is the leading cause of mortality due to pneumonia. Transmission of S. pneumoniae primarily occurs through direct contact with respiratory secretions, although the host and bacterial factors underlying transmission are poorly understood. We examined transmission dynamics of S. pneumoniae in an infant mouse model and here show that S. pneumoniae colonization of the upper respiratory tract stimulates host inflammatory pathways commonly associated with viral infections. Amplification of this response was shown to be a critical host factor driving shedding and transmission of both S. pneumoniae and influenza A virus, with infection stimulating expression of a wide variety of genes, including those involved in the biosynthesis of mucin, a major component of respiratory secretions. Our findings suggest a mechanism facilitating S. pneumoniae contagion that is shared by viral infection.
Collapse
Affiliation(s)
- Tonia Zangari
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Mila B Ortigoza
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Medicine, Division of Infectious Diseases, New York University Grossman School of Medicine, New York, New York, USA
| | - Kristen L Lokken-Toyli
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Jeffrey N Weiser
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
132
|
Sponchiado M, Liao YS, Atanasova KR, Collins EN, Schurmann V, Bravo L, Reznikov LR. Overexpression of Substance P in pig airways increases MUC5AC through an NF-kβ pathway. Physiol Rep 2021; 9:e14749. [PMID: 33580593 PMCID: PMC7881348 DOI: 10.14814/phy2.14749] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 01/13/2021] [Indexed: 01/04/2023] Open
Abstract
Substance P (SP) is a tachykinin that regulates airway mucous secretion in both health and disease. Our study aimed to determine whether overexpression of SP without pre‐existing inflammation was sufficient to induce changes in mucin secretion and transport in small airways. Utilizing porcine precision‐cut lung slices, we measured the impact of AAV‐mediated overexpression of SP on airway physiology ex vivo. Immunofluorescence signal intensity for MUC5AC was significantly increased in SP‐overexpressed precision‐cut lung slices compared to GFP controls. No difference in MUC5B signal intensity between treatments was detected. SP‐overexpressed precision‐cut lung slices also exhibited decreased IL10 mRNA, an important inhibitor of mucous cell metaplasia. Overt deficits in mucociliary transport were not noted, though a trend for decreased mean transport speed was detected in methacholine‐challenged airways overexpressing SP compared to GFP controls. Pharmacologic inhibition of the NF‐kβ pathway abrogated the effects of overexpression of SP on both MUC5AC and IL10. Collectively, these data suggest that overexpression of SP in the absence of existing inflammation increases MUC5AC via activation of the NF‐kβ pathway. Thus, these data further highlight SP as a key driver of abnormal mucous secretion and underscore NF‐kβ signaling as a pathway of potential therapeutic intervention.
Collapse
Affiliation(s)
- Mariana Sponchiado
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Yan-Shin Liao
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Kalina R Atanasova
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA.,Department of Medicinal Chemistry and Center for Natural Products, Drug Discovery and Development, University of Florida, Gainesville, FL, USA
| | - Emily N Collins
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Veronica Schurmann
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Laura Bravo
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Leah R Reznikov
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| |
Collapse
|
133
|
Bastola R, Young PM, Das SC. Simulation of respiratory tract lining fluid for in vitro dissolution study. Expert Opin Drug Deliv 2021; 18:1091-1100. [PMID: 33504235 DOI: 10.1080/17425247.2021.1882991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Drug particles inhaled via the respiratory system must first dissolve in the respiratory tract lining fluid (RTLF) that lies on the surfaces of airways and alveoli, so that they are absorbed and have therapeutic action. Artificial simulated RTLFs are often used for in vitro dissolution studies to determine the solubility and dissolution of inhaled drug particles. Such studies can be used to predict bioavailability minimizing the requirement for in vivo studies. Numerous studies have been conducted to develop bio-relevant simulated RTLFs; however, to date, there is no singular simulated RTLF that closely resembles human RTLF.Areas covered: This review focuses on the composition of natural and simulated RTLFs and their use in in vitro dissolution studies.Expert opinion: There is variation in the composition and thickness of RTLF along the respiratory tract. Identification of the actual concentration of components of endogenous RTLF present in different areas of the respiratory tract helps in the development of region-specific simulated RTLFs. It is recommended that region-specific simulated RTLFs can be prepared by varying concentration of major RTLF components like mucus/gel simulants, lipids/surfactants, peptides/proteins, and inorganic/organic salts.
Collapse
Affiliation(s)
- Rakesh Bastola
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Paul M Young
- Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, Glebe, Australia
| | - Shyamal C Das
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
134
|
Lee SU, Kim MO, Kang MJ, Oh ES, Ro H, Lee RW, Song YN, Jung S, Lee JW, Lee SY, Bae T, Hong ST, Kim TD. Transforming Growth Factor β Inhibits MUC5AC Expression by Smad3/HDAC2 Complex Formation and NF-κB Deacetylation at K310 in NCI-H292 Cells. Mol Cells 2021; 44:38-49. [PMID: 33510050 PMCID: PMC7854180 DOI: 10.14348/molcells.2020.0188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/29/2020] [Accepted: 01/12/2021] [Indexed: 01/17/2023] Open
Abstract
Airway mucus secretion is an essential innate immune response for host protection. However, overproduction and hypersecretion of mucus, mainly composed of the gel- forming MUC5AC protein, are significant risk factors for patients with asthma and chronic obstructive pulmonary disease (COPD). The transforming growth factor β (TGFβ) signaling pathway negatively regulates MUC5AC expression; however, the underlying molecular mechanism is not fully understood. Here, we showed that TGFβ significantly reduces the expression of MUC5AC mRNA and its protein in NCI-H292 cells, a human mucoepidermoid carcinoma cell line. This reduced MUC5AC expression was restored by a TGFβ receptor inhibitor (SB431542), but not by the inhibition of NF-κB (BAY11-7082 or Triptolide) or PI3K (LY294002) activities. TGFβ-activated Smad3 dose-dependently bound to MUC5AC promoter. Notably, TGFβ-activated Smad3 recruited HDAC2 and facilitated nuclear translocation of HDAC2, thereby inducing the deacetylation of NF-κB at K310, which is essential for a reduction in NF-κB transcriptional activity. Both TGFβ-induced nuclear translocation of Smad3/HDAC2 and deacetylation of NF-κB at K310 were suppressed by a Smad3 inhibitor (SIS3). These results suggest that the TGFβ-activated Smad3/HDAC2 complex is an essential negative regulator for MUC5AC expression and an epigenetic regulator for NF-κB acetylation. Therefore, these results collectively suggest that modulation of the TGFβ1/Smad3/HDAC2/NF-κB pathway axis can be a promising way to improve lung function as a treatment strategy for asthma and COPD.
Collapse
Affiliation(s)
- Su Ui Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Mun-Ock Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Myung-Ji Kang
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Eun Sol Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea
| | - Ro Woon Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea
| | - Yu Na Song
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea
| | - Sunin Jung
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Jae-Won Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Soo Yun Lee
- Immunotherapy Research Center, KRIBB, Daejeon 34141, Korea
| | - Taeyeol Bae
- Immunotherapy Research Center, KRIBB, Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Sung-Tae Hong
- Department of Anatomy & Cell Biology, Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
- Chungnam National University Hospital, Daejeon 35015, Korea
| | - Tae-Don Kim
- Immunotherapy Research Center, KRIBB, Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| |
Collapse
|
135
|
Adivitiya, Kaushik MS, Chakraborty S, Veleri S, Kateriya S. Mucociliary Respiratory Epithelium Integrity in Molecular Defense and Susceptibility to Pulmonary Viral Infections. BIOLOGY 2021; 10:95. [PMID: 33572760 PMCID: PMC7911113 DOI: 10.3390/biology10020095] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 01/08/2023]
Abstract
Mucociliary defense, mediated by the ciliated and goblet cells, is fundamental to respiratory fitness. The concerted action of ciliary movement on the respiratory epithelial surface and the pathogen entrapment function of mucus help to maintain healthy airways. Consequently, genetic or acquired defects in lung defense elicit respiratory diseases and secondary microbial infections that inflict damage on pulmonary function and may even be fatal. Individuals living with chronic and acute respiratory diseases are more susceptible to develop severe coronavirus disease-19 (COVID-19) illness and hence should be proficiently managed. In light of the prevailing pandemic, we review the current understanding of the respiratory system and its molecular components with a major focus on the pathophysiology arising due to collapsed respiratory epithelium integrity such as abnormal ciliary movement, cilia loss and dysfunction, ciliated cell destruction, and changes in mucus rheology. The review includes protein interaction networks of coronavirus infection-manifested implications on the molecular machinery that regulates mucociliary clearance. We also provide an insight into the alteration of the transcriptional networks of genes in the nasopharynx associated with the mucociliary clearance apparatus in humans upon infection by severe acute respiratory syndrome coronavirus-2.
Collapse
Affiliation(s)
- Adivitiya
- Laboratory of Optobiology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India; (A.); (M.S.K.); (S.C.)
| | - Manish Singh Kaushik
- Laboratory of Optobiology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India; (A.); (M.S.K.); (S.C.)
| | - Soura Chakraborty
- Laboratory of Optobiology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India; (A.); (M.S.K.); (S.C.)
| | - Shobi Veleri
- Drug Safety Division, ICMR-National Institute of Nutrition, Hyderabad 500007, India;
| | - Suneel Kateriya
- Laboratory of Optobiology, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India; (A.); (M.S.K.); (S.C.)
| |
Collapse
|
136
|
Liu D, Qian T, Sun S, Jiang JJ. Laryngopharyngeal Reflux and Inflammatory Responses in Mucosal Barrier Dysfunction of the Upper Aerodigestive Tract. J Inflamm Res 2021; 13:1291-1304. [PMID: 33447069 PMCID: PMC7801919 DOI: 10.2147/jir.s282809] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/16/2020] [Indexed: 12/29/2022] Open
Abstract
The upper aerodigestive tract (UAT) is the first line of defense against environmental stresses such as antigens, microbes, inhalants, foods, etc., and mucins, intracellular junctions, epithelial cells, and immune cells are the major constituents of this defensive mucosal barrier. Laryngopharyngeal reflux (LPR) is recognized as an independent risk factor for UAT mucosal disorders, and in this review, we describe the components and functions of the mucosal barrier and the results of LPR-induced mucosal inflammation in the UAT. We discuss the interactions between the refluxate and the mucosal components and the mechanisms through which these damaging events disrupt and alter the mucosal barriers. In addition, we discuss the dynamic alterations in the mucosal barrier that might be potential therapeutic targets for LPR-induced disorders.
Collapse
Affiliation(s)
- Danling Liu
- Otorhinolaryngology Department, ENT Institute, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai 200032, People's Republic of China
| | - Tingting Qian
- Otorhinolaryngology Department, ENT Institute, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai 200032, People's Republic of China
| | - Shan Sun
- Otorhinolaryngology Department, ENT Institute, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai 200032, People's Republic of China
| | - Jack J Jiang
- Otorhinolaryngology Department, ENT Institute, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai 200032, People's Republic of China.,Department of Surgery, Division of Otolaryngology Head and Neck Surgery, University of Wisconsin Medical School, Madison, WI 53792-7375, USA
| |
Collapse
|
137
|
Ishibashi J, Saito K, Ishizaki T, Horie I, Isohama Y. Ibudilast Suppresses MUC5AC Mucus Production through Inhibition of ERK1/2 Phosphorylation. Biol Pharm Bull 2021; 44:404-409. [PMID: 33642548 DOI: 10.1248/bpb.b20-00798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mucus hypersecretion is a hallmark of respiratory diseases, and excess airway mucus can worsen these conditions. Therefore, it is important to control the production of airway mucus in the treatment of respiratory diseases. The phosphodiesterase inhibitor ibudilast has been reported to be effective in treating sputum and postnasal drip in patients with chronic airway inflammation. On the basis of the hypothesis that ibudilast could inhibit mucus production in the airway, in the present study, we examined the effects of ibudilast on the production of MUC5AC, a major protein component of mucus. In in vitro studies using NCI-H292 cells, ibudilast suppressed MUC5AC production induced by various stimuli. In addition, ibudilast inhibited extracellular signal-regulated kinase (ERK)1/2 phosphorylation and MUC5AC gene transcription. Furthermore, it attenuated MUC5AC production and Muc5ac mRNA expression in lipopolysaccharide-treated mice in vivo. Collectively, these findings demonstrate that ibudilast has an inhibitory effect on mucus production, which could at least partly be attributed to the inhibition of ERK1/2 phosphorylation and the repression of MUC5AC gene transcription.
Collapse
Affiliation(s)
- Jumpei Ishibashi
- Laboratory of Applied Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Kana Saito
- Laboratory of Applied Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Takako Ishizaki
- Laboratory of Applied Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Ichiro Horie
- Laboratory for Systems Immunology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University
| | - Yoichiro Isohama
- Laboratory of Applied Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| |
Collapse
|
138
|
Ra SW, Kim SY, Lim YY, Park SJ, Rhee CK, Kim DK, Park YB, Lee CY, Yoon HK, Park JW, Yoo KH. The safety and efficacy of CKD-497 in patients with acute upper respiratory tract infection and bronchitis symptoms: a multicenter, double-blind, double-dummy, randomized, controlled, phase II clinical trial. J Thorac Dis 2021; 13:1-9. [PMID: 33569179 PMCID: PMC7867795 DOI: 10.21037/jtd-20-1567] [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: 05/22/2020] [Accepted: 10/22/2020] [Indexed: 11/06/2022]
Abstract
BACKGROUND Acute upper respiratory tract infection (AURI) together with acute bronchitis is the most common illness worldwide. Botanical medicines used as expectorants and antitussives have proven to be effective while also having excellent safety margins. We aimed at evaluating the efficacy and safety of a new botanical drug, CKD-497, in patients with AURI and acute bronchitis. METHODS In this phase 2 study, 225 patients were enrolled and randomly assigned to one of four treatment groups: placebo (n=55), Synatura® (n=49), CKD-497 200 mg (n=68), or CKD-497 300 mg (n=53). The study drugs were administered three times daily over the course of 7 days. Primary endpoint was the change in the bronchitis severity score (BSS) from baseline to day 7. Secondary endpoint was evaluated based on clinical response rates on days 4 and 7. A safety analysis was also performed. RESULTS Between baseline and day 7, the mean BSS scores decreased significantly in each group (P<0.001): -4.04±1.85, -4.31±1.47, -4.09±1.48, and -4.28±1.69. However, neither the CKD-497 nor Synatura® group showed any significant effect on the difference in BSS change (P=0.75). The rate of clinical response was higher in the CKD-497 300 mg group as compared to the placebo only on day 4 (36% vs. 18%; P<0.05) and those having more severe bronchitis (phlegm score ≥3) showed a significant reduction of total BSS in the Synatura® and CKD-497 groups (P=0.042). No significant adverse events were observed in either of the CKD-497 groups. CONCLUSIONS CKD-497 and even the positive control drug had no significant effect on BSS change in this phase 2 clinical trial. However, CKD-497 300 mg had a mild but significant clinical improvement in early bronchitis patients with more severe phlegm. Considering both efficacy and safety, a future study using 300 mg of CKD-497 with a shorter-term endpoint is warranted in patients with more severe bronchitis symptoms.
Collapse
Affiliation(s)
- Seung Won Ra
- Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Sun Young Kim
- Department of Botanical Drug, Chong Kun Dang (CKD) Pharm Research Institute, Yongin-si, Gyeonggi-do, Korea
| | - Yun Young Lim
- Department of Botanical Drug, Chong Kun Dang (CKD) Pharm Research Institute, Yongin-si, Gyeonggi-do, Korea
| | - Shin Jung Park
- Department of Botanical Drug, Chong Kun Dang (CKD) Pharm Research Institute, Yongin-si, Gyeonggi-do, Korea
| | - Chin Kook Rhee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Deog Kyeom Kim
- Department of Internal Medicine, Seoul National University Seoul Metropolitan Government Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Bum Park
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Hallym University Kangdong Sacred Heart Hospital, Hallym University Medical Center, Seoul, Korea
| | - Chang Youl Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chuncheon Sacred Heart Hospital, Hallym University Medical Center, Chuncheon, Korea
| | - Hyoung Kyu Yoon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yeouido St Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jeong-Woong Park
- Division of Pulmonology and Allergy, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Korea
| | - Kwang Ha Yoo
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
139
|
Miya C, Cueno ME, Suzuki R, Maruoka S, Gon Y, Kaneko T, Yonehara Y, Imai K. Porphyromonas gingivalis gingipains potentially affect MUC5AC gene expression and protein levels in respiratory epithelial cells. FEBS Open Bio 2020; 11:446-455. [PMID: 33332733 PMCID: PMC7876492 DOI: 10.1002/2211-5463.13066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/02/2020] [Accepted: 12/15/2020] [Indexed: 01/08/2023] Open
Abstract
Porphyromonas gingivalis (Pg) is a periodontopathic pathogen that may affect MUC5AC‐related mucus hypersecretion along airway epithelial cells. Here, we attempted to establish whether Pg virulence factors (lipopolysaccharide, FimA fimbriae, gingipains) affect MUC5AC in immortalized and primary bronchial cells. We report that MUC5AC gene expression and protein levels are affected by Pg culture supernatant, but not by lipopolysaccharide or FimA fimbriae. Cells treated with either Pg single (Kgp or Rgp) or double (Kgp/Rgp) mutants had altered levels of MUC5AC gene expression and protein levels, and MUC5AC staining of double mutant‐treated mouse lung cells showed that MUC5AC protein levels were unaffected. Taken together, we propose that Pg gingipains may be the primary virulence factor that influences both MUC5AC gene expression and protein levels.
Collapse
Affiliation(s)
- Chihiro Miya
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan.,Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Marni E Cueno
- Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Ryuta Suzuki
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan.,Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Shuichiro Maruoka
- Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Yasuhiro Gon
- Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Tadayoshi Kaneko
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan
| | - Yoshiyuki Yonehara
- Department of Oral and Maxillofacial Surgery II, Nihon University School of Dentistry, Tokyo, Japan
| | - Kenichi Imai
- Department of Microbiology, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
140
|
A bird eye view on cystic fibrosis: An underestimated multifaceted chronic disorder. Life Sci 2020; 268:118959. [PMID: 33383045 DOI: 10.1016/j.lfs.2020.118959] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 01/19/2023]
Abstract
Cystic fibrosis (CF) is an autosomal recessive disease which involves the mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. CF involves in the inflammatory processes and is considered as a multisystem disorder that is not confined to lungs, but it also affects other vital organs that leads to numerous co-morbidities. The respiratory disorder in the CF results in mortality and morbidity which is characterized by series of serious events involving mucus hypersecretion, microbial infections, airways obstruction, inflammation, destruction of epithelium, tissue remodeling and terminal lung diseases. Mucins are the high molecular weight glycoproteins important for the viscoelastic properties of the mucus, play a significant role in the disease mechanisms. Determining the functional association between the CFTR and mucins might help to identify the putative target for specific therapeutic approach. In fact, furin enzyme which helps in the entry of novel COVID-19 virus into the cell, is upregulated in CF and this can also serve as a potential target for CF treatment. Moreover, the use of nano-formulations for CF treatment is an area of research being widely studied as they have also demonstrated promising outcomes. The in-depth knowledge of non-coding RNAs like miRNAs and lncRNAs and their functional association with CFTR gene expression and mutation can provide a different range of opportunity to identify the promising therapeutic approaches for CF.
Collapse
|
141
|
Autoantibodies against tumor-associated antigens in sputum as biomarkers for lung cancer. Transl Oncol 2020; 14:100991. [PMID: 33333369 PMCID: PMC7736713 DOI: 10.1016/j.tranon.2020.100991] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/28/2022] Open
Abstract
Tumor antigens (TAs) can initiate host immune responses and produce TA-associated autoantibody (TAAbs), potential cancer biomarkers. Sputum is directly generated from the upper and lower airways, and thus can be used as a surrogate sample for the diagnosis of lung cancer based on molecular analysis. To develop sputum TAAb biomarkers for the early detection of lung cancer, the leading cause of cancer death, we probed a protein microarray containing more than 9,000 antigens with sputum supernatants of a discovery set of 30 lung cancer patients and 30 cancer-free smokers. Twenty-eight TAs with higher reactivity in sputum of lung cancer cases vs. controls were identified. The diagnostic significance of TAAbs against the TAs was determined by enzyme-linked immunosorbent assays (ELISAs) in sputum of the discovery set and additional 166 lung cancer patients and 213 cancer-free smokers (validation set). Three sputum TAAbs against DDX6, ENO1, and 14-3-3ζ were developed as a biomarker panel with 81% sensitivity and 83% specificity for diagnosis of lung cancer, regardless of stages, locations, and histological types of lung tumors. This study provides the first evidence that sputum TAAbs could be used as biomarkers for the early detection of lung cancer.
Collapse
|
142
|
Wu X, Yao C, Kong J, Tian Y, Fan Y, Zhang Z, Han J, Wu S. Molecular mechanism underlying miR‑130b‑Sp1 transcriptional regulation in LPS‑induced upregulation of MUC5AC in the bile duct epithelium. Mol Med Rep 2020; 23:106. [PMID: 33300069 PMCID: PMC7723072 DOI: 10.3892/mmr.2020.11745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 10/16/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatolithiasis is a common disease that represents a serious health threat to the Chinese population. The pathological mechanism underlying hepatolithiasis is closely related to bacterial infections of the intrahepatic bile duct, followed by chronic inflammation and the overexpression of mucin 5AC (MUC5AC). However, the exact mechanism responsible for the lipopolysaccharide (LPS)-induced upregulation of MUC5AC has yet to be elucidated. Specificity protein 1 (Sp1) is a ubiquitous transcription factor that plays a vital role in the regulation of a number of genes that are responsible for normal cellular function. microRNA (miR/miRNA)-130b is a member of the miRNA family. miRNAs can bind to the 3′-untralsated region (3′-UTR) of a target gene and influence its expression levels. The present study found that LPS increases the expression of MUC5AC by influencing Sp1 secretion. Chromatin immunoprecipitation-quantitative PCR experiments further verified three Sp1 binding sites in the MUC5AC promoter sequence that can regulate the expression of MUC5AC. Further analysis demonstrated that Sp1 expression was regulated by miR-130b. Luciferase experiments identified one miR-130b binding site in the Sp1 3′-UTR region. In vivo experiments also confirmed the role of the miR-130b-Sp1-MUC5AC signaling pathway in the formation of biliary stones and indicated that this pathway may provide targeted therapeutic strategies for the treatment of intrahepatic bile duct stones.
Collapse
Affiliation(s)
- Xiaodong Wu
- Department of Secondary General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Chenhui Yao
- Department of Secondary General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jing Kong
- Department of Secondary General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yu Tian
- Department of Secondary General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Ying Fan
- Department of Secondary General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Zhen Zhang
- Department of Secondary General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jinyan Han
- Department of Secondary General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Shuodong Wu
- Department of Secondary General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
143
|
Zhang Y, Song A, Liu J, Dai J, Lin J. Therapeutic effect of nebulized hypertonic saline for muco-obstructive lung diseases: a systematic review and meta-analysis with trial sequential analysis. J Investig Med 2020; 69:742-748. [PMID: 33272932 DOI: 10.1136/jim-2020-001479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2020] [Indexed: 11/04/2022]
Abstract
Overproduction of mucus and impaired clearance play important roles in the pathogenesis of muco-obstructive lung diseases (MOLDs). This study aims to evaluate the therapeutic effect and safety of nebulized hypertonic saline (HS) on MOLDs. Five electronic databases including PubMed, Excerpt Medica Database (EMBASE), Cochrane Central Register of Controlled Trials, ClinicalTrials.gov and International Standard Randomized Controlled Trial Number Register were searched until June 2019. Randomized controlled trials or randomized controlled crossover trials which investigated the therapeutic effect of HS versus non-HS for MOLDs were included. Twenty-one studies met the eligibility criteria. For cystic fibrosis (CF), although the forced expiratory volume in the first second and forced vital capacity did not improve significantly (mean difference (MD) -0.48, 95% CI -3.72 to 2.76), (MD 1.85, 95% CI -4.31 to 8.01), respectively), the clearance capability of lung and quality of life (QOL) improved significantly in the HS group ((standard mean difference 0.44, 95% CI 0.02 to 0.87), (MD -0.64, 95% CI -)1.14, to 0.13), respectively). However, the results of trial sequential analysis showed the evidence needed more researches to support. The effect of nebulized HS on non-CF bronchiectasis, chronic obstructive pulmonary disease, and primary ciliary dyskinesia also need more evidence to conclude, since current studies are limited and results are inconsistent. Most adverse events of nebulized HS were mild and transient. In summary, the current available evidence suggests that nebulized HS may increase the QOL in CF, but there was no significant improvement in lung function. However, it is not possible to draw firm conclusions for other MOLDs due to limited data.
Collapse
Affiliation(s)
- Yin Zhang
- Department of Respiratory Disease, Chongqing Medical University Affiliated Children's Hospital, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Anchao Song
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Jingyue Liu
- Department of Respiratory Disease, Chongqing Medical University Affiliated Children's Hospital, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jihong Dai
- Department of Respiratory Disease, Chongqing Medical University Affiliated Children's Hospital, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jilei Lin
- Department of Respiratory Disease, Chongqing Medical University Affiliated Children's Hospital, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
144
|
Deng F, Wu S, Wu Y, Liu X, Wu P, Zhai Z. Identification of mucins and their expression in the vector mosquito Aedes albopictus. JOURNAL OF VECTOR ECOLOGY : JOURNAL OF THE SOCIETY FOR VECTOR ECOLOGY 2020; 45:297-305. [PMID: 33207050 DOI: 10.1111/jvec.12400] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/04/2020] [Indexed: 06/11/2023]
Abstract
Mucins, the main structural components of vertebrate respiratory, digestive and reproductive tract mucus, as well as insect peritrophic matrix, play important roles in protecting host cells from invading microbes and difficult external environments. Mucins are characterized by highly glycosylated proteins constituting the mucin domain that is rich in repetitive sequences of threonine, serine, and proline (PTS). Despite potential important roles, mosquito mucins remain largely uncharacterized. Here, we performed bioinformatics analyses to identify proteins with PTS repeat domain and predicted 43 mucins or mucin-related proteins in Aedes albopictus. Gene expression analysis revealed that these mucins are dynamically expressed across different development stages and in different organs of Aedes albopictus. Of note, blood feeding upregulated AALF016448 and AALF013291 expression in the midgut, fat body, and ovary, raising the possibility that these mucins play potential roles in reproduction, digestion, and intestinal defense against invading pathogens upon blood feeding. Our in silico identification, followed by expressional validation, thus established a valuable resource for further dissecting the functions of mucins for vector control.
Collapse
Affiliation(s)
- Fangqing Deng
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Science, Hunan Normal University, Changsha, 410081, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory for Microbial Molecular Biology, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Si Wu
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Science, Hunan Normal University, Changsha, 410081, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory for Microbial Molecular Biology, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yan Wu
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Science, Hunan Normal University, Changsha, 410081, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory for Microbial Molecular Biology, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Xinyi Liu
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Science, Hunan Normal University, Changsha, 410081, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory for Microbial Molecular Biology, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Pa Wu
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Science, Hunan Normal University, Changsha, 410081, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory for Microbial Molecular Biology, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Zongzhao Zhai
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Science, Hunan Normal University, Changsha, 410081, China
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory for Microbial Molecular Biology, College of Life Science, Hunan Normal University, Changsha, 410081, China
| |
Collapse
|
145
|
Coelho B, Mazzarino L, Pitz HS, Feltrin C, Voytena APL, Coelho DS, Schneider NFZ, Neubert EO, SimÕes CMO, Maraschin M. Development of nanoparticles coated with cassava bagasse pectin (Manihot esculenta Crantz) containing β-carotene for mucoadhesive applications. AN ACAD BRAS CIENC 2020; 92:e20200134. [PMID: 33237141 DOI: 10.1590/0001-3765202020190134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 05/04/2020] [Indexed: 12/17/2022] Open
Abstract
Pectin (PC) extracted from a solid residue from cassava roots (Manihot esculenta Crantz) was used to coat nanoparticles (NP) containing β-carotene (BC) aiming at the gastrointestinal administration of this lipophilic nutraceutical. The NP were prepared by spontaneous emulsification method using food grade components. Pectin-coated NP have been successfully prepared as confirmed by the increased particle size and negative surface charges due to the pectin's anionic nature. NP showed spherical shape and monodisperse distribution, with a mean size of 21.3 nm (polydispersity index (PDI) 0.29) for BC PC T80-NP (nanoparticle with β-carotene, pectin and Tween 80) and 261.4 nm (PDI 0.1) for BC PC T20-NP (nanoparticle with β-carotene, pectin and Tween 20). BC was encapsulated at amounts of 530 and 324 µg/ml for BC PC T80-NP and BC PC T20-NP, respectively, with high encapsulation efficiency (> 95%), increasing its antioxidant capacity in vitro, besides no cytotoxic effect. However, only BC PC T20-NP was stable over a 90 days storage period (4°C) and revealed a strong interaction between pectin and mucin. These results suggest that pectin-coated BC PC T20-NP is a promising strategy to improve the bioavailability and permeation of BC for administration through mucosal surfaces.
Collapse
Affiliation(s)
- Bianca Coelho
- Universidade Federal de Santa Catarina, Laboratório de Morfogênese e Bioquímica Vegetal, Rodovia Admar Gonzaga, 1346, Caixa Postal 476, 88034-000 Florianópolis, SC, Brazil.,Universidade Federal de Santa Catarina, Laboratório NanoBioMat, Rodovia Virgílio Várzea, 2600, Saco Grande, 88032-001 Florianópolis, SC, Brazil
| | - LetÍcia Mazzarino
- Universidade Federal de Santa Catarina, Laboratório de Morfogênese e Bioquímica Vegetal, Rodovia Admar Gonzaga, 1346, Caixa Postal 476, 88034-000 Florianópolis, SC, Brazil.,Universidade Federal de Santa Catarina, Laboratório NanoBioMat, Rodovia Virgílio Várzea, 2600, Saco Grande, 88032-001 Florianópolis, SC, Brazil
| | - HeloÍsa S Pitz
- Universidade Federal de Santa Catarina, Laboratório de Morfogênese e Bioquímica Vegetal, Rodovia Admar Gonzaga, 1346, Caixa Postal 476, 88034-000 Florianópolis, SC, Brazil
| | - Clarissa Feltrin
- Universidade Federal de Santa Catarina, Laboratório de Virologia Aplicada, Avenida Professor Henrique da Silva Fontes, 2754, 88040-970 Florianópolis, SC, Brazil
| | - Ana Paula L Voytena
- Universidade Federal de Santa Catarina, Laboratório de Morfogênese e Bioquímica Vegetal, Rodovia Admar Gonzaga, 1346, Caixa Postal 476, 88034-000 Florianópolis, SC, Brazil
| | - Daniela S Coelho
- Universidade Federal de Santa Catarina, Laboratório de Morfogênese e Bioquímica Vegetal, Rodovia Admar Gonzaga, 1346, Caixa Postal 476, 88034-000 Florianópolis, SC, Brazil.,Universidade Federal de Santa Catarina, Laboratório NanoBioMat, Rodovia Virgílio Várzea, 2600, Saco Grande, 88032-001 Florianópolis, SC, Brazil
| | - Naira F Z Schneider
- Universidade Federal de Santa Catarina, Laboratório de Virologia Aplicada, Avenida Professor Henrique da Silva Fontes, 2754, 88040-970 Florianópolis, SC, Brazil
| | - Enilto O Neubert
- Empresa de Pesquisa Agropecuária e Extensão Rural de Santa Catarina (EPAGRI), Estação Experimental de Urussanga, 1563, Rodovia SC 108-Km 353, 88840-000 Urussanga, SC, Brazil
| | - ClÁudia M O SimÕes
- Universidade Federal de Santa Catarina, Laboratório de Virologia Aplicada, Avenida Professor Henrique da Silva Fontes, 2754, 88040-970 Florianópolis, SC, Brazil
| | - Marcelo Maraschin
- Universidade Federal de Santa Catarina, Laboratório de Morfogênese e Bioquímica Vegetal, Rodovia Admar Gonzaga, 1346, Caixa Postal 476, 88034-000 Florianópolis, SC, Brazil.,Universidade Federal de Santa Catarina, Laboratório NanoBioMat, Rodovia Virgílio Várzea, 2600, Saco Grande, 88032-001 Florianópolis, SC, Brazil
| |
Collapse
|
146
|
Hu J, Kang H, Chen H, Yao J, Yi X, Tang W, Wan M. Targeting neutrophil extracellular traps in severe acute pancreatitis treatment. Therap Adv Gastroenterol 2020; 13:1756284820974913. [PMID: 33281940 PMCID: PMC7692350 DOI: 10.1177/1756284820974913] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/26/2020] [Indexed: 02/05/2023] Open
Abstract
Severe acute pancreatitis (SAP) is a critical abdominal disease associated with high death rates. A systemic inflammatory response promotes disease progression, resulting in multiple organ dysfunction. The functions of neutrophils in the pathology of SAP have been presumed traditionally to be activation of chemokine and cytokine cascades accompanying the inflammatory process. Recently, since their discovery, a new type of antimicrobial mechanism, neutrophil extracellular traps (NETs), and their role in SAP, has attracted widespread attention from the scientific community. Significantly different from phagocytosis and degranulation, NETs kill extracellular microorganisms by releasing DNA fibers decorated with granular proteins. In addition to their strong antimicrobial functions, NETs participate in the pathophysiological process of many noninfectious diseases. In SAP, NETs injure normal tissues under inflammatory stress, which is associated with the activation of inflammatory cells, to cause an inflammatory cascade, and SAP products also trigger NET formation. Thus, due to the interaction between NET generation and SAP, a treatment targeting NETs might become a key point in SAP therapy. In this review, we summarize the mechanism of NETs in protecting the host from pathogen invasion, the stimulus that triggers NET formation, organ injury associated with SAP involving NETs, methods to interrupt the harmful effects of NETs, and different therapeutic strategies to preserve the organ function of patients with SAP by targeting NETs.
Collapse
Affiliation(s)
| | | | - Huan Chen
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaqi Yao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaolin Yi
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Wenfu Tang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | | |
Collapse
|
147
|
Kameyama A, Thet Tin WW, Nishijima R, Yamakoshi K. Alteration of mucins in the submandibular gland during aging in mice. Arch Oral Biol 2020; 121:104967. [PMID: 33197804 DOI: 10.1016/j.archoralbio.2020.104967] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/08/2020] [Accepted: 10/23/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Mucins are large glycosylated glycoproteins that are produced in the salivary glands, and their changes may contribute to the development of xerostomia due to aging and the accompanying deterioration of oral hygiene. This study aimed to characterize the changes in the mucins produced in submandibular gland (SMG) during the aging process. METHODS SMG mucins derived from mice of each age were separated using supported molecular matrix electrophoresis (SMME). Subsequently, the membranes were stained with Alcian blue (AB) or blotted with MAL-II lectin. The SMME membranes stained with AB were subjected to densitometric analysis and glycan analysis. The detailed structures of O-glycan were investigated by tandem mass spectrometry (MS/MS). RESULTS The SMG of mice secreted three mucins with different glycan profiles: age-specific mucin, youth-specific mucin, and a mucin expressed throughout life, and the expression patterns of these mucins change during aging. Additionally, age-specific mucin began to be detected at about 12 months of age. A mucin expressed throughout life and age-specific mucin had the same mass of major glycans but different structures. Furthermore, the proportion of mucin glycan species expressed throughout life changed during the aging process, and aging tended to decrease the proportion of fucosylated glycans and increase the proportion of sialoglycans. CONCLUSION There are three secretory mucins with different glycan profiles in the SMG of mice, and their expression patterns change according to the period of the aging process. The proportion of glycan species of mucin expressed throughout life also changes during the aging process.
Collapse
Affiliation(s)
- Akihiko Kameyama
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8565, Japan
| | - Wai Wai Thet Tin
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8565, Japan
| | - Risa Nishijima
- Department of Mechanism of Aging, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Kimi Yamakoshi
- Department of Mechanism of Aging, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan.
| |
Collapse
|
148
|
Linthorst J, Meert W, Hestand MS, Korlach J, Vermeesch JR, Reinders MJT, Holstege H. Extreme enrichment of VNTR-associated polymorphicity in human subtelomeres: genes with most VNTRs are predominantly expressed in the brain. Transl Psychiatry 2020; 10:369. [PMID: 33139705 PMCID: PMC7608644 DOI: 10.1038/s41398-020-01060-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/27/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
The human genome harbors numerous structural variants (SVs) which, due to their repetitive nature, are currently underexplored in short-read whole-genome sequencing approaches. Using single-molecule, real-time (SMRT) long-read sequencing technology in combination with FALCON-Unzip, we generated a de novo assembly of the diploid genome of a 115-year-old Dutch cognitively healthy woman. We combined this assembly with two previously published haploid assemblies (CHM1 and CHM13) and the GRCh38 reference genome to create a compendium of SVs that occur across five independent human haplotypes using the graph-based multi-genome aligner REVEAL. Across these five haplotypes, we detected 31,680 euchromatic SVs (>50 bp). Of these, ~62% were comprised of repetitive sequences with 'variable number tandem repeats' (VNTRs), ~10% were mobile elements (Alu, L1, and SVA), while the remaining variants were inversions and indels. We observed that VNTRs with GC-content >60% and repeat patterns longer than 15 bp were 21-fold enriched in the subtelomeric regions (within 5 Mb of the ends of chromosome arms). VNTR lengths can expand to exceed a critical length which is associated with impaired gene transcription. The genes that contained most VNTRs, of which PTPRN2 and DLGAP2 are the most prominent examples, were found to be predominantly expressed in the brain and associated with a wide variety of neurological disorders. Repeat-induced variation represents a sizeable fraction of the genetic variation in human genomes and should be included in investigations of genetic factors associated with phenotypic traits, specifically those associated with neurological disorders. We make available the long and short-read sequence data of the supercentenarian genome, and a compendium of SVs as identified across 5 human haplotypes.
Collapse
Affiliation(s)
- Jasper Linthorst
- grid.484519.5Department of Clinical Genetics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands ,grid.5292.c0000 0001 2097 4740Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands
| | - Wim Meert
- grid.5596.f0000 0001 0668 7884Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Matthew S. Hestand
- grid.5596.f0000 0001 0668 7884Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Jonas Korlach
- grid.423340.20000 0004 0640 9878Pacific Biosciences, Menlo Park, CA USA
| | | | - Marcel J. T. Reinders
- grid.5292.c0000 0001 2097 4740Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands
| | - Henne Holstege
- Department of Clinical Genetics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands. .,Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands. .,Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| |
Collapse
|
149
|
Kennel C, Gould EA, Larson ED, Salcedo E, Vickery T, Restrepo D, Ramakrishnan VR. Differential Expression of Mucins in Murine Olfactory Versus Respiratory Epithelium. Chem Senses 2020; 44:511-521. [PMID: 31300812 DOI: 10.1093/chemse/bjz046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mucins are a key component of the surface mucus overlying airway epithelium. Given the different functions of the olfactory and respiratory epithelia, we hypothesized that mucins would be differentially expressed between these 2 areas. Secondarily, we evaluated for potential changes in mucin expression with radiation exposure, given the clinical observations of nasal dryness, altered mucus rheology, and smell loss in radiated patients. Immunofluorescence staining was performed to evaluate expression of mucins 1, 2, 5AC, and 5B in nasal respiratory and olfactory epithelia of control mice and 1 week after exposure to 8 Gy of radiation. Mucins 1, 5AC, and 5B exhibited differential expression patterns between olfactory and respiratory epithelium (RE) while mucin 2 showed no difference. In the olfactory epithelium (OE), mucin 1 was located in a lattice-like pattern around gaps corresponding to dendritic knobs of olfactory sensory neurons, whereas in RE it was intermittently expressed by surface goblet cells. Mucin 5AC was expressed by subepithelial glands in both epithelial types but to a higher degree in the OE. Mucin 5B was expressed by submucosal glands in OE and by surface epithelial cells in RE. At 1-week after exposure to single-dose 8 Gy of radiation, no qualitative effects were seen on mucin expression. Our findings demonstrate that murine OE and RE express mucins differently, and characteristic patterns of mucins 1, 5AC, and 5B can be used to define the underlying epithelium. Radiation (8 Gy) does not appear to affect mucin expression at 1 week. LEVEL OF EVIDENCE N/A (Basic Science Research).IACUC-approved study [Protocol 200065].
Collapse
Affiliation(s)
- Christopher Kennel
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Elizabeth A Gould
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA.,Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, CO, USA.,Neuroscience Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Eric D Larson
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ernesto Salcedo
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Thad Vickery
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Diego Restrepo
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA.,Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, CO, USA.,Neuroscience Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Vijay R Ramakrishnan
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, CO, USA.,Rocky Mountain Taste and Smell Center, University of Colorado School of Medicine, Aurora, CO, USA.,Department of Neurosurgery, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
150
|
Li J, Ye Z. The Potential Role and Regulatory Mechanisms of MUC5AC in Chronic Obstructive Pulmonary Disease. Molecules 2020; 25:molecules25194437. [PMID: 32992527 PMCID: PMC7582261 DOI: 10.3390/molecules25194437] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/14/2020] [Accepted: 09/21/2020] [Indexed: 12/15/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is associated with high morbidity and mortality globally. Studies show that airway mucus hypersecretion strongly compromises lung function, leading to frequent hospitalization and mortality, highlighting an urgent need for effective COPD treatments. MUC5AC is known to contribute to severe muco-obstructive lung diseases, worsening COPD pathogenesis. Various pathways are implicated in the aberrant MUC5AC production and secretion MUC5AC. These include signaling pathways associated with mucus-secreting cell differentiation [nuclear factor-κB (NF-κB)and IL-13-STAT6- SAM pointed domain containing E26 transformation-specific transcription factor (SPDEF), as well as epithelial sodium channel (ENaC) and cystic fibrosis transmembrane conductance regulator (CFTR)], and signaling pathways related to mucus transport and excretion-ciliary beat frequency (CBF). Various inhibitors of mucus hypersecretion are in clinical use but have had limited benefits against COPD. Thus, novel therapies targeting airway mucus hypersecretion should be developed for effective management of muco-obstructive lung disease. Here, we systematically review the mechanisms and pathogenesis of airway mucus hypersecretion, with emphasis on multi-target and multi-link intervention strategies for the elucidation of novel inhibitors of airway mucus hypersecretion.
Collapse
Affiliation(s)
- Jingyuan Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China;
| | - Zuguang Ye
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China;
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Correspondence: ; Tel./Fax: +86-10-8425-2805
| |
Collapse
|