1
|
Papi C, Gasparello J, Marzaro G, Macone A, Zurlo M, Di Padua F, Fino P, Agostinelli E, Gambari R, Finotti A. Aged garlic extract major constituent S-1-propenyl-l-cysteine inhibits proinflammatory mRNA expression in bronchial epithelial IB3-1 cells exposed to the BNT162b2 vaccine. Exp Ther Med 2025; 30:153. [PMID: 40529387 PMCID: PMC12171798 DOI: 10.3892/etm.2025.12903] [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: 10/07/2024] [Accepted: 04/15/2025] [Indexed: 06/20/2025] Open
Abstract
A simple experimental model system was developed and validated for the identification and characterization of molecules exhibiting the ability to inhibit the expression of genes activated during the coronavirus disease 2019 (COVID-19) 'cytokine storm' for the present study. Biomolecules derived from herbal medicinal extracts have been proposed as anti-inflammatory strategies for reducing COVID-19 'cytokine storm' and the associated Acute Respiratory Distress Syndrome. Considering this, the present study focused on a major component of Aged Garlic Extract (AGE), S-1-propenylcysteine (S1PC). The human bronchial epithelial IB3-1 cell line was used to upregulate the expression of proinflammatory genes after exposure to the COVID-19 BNT162b2 vaccine. The effects of S1PC were then studied following continuous treatment for 2 days in BNT162b2-exposed IB3-1 cells. The concentrations of S1PC were 1, 5, 10, 25, 50 and 100 µM. GC-MS analysis was performed in order to characterize the S1PC used in the experiments. Reverse-transcription-quantitative PCR and western blotting analysis revealed the accumulation of Spike mRNA and protein in BNT162b2-exposed IB3-1 cells. Subsequently, the effects of S1PC on the several biological and biochemical parameters were analyzed, including cell viability, apoptosis, the NF-κB pathway and the expression of proinflammatory factors. Molecular docking analysis was performed to obtain preliminary information on the putative mechanism(s) of action of S1PC. The results of the present study demonstrate that exposure of epithelial IB3-1 cells to the COVID-19 BNT162b2 vaccine is associated with a sharp increase in the expression of the transcription factor NF-κB and NF-κB-regulated genes, including IL-6, IL-8 and granulocyte-colony stimulation factor 9 (G-CSF). Treatment with S-1-propenyl-l-cysteine (S1PC) was found to reverse the BNT162b2-induced upregulation of NF-κB, IL-6, IL-8 and G-CSF. These effects were not associated with inhibition of cell viability, induction of apoptosis or a decrease of the cell growth rate, as demonstrated by the results based on the analysis of cell number and the proportion of early and late apoptotic cells within the cell population. With respect to possible mechanisms of action, molecular docking and molecular dynamics simulations strongly suggest that S1PC interacts with Toll-like receptor-4, possibly explaining the inhibitory effects on NF-κB and NF-κB-regulated genes. Therefore, S1PC should be further evaluated as a potential inhibitor of this COVID-19 'cytokine storm'. However, further experimental studies are needed to identify other agents that can also able to inhibit gene expression induced by the COVID-19 BNT162b2 vaccine and to verify whether combined treatments with S1PC could be proposed to obtain even superior inhibitory effects.
Collapse
Affiliation(s)
- Chiara Papi
- Department of Life Sciences and Biotechnology, Ferrara University, I-44121 Ferrara, Italy
| | - Jessica Gasparello
- Department of Life Sciences and Biotechnology, Ferrara University, I-44121 Ferrara, Italy
| | - Giovanni Marzaro
- Department of Diagnostics and Public Health, University of Verona, I-37134 Verona, Italy
| | - Alberto Macone
- Department of Biochemical Sciences ‘A. Rossi Fanelli’, Sapienza University of Rome, I-00185 Rome, Italy
| | - Matteo Zurlo
- Department of Life Sciences and Biotechnology, Ferrara University, I-44121 Ferrara, Italy
| | - Federica Di Padua
- Department of Life Sciences and Biotechnology, Ferrara University, I-44121 Ferrara, Italy
| | - Pasquale Fino
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, I-00161 Rome, Italy
| | - Enzo Agostinelli
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, I-00161 Rome, Italy
- International Polyamines Foundation ‘ETS-ONLUS’, I-00159 Rome, Italy
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, Ferrara University, I-44121 Ferrara, Italy
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, Ferrara University, I-44121 Ferrara, Italy
| |
Collapse
|
2
|
Vailionytė A, Uogintė I, Pajarskienė J, Bagdonas E, Jelinskas T, Ignatjev I, Byčenkienė S, Aldonytė R. In vitro effects of aged low-density polyethylene micro(nano)plastic particles on human airway epithelial cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 374:126186. [PMID: 40185180 DOI: 10.1016/j.envpol.2025.126186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/13/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Airborne micro(nano)plastic (MNP) pollution has emerged as a major global concern due to its increasingly worrying adverse health effects. Environmental weathering and UV irradiation of plastic waste, together with tire wear, generate airborne MNPs with irregular shapes and varied sizes, with low-density polyethylene (LDPE) being the predominant plastic type. However, knowledge of MNPs' toxicological effects remains scarce, as current in vitro research mainly focuses on commercial polystyrene beads. In this study, we investigated for the first time the toxicological effects of environmentally relevant aged LDPE MNPs on human bronchial epithelial cells (BEAS-2B). UV-aged LDPE fragments of irregular sizes and shapes were used to mimic real atmospheric particles, and BEAS-2B cells were exposed to 10-1000 μg/cm2 of LDPE MNPs. Our results showed that MNPs were internalized by BEAS-2B cells and promoted epithelial-to-mesenchymal transition (EMT), characterized by reduced β-catenin and increased vimentin expression, enhanced motility, and disturbed cell cycle. Moreover, exposure to aged LDPE MNPs significantly increased intracellular ROS levels and reduced cell proliferation rate at the highest dose. LDPE MNPs triggered oxidative stress in BEAS-2B cells through activation of the NRF2 signaling pathway, with impaired autophagic flux indicated by increased expression of p62 and LC3A/B. Importantly, LDPE MNP exposure significantly increased the secretion of pro-inflammatory mediators (CD62E, CD62P, ICAM-1, IL-6, IL-8), accompanied by suppressive effects on mitochondrial respiration and glycolytic function at 1000 μg/cm2. Taken together, our findings suggest that inhalation of LDPE MNPs could impact the morphology and function of the human airway epithelium and respiratory health in general.
Collapse
Affiliation(s)
- Agnė Vailionytė
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, 08406, Lithuania; Department of Nanoengineering, State Research Institute Center for Physical Sciences and Technology, Vilnius, 02300, Lithuania.
| | - Ieva Uogintė
- Department of Environmental Research, State Research Institute Center for Physical Sciences and Technology, Vilnius, 10257, Lithuania
| | - Justina Pajarskienė
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, 08406, Lithuania
| | - Edvardas Bagdonas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, 08406, Lithuania
| | - Tadas Jelinskas
- Department of Nanoengineering, State Research Institute Center for Physical Sciences and Technology, Vilnius, 02300, Lithuania
| | - Ilja Ignatjev
- Department of Organic Chemistry, State Research Institute Center for Physical Sciences and Technology, Vilnius, 10257, Lithuania
| | - Steigvilė Byčenkienė
- Department of Environmental Research, State Research Institute Center for Physical Sciences and Technology, Vilnius, 10257, Lithuania
| | - Rūta Aldonytė
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, 08406, Lithuania
| |
Collapse
|
3
|
Kopa-Stojak PN, Kleniewska P, Hoffmann A, Pawliczak R. The potential effect of gut microbiota on the secretion of selected cytokines by human cells in vitro. Sci Rep 2025; 15:17367. [PMID: 40389545 PMCID: PMC12089611 DOI: 10.1038/s41598-025-01581-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
Colonization of the gut early in life plays a significant role in guiding the development of the immune system. The effect of individual intestinal bacterial strains on the asthma development is the subject of numerous scientific studies. The aim of the study was to determine the role and the potential mechanism of action of intestinal microflora on secretion of cytokines and potential predisposition to asthma development. The effect of Parabacteroides distasonis (PD), Bacteroides vulgatus (BV) Clostridium perfringens (CP) and Ruminococcus albus (RA) lysates on the secretion of IL-4, IL-5, IL-8 (CXCL8) and IL-13 in human peripheral blood mononuclear cells, monocytes and HT-29 cells has been analyzed by enzyme-linked immunosorbent assays. RA and PD 400 µg lysates significantly increased secretion of IL-5 by PBMC compared to control (p < 0.05). In addition, BV, CP, PD and RA 100 µg lysates significantly increased IL-8 secretion by PBMC, as well as BV, PD and RA 100 µg lysates by monocytes compared to control (p < 0.05). Moreover, PD 100 µg and 400 µg lysates significantly increased secretion of IL-8 by HT-29 cells compared to BV 100 µg and 400 µg lysates (p < 0.05). CP, BV, PD and RA 100 µg lysates significantly lowered IL-13 secretion by PBMC vs. control (p < 0.001). For a better understanding of the mechanisms of action of gut microflora and their impact on parameters important in asthma, complex studies which compare the asthma and control samples should be carried out in the future.
Collapse
Affiliation(s)
- Paulina Natalia Kopa-Stojak
- Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, Zeligowskiego 7/9, (bldg 2 Rm 177), 90-752, Lodz, Poland
| | - Paulina Kleniewska
- Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, Zeligowskiego 7/9, (bldg 2 Rm 177), 90-752, Lodz, Poland
| | - Arkadiusz Hoffmann
- Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, Zeligowskiego 7/9, (bldg 2 Rm 177), 90-752, Lodz, Poland
| | - Rafał Pawliczak
- Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, Zeligowskiego 7/9, (bldg 2 Rm 177), 90-752, Lodz, Poland.
| |
Collapse
|
4
|
Arrigo E, Carriero V, Bertolini F, Levra S, Demasi C, Di Stefano A, Miglietta D, Villetti G, Ricciardolo FLM. Differential Expression of Type 1 and Type 17 Immune Pathways and Chemokines in Mild-to-Severe Asthma With Mid to High Neutrophilic Inflammation. Clin Exp Allergy 2025. [PMID: 40165433 DOI: 10.1111/cea.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/31/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025]
Affiliation(s)
- Elisa Arrigo
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Vitina Carriero
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Francesca Bertolini
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Stefano Levra
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Camilla Demasi
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Antonino Di Stefano
- Divisione di Pneumologia e Laboratorio di Citoimmunopatologia dell'Apparato Cardio Respiratorio, Istituti Clinici Scientifici Maugeri, IRCCS, Respiratory Rehabilitation Unit of Gattico-Veruno, Veruno, Italy
| | - Daniela Miglietta
- Chiesi Farmaceutici S.P.A., Global Research and Preclinical Development, Parma, Italy
| | - Gino Villetti
- Chiesi Farmaceutici S.P.A., Global Research and Preclinical Development, Parma, Italy
| | - Fabio Luigi Massimo Ricciardolo
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- Severe Asthma, Rare Lung Disease and Respiratory Pathophysiology Unit, San Luigi Gonzaga University Hospital, Torino, Italy
- Institute of Translational Pharmacology, National Research Council (IFT-CNR), Section of Palermo, Palermo, Italy
| |
Collapse
|
5
|
Jin YY, Guo Y, Xiong SW, Zhang N, Chen JH, Liu F. BALF editome profiling reveals A-to-I RNA editing associated with severity and complications of Mycoplasma pneumoniae pneumonia in children. mSphere 2025; 10:e0101224. [PMID: 39998235 PMCID: PMC11934315 DOI: 10.1128/msphere.01012-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/09/2025] [Indexed: 02/26/2025] Open
Abstract
Mycoplasma pneumoniae is an important human respiratory pathogen that causes mild-to-moderate community-acquired M. pneumoniae pneumonia (MPP), particularly in children. RNA editing plays a vital role in pathogen infection and host immune response, but it remains largely unknown how it could be involved in the epigenetic regulation of host response to M. pneumoniae infection. In the present study, we performed an epitranscriptomic analysis of adenosine to inosine (A-to-I) editing in 39 bronchoalveolar lavage fluid (BALF) samples from the severe side (SS) and the opposite side (OS) of patients with MPP. Our editome profiling identified 87 differential RNA editing (DRE) events in 50 genes, especially missense editing events that recoded C-C motif chemokine receptor-like 2 (CCRL2, p.K147R) and cyclin I (CCNI, p.R75G). The expression of adenosine deaminase acting on RNA (ADAR) significantly increased on SS compared to OS and positively correlated with the average RNA editing level and individual DRE events. Meanwhile, functional enrichment analysis showed that DRE was observed in genes primarily associated with the negative regulation of innate immune response, type I interferon production, and cytokine production. Further comparison of SS between complicated MPP (CMPP) and non-complicated MPP (NCMPP) revealed RNA editing events associated with MPP complications, with a higher ADAR expression in CMPP than NCMPP. By identifying DRE events as biomarkers associated with MPP severity and complications, our editome profiling provides new insight into the potential role played by A-to-I RNA editing in modulating the host's immune system during M. pneumoniae infection.IMPORTANCEOur research investigates how Mycoplasma pneumoniae, a common respiratory pathogen, influences how our cells modify their genetic instructions. By studying RNA editing changes in bronchoalveolar lavage fluid from patients with M. pneumoniae pneumonia, we aim to investigate how M. pneumoniae infection alters epigenetics and contributes to the disease severity and complications. Understanding such epigenetic alterations not only sheds light on the mechanisms underlying M. pneumoniae infection but also holds potential implications for developing better diagnostic tools and therapies. Ultimately, this work may facilitate the design of more targeted treatments to alleviate the impact of respiratory infections caused by the pathogen. Our findings may also offer broader insights into how microbial infections reshape immune processes, highlighting the importance of RNA editing in host-pathogen interactions.
Collapse
Affiliation(s)
- Yun-Yun Jin
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yun Guo
- Department of Respiratory Medicine & Clinical Allergy Center, Affiliated Children’s Hospital of Jiangnan University (Wuxi Children’s Hospital), Wuxi, Jiangsu, China
| | - Su-Wan Xiong
- Department of Respiratory Medicine & Clinical Allergy Center, Affiliated Children’s Hospital of Jiangnan University (Wuxi Children’s Hospital), Wuxi, Jiangsu, China
| | - Na Zhang
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Department of Ophthalmology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Jian-Huan Chen
- Laboratory of Genomic and Precision Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Joint Primate Research Center for Chronic Diseases, Institute of Zoology of Guangdong Academy of Science, Jiangnan University, Wuxi, Jiangsu, China
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Feng Liu
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
6
|
Hussain MS, Goyal A, Goyal K, S. RJ, Nellore J, Shahwan M, Rekha A, Ali H, Dhanasekaran M, MacLoughlin R, Dua K, Gupta G. Targeting CXCR2 signaling in inflammatory lung diseases: neutrophil-driven inflammation and emerging therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025. [DOI: 10.1007/s00210-025-03970-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 02/20/2025] [Indexed: 05/04/2025]
|
7
|
Lee SM, Ryu HW, Kim HG, Jo YH, Park KJ, Lee SU, Oh ES, Lee SW, Choi S, Li WY, Hwang BY, Oh SR. Anti-Inflammatory Dimeric and Trimeric Flavonoids from the Roots of Pistacia weinmannifolia. JOURNAL OF NATURAL PRODUCTS 2025; 88:314-321. [PMID: 39931765 DOI: 10.1021/acs.jnatprod.4c01009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
As part of an ongoing search for new anti-inflammatory agents from medicinal plants, five new dimeric and trimeric flavonoids (1-5) were isolated from the roots of Pistacia weinmannifolia. The structures of pistachalcone A (1), pistachalcone B (2), pistaflavanone A (3), pistachalcone C (4), and pistachalcone D (5) were elucidated by the analysis of spectroscopic data. The known compounds rhuschalcone II (6), rhuschalcone VI (7), and pauferrol B (8) were also isolated and identified. Our in vitro analysis found that compounds isolated from P. weinmannifolia root extract exert anti-inflammatory effects in phorbol myristate acetate (PMA)-induced NCI-H292 airway epithelial cells by the suppression of expression levels such as interleukin-8 (IL-8) and mucin 5AC (MUC5AC), which are closely related to the pulmonary inflammatory response in the pathogenesis of COPD. Therefore, these dihydrochalcone derivatives may have value as new starting materials for the development of drug candidates against COPD.
Collapse
Affiliation(s)
- Seong Mi Lee
- Natural Product Research Center and Natural Product Central Bank, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Chungcheungbuk-do 28116, Republic of Korea
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Republic of Korea
- Herbal Medicine Research Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Chungju 28159, Korea
| | - Hyung Won Ryu
- Natural Product Research Center and Natural Product Central Bank, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Chungcheungbuk-do 28116, Republic of Korea
| | - Hyoung-Geun Kim
- Natural Product Research Center and Natural Product Central Bank, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Chungcheungbuk-do 28116, Republic of Korea
| | - Yang Hee Jo
- Natural Product Research Center and Natural Product Central Bank, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Chungcheungbuk-do 28116, Republic of Korea
| | - Kyoung Jin Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Su Ui Lee
- Natural Product Research Center and Natural Product Central Bank, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Chungcheungbuk-do 28116, Republic of Korea
| | - Eun Sol Oh
- Natural Product Research Center and Natural Product Central Bank, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Chungcheungbuk-do 28116, Republic of Korea
| | - Sang Woo Lee
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Sangho Choi
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Wan-Yi Li
- Institute of Medicinal Plants, Yunnan Academy of Agricultural Sciences, Kunming 650200, China
| | - Bang Yeon Hwang
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Sei-Ryang Oh
- Natural Product Research Center and Natural Product Central Bank, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Chungcheungbuk-do 28116, Republic of Korea
| |
Collapse
|
8
|
Ivaneev AI, Ermolin MS, Zinovkin RA, Dashkevich AA, Zinovkina LA, Chernyak BV, Fedotov PS. Separation and preparation of nanoparticles of urban dust for biological studies. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:440-449. [PMID: 39648879 DOI: 10.1039/d4ay01592d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Nanoparticles (NPs) of urban dust pose a potential threat to public health. Nevertheless, this issue remains largely unexplored due to a lack of biological research related to these NPs. This may be attributed to the complexity of the separation, characterization, analysis, and subsequent preparation of NPs of urban dust for biological studies. In the present work, the methodology for the separation and preparation of NPs of urban dust for biological assays has been developed. The isolation of NPs from bulk samples of urban dust has been carried out using coiled tube field-flow fractionation, which allows one to recover a fraction of NPs, which is sufficient for further research. The weight of the recovered fraction of NPs was 0.42 ± 15 mg; the mean size of particles in the fraction was 220 nm. Albumin was employed as a stabilizing agent for NPs, a phosphate buffer solution simulated physiological salt concentrations. The isolated NPs were found to contain microorganisms, and a sterilization procedure was therefore applied. A 5 min UV treatment ensured sterilization of the suspension of NPs. Ultrafiltration was used to pre-concentrate NPs of urban dust required for biological studies; the ultrafiltration procedure did not affect the stability of the NPs suspension. The concentrations of toxic elements like Cu, Zn, As, Mo, Cd, Sn, Sb, Hg, Pb and Bi in the obtained NPs were found to be up to 10 times higher than in the bulk dust samples, indicating a potential health threat. The proposed procedure for the separation and preparation of NPs of urban dust can serve as a reliable basis for further biological studies.
Collapse
Affiliation(s)
- Alexandr I Ivaneev
- Vernadsky Institute of Geochemistry and Analytical Chemistry, Russian Academy of Sciences, 19 Kosygin Street, Moscow 119991, Russia
| | - Mikhail S Ermolin
- Vernadsky Institute of Geochemistry and Analytical Chemistry, Russian Academy of Sciences, 19 Kosygin Street, Moscow 119991, Russia
| | - Roman A Zinovkin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Anna A Dashkevich
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Ludmila A Zinovkina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Boris V Chernyak
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Petr S Fedotov
- Vernadsky Institute of Geochemistry and Analytical Chemistry, Russian Academy of Sciences, 19 Kosygin Street, Moscow 119991, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia
| |
Collapse
|
9
|
Barbeta E, Ferrando C, López-Aladid R, Motos A, Bueno-Freire L, Fernández-Barat L, Soler-Comas A, Palomeque A, Gabarrús A, Artigas A, Camprubí-Rimblas M, Li Bassi G, López-Sobrino T, Sandoval E, Toapanta D, Fernández S, Mellado-Artigas R, Zattera L, Vallverdú J, Laffey JG, Ferrer M, Torres A. Association between driving pressure, systemic inflammation and non-pulmonary organ dysfunction in patients with acute respiratory distress syndrome, a prospective pathophysiological study. Anaesth Crit Care Pain Med 2025; 44:101458. [PMID: 39710229 DOI: 10.1016/j.accpm.2024.101458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Driving pressure is thought to determine the effect of low tidal ventilation on survival in patients with acute respiratory distress syndrome. The leading cause of mortality in these patients is non-pulmonary multiorgan dysfunction, which is believed to worsen due to the biological response to mechanical ventilation (biotrauma). Therefore, we aimed to analyze the association between driving pressure, biotrauma, and non-pulmonary multiorgan dysfunction. Additionally, we analyzed this relationship for tidal volume/predicted body weight. METHODS Observational study that included adult patients with acute respiratory distress syndrome undergoing invasive mechanical ventilation admitted to the Hospital Clinic of Barcelona, Spain, between June 2019 and February 2021. We conducted mixed-effects models to assess the effects of driving pressure and tidal volume/predicted body weight on the evolution of 22 log-transformed biomarker variables during the first, third, and fifth days after study enrollment. These 22 systemic biomarkers characterized epithelial and endothelial pulmonary dysfunction, inflammation, and coagulation disorders in the included patients. In the same fashion, the association between driving pressure and non-pulmonary multiorgan dysfunction was evaluated by the non-pulmonary sequential organ failure assessment score (non-pulmonary SOFA) and its associated variables. Finally, we performed mediation analyses to assess whether the relationship between biomarkers and driving pressure was mediated by other ventilator-induced lung injury parameters. RESULTS Thirty-eight patients were included. Driving pressure was independently associated with soluble Receptor for advanced glycation end-products, Interleukin (IL)-8, IL-6, IL-10, IL-17, Interferon-ɣ, Chemokine (C-C motif)-2, Vascular endothelial growth factor, Tissue factor, Protein C, Protein S, and higher dose of norepinephrine. However, this relationship attenuated over time. In contrast, tidal volume/predicted body weight was not associated with any of the 22 biomarkers tested . A concomitant increase in positive end-inspiratory plateau pressure or tidal volume did not mediate the effect of driving pressure on biomarkers. Conversely, the association between compliance of the respiratory system and pulmonary epithelial dysfunction was primarily mediated by driving pressure. CONCLUSIONS Driving pressure, but not tidal volume/predicted body weight, was correlated with epithelial and endothelial pulmonary dysfunction, inflammation, coagulation disorders, and hemodynamic dysfunction. However, this relationship diminished over time.
Collapse
Affiliation(s)
- Enric Barbeta
- Surgical Intensive Care Unit, Hospital Clínic de Barcelona, Barcelona, Spain; CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona (UB), Barcelona, Spain
| | - Carlos Ferrando
- Surgical Intensive Care Unit, Hospital Clínic de Barcelona, Barcelona, Spain; CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona (UB), Barcelona, Spain
| | - Rubén López-Aladid
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona (UB), Barcelona, Spain
| | - Anna Motos
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona (UB), Barcelona, Spain
| | - Letícia Bueno-Freire
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona (UB), Barcelona, Spain
| | - Laia Fernández-Barat
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona (UB), Barcelona, Spain
| | - Alba Soler-Comas
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona (UB), Barcelona, Spain
| | - Andrea Palomeque
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; Respiratory Intensive Care Unit, Pneumology, Respiratory Institute, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Albert Gabarrús
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona (UB), Barcelona, Spain
| | - Antonio Artigas
- Critical Care Center, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Marta Camprubí-Rimblas
- Critical Care Center, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Gianluigi Li Bassi
- Division of Animal Experimentation, Critical Care Research Group, The Prince Charles Hospital, Chermside, Australia
| | - Teresa López-Sobrino
- Cardiovascular Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Elena Sandoval
- Cardiovascular Surgery, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - David Toapanta
- Liver ICU, Liver Unit, Hospital Clinic, Barcelona, Spain
| | - Sara Fernández
- Medical Intensive Care Unit, Hospital Clinic, Barcelona, Spain
| | - Ricard Mellado-Artigas
- Surgical Intensive Care Unit, Hospital Clínic de Barcelona, Barcelona, Spain; CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona (UB), Barcelona, Spain
| | - Luigi Zattera
- Surgical Intensive Care Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Jordi Vallverdú
- Surgical Intensive Care Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - John G Laffey
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland; Anesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland; Regenerative Medicine Institute (REMEDI) at CÚRAM Center for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| | - Miquel Ferrer
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona (UB), Barcelona, Spain; Respiratory Intensive Care Unit, Pneumology, Respiratory Institute, Hospital Clinic of Barcelona, Barcelona, Spain.
| | - Antoni Torres
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; University of Barcelona (UB), Barcelona, Spain; Respiratory Intensive Care Unit, Pneumology, Respiratory Institute, Hospital Clinic of Barcelona, Barcelona, Spain.
| |
Collapse
|
10
|
Flögel L, Kaiser E, Hans MC, Goedicke-Fritz S, Bous M, Abdul-Khaliq H, Poryo M, Zemlin M, Weber R. Immunological characterization of pleural effusions in pediatric patients. Front Immunol 2024; 15:1506073. [PMID: 39737183 PMCID: PMC11682977 DOI: 10.3389/fimmu.2024.1506073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/02/2024] [Indexed: 01/01/2025] Open
Abstract
Background The pleural cavity represents a unique immunological compartment that can mount inflammatory reactions during infections, after surgery and in chronic immunological diseases. The connection between systemic immune reactions in the blood and local immune reactions in pleural effusions remains unclear. This study provides the first comprehensive immunological characterization of paired blood and pleural effusion samples, utilizing combined cell and cytokine analyses in pediatric patients undergoing cardiac surgery. Methods In 30 pediatric patients (median age: 22 months) with pleural effusion after cardiac surgery for congenital heart defects, corresponding peripheral blood and pleural effusion samples were analyzed for their immune response. We used flow cytometry and multiplex immunoassays to quantify 14 T cell subpopulations and 12 T cell associated cytokines in each biosample. Results IL-6, IL-8, IL-10, TNF (p<0.0001) levels were significantly higher in pleural effusion compared to plasma. In contrast, IFN-γ, GM-CSF, IL-17A levels were lower in pleural effusion than in plasma (p ≤ 0.0005). In comparison to peripheral blood, there was a significantly higher proportion of T helper cells 1 (Th1, p=0.0023), T helper cells 17 (Th17, p=0.0334) and memory effector cytotoxic T cells (CD3+CD8+CD45RO+CD62L-, p=0.0449) in pleural effusion and the same trend was observed for memory effector Th cells (CD3+CD4+CD45RO+CD62L-, p=0.0633) and double-negative T cells (CD3+CD4-CD8-) (p=0.1085). Naïve Th cells (CD3+CD4+CD45RO-CD62L+) and naïve cytotoxic T cells (CD3+CD8+CD45RO-CD62L+) were slightly reduced in pleural effusion compared to peripheral blood (not significant). Conclusion Immunological factors in pleural effusions differed significantly from the corresponding blood samples in pediatric patients after cardiac surgery. The results suggest localized production of specific cytokines within the pleural space, while the distribution of other cytokines in pleural effusions appears to be more reflective of the systemic immune response. We found evidence that on the cellular level, the surface marker CD62L may play a key role in navigating T cells between the blood and pleural effusion. This study confirms that the pleural cavity harbors a unique lymphatic compartment, the analysis of which may be useful for both diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Luca Flögel
- Department of General Pediatrics and Neonatology, Saarland University, Campus Homburg, Homburg, Germany
| | - Elisabeth Kaiser
- Department of General Pediatrics and Neonatology, Saarland University, Campus Homburg, Homburg, Germany
| | - Muriel Charlotte Hans
- Department of General Pediatrics and Neonatology, Saarland University, Campus Homburg, Homburg, Germany
| | - Sybelle Goedicke-Fritz
- Department of General Pediatrics and Neonatology, Saarland University, Campus Homburg, Homburg, Germany
| | - Michelle Bous
- Department of General Pediatrics and Neonatology, Saarland University, Campus Homburg, Homburg, Germany
| | - Hashim Abdul-Khaliq
- Department of Pediatric Cardiology, Saarland University Medical Center, Homburg, Germany
| | - Martin Poryo
- Department of Pediatric Cardiology, Saarland University Medical Center, Homburg, Germany
| | - Michael Zemlin
- Department of General Pediatrics and Neonatology, Saarland University, Campus Homburg, Homburg, Germany
| | - Regine Weber
- Department of General Pediatrics and Neonatology, Saarland University, Campus Homburg, Homburg, Germany
| |
Collapse
|
11
|
Zeng T, Liu L, Xu D, Wang T, Wu Y, Qin J, Gao L, Chen M, Li X, Li D, Chen J, Shen Y, Wen F. The Mitochondrial Fusion Promoter M1 Mitigates Cigarette Smoke-Induced Airway Inflammation and Oxidative Stress via the PI3K-AKT Signaling Pathway. Lung 2024; 203:12. [PMID: 39625494 DOI: 10.1007/s00408-024-00766-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/12/2024] [Indexed: 03/17/2025]
Abstract
PURPOSE This study investigated the efficacy and underlying mechanism of the mitochondrial fusion promoter M1 in mitigating cigarette smoking (CS)-induced airway inflammation and oxidative stress both in vitro and in vivo models. METHODS Cigarette smoke extract (CSE)-treated airway epithelial cells (BEAS-2B) and CS-exposed mice were pretreated with M1, followed by the measurement of proinflammatory cytokines, oxidative stress, mitochondrial fusion proteins (MFN2 and OPA1) and fission proteins (DRP1 and MFF). Molecular pathways were elucidated through transcriptomic analysis and Western blotting. RESULTS M1 pretreatment in CSE-treated cells significantly reduced the release of inflammatory cytokines (interleukin (IL)-6, IL-8 and tumor necrosis factor (TNF)-α); reduced malondialdehyde (MDA) and reactive oxygen species (ROS) levels; increased superoxide dismutase (SOD) activity; protected mitochondrial function by increasing the expression of mitochondrial fusion proteins (MFN2 and OPA1) and decreasing the expression of mitochondrial fission proteins (DRP1 and MFF). M1 attenuated CS-induced lung histologic damage and mucus hypersecretion in mice, relieved high oxidative stress and reduced the release of IL-6 and IL-8 in BALF. Similarly, it also protected mitochondrial function by regulating the CS-induced imbalance of mitochondrial dynamic proteins. Transcriptome sequencing and Western blotting showed that M1 inhibited CSE- or CS-induced activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB/AKT) signaling pathway. CONCLUSION M1 plays a protective role in inflammation, oxidative stress and mitochondrial dynamics dysfunction caused by CS by inhibiting the PI3K-AKT signaling pathway; thus, it has therapeutic potential for the treatment of CS-induced airway disorders.
Collapse
Affiliation(s)
- Tingting Zeng
- State Key Laboratory of Biotherapy of China, Division of Pulmonary Diseases, Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lian Liu
- State Key Laboratory of Biotherapy of China, Division of Pulmonary Diseases, Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dan Xu
- State Key Laboratory of Biotherapy of China, Division of Pulmonary Diseases, Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Wang
- State Key Laboratory of Biotherapy of China, Division of Pulmonary Diseases, Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanqiu Wu
- State Key Laboratory of Biotherapy of China, Division of Pulmonary Diseases, Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiangyue Qin
- State Key Laboratory of Biotherapy of China, Division of Pulmonary Diseases, Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lijuan Gao
- State Key Laboratory of Biotherapy of China, Division of Pulmonary Diseases, Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mei Chen
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Xiaohua Li
- Department of Respiratory and Critical Care Medicine, Sixth People's Hospital of Chengdu, Chengdu, 610051, China
| | - Diandian Li
- State Key Laboratory of Biotherapy of China, Division of Pulmonary Diseases, Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jun Chen
- State Key Laboratory of Biotherapy of China, Division of Pulmonary Diseases, Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yongchun Shen
- State Key Laboratory of Biotherapy of China, Division of Pulmonary Diseases, Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Fuqiang Wen
- State Key Laboratory of Biotherapy of China, Division of Pulmonary Diseases, Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
12
|
Norheim KL, Ben Ezra M, Heckenbach I, Andreasson LM, Eriksen LL, Dyhre-Petersen N, Damgaard MV, Berglind M, Pricolo L, Sampson D, Dellinger RW, Sverrild A, Treebak JT, Ditlev SB, Porsbjerg C, Scheibye-Knudsen M. Effect of nicotinamide riboside on airway inflammation in COPD: a randomized, placebo-controlled trial. NATURE AGING 2024; 4:1772-1781. [PMID: 39548320 PMCID: PMC11645284 DOI: 10.1038/s43587-024-00758-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive, incurable disease associated with smoking and advanced age, ranking as the third leading cause of death worldwide. DNA damage and loss of the central metabolite nicotinamide adenine dinucleotide (NAD+) may contribute to both aging and COPD, presenting a potential avenue for interventions. In this randomized, double-blind, placebo-controlled clinical trial, we treated patients with stable COPD (n = 40) with the NAD+ precursor nicotinamide riboside (NR) for 6 weeks and followed-up 12 weeks later. The primary outcome was change in sputum interleukin-8 (IL-8) from baseline to week 6. The estimated treatment difference between NR and placebo in IL-8 after 6 weeks was -52.6% (95% confidence interval (CI): -75.7% to -7.6%; P = 0.030). This effect persisted until the follow-up 12 weeks after the end of treatment (-63.7%: 95% CI -85.7% to -7.8%; P = 0.034). For secondary outcomes, NR treatment increased NAD+ levels by more than twofold in whole blood, whereas IL-6 levels in plasma remained unchanged. In exploratory analyses, treatment with NR showed indications of upregulated gene pathways related to genomic integrity in the airways and reduced epigenetic aging, possibly through a reduction in cellular senescence. These exploratory analyses need to be confirmed in future trials. ClinicalTrials.gov identifier: NCT04990869 .
Collapse
Affiliation(s)
- Kristoffer L Norheim
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Michael Ben Ezra
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | | | - Louise Munkholm Andreasson
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Lise Lotte Eriksen
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Nanna Dyhre-Petersen
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Mads Vargas Damgaard
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Magnus Berglind
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Luca Pricolo
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Asger Sverrild
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sisse Bolm Ditlev
- Copenhagen Center for Translational Research, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Celeste Porsbjerg
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Morten Scheibye-Knudsen
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
13
|
Boosting NAD + in patients with COPD reduces airway inflammation. NATURE AGING 2024; 4:1676-1677. [PMID: 39567759 DOI: 10.1038/s43587-024-00765-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
|
14
|
Sim S, Choi Y, Park HS. Update on Inflammatory Biomarkers for Defining Asthma Phenotype. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:462-472. [PMID: 39363766 PMCID: PMC11450439 DOI: 10.4168/aair.2024.16.5.462] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 10/05/2024]
Abstract
Asthma is a chronic heterogeneous disease characterized by various symptoms and persistent airway inflammation, resulting in progressive lung function decline. Classifying asthma phenotypes/endotypes is crucial because the underlying mechanisms and long-term outcomes vary from patient to patient. Recent trials have identified several biomarkers for classifying asthma phenotypes/endotypes, and current treatments have been developed on the basis of these biomarkers. Conventional biomarkers, including immunoglobulin E, blood/sputum eosinophil counts, airway obstruction or reversibility, and fractional exhaled nitric oxide, are widely used to diagnose asthma. However, these markers have some limitations, necessitating the discovery of additional biomarkers. Therefore, this review summarizes recently suggested biomarkers for representing type 2-high (eosinophilic) vs. type 2-low (neutrophilic) asthma, non-steroidal anti-inflammatory drug-exacerbated respiratory disease, and severe asthma. Additionally, we discuss the potential benefits of these biomarkers in classifying specific phenotypes/endotypes and managing asthmatic patients.
Collapse
Affiliation(s)
- Soyoon Sim
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Youngwoo Choi
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea.
| |
Collapse
|
15
|
Kim SR, Um YJ, Chung SI, Jeong KY, Park HJ, Park KH, Park JW, Park SG, Lee JH. Anti-aminoacyl-tRNA synthetase-interacting multifunctional protein-1 antibody improves airway inflammation in mice with house dust mite induced asthma. World Allergy Organ J 2024; 17:100956. [PMID: 39262899 PMCID: PMC11388501 DOI: 10.1016/j.waojou.2024.100956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 09/13/2024] Open
Abstract
Background Several biologics have been developed and used to treat severe asthma. However, commercialized biologics have limitations in treating T2-low asthma because their main target is the T2 inflammation marker. Therefore, there is an unmet need for treating T2-low severe asthma. Aminoacyl-tRNA synthetase-interacting multifunctional protein 1 (AIMP1) is an auxiliary protein in the mammalian multi-aminoacyl-tRNA synthetase complex. AIMP1 also acts as a cytokine and induces the secretion of proinflammatory cytokines. Since anti-AIMP1 has been shown to reduce interleukin (IL)-6, tumor necrosis factor-α, and IL-17A levels in a mouse model, it could be effective in the treatment of T2-low severe asthma. Methods Wild-type BALB/c mice were sensitized and challenged with intranasal inoculation of a crude HDM extract. Atliximab, a chimeric AIMP1 antibody, was administered once (20 μg, 40 μg, 100 μg) on Day 14. We evaluated airway hyperresponsiveness (AHR), performed cellular analyses of the bronchoalveolar lavage fluid (BALF), measured inflammatory cytokine levels, and examined peribronchial histological features. Results Atliximab reduced AIMP1 levels in asthmatic mice in a dose-dependent manner. AHR and Inflammatory cells such as neutrophils and eosinophils in the BALF decreased in asthmatic mice treated with atliximab. The levels of IL-6, IL-13, and transforming growth factor-β (TGF-β) in the lung tissue decreased in asthmatic mice treated with a high dose of atliximab (100 μg). Atliximab also reduced goblet cell hyperplasia and peribronchial fibrosis. Conclusions Atliximab improved asthmatic airway inflammation including neutrophilic inflammation in HDM-induced asthma mice. These data suggest that anti-AIMP1 plays an important role in the treatment of severe T2-low asthma.
Collapse
Affiliation(s)
- Sung-Ryeol Kim
- Yongin Severance Hospital, Yonsei University College of Medicine, Division of Pulmonology, Allergy and Critical Care Medicine, Department of Internal Medicine, Kyounggi-do, Republic of Korea
| | - Yun Jung Um
- College of Pharmacy, Ajou University, Suwon, Gyeonggi-do, Republic of Korea
| | - Sook In Chung
- Institute of Allergy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyoung Yong Jeong
- Institute of Allergy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Jung Park
- Gangnam Severance Hospital, Yonsei University College of Medicine, Department of Internal Medicine, Seoul, Republic of Korea
| | - Kyung Hee Park
- Institute of Allergy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei University College of Medicine, Division of Allergy and Immunology, Department of Internal Medicine, Seoul, Republic of Korea
| | - Jung-Won Park
- Institute of Allergy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei University College of Medicine, Division of Allergy and Immunology, Department of Internal Medicine, Seoul, Republic of Korea
| | - Sang Gyu Park
- College of Pharmacy, Ajou University, Suwon, Gyeonggi-do, Republic of Korea
| | - Jae-Hyun Lee
- Institute of Allergy, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei University College of Medicine, Division of Allergy and Immunology, Department of Internal Medicine, Seoul, Republic of Korea
| |
Collapse
|
16
|
Valera P, Henriques-Pereira M, Wagner M, Gaspar VM, Mano JF, Liz-Marzán LM. Surface-Enhanced Raman Scattering Monitoring of Tryptophan Dynamics in 3D Pancreatic Tumor Models. ACS Sens 2024; 9:4236-4247. [PMID: 39038809 PMCID: PMC11348414 DOI: 10.1021/acssensors.4c01210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/24/2024]
Abstract
In the intricate landscape of the tumor microenvironment, both cancer and stromal cells undergo rapid metabolic adaptations to support their growth. Given the relevant role of the metabolic secretome in fueling tumor progression, its unique metabolic characteristics have gained prominence as potential biomarkers and therapeutic targets. As a result, rapid and accurate tools have been developed to track metabolic changes in the tumor microenvironment with high sensitivity and resolution. Surface-enhanced Raman scattering (SERS) is a highly sensitive analytical technique and has been proven efficient toward the detection of metabolites in biological media. However, profiling secreted metabolites in complex cellular environments such as those in tumor-stroma 3D in vitro models remains challenging. To address this limitation, we employed a SERS-based strategy to investigate the metabolic secretome of pancreatic tumor models within 3D cultures. We aimed to monitor the immunosuppressive potential of stratified pancreatic cancer-stroma spheroids as compared to 3D cultures of either pancreatic cancer cells or cancer-associated fibroblasts, focusing on the metabolic conversion of tryptophan into kynurenine by the IDO-1 enzyme. We additionally sought to elucidate the dynamics of tryptophan consumption in correlation with the size, temporal evolution, and composition of the spheroids, as well as assessing the effects of different drugs targeting the IDO-1 machinery. As a result, we confirm that SERS can be a valuable tool toward the optimization of cancer spheroids, in connection with their tryptophan metabolizing capacity, potentially allowing high-throughput spheroid analysis.
Collapse
Affiliation(s)
- Pablo
S. Valera
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San
Sebastián, Spain
- CIC
bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
- Departamento
de Química Aplicada, Universidad
del País Vasco/Euskal Herriko Universitatea (UPV/EHU), 20018 Donostia-San
Sebastián, Spain
| | - Margarida Henriques-Pereira
- Department
of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Marita Wagner
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Departamento
de Química Aplicada, Universidad
del País Vasco/Euskal Herriko Universitatea (UPV/EHU), 20018 Donostia-San
Sebastián, Spain
- CIC nanoGUNE,
Basque Research and Technology Alliance (BRTA), 20018 Donostia-San Sebastián, Spain
| | - Vítor M. Gaspar
- Department
of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - João F. Mano
- Department
of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Luis M. Liz-Marzán
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San
Sebastián, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
- Cinbio, Universidade de Vigo, 36310 Vigo, Spain
| |
Collapse
|
17
|
Carpi S, Polini B, Nieri D, Doccini S, Conti M, Bazzan E, Pagnini M, Santorelli FM, Cecchini M, Nieri P, Celi A, Neri T. Extracellular Vesicles Induce Nuclear Factor-κB Activation and Interleukin-8 Synthesis through miRNA-191-5p Contributing to Inflammatory Processes: Potential Implications in the Pathogenesis of Chronic Obstructive Pulmonary Disease. Biomolecules 2024; 14:1030. [PMID: 39199417 PMCID: PMC11352467 DOI: 10.3390/biom14081030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 09/01/2024] Open
Abstract
Extracellular vesicles (EVs) play a pivotal role in a variety of physiologically relevant processes, including lung inflammation. Recent attention has been directed toward EV-derived microRNAs (miRNAs), such as miR-191-5p, particularly in the context of inflammation. Here, we investigated the impact of miR-191-5p-enriched EVs on the activation of NF-κB and the expression of molecules associated with inflammation such as interleukin-8 (IL-8). To this aim, cells of bronchial epithelial origin, 16HBE, were transfected with miR-191-5p mimic and inhibitor and subsequently subjected to stimulations to generate EVs. Then, bronchial epithelial cells were exposed to the obtained EVs to evaluate the activation of NF-κB and IL-8 levels. Additionally, we conducted a preliminary investigation to analyze the expression profiles of miR-191-5p in EVs isolated from the plasma of patients diagnosed with chronic obstructive pulmonary disease (COPD). Our initial findings revealed two significant observations. First, the exposure of bronchial epithelial cells to miR-191-5p-enriched EVs activated the NF-kB signaling and increased the synthesis of IL-8. Second, we discovered the presence of miR-191-5p in peripheral blood-derived EVs from COPD patients and noted a correlation between miR-191-5p levels and inflammatory and functional parameters. Collectively, these data corroborate and further expand the proinflammatory role of EVs, with a specific emphasis on miR-191-5p as a key cargo involved in this process. Consequently, we propose a model in which miR-191-5p, carried by EVs, plays a role in airway inflammation and may contribute to the pathogenesis of COPD.
Collapse
Affiliation(s)
- Sara Carpi
- Department of Health Sciences, University ‘Magna Græcia’ of Catanzaro, 88100 Catanzaro, Italy;
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy;
| | - Beatrice Polini
- Department of Pathology, University of Pisa, 56100 Pisa, Italy;
| | - Dario Nieri
- Centre for Cardio-Respiratory Cell Biology, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, 56100 Pisa, Italy; (D.N.); (M.P.); (T.N.)
| | - Stefano Doccini
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (S.D.); (F.M.S.)
| | - Maria Conti
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35122 Padua, Italy; (M.C.); (E.B.)
| | - Erika Bazzan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35122 Padua, Italy; (M.C.); (E.B.)
| | - Marta Pagnini
- Centre for Cardio-Respiratory Cell Biology, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, 56100 Pisa, Italy; (D.N.); (M.P.); (T.N.)
| | - Filippo Maria Santorelli
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (S.D.); (F.M.S.)
| | - Marco Cecchini
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy;
| | - Paola Nieri
- Department of Pharmacy, University of Pisa, 56100 Pisa, Italy;
| | - Alessandro Celi
- Centre for Cardio-Respiratory Cell Biology, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, 56100 Pisa, Italy; (D.N.); (M.P.); (T.N.)
| | - Tommaso Neri
- Centre for Cardio-Respiratory Cell Biology, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, 56100 Pisa, Italy; (D.N.); (M.P.); (T.N.)
| |
Collapse
|
18
|
Vitucci ECM, Simmons AE, Martin EM, McCullough SD. Epithelial MAPK signaling directs endothelial NRF2 signaling and IL-8 secretion in a tri-culture model of the alveolar-microvascular interface following diesel exhaust particulate (DEP) exposure. Part Fibre Toxicol 2024; 21:15. [PMID: 38468337 PMCID: PMC10926573 DOI: 10.1186/s12989-024-00576-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 02/27/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Particulate matter 2.5 (PM2.5) deposition in the lung's alveolar capillary region (ACR) is significantly associated with respiratory disease development, yet the molecular mechanisms are not completely understood. Adverse responses that promote respiratory disease development involve orchestrated, intercellular signaling between multiple cell types within the ACR. We investigated the molecular mechanisms elicited in response to PM2.5 deposition in the ACR, in an in vitro model that enables intercellular communication between multiple resident cell types of the ACR. METHODS An in vitro, tri-culture model of the ACR, incorporating alveolar-like epithelial cells (NCI-H441), pulmonary fibroblasts (IMR90), and pulmonary microvascular endothelial cells (HULEC) was developed to investigate cell type-specific molecular responses to a PM2.5 exposure in an in-vivo-like model. This tri-culture in vitro model was termed the alveolar capillary region exposure (ACRE) model. Alveolar epithelial cells in the ACRE model were exposed to a suspension of diesel exhaust particulates (DEP) (20 µg/cm2) with an average diameter of 2.5 µm. Alveolar epithelial barrier formation, and transcriptional and protein expression alterations in the directly exposed alveolar epithelial and the underlying endothelial cells were investigated over a 24 h DEP exposure. RESULTS Alveolar epithelial barrier formation was not perturbed by the 24 h DEP exposure. Despite no alteration in barrier formation, we demonstrate that alveolar epithelial DEP exposure induces transcriptional and protein changes in both the alveolar epithelial cells and the underlying microvascular endothelial cells. Specifically, we show that the underlying microvascular endothelial cells develop redox dysfunction and increase proinflammatory cytokine secretion. Furthermore, we demonstrate that alveolar epithelial MAPK signaling modulates the activation of NRF2 and IL-8 secretion in the underlying microvascular endothelial cells. CONCLUSIONS Endothelial redox dysfunction and increased proinflammatory cytokine secretion are two common events in respiratory disease development. These findings highlight new, cell-type specific roles of the alveolar epithelium and microvascular endothelium in the ACR in respiratory disease development following PM2.5 exposure. Ultimately, these data expand our current understanding of respiratory disease development following particle exposures and illustrate the utility of multicellular in vitro systems for investigating respiratory tract health.
Collapse
Affiliation(s)
- Eva C M Vitucci
- Interdisciplinary Faculty of Toxicology, School of Public Health, Texas A&M University, College Station, TX, USA
- Curriculum in Toxicology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- The Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Alysha E Simmons
- Curriculum in Toxicology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Elizabeth M Martin
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Shaun D McCullough
- Exposure and Protection, RTI International, 3040 East Cornwallis Road, Durham, NC, USA.
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Chapel Hill, NC, USA.
| |
Collapse
|
19
|
Situmorang PC, Ilyas S, Syahputra RA, Nugraha AP, Putri MSS, Rumahorbo CGP. Rhodomyrtus tomentosa (Aiton) Hassk. (haramonting) protects against allethrin-exposed pulmo damage in rats: mechanistic interleukins. Front Pharmacol 2024; 15:1343936. [PMID: 38379903 PMCID: PMC10877004 DOI: 10.3389/fphar.2024.1343936] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/25/2024] [Indexed: 02/22/2024] Open
Abstract
Inhaling Allethrin (C19H26O3) may induce oxidative stress in lung cells by causing the formation of free radi-cals. Interleukins (IL) are a group of secreted cytokines or proteins and signaling molecules initially produced as an immune response by leukocytes. Rhodomyrtus tomentosa (Aiton) Hassk. (haramonting) contains antioxidants that may prevent lung damage induced by allethrin-containing electric mosquito repellents. In this study, six groups of rats were exposed to allethrin via an electric mosquito repellent, including positive, negative, and comparison control groups and three groups were administered Rhodomyrtus tomentosa (Aiton) Hassk at 100 mg/kg BW, 200 mg/kg BW, and 300 mg/kg BW. After 30 days, the pulmonary tissue and the blood were taken for immunohisto-chemical and ELISA analysis. The accumulation of inflammatory cells causes the thickening of the alveolar wall structures. Injuries were more prevalent in the A+ group than in the other groups. The connection between the alveoli and blood capillaries, which can interfere with alveolar gas exchange, is not regulated, and the lu-minal morphology is aberrant, causing damage to the alveolar epithelial cells. Exposure to electric mosquito coils containing allethrin can increase the expression of interleukin-1, interleukin-8, interleukin-9, and interleu-kin-18 in blood serum and tissues while decreasing the expression of interleukin-6 and interleukin-10. Like the Vitamin C group, Rhodomyrtus tomentosa can increase alveolar histological alterations by decreasing the ex-pression of IL-1β, IL-8, IL-9, and IL-18 while increasing IL-6 and IL-10. So that this plant can be developed in the future as a drug to prevent lung harm from exposure.
Collapse
Affiliation(s)
- Putri Cahaya Situmorang
- Study Program of Biology, Faculty of Mathematics and Natural Sciences, Universitas Sumatera Utara, Medan, Indonesia
| | - Syafruddin Ilyas
- Study Program of Biology, Faculty of Mathematics and Natural Sciences, Universitas Sumatera Utara, Medan, Indonesia
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Alexander Patera Nugraha
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Mimmy Sari Syah Putri
- Study Program of Biology, Faculty of Mathematics and Natural Sciences, Universitas Sumatera Utara, Medan, Indonesia
| | | |
Collapse
|
20
|
Tamayo JM, Osman HC, Schwartzer JJ, Ashwood P. The influence of asthma on neuroinflammation and neurodevelopment: From epidemiology to basic models. Brain Behav Immun 2024; 116:218-228. [PMID: 38070621 DOI: 10.1016/j.bbi.2023.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 11/08/2023] [Accepted: 12/04/2023] [Indexed: 12/18/2023] Open
Abstract
Asthma is a highly heterogeneous inflammatory disease that can have a significant effect on both the respiratory system and central nervous system. Population based studies and animal models have found asthma to be comorbid with a number of neurological conditions, including depression, anxiety, and neurodevelopmental disorders. In addition, maternal asthma during pregnancy has been associated with neurodevelopmental disorders in the offspring, such as autism spectrum disorders and attention deficit hyperactivity disorder. In this article, we review the most current epidemiological studies of asthma that identify links to neurological conditions, both as it relates to individuals that suffer from asthma and the impacts asthma during pregnancy may have on offspring neurodevelopment. We also discuss the relevant animal models investigating these links, address the gaps in knowledge, and explore the potential future directions in this field.
Collapse
Affiliation(s)
- Juan M Tamayo
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, CA 95817, USA
| | - Hadley C Osman
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, CA 95817, USA
| | - Jared J Schwartzer
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, and the M.I.N.D. Institute, University of California at Davis, CA 95817, USA.
| |
Collapse
|
21
|
Mohanty D, Padhee S, Sahoo C, Jena S, Sahoo A, Chandra Panda P, Nayak S, Ray A. Integrating network pharmacology and experimental verification to decipher the multitarget pharmacological mechanism of Cinnamomum zeylanicum essential oil in treating inflammation. Heliyon 2024; 10:e24120. [PMID: 38298712 PMCID: PMC10828654 DOI: 10.1016/j.heliyon.2024.e24120] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
Inflammatory diseases contribute to more than 50 % of global deaths. Research suggests that network pharmacology can reveal the biological mechanisms underlying inflammatory diseases and drug effects at the molecular level. The aim of the study was to clarify the biological mechanism of Cinnamomum zeylanicum essential oil (CZEO) and predict molecular targets of CZEO against inflammation by employing network pharmacology and in vitro assays. First, the genes related to inflammation were identified from the Genecards and Online Mendelian Inheritance in Man (OMIM) databases. The CZEO targets were obtained from the SwissTargetPrediction and Similarity Ensemble Approach (SEA) database. A total of 1057 CZEO and 526 anti-inflammation targets were obtained. The core hub target of CZEO anti-inflammatory was obtained using the protein-protein interaction network. KEGG pathway analysis suggested CZEO to exert anti-inflammatory effect mainly through Tumor necrosis factor, Toll-like receptor and IL-17 signalling pathway. Molecular docking of active ingredients-core targets interactions was modelled using Pyrx software. Docking and simulation studies revealed benzyl benzoate to exhibit good binding affinity towards IL8 protein. MTT assay revealed CZEO to have non-cytotoxic effect on RAW 264.7 cells. CZEO also inhibited the production of NO, PGE2, IL-6, IL-1β and TNF-α and promoted the activity of endogenous antioxidant enzymes in LPS-stimulated RAW 264.7 cells. Additionally, CZEO inhibited intracellular ROS generation, NF-kB nuclear translocation and modulated the expression of downstream genes involved in Toll-like receptor signalling pathway. The results deciphered the mechanism of CZEO in treating inflammation and provided a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Debajani Mohanty
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| | - Sucheesmita Padhee
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| | - Chiranjibi Sahoo
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| | - Sudipta Jena
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| | - Ambika Sahoo
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| | - Pratap Chandra Panda
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| | - Sanghamitra Nayak
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| | - Asit Ray
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar-751003, Odisha, India
| |
Collapse
|
22
|
Zhuang Y, Yang Y, Peng L. Circ_0026579 knockdown ameliorates lipopolysaccharide-induced human lung fibroblast cell injury by regulating CXCR1 via miR-370-3p. Clin Exp Pharmacol Physiol 2023; 50:992-1004. [PMID: 37786235 DOI: 10.1111/1440-1681.13826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 10/04/2023]
Abstract
Pneumonia is an inflammatory disease in lower respiratory tracts and its development involves the regulation of RNAs. Circular RNAs are a class of RNA subgroups that can mediate the progression of pneumonia. However, the molecular mechanism of circ_0026579 in regulating pneumonia occurrence remains unclear. The study is designed to reveal the role of circ_0026579 in lipopolysaccharide (LPS)-induced human lung fibroblast cell injury and the underlying mechanism. The expression levels of circ_0026579, miR-370-3p and C-X-C motif chemokine receptor 1 (CXCR1) were detected by quantitative real-time polymerase chain reaction or by western blotting. The production of tumour necrosis factor-α, interleukin (IL)-1β and IL-6 was assessed by enzyme-linked immunosorbent assays. Malondialdehyde and superoxide dismutase levels were analysed using commercial kits. Cell viability, proliferation and apoptosis were analysed by cell counting kit-8 assay, 5-Ethynyl-2'-deoxyuridine assay and flow cytometry analysis, respectively. The binding relationship between miR-370-3p and circ_0026579 or CXCR1 was identified by dual-luciferase reporter assay, RNA immunoprecipitation assay and RNA pull-down assay. Circ_0026579 and CXCR1 expression were significantly upregulated, whereas miR-370-3p was downregulated in the serum of pneumonia patients. LPS treatment induced inflammatory response, oxidative stress and cell apoptosis and inhibited cell proliferation in MRC-5 cells; however, these effects were reversed after circ_0026579 depletion. In terms of the mechanism, circ_0026579 acted as a miR-370-3p sponge, and miR-370-3p combined with CXCR1. Additionally, circ_0026579 depletion ameliorated LPS-induced MRC-5 cell disorder by increasing miR-370-3p expression. CXCR1 overexpression also relieved the miR-370-3p-mediated effects in LPS-treated MRC-5 cells. Further, circ_0026579 induced CXCR1 expression by interacting with miR-370-3p. Circ_0026579 absence ameliorated MRC-5 cell dysfunction induced by LPS through the regulation of the miR-370-3p/CXCR1 axis.
Collapse
Affiliation(s)
- Yuanhong Zhuang
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital Xiamen University, Xiamen, China
| | - Yuyun Yang
- Department of Geriatrics, Zhongshan Hospital Xiamen University, Xiamen, China
| | - Lihong Peng
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital Xiamen University, Xiamen, China
| |
Collapse
|
23
|
Paudel I, Barutcu AR, Samuel R, Moreau M, Slattery SD, Scaglione J, Recio L. Increasing confidence in new approach methodologies for inhalation risk assessment with multiple end point assays using 5-day repeated exposure to 1,3-dichloropropene. Toxicology 2023; 499:153642. [PMID: 37863466 DOI: 10.1016/j.tox.2023.153642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/29/2023] [Accepted: 10/09/2023] [Indexed: 10/22/2023]
Abstract
New Approach Methodologies (NAMs) are being widely used to reduce, refine, and replace, animal use in studying toxicology. For respiratory toxicology, this includes both in silico and in vitro alternatives to replace traditional in vivo inhalation studies. 1,3-Dichloropropene (1,3-DCP) is a volatile organic compound that is widely used in agriculture as a pre-planting fumigant. Short-term exposure of humans to 1,3-DCP can result in mucous membrane irritation, chest pain, headache, and dizziness. In our previous work, we exposed differentiated cells representing different parts of the respiratory epithelium to 1,3-DCP vapor, measured cytotoxicity, and did In Vitro to In Vivo Extrapolation (IVIVE). We have extended our previous study with 1,3-DCP vapors by conducting transcriptomics on acutely exposed nasal cultures and have implemented a separate 5-day repeated exposure with multiple endpoints to gain further molecular insight into our model. MucilAir™ Nasal cell culture models, representing the nasal epithelium, were exposed to six sub-cytotoxic concentrations of 1,3-DCP vapor at the air-liquid interface, and the nasal cultures were analyzed by different methodologies, including histology, transcriptomics, and glutathione (GSH) -depletion assays. We observed the dose-dependent effect of 1,3-DCP in terms of differential gene expression, change in cellular morphology from pseudostratified columnar epithelium to squamous epithelium, and depletion of GSH in MucilAir™ nasal cultures. The MucilAir™ nasal cultures were also exposed to 3 concentrations of 1,3-DCP using repeated exposure 4 h per day for 5 days and the histological analyses indicated changes in cellular morphology and a decrease in ciliated bodies and an increase in apoptotic bodies, with increasing concentrations of 1,3-DCP. Altogether, our results suggest that sub-cytotoxic exposures to 1,3-DCP lead to several molecular and cellular perturbations, providing significant insight into the mode-of-action (MoA) of 1,3-DCP using an innovative NAM model.
Collapse
Affiliation(s)
- Iru Paudel
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, NC 27709, USA
| | - A Rasim Barutcu
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, NC 27709, USA
| | - Raymond Samuel
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, NC 27709, USA
| | - Marjory Moreau
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, NC 27709, USA
| | - Scott D Slattery
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, NC 27709, USA
| | - Jamie Scaglione
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, NC 27709, USA
| | - Leslie Recio
- ScitoVation, LLC, 6 Davis Drive, Suite 146, Durham, NC 27709, USA.
| |
Collapse
|
24
|
Calvert BA, Quiroz EJ, Lorenzana Z, Doan N, Kim S, Senger CN, Anders JJ, Wallace WD, Salomon MP, Henley J, Ryan AL. Neutrophilic inflammation promotes SARS-CoV-2 infectivity and augments the inflammatory responses in airway epithelial cells. Front Immunol 2023; 14:1112870. [PMID: 37006263 PMCID: PMC10061003 DOI: 10.3389/fimmu.2023.1112870] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction In response to viral infection, neutrophils release inflammatory mediators as part of the innate immune response, contributing to pathogen clearance through virus internalization and killing. Pre- existing co-morbidities correlating to incidence to severe COVID-19 are associated with chronic airway neutrophilia. Furthermore, examination of COVID-19 explanted lung tissue revealed a series of epithelial pathologies associated with the infiltration and activation of neutrophils, indicating neutrophil activity in response to SARS-CoV-2 infection. Methods To determine the impact of neutrophil-epithelial interactions on the infectivity and inflammatory responses to SARS-CoV-2 infection, we developed a co-culture model of airway neutrophilia. This model was infected with live SARS-CoV-2 virus the epithelial response to infection was evaluated. Results SARS-CoV-2 infection of airway epithelium alone does not result in a notable pro-inflammatory response from the epithelium. The addition of neutrophils induces the release of proinflammatory cytokines and stimulates a significantly augmented proinflammatory response subsequent SARS-CoV-2 infection. The resulting inflammatory responses are polarized with differential release from the apical and basolateral side of the epithelium. Additionally, the integrity of the \epithelial barrier is impaired with notable epithelial damage and infection of basal stem cells. Conclusions This study reveals a key role for neutrophil-epithelial interactions in determining inflammation and infectivity.
Collapse
Affiliation(s)
- Ben A. Calvert
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa, IA, United States
| | - Erik J. Quiroz
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa, IA, United States
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Zareeb Lorenzana
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ngan Doan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Seongjae Kim
- The Salk Institute of Biological Studies, La Jolla, CA, United States
| | - Christiana N. Senger
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jeffrey J. Anders
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa, IA, United States
| | - Wiliam D. Wallace
- Department of Pathology, University of Southern California, Los Angeles, CA, United States
| | - Matthew P. Salomon
- Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jill Henley
- Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa, IA, United States
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
25
|
Szymczak K, Pelletier MGH, Mackay JM, Reid D, Gaines PCW. CXCR2 Antagonist RIST4721 Acts as a Potent Chemotaxis Inhibitor of Mature Neutrophils Derived from Ex Vivo-Cultured Mouse Bone Marrow. Biomedicines 2023; 11:biomedicines11020479. [PMID: 36831016 PMCID: PMC9953560 DOI: 10.3390/biomedicines11020479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/24/2023] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
Neutrophils act as critical mediators of innate immunity, which depends on their rapid responses to chemokines followed by their migration towards sites of infection during chemotaxis. Chemokine receptors expressed on the surface of neutrophils mediate chemotaxis by activating contractile machinery as the cells escape from capillary beds and then attack pathogens. Neutrophils also contribute to inflammatory responses, which support pathogen destruction but can lead to acute and chronic inflammatory disorders. CXCR2, a G-protein-coupled chemokine receptor expressed on both myeloid and epithelial cells, is well-characterized for its capacities to bind multiple chemokines, including interleukin-8 and growth-related oncogene alpha in humans or keratinocyte chemokine (KC) in mice. Here we show that a small molecule CXCR2 antagonist termed RIST4721 can effectively inhibit KC-stimulated chemotaxis by neutrophils derived from ex vivo-cultured mouse bone marrow in a potent and dose-dependent manner. Antagonistic properties of RIST4721 are thoroughly characterized, including the maximal, half-maximal and minimum concentrations required to inhibit chemotaxis. Importantly, RIST4721-treated neutrophils exhibit robust phagocytosis and reactive oxygen species production, confirming drug specificity to chemotaxis inhibition. Together our data indicate that RIST4721 acts to inhibit inflammation mediated and potentiated by neutrophils and therefore promises to facilitate treatment of a host of inflammatory conditions.
Collapse
Affiliation(s)
- Klaudia Szymczak
- Department of Biological Sciences, Biomedical Engineering and Biotechnology Program, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Margery G. H. Pelletier
- Department of Biological Sciences, Biomedical Engineering and Biotechnology Program, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - James M. Mackay
- Aristea Therapeutics, 12770 High Bluff Drive, #380, San Diego, CA 92130, USA
| | - DeAnne Reid
- Aristea Therapeutics, 12770 High Bluff Drive, #380, San Diego, CA 92130, USA
| | - Peter C. W. Gaines
- Department of Biological Sciences, Biomedical Engineering and Biotechnology Program, University of Massachusetts Lowell, Lowell, MA 01854, USA
- Correspondence: ; Tel.: +978-934-2894
| |
Collapse
|
26
|
Eckhardt CM, Gambazza S, Bloomquist TR, De Hoff P, Vuppala A, Vokonas PS, Litonjua AA, Sparrow D, Parvez F, Laurent LC, Schwartz J, Baccarelli AA, Wu H. Extracellular Vesicle-Encapsulated microRNAs as Novel Biomarkers of Lung Health. Am J Respir Crit Care Med 2023; 207:50-59. [PMID: 35943330 PMCID: PMC9952856 DOI: 10.1164/rccm.202109-2208oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 08/08/2022] [Indexed: 02/03/2023] Open
Abstract
Rationale: Early detection of respiratory diseases is critical to facilitate delivery of disease-modifying interventions. Extracellular vesicle-enriched microRNAs (EV-miRNAs) may represent reliable markers of early lung injury. Objectives: Evaluate associations of plasma EV-miRNAs with lung function. Methods: The prospective NAS (Normative Aging Study) collected plasma EV-miRNA measurements from 1996-2015 and spirometry every 3-5 years through 2019. Associations of EV-miRNAs with baseline lung function were modeled using linear regression. To complement the individual miRNA approach, unsupervised machine learning was used to identify clusters of participants with distinct EV-miRNA profiles. Associations of EV-miRNA profiles with multivariate latent longitudinal lung function trajectories were modeled using log binomial regression. Biological functions of significant EV-miRNAs were explored using pathway analyses. Results were replicated in an independent sample of NAS participants and in the HEALS (Health Effects of Arsenic Longitudinal Study). Measurements and Main Results: In the main cohort of 656 participants, 51 plasma EV-miRNAs were associated with baseline lung function (false discovery rate-adjusted P value < 0.05), 28 of which were replicated in the independent NAS sample and/or in the HEALS cohort. A subset of participants with distinct EV-miRNA expression patterns had increased risk of declining lung function over time, which was replicated in the independent NAS sample. Significant EV-miRNAs were shown in pathway analyses to target biological pathways that regulate respiratory cellular immunity, the lung inflammatory response, and airway structural integrity. Conclusions: Plasma EV-miRNAs may represent a robust biomarker of subclinical lung injury and may facilitate early identification and treatment of patients at risk of developing overt lung disease.
Collapse
Affiliation(s)
- Christina M. Eckhardt
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Irving Medical Center, New York, New York
| | - Simone Gambazza
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Healthcare Professions Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Tessa R. Bloomquist
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
| | - Peter De Hoff
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California
| | - Aishwarya Vuppala
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California
| | - Pantel S. Vokonas
- Veterans Affairs Normative Aging Study, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Augusto A. Litonjua
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; and
| | - David Sparrow
- Veterans Affairs Normative Aging Study, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Faruque Parvez
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
| | - Louise C. Laurent
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California
| | - Joel Schwartz
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Andrea A. Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
| | - Haotian Wu
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
| |
Collapse
|
27
|
Kong W, Tao Y, Fan Q, Xie L, Chen C, Du K, Wei W. Changes and Treatment Prognosis of Aqueous Humor Cytokine Concentrations of Patients with Acquired Immune Deficiency Syndrome Complicated by Cytomegalovirus Retinitis. J Ocul Pharmacol Ther 2022; 38:695-702. [PMID: 36378859 DOI: 10.1089/jop.2022.0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Purpose: The purposes of this study were to investigate cytokine changes in the aqueous humor after treatment of acquired immune deficiency syndrome (AIDS) complicated with cytomegalovirus retinitis (CMVR) and to determine whether these changes are useful prognostic indicators. Methods: This study included 12 patients (15 eyes) undergoing treatment for AIDS and CMVR. The patients received intravitreal injections and systemic intravenous treatment with ganciclovir and foscarnet sodium. The aqueous humor of each eye was sampled before treatment and before the third and fifth injections. The samples were tested to determine the concentrations of each of 27 cytokines using the Luminex 200™ liquid phase chip. Results: The concentrations of cytokines interleukin (IL)-1rα (P = 0.002), IL-1b (P = 0.001), IL-8 (P = 0.001), basic fibroblast growth factor (bFGF) (P < 0.001), interferon γ-induced protein 10 (IP-10) (P = 0.001), and tumor necrosis factor (TNF)-α (P = 0.004) in the aqueous humor before the third and fifth injections were significantly lower after than before treatment. The reductions in TNF-α (P = 0.028) and IL-1b (P = 0.028) concentrations after treatment were statistically significant compared with the postoperative visual acuity improvement (≥3 lines and <3 lines). The difference in TNF-α (P = 0.018) level before and after treatment (the difference between before treatment and before the fifth intravitreal injection) was also statistically significant compared with the number of injections (≥6 times and <6 times). Conclusion: The cytokines IL-1rα, IL-1b, IL-8, bFGF, IP-10, and TNF-α may offer new avenues for evaluation of therapeutic effect, and TNF-α and IL-1b may be important cytokines for prognostic evaluation (based on visual acuity and the number of injections) in patients suffering from AIDS and CMVR. Clinical Trial Registration: Number: ChiCTR2200056955.
Collapse
Affiliation(s)
- Wenjun Kong
- Department of Opthalmology, Beijing You'an Hosptial, Capital Medical University, Beijing, China
| | - Yong Tao
- Department of Opthalmology, Beijing Chaoyang Hosptial, Capital Medical University, Beijing, China
| | - Qian Fan
- Department of Opthalmology, Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Nankai University Affiliated Eye Hospital, Tianjin, China
| | - Lianyong Xie
- Department of Opthalmology, Beijing You'an Hosptial, Capital Medical University, Beijing, China
| | - Chao Chen
- Department of Opthalmology, Beijing You'an Hosptial, Capital Medical University, Beijing, China
| | - Kuifang Du
- Department of Opthalmology, Beijing You'an Hosptial, Capital Medical University, Beijing, China
| | - Wenbin Wei
- Department of Opthalmology, Beijing Tongren Hosptial, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Dey D, Mondal P, Moitra S, Saha GK, Podder S. Association of Interleukin 6 and Interleukin 8 genes polymorphisms with house dust mite-induced nasal-bronchial allergy in a sample of Indian patients. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00348-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Genetic background of nasal-bronchial allergy (NBA) is well documented. House Dust Mites (HDMs) are reported to elicit NBA symptoms. Susceptibility to HDM sensitization varies considerably from person to person. Interleukin 6 (IL 6) and Interleukin 8 (IL 8) are studied previously for genetic association with several diseases. To the best of our knowledge, the genetic association of HDM-induced NBA has not been largely reported from India. The aim of our present study was to evaluate any possible association of IL 6 and IL 8 gene polymorphisms with HDM-induced NBA in an Indian population.
Methods
IL 6 (− 572G/C, − 597G/A) and IL 8 polymorphisms (− 251A/T, + 781C/T) were analyzed in a HDM-sensitized group (N = 372) and a control group (N = 110). Polymerase Chain Reaction-Restriction Fragment Length Polymorphism (PCR–RFLP) based genotyping was done. Chi-square test and Fisher’s exact tests were applied for statistical analysis.
Results
IL 6 − 597G/A and IL 8 + 781C/T were not associated with HDM-sensitization, while IL 6 − 72G/C and IL 8 − 51A/T showed significant associations in terms of both genotype and allele frequencies. For both the SNPs, minor allele frequencies were significantly higher in the patients compared to the control. Moreover, IL 6 -572G/C and IL 8 -251A/T were found to be strongly linked with HDM sensitization and severity.
Conclusion
This is probably the pioneer study to describe the association of IL 6 and IL 8 polymorphisms with HDM sensitization in any Indian population. The results suggested that IL 6 -572G/C and IL 8 -251A/T may exert a risk of HDM sensitization leading to NBA.
Collapse
|
29
|
Calvert BA, Quiroz EJ, Lorenzana Z, Doan N, Kim S, Senger CN, Wallace WD, Salomon MP, Henley J, Ryan AL. Neutrophilic inflammation promotes SARS-CoV-2 infectivity and augments the inflammatory responses in airway epithelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2021.08.09.455472. [PMID: 34401877 PMCID: PMC8366793 DOI: 10.1101/2021.08.09.455472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In response to viral infection, neutrophils release inflammatory mediators as part of the innate immune response, contributing to pathogen clearance through virus internalization and killing. Pre-existing co- morbidities correlating to incidence of severe COVID-19 are associated with chronic airway neutrophilia. Furthermore, examination of COVID-19 explanted lung tissue revealed a series of epithelial pathologies associated with the infiltration and activation of neutrophils, indicating neutrophil activity in response to SARS- CoV-2 infection. To determine the impact of neutrophil-epithelial interactions on the infectivity and inflammatory responses to SARS-CoV-2 infection, we developed a co-culture model of airway neutrophilia. SARS-CoV-2 infection of the airway epithelium alone does not result in a notable pro-inflammatory response from the epithelium. The addition of neutrophils induces the release of proinflammatory cytokines and stimulates a significantly augmented pro-inflammatory response subsequent SARS-CoV-2 infection. The resulting inflammatory response is polarized with differential release from the apical and basolateral side of the epithelium. Additionally, the integrity of the epithelial barrier is impaired with notable epithelial damage and infection of basal stem cells. This study reveals a key role for neutrophil-epithelial interactions in determining inflammation and infectivity in response to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- BA Calvert
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - EJ Quiroz
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Z Lorenzana
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - N Doan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - S Kim
- The Salk Institute of Biological Studies, 10010 North Torey Pines Road, La Jolla, Ca, USA
| | - CN Senger
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - WD Wallace
- Department of Pathology, University of Southern California, Los Angeles, CA, USA
| | - MP Salomon
- Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - J Henley
- Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - AL Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
30
|
Repeated exposure of bronchial epithelial cells to particular matter increases allergen-induced cytokine release and permeability. Cytokine 2022; 154:155878. [DOI: 10.1016/j.cyto.2022.155878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 04/03/2022] [Indexed: 11/22/2022]
|
31
|
Liew KY, Koh SK, Hooi SL, Ng MKL, Chee HY, Harith HH, Israf DA, Tham CL. Rhinovirus-Induced Cytokine Alterations With Potential Implications in Asthma Exacerbations: A Systematic Review and Meta-Analysis. Front Immunol 2022; 13:782936. [PMID: 35242128 PMCID: PMC8886024 DOI: 10.3389/fimmu.2022.782936] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 01/13/2022] [Indexed: 12/01/2022] Open
Abstract
Background Rhinovirus (RV) infections are a major cause of asthma exacerbations. Unlike other respiratory viruses, RV causes minimal cytotoxic effects on airway epithelial cells and cytokines play a critical role in its pathogenesis. However, previous findings on RV-induced cytokine responses were largely inconsistent. Thus, this study sought to identify the cytokine/chemokine profiles induced by RV infection and their correlations with airway inflammatory responses and/or respiratory symptoms using systematic review, and to determine whether a quantitative difference exists in cytokine levels between asthmatic and healthy individuals via meta-analysis. Methods Relevant articles were obtained from PubMed, Scopus, and ScienceDirect databases. Studies that compared RV-induced cytokine responses between asthmatic and healthy individuals were included in the systematic review, and their findings were categorized based on the study designs, which were ex vivo primary bronchial epithelial cells (PBECs), ex vivo peripheral blood mononuclear cells (PBMCs), and human experimental studies. Data on cytokine levels were also extracted and analyzed using Review Manager 5.4. Results Thirty-four articles were included in the systematic review, with 18 of these further subjected to meta-analysis. Several studies reported the correlations between the levels of cytokines, such as IL-8, IL-4, IL-5, and IL-13, and respiratory symptoms. Evidence suggests that IL-25 and IL-33 may be the cytokines that promote type 2 inflammation in asthmatics after RV infection. Besides that, a meta-analysis revealed that PBECs from children with atopic asthma produced significantly lower levels of IFN-β [Effect size (ES): -0.84, p = 0.030] and IFN-λ (ES: -1.00, p = 0.002), and PBECs from adult atopic asthmatics produced significantly lower levels of IFN-β (ES: -0.68, p = 0.009), compared to healthy subjects after RV infection. A trend towards a deficient production of IFN-γ (ES: -0.56, p = 0.060) in PBMCs from adult atopic asthmatics was observed. In lower airways, asthmatics also had significantly lower baseline IL-15 (ES: -0.69, p = 0.020) levels. Conclusion Overall, RV-induced asthma exacerbations are potentially caused by an imbalance between Th1 and Th2 cytokines, which may be contributed by defective innate immune responses at cellular levels. Exogenous IFNs delivery may be beneficial as a prophylactic approach for RV-induced asthma exacerbations. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=184119, identifier CRD42020184119.
Collapse
Affiliation(s)
- Kong Yen Liew
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Sue Kie Koh
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Suet Li Hooi
- School of Science, Monash University Malaysia, Subang Jaya, Malaysia
| | | | - Hui-Yee Chee
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Hanis Hazeera Harith
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Daud Ahmad Israf
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
32
|
Xu J, Ghadiri M, Svolos M, McParland B, Traini D, Ong HX, Young PM. Investigating Potential TRPV1 Positive Feedback to Explain TRPV1 Upregulation in Airway Disease States. Drug Dev Ind Pharm 2022; 47:1924-1934. [PMID: 35473456 DOI: 10.1080/03639045.2022.2070759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE The airway epithelium is a potential source of pathophysiology through activation of transient potential receptor vallinoid type 1 (TRPV1) channel. A positive feedback cycle caused by TRPV1 activity is hypothesised to induce upregulation and production of inflammatory cytokines, leading to exacerbations of chronic airway diseases. These cytokine and protein regulation effects were investigated in this study. METHODS Healthy (BEAS-2B) and cancer-derived (Calu-3) airway epithelial cell lines were assessed for changes to TRPV1 protein expression and mRNA expression following exposure to capsaicin (5 µM to 50 µM), and TRPV1 modulators including heat (43 °C), and hydrochloric acid (pH 3.4 to pH 6.4). Cytotoxicity was measured to determine the working concentration ranges of treatment. Subsequent bronchoconstriction by TRPV1 activation with capsaicin was measured on guinea pig airway tissue to confirm locally mediated activity without the action of known neuronal inputs. RESULTS TRPV1 protein expression was not different for all capsaicin, acidity, and heat exposures (P > 0.05), and was replicated in mRNA protein expression (P > 0.05). IL-6 and IL-8 expression were lower in BEAS-2B and Calu-3 cell lines exposed with acidity and heat (P < 0.05), but not consistently with capsaicin exposure, with potential cytotoxic effects possible. CONCLUSIONS TRPV1 expression was present in airway epithelial cells but its expression was not changed after activation by TRPV1 activators. Thus, it was not apparent the reason for reported TRPV1 upregulation in patients with airway disease states. More complex mechanisms are likely involved and will require further investigation.
Collapse
Affiliation(s)
- Jesse Xu
- Respiratory Technology Group, Woolcock Institute of Medical Research, 431 Glebe Point Road, Glebe NSW 2037, Australia.,School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Camperdown NSW 2006, Australia
| | - Maliheh Ghadiri
- Respiratory Technology Group, Woolcock Institute of Medical Research, 431 Glebe Point Road, Glebe NSW 2037, Australia
| | - Maree Svolos
- Respiratory Technology Group, Woolcock Institute of Medical Research, 431 Glebe Point Road, Glebe NSW 2037, Australia
| | - Brent McParland
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Camperdown NSW 2006, Australia
| | - Daniela Traini
- Respiratory Technology Group, Woolcock Institute of Medical Research, 431 Glebe Point Road, Glebe NSW 2037, Australia.,Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Australia
| | - Hui Xin Ong
- Respiratory Technology Group, Woolcock Institute of Medical Research, 431 Glebe Point Road, Glebe NSW 2037, Australia.,Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Australia
| | - Paul M Young
- Respiratory Technology Group, Woolcock Institute of Medical Research, 431 Glebe Point Road, Glebe NSW 2037, Australia.,Department of Marketing, Macquarie Business School, Macquarie University, NSW 2109, Australia
| |
Collapse
|
33
|
Yoshioka K, Sato H, Kawasaki T, Ishii D, Imamoto T, Abe M, Hasegawa Y, Ohara O, Tatsumi K, Suzuki T. Transcriptome Analysis of Peripheral Blood Mononuclear Cells in Pulmonary Sarcoidosis. Front Med (Lausanne) 2022; 9:822094. [PMID: 35141260 PMCID: PMC8818883 DOI: 10.3389/fmed.2022.822094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/03/2022] [Indexed: 12/14/2022] Open
Abstract
Background Sarcoidosis is a granulomatous systemic disease of unknown etiology. Mononuclear cells such as macrophages or lymphocytes in lung tissue and hilar or mediastinal lymph nodes have been recognized to play an essential role in granuloma formation in pulmonary sarcoidosis. Peripheral blood mononuclear cells (PBMCs) consist of several immunocompetent cells and have been shown to play a mechanistic role in the pathogenesis of sarcoidosis. However, the genetic modifications that occur in bulk PBMCs of sarcoidosis remain to be elucidated. Purpose This study aimed to explore the pathobiological markers of sarcoidosis in PBMCs by comparing the transcriptional signature of PBMCs from patients with pulmonary sarcoidosis with those of healthy controls by RNA sequencing. Methods PBMC samples were collected from subjects with pulmonary sarcoidosis with no steroid/immunosuppressant drugs (n = 8) and healthy controls (n = 11) from August 2020 to April 2021, and RNA sequencing was performed with the PBMC samples. Results Principal component analysis using RNA sequencing datasets comparing pulmonary sarcoidosis with healthy controls revealed that the two groups appeared to be differentiated, in which 270 differentially expressed genes were found in PBMCs between sarcoidosis and healthy controls. Enrichment analysis for gene ontology suggested that some biological processes related to the pathobiology of sarcoidosis, such as cellular response to interleukin (IL)-1 and IFN-γ, regulation of IL-6 production, IL-8 secretion, regulation of mononuclear cell migration, and response to lipopolysaccharide, were involved. Enrichment analysis of the KEGG pathway indicated the involvement of tumor necrosis factor (TNF), toll-like receptor signaling, IL-17 signaling pathways, phagosomes, and ribosomes. Most of the genes involved in TNF and IL-17 signaling pathways and phagosomes were upregulated, while most of the ribosome-related genes were downregulated. Conclusion The present study demonstrated that bulk gene expression patterns in PBMCs were different between patients with pulmonary sarcoidosis and healthy controls. The changes in the gene expression pattern of PBMCs could reflect the existence of sarcoidosis lesions and influence granuloma formation in sarcoidosis. These new findings are important to strengthen our understanding of the etiology and pathobiology of sarcoidosis and indicate a potential therapeutic target for sarcoidosis.
Collapse
Affiliation(s)
- Keiichiro Yoshioka
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hironori Sato
- Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan.,Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takeshi Kawasaki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Daisuke Ishii
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takuro Imamoto
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Mitsuhiro Abe
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yoshinori Hasegawa
- Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan
| | - Osamu Ohara
- Department of Applied Genomics, Kazusa DNA Research Institute, Chiba, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takuji Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
34
|
Meng H, Long Q, Wang R, Zhou X, Su H, Wang T, Li Y. Identification of the Key Immune-Related Genes in Chronic Obstructive Pulmonary Disease Based on Immune Infiltration Analysis. Int J Chron Obstruct Pulmon Dis 2022; 17:13-24. [PMID: 35018096 PMCID: PMC8742581 DOI: 10.2147/copd.s333251] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose Chronic obstructive pulmonary disease (COPD) is a major cause of death and morbidity worldwide. A better understanding of new biomarkers for COPD patients and their complex mechanisms in the progression of COPD are needed. Methods An algorithm was conducted to reveal the proportions of 22 subsets of immune cells in COPD samples. Differentially expressed immune-related genes (DE-IRGs) were obtained based on the differentially expressed genes (DEGs) of the GSE57148 dataset, and 1509 immune-related genes (IRGs) were downloaded from the ImmPort database. Functional enrichment analyses of DE-IRGs were conducted by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses and Ingenuity Pathway Analysis (IPA). We defined the DE-IRGs that had correlations with immune cells as hub genes. The potential interactions among the hub genes were explored by a protein-protein interaction (PPI) network. Results The CIBERSORT results showed that lung tissue of COPD patients contained a greater number of resting NK cells, activated dendritic cells, and neutrophils than normal samples. However, the fractions of follicular helper T cells and resting dendritic cells were relatively lower. Thirty-eight DE-IRGs were obtained for further analysis. Functional enrichment analysis revealed that these DE-IRGs were significantly enriched in several immune-related biological processes and pathways. Notably, we also observed that DE-IRGs were associated with the coronavirus disease COVID-19 in the progression of COPD. After correlation analysis, six DE-IRGs associated with immune cells were considered hub genes, including AHNAK, SLIT2 TNFRRSF10C, CXCR1, CXCR2, and FCGR3B. Conclusion In the present study, we investigated immune-related genes as novel diagnostic biomarkers and explored the potential mechanism for COPD based on CIBERSORT analysis, providing a new understanding for COPD treatment.
Collapse
Affiliation(s)
- Hongqiong Meng
- Department of General Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, People's Republic of China
| | - Qionghua Long
- Department of General Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, People's Republic of China
| | - Ruiping Wang
- Department of General Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, People's Republic of China
| | - Xian Zhou
- Department of General Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, People's Republic of China
| | - Huipeng Su
- Department of General Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, People's Republic of China
| | - Tingting Wang
- Department of General Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, People's Republic of China
| | - Ya Li
- Department of Respiratory and Critical Care Medicine, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, People's Republic of China
| |
Collapse
|
35
|
Lee JW, Kim MN, Kim EG, Leem JS, Baek SM, Kim MJ, Kim KW, Sohn MH. Chitinase 3-like 1 is involved in the induction of IL-8 expression by double-stranded RNA in airway epithelial cells. Biochem Biophys Res Commun 2022; 592:106-112. [PMID: 35033868 DOI: 10.1016/j.bbrc.2022.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 11/25/2022]
Abstract
Viral respiratory infection causes inflammatory lung disease. Chitinase 3-like 1 (CHI3L1) contributes to airway inflammation, but its role in human airway epithelial cells following viral infection is unclear. Thus, we investigated whether CHI3L1 regulates inflammatory responses caused by viral infections in airway epithelial cells. Human bronchial epithelial cells, BEAS-2B, were stimulated with a synthetic analog of viral double-stranded RNA, polyinosinic:polycytidylic acid (poly(I:C)). To confirm the specific role of CHI3L1, CHI3L1 was knocked down in BEAS-2B cells using shRNA lentivirus. The expression of CHI3L1 and proinflammatory cytokines such as IL-8 and phosphorylation of mitogen-activated protein kinase (MAPK) pathways were analyzed. In addition to poly(I:C), BEAS-2B cells were infected with the human respiratory syncytial virus (RSV) A2 strain, and CHI3L1 and IL-8 expression was analyzed. Stimulating the cells with poly(I:C) increased CHI3L1 and IL-8 expression, whereas IL-8 expression was abrogated in CHI3L1 knockdown BEAS-2B cells. Poly(I:C) stimulation of BEAS-2B cells resulted in phosphorylation of MAPK pathways, and inhibition of MAPK pathways significantly abolished IL-8 secretion. Phosphorylation of MAPK pathways was diminished in CHI3L1 knockdown BEAS-2B cells. Infection with RSV increased CHI3L1 and IL-8 expression. IL-8 expression induced by RSV infection was abrogated in CHI3L1 knockdown cells. In conclusion, CHI3L1 may be involved in IL-8 secretion by regulating MAPK pathways during respiratory viral infections in airway epithelial cells.
Collapse
Affiliation(s)
- Jae Woo Lee
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul, 03722, South Korea.
| | - Mi Na Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul, 03722, South Korea.
| | - Eun Gyul Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul, 03722, South Korea.
| | - Ji Su Leem
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul, 03722, South Korea.
| | - Seung Min Baek
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul, 03722, South Korea.
| | - Min Jung Kim
- Department of Pediatrics, Yonsei University Yongin Severance Hospital, 363 Dongbaekjukjeon-daero, Giheung-gu, Yongin-si, Gyeonggi-do, 16995, South Korea.
| | - Kyung Won Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul, 03722, South Korea.
| | - Myung Hyun Sohn
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul, 03722, South Korea.
| |
Collapse
|
36
|
Classical swine fever virus NS4B protein interacts with MAVS and inhibits IL-8 expression in PAMs. Virus Res 2022; 307:198622. [PMID: 34762991 DOI: 10.1016/j.virusres.2021.198622] [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] [Received: 08/13/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/27/2022]
Abstract
Classical swine fever virus (CSFV) infection causes a severe disease of pigs, resulting in significant economic losses. The CSFV NS4B protein is crucial for viral replication and pathogenicity. Interleukin 8 (IL-8), a main chemokine, is induced by multiple cell types and plays an essential role in host defense mechanisms against numerous viruses. It has been reported that NS4A of CSFV is involved in the induction of IL-8 expression in swine umbilical vein endothelial cells. However, the effect of CSFV NS4B on IL-8 expression is unknown. In this study, we showed that CSFV NS4B inhibited IL-8 expression in porcine alveolar macrophages (PAMs), and NS4B inhibited mitochondrial antiviral signaling protein (MAVS)-induced IL-8 expression. Moreover, CSFV NS4B interacted with MAVS. However, NS4B did not alter MAVS expression. Subsequently, we demonstrated that IRF3 knockdown or NF-κB inhibition reduced MAVS-induced IL-8 expression. Furthermore, the IRF3 and NF-κB pathways were activated by MAVS expression. However, CSFV NS4B inhibited MAVS-mediated NF-κB activation and IRF3 expression. Finally, CSFV NS4B inhibited IRF3 expression. Our findings reveal that CSFV NS4B interacts with MAVS and inhibits IL-8 expression by blocking the activation of IRF3 and NF-κB. Taken together, this study provides insights into the mechanism of NS4B-inhibited IL-8 expression.
Collapse
|
37
|
Abstract
Mycoplasmas are small, genome-reduced bacteria. They are obligate parasites that can be found in a wide range of host species, including the majority of livestock animals and humans. Colonization of the host can result in a wide spectrum of outcomes. In many cases, these successful parasites are considered commensal, as they are found in the microbiota of asymptomatic carriers. Conversely, mycoplasmas can also be pathogenic, as they are associated with a range of both acute and chronic inflammatory diseases which are problematic in veterinary and human medicine. The chronicity of mycoplasma infections and the ability of these bacteria to infect even recently vaccinated individuals clearly indicate that they are able to successfully evade their host’s humoral immune response. Over the years, multiple strategies of immune evasion have been identified in mycoplasmas, with a number of them aimed at generating important antigenic diversity. More recently, mycoplasma-specific anti-immunoglobulin strategies have also been characterized. Through the expression of the immunoglobulin-binding proteins protein M or mycoplasma immunoglobulin binding (MIB), mycoplasmas have the ability to target the host’s antibodies and to prevent them from interacting with their cognate antigens. In this review, we discuss how these discoveries shed new light on the relationship between mycoplasmas and their host’s immune system. We also propose that these strategies should be taken into consideration for future studies, as they are key to our understanding of mycoplasma diseases' chronic and inflammatory nature and are probably a contributing factor to reduce vaccine efficacy.
Collapse
|
38
|
Lu D, Zhang H, Zhang Y, Zhao G, Anwar Khan F, Chen Y, Hu C, Yang L, Chen H, Guo A. Secreted MbovP0145 Promotes IL-8 Expression through Its Interactive β-Actin and MAPK Activation and Contributes to Neutrophil Migration. Pathogens 2021; 10:pathogens10121628. [PMID: 34959583 PMCID: PMC8707762 DOI: 10.3390/pathogens10121628] [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: 10/20/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 11/16/2022] Open
Abstract
Mycoplasma bovis (M. bovis) is an important pathogen of cattle responsible for huge economic losses in the dairy and beef industries worldwide. The proteins secreted by M. bovis are mainly related to its adhesion, invasion, virulence, and intracellular survival and play a role in mycoplasma-host interactions. In our previous study, we found MbovP0145, a secreted protein present in the M. bovis secretome, but little is known about its function. In this study, we assessed the inflammatory characteristics and underlined mechanism of this inflammation of recombinant MbovP0145 (rMbovP0145). For this, bovine lung epithelial cells (EBL) were stimulated by rMbovP0145 to see the IL-8 production in a time- and dose-dependent manner. We observed that rMbovP0145 increased the production of IL-8 via ERK1/2 and P38 pathway activation. Further, the effect of the M. bovis ΔMbov_0145 mutant and its complementary strain on IL-8 mRNA expression was also confirmed. A pulldown assay of the GST-tagged MbovP0145 protein with mass spectrometry demonstrated that β-actin could specifically interact with rMbovP0145 to mediate the IL-8 signaling. As knockdown of β-actin expression with RNA interference in EBL cells decreased the mRNA expression of IL-8 and the phosphorylated ERK1/2 and P38 proteins, whereas disrupted actin polymerization by cytochalasin D led to a significantly higher IL-8 expression and MAPK phosphorylation in rMbovP0145-stimulated cells. Compared to M. bovis HB0801 and its complementary strain, the culture supernatant of EBL cells infected with the M. bovis ΔMbov_0145 mutant induced less neutrophil migration to the lower chamber in a transwell system. In conclusion, MbovP0145 promoted IL-8 expression by interacting with β-actin through activation of the MAPK pathway, thus contributing to neutrophil migration.
Collapse
Affiliation(s)
- Doukun Lu
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (D.L.); (H.Z.); (Y.Z.); (G.Z.); (Y.C.); (C.H.); (H.C.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Hui Zhang
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (D.L.); (H.Z.); (Y.Z.); (G.Z.); (Y.C.); (C.H.); (H.C.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yiqiu Zhang
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (D.L.); (H.Z.); (Y.Z.); (G.Z.); (Y.C.); (C.H.); (H.C.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Gang Zhao
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (D.L.); (H.Z.); (Y.Z.); (G.Z.); (Y.C.); (C.H.); (H.C.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Farhan Anwar Khan
- Department of Animal Health, The University of Agriculture, Peshawar 25120, Pakistan;
| | - Yingyu Chen
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (D.L.); (H.Z.); (Y.Z.); (G.Z.); (Y.C.); (C.H.); (H.C.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Changmin Hu
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (D.L.); (H.Z.); (Y.Z.); (G.Z.); (Y.C.); (C.H.); (H.C.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Liguo Yang
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China;
| | - Huanchun Chen
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (D.L.); (H.Z.); (Y.Z.); (G.Z.); (Y.C.); (C.H.); (H.C.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan 430070, China
| | - Aizhen Guo
- The State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (D.L.); (H.Z.); (Y.Z.); (G.Z.); (Y.C.); (C.H.); (H.C.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China
- Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan 430070, China
- Correspondence: ; Tel.: +86-27-87287115
| |
Collapse
|
39
|
Facchinetti F, Civelli M, Singh D, Papi A, Emirova A, Govoni M. Tanimilast, A Novel Inhaled Pde4 Inhibitor for the Treatment of Asthma and Chronic Obstructive Pulmonary Disease. Front Pharmacol 2021; 12:740803. [PMID: 34887752 PMCID: PMC8650159 DOI: 10.3389/fphar.2021.740803] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic respiratory diseases are the third leading cause of death, behind cardiovascular diseases and cancer, affecting approximately 550 million of people all over the world. Most of the chronic respiratory diseases are attributable to asthma and chronic obstructive pulmonary disease (COPD) with this latter being the major cause of deaths. Despite differences in etiology and symptoms, a common feature of asthma and COPD is an underlying degree of airways inflammation. The nature and severity of this inflammation might differ between and within different respiratory conditions and pharmacological anti-inflammatory treatments are unlikely to be effective in all patients. A precision medicine approach is needed to selectively target patients to increase the chance of therapeutic success. Inhibitors of the phosphodiesterase 4 (PDE4) enzyme like the oral PDE4 inhibitor roflumilast have shown a potential to reduce inflammatory-mediated processes and the frequency of exacerbations in certain groups of COPD patients with a chronic bronchitis phenotype. However, roflumilast use is dampened by class related side effects as nausea, diarrhea, weight loss and abdominal pain, resulting in both substantial treatment discontinuation in clinical practice and withdrawal from clinical trials. This has prompted the search for PDE4 inhibitors to be given by inhalation to reduce the systemic exposure (and thus optimize the systemic safety) and maximize the therapeutic effect in the lung. Tanimilast (international non-proprietary name of CHF6001) is a novel highly potent and selective inhaled PDE4 inhibitor with proven anti-inflammatory properties in various inflammatory cells, including leukocytes derived from asthma and COPD patients, as well as in experimental rodent models of pulmonary inflammation. Inhaled tanimilast has reached phase III clinical development by showing promising pharmacodynamic results associated with a good tolerability and safety profile, with no evidence of PDE4 inhibitors class-related side effects. In this review we will discuss the main outcomes of preclinical and clinical studies conducted during tanimilast development, with particular emphasis on the characterization of the pharmacodynamic profile that led to the identification of target populations with increased therapeutic potential in inflammatory respiratory diseases.
Collapse
Affiliation(s)
| | | | - Dave Singh
- Medicines Evaluation Unit, Manchester University NHS Foundation Hospital Trust, Manchester, United Kingdom
| | - Alberto Papi
- Respiratory Medicine, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Aida Emirova
- Global Clinical Development, Chiesi, Parma, Italy
| | - Mirco Govoni
- Global Clinical Development, Chiesi, Parma, Italy
| |
Collapse
|
40
|
Yang J, Yang Q, Zhang J, Gao X, Luo R, Xie K, Wang W, Li J, Huang X, Yan Z, Wang P, Gun S. N6-Methyladenosine Methylation Analysis of Long Noncoding RNAs and mRNAs in IPEC-J2 Cells Treated With Clostridium perfringens beta2 Toxin. Front Immunol 2021; 12:769204. [PMID: 34880865 PMCID: PMC8646102 DOI: 10.3389/fimmu.2021.769204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/04/2021] [Indexed: 12/13/2022] Open
Abstract
Background The n6-methyladenosine (m6A) modification is present widely in mRNAs and long non-coding RNAs (lncRNAs), and is related to the occurrence and development of certain diseases. However, the role of m6A methylation in Clostridium perfringens type C infectious diarrhea remains unclear. Methods Here, we treated intestinal porcine jejunum epithelial cells (IPEC-J2 cells) with Clostridium perfringens beta2 (CPB2) toxin to construct an in vitro model of Clostridium perfringens type C (C. perfringens type C) infectious diarrhea, and then used methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) to identify the methylation profiles of mRNAs and lncRNAs in IPEC-J2 cells. Results We identified 6,413 peaks, representing 5,825 m6A-modified mRNAs and 433 modified lncRNAs, of which 4,356 m6A modified mRNAs and 221 m6A modified lncRNAs were significantly differential expressed between the control group and CPB2 group. The motif GGACU was enriched significantly in both the control group and the CPB2 group. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) annotation analysis showed that the differentially methylated modified mRNAs were mainly enriched in Hippo signaling pathway and Wnt signaling pathway. In addition, the target genes of the differentially m6A modified lncRNAs were related to defense response to virus and immune response. For example, ENSSSCG00000042575, ENSSSCG00000048701 and ENSSSCG00000048785 might regulate the defense response to virus, immune and inflammatory response to resist the harmful effects of viruses on cells. Conclusion In summary, this study established the m6A transcription profile of mRNAs and lncRNAs in IPEC-J2 cells treated by CPB2 toxin. Further analysis showed that m6A-modified RNAs were related to defense against viruses and immune response after CPB2 toxin treatment of the cells. Threem6A-modified lncRNAs, ENSSSCG00000042575, ENSSSCG00000048785 and ENSSSCG00000048701, were most likely to play a key role in CPB2 toxin-treated IPEC-J2 cells. The results provide a theoretical basis for further research on the role of m6A modification in piglet diarrhea.
Collapse
Affiliation(s)
- Jiaojiao Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Qiaoli Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Juanli Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xiaoli Gao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Ruirui Luo
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Kaihui Xie
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Wei Wang
- College of Animal Science and Technology, Northwest A&F University, Xian, China
| | - Jie Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xiaoyu Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Zunqiang Yan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Pengfei Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Shuangbao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
- Gansu Research Center for Swine Production Engineering and Technology, Lanzhou, China
| |
Collapse
|
41
|
Lee CW, Vo TTT, Wee Y, Chiang YC, Chi MC, Chen ML, Hsu LF, Fang ML, Lee KH, Guo SE, Cheng HC, Lee IT. The Adverse Impact of Incense Smoke on Human Health: From Mechanisms to Implications. J Inflamm Res 2021; 14:5451-5472. [PMID: 34712057 PMCID: PMC8548258 DOI: 10.2147/jir.s332771] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Incense burning is a very popular activity in daily life among many parts all over the world. A growing body of both epidemiological and experimental evidences has reported the negative effects of incense use on human well-being, posing a potential threat at public significance. This work is a comprehensive review that covers the latest findings regarding the adverse impact of incense smoke on our health, providing a panoramic visualization ranging from mechanisms to implications. The toxicities of incense smoke come directly from its harmful constituents and deposition capacity in the body. Besides, reactive oxygen species-driven oxidative stress and associated inflammation seem to be plausible underlying mechanisms, eliciting various unfavorable responses. Although our current knowledge remains many gaps, this issue still has some important implications.
Collapse
Affiliation(s)
- Chiang-Wen Lee
- Department of Nursing, Division of Basic Medical Sciences, Chronic Diseases and Health Promotion Research Center and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County, Taiwan
- Department of Safety Health and Environmental Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Thi Thuy Tien Vo
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yinshen Wee
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Yao-Chang Chiang
- Department of Nursing, Division of Basic Medical Sciences, Chronic Diseases and Health Promotion Research Center and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County, Taiwan
| | - Miao-Ching Chi
- Chronic Disease and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan
- Division of Pulmonary and Critical Care Medicine, Chiayi Chang Gung Memorial Hospital, Puzi City, Chiayi County, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan
| | - Min-Li Chen
- Department of Nursing, Chang Gung Memorial Hospital, Puzi City, Chiayi County, Taiwan
- Graduate Institute of Nursing, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan
| | - Lee-Fen Hsu
- Department of Respiratory Care, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan
- Division of Neurosurgery, Department of Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County, Taiwan
| | - Mei-Ling Fang
- Center for Environmental Toxin and Emerging-Contaminant Research, Cheng Shiu University, Kaohsiung, Taiwan
- Super Micro Research and Technology Center, Cheng Shiu University, Kaohsiung, Taiwan
| | - Kuan-Han Lee
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Su-Er Guo
- Graduate Institute of Nursing, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan
| | - Hsin-Chung Cheng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Dentistry, Taipei Medical University Hospital, Taipei, Taiwan
| | - I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
42
|
Elevated Monocytic Interleukin-8 Expression under Intermittent Hypoxia Condition and in Obstructive Sleep Apnea Patients. Int J Mol Sci 2021; 22:ijms222111396. [PMID: 34768826 PMCID: PMC8583894 DOI: 10.3390/ijms222111396] [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: 08/30/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 01/08/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a disease with great cardiovascular risk. Interleukin-8 (IL-8), an important chemokine for monocyte chemotactic migration, was studied under intermittent hypoxia condition and in OSA patients. Monocytic THP-1 cells were used to investigate the effect of intermittent hypoxia on the regulation of IL-8 by an intermittent hypoxic culture system. The secreted protein and mRNA levels were studied by means of enzyme-linked immunosorbent assay and RT/real-time PCR. The chemotactic migration of monocytes toward a conditioned medium containing IL-8 was performed by means of the transwell filter migration assay. Peripheral venous blood was collected from 31 adult OSA patients and RNA was extracted from the monocytes for the analysis of IL-8 expression. The result revealed that intermittent hypoxia enhanced the monocytic THP-1 cells to actively express IL-8 at both the secreted protein and mRNA levels, which subsequently increased the migration ability of monocytes toward IL-8. The ERK, PI3K and PKC pathways were demonstrated to contribute to the activation of IL-8 expression by intermittent hypoxia. In addition, increased monocytic IL-8 expression was found in OSA patients, with disease severity dependence and diurnal changes. This study concluded the monocytic IL-8 gene expression can be activated by intermittent hypoxia and increased in OSA patients.
Collapse
|
43
|
McKenna S, Giblin SP, Bunn RA, Xu Y, Matthews SJ, Pease JE. A highly efficient method for the production and purification of recombinant human CXCL8. PLoS One 2021; 16:e0258270. [PMID: 34653205 PMCID: PMC8519433 DOI: 10.1371/journal.pone.0258270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/22/2021] [Indexed: 12/02/2022] Open
Abstract
Chemokines play diverse and fundamental roles in the immune system and human disease, which has prompted their structural and functional characterisation. Production of recombinant chemokines that are folded and bioactive is vital to their study but is limited by the stringent requirements of a native N-terminus for receptor activation and correct disulphide bonding required to stabilise the chemokine fold. Even when expressed as fusion proteins, overexpression of chemokines in E. coli tends to result in the formation of inclusion bodies, generating the additional steps of solubilisation and refolding. Here we present a novel method for producing soluble chemokines in relatively large amounts via a simple two-step purification procedure with no requirements for refolding. CXCL8 produced by this method has the correct chemokine fold as determined by NMR spectroscopy and in chemotaxis assays was indistinguishable from commercially available chemokines. We believe that this protocol significantly streamlines the generation of recombinant chemokines.
Collapse
Affiliation(s)
- Sophie McKenna
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Sean Patrick Giblin
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Rosemarie Anne Bunn
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Yingqi Xu
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | | | - James Edward Pease
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
44
|
de Cevins C, Luka M, Smith N, Meynier S, Magérus A, Carbone F, García-Paredes V, Barnabei L, Batignes M, Boullé A, Stolzenberg MC, Pérot BP, Charbit B, Fali T, Pirabakaran V, Sorin B, Riller Q, Abdessalem G, Beretta M, Grzelak L, Goncalves P, Di Santo JP, Mouquet H, Schwartz O, Zarhrate M, Parisot M, Bole-Feysot C, Masson C, Cagnard N, Corneau A, Brunaud C, Zhang SY, Casanova JL, Bader-Meunier B, Haroche J, Melki I, Lorrot M, Oualha M, Moulin F, Bonnet D, Belhadjer Z, Leruez M, Allali S, Gras-Leguen C, de Pontual L, Fischer A, Duffy D, Rieux-Laucat F, Toubiana J, Ménager MM. A monocyte/dendritic cell molecular signature of SARS-CoV-2-related multisystem inflammatory syndrome in children with severe myocarditis. MED 2021; 2:1072-1092.e7. [PMID: 34414385 PMCID: PMC8363470 DOI: 10.1016/j.medj.2021.08.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/12/2021] [Accepted: 08/09/2021] [Indexed: 12/28/2022]
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in children is generally milder than in adults, but a proportion of cases result in hyperinflammatory conditions often including myocarditis. Methods To better understand these cases, we applied a multiparametric approach to the study of blood cells of 56 children hospitalized with suspicion of SARS-CoV-2 infection. Plasma cytokine and chemokine levels and blood cellular composition were measured, alongside gene expression at the bulk and single-cell levels. Findings The most severe forms of multisystem inflammatory syndrome in children (MIS-C) related to SARS-CoV-2 that resulted in myocarditis were characterized by elevated levels of pro-angiogenesis cytokines and several chemokines. Single-cell transcriptomics analyses identified a unique monocyte/dendritic cell gene signature that correlated with the occurrence of severe myocarditis characterized by sustained nuclear factor κB (NF-κB) activity and tumor necrosis factor alpha (TNF-α) signaling and associated with decreased gene expression of NF-κB inhibitors. We also found a weak response to type I and type II interferons, hyperinflammation, and response to oxidative stress related to increased HIF-1α and Vascular endothelial growth factor (VEGF) signaling. Conclusions These results provide potential for a better understanding of disease pathophysiology. Funding Agence National de la Recherche (Institut Hospitalo-Universitaire Imagine, grant ANR-10-IAHU-01; Recherche Hospitalo-Universitaire, grant ANR-18-RHUS-0010; Laboratoire d’Excellence ‘‘Milieu Intérieur,” grant ANR-10-LABX-69-01; ANR-flash Covid19 “AIROCovid” and “CoVarImm”), Institut National de la Santé et de la Recherche Médicale (INSERM), and the “URGENCE COVID-19” fundraising campaign of Institut Pasteur. Children with SARS-CoV-2 infection were initially thought to have only mild COVID-19 symptoms. However, several weeks into the first wave of SARS-CoV-2 infections, there was a surge of a postacute pathology called multisystem inflammatory syndrome in children (MIS-C). The authors recruited a cohort of children with suspicion of SARS-CoV-2 infection and uncovered hyperinflammation, hypoxic conditions, exacerbation of TNF-α signaling via NF-κB, and absence of responses to type I and type II IFN secretion in the most severe forms of MIS-C with severe myocarditis. This work led the authors to identify in monocytes and validate in peripheral blood mononuclear cells a molecular signature of 25 genes that allows discrimination of the most severe forms of MIS-C with myocarditis.
Collapse
Affiliation(s)
- Camille de Cevins
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Molecular Biology and Genomics, Translational Sciences, Sanofi R&D, Chilly-Mazarin, France
| | - Marine Luka
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Nikaïa Smith
- Translational Immunology Lab, Department of Immunology, Institut Pasteur, 75015 Paris, France
| | - Sonia Meynier
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Aude Magérus
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Francesco Carbone
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Víctor García-Paredes
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Laura Barnabei
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Maxime Batignes
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Alexandre Boullé
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Marie-Claude Stolzenberg
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Brieuc P Pérot
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Bruno Charbit
- Cytometry and Biomarkers UTechS, CRT, Institut Pasteur, 75015, Paris, France
| | - Tinhinane Fali
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Vithura Pirabakaran
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Boris Sorin
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Quentin Riller
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Ghaith Abdessalem
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Maxime Beretta
- Humoral Immunology Laboratory, Department of Immunology, Institut Pasteur, 75015, Paris, France
- INSERM U1222, Institut Pasteur, 75015, Paris, France
| | - Ludivine Grzelak
- Virus and Immunity Unit, Department of Virology, Institut Pasteur, 75015, Paris, France
| | - Pedro Goncalves
- INSERM U1223, Institut Pasteur, 75015, Paris, France
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, 75015, Paris, France
| | - James P Di Santo
- INSERM U1223, Institut Pasteur, 75015, Paris, France
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, 75015, Paris, France
| | - Hugo Mouquet
- Humoral Immunology Laboratory, Department of Immunology, Institut Pasteur, 75015, Paris, France
- INSERM U1222, Institut Pasteur, 75015, Paris, France
| | - Olivier Schwartz
- Virus and Immunity Unit, Department of Virology, Institut Pasteur, 75015, Paris, France
| | - Mohammed Zarhrate
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UMS3633, Paris Descartes Sorbonne Paris Cite University, Paris, France
| | - Mélanie Parisot
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UMS3633, Paris Descartes Sorbonne Paris Cite University, Paris, France
| | - Christine Bole-Feysot
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UMS3633, Paris Descartes Sorbonne Paris Cite University, Paris, France
| | - Cécile Masson
- Bioinformatics Platform, Structure Fédérative de Recherche Necker, INSERM UMR1163, Université de Paris, Imagine Institute, Paris, France
| | - Nicolas Cagnard
- Bioinformatics Platform, Structure Fédérative de Recherche Necker, INSERM UMR1163, Université de Paris, Imagine Institute, Paris, France
| | - Aurélien Corneau
- Sorbonne Université, UMS037, PASS, Plateforme de Cytométrie de la Pitié-Salpêtrière CyPS, 75013 Paris, France
| | - Camille Brunaud
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Shen-Ying Zhang
- Université de Paris, Imagine Institute, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Jean-Laurent Casanova
- Université de Paris, Imagine Institute, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Department of Paediatric Immuno-Haematology and Rheumatology, Reference Center for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), Hôpital Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), 75015 Paris, France
| | - Brigitte Bader-Meunier
- Department of Paediatric Immuno-Haematology and Rheumatology, Reference Center for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), Hôpital Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), 75015 Paris, France
| | - Julien Haroche
- Department of Immunology and Infectious Disease (CIMI-Paris), Pitié-Salpêtrière University Hospital, Sorbonne Université, AP-HP, 75013 Paris, France
| | - Isabelle Melki
- Department of Paediatric Immuno-Haematology and Rheumatology, Reference Center for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), Hôpital Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), 75015 Paris, France
- Department of Pediatrics, Robert-Debré University Hospital, AP-HP, Université de Paris, Paris, France
| | - Mathie Lorrot
- Department of Pediatrics, Armand-Trousseau University Hospital, AP-HP, 75012 Paris, France
| | - Mehdi Oualha
- Pediatric Intensive Care Unit, Necker-Enfants Malades University Hospital, AP-HP, Université de Paris, 75015 Paris, France
| | - Florence Moulin
- Pediatric Intensive Care Unit, Necker-Enfants Malades University Hospital, AP-HP, Université de Paris, 75015 Paris, France
| | | | | | - Marianne Leruez
- Virology Laboratory, Necker-Enfants Malades University Hospital, AP-HP, Université de Paris, 75015 Paris, France
| | - Slimane Allali
- Department of General Paediatrics and Paediatric Infectious Diseases, Necker-Enfants Malades University Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Université de Paris, 75015 Paris, France
| | - Christèle Gras-Leguen
- Pediatric Department, Nantes University Hospital, CIC 1413, INSERM, 44000 Nantes, France
| | - Loïc de Pontual
- Department of Pediatrics, Jean Verdier Hospital, Assistance Publique-Hôpitaux de Paris, Paris 13 University, Bondy, France
| | - Alain Fischer
- Department of Paediatric Immuno-Haematology and Rheumatology, Reference Center for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), Hôpital Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), 75015 Paris, France
- Université de Paris, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
- Collège de France, Paris, France
| | - Darragh Duffy
- Translational Immunology Lab, Department of Immunology, Institut Pasteur, 75015 Paris, France
- Cytometry and Biomarkers UTechS, CRT, Institut Pasteur, 75015, Paris, France
| | - Fredéric Rieux-Laucat
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Julie Toubiana
- Department of General Paediatrics and Paediatric Infectious Diseases, Necker-Enfants Malades University Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Université de Paris, 75015 Paris, France
- Institut Pasteur, Biodiversity and Epidemiology of Bacterial Pathogens, Paris, France
| | - Mickaël M Ménager
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| |
Collapse
|
45
|
Effect of tiger milk mushroom (Lignosus rhinocerus) supplementation on respiratory health, immunity and antioxidant status: an open-label prospective study. Sci Rep 2021; 11:11781. [PMID: 34083710 PMCID: PMC8175741 DOI: 10.1038/s41598-021-91256-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/25/2021] [Indexed: 11/21/2022] Open
Abstract
Tiger milk mushroom (TMM; Lignosus rhinocerus) have been used for a long time by indigenous communities in South East Asia regions as traditional medicine for different ailments, including respiratory disorders. The beneficial effects of TMM have been proven through in vivo and in vitro models, but these effects have yet to be validated in a clinical study. In this study, the beneficial effects of TMM supplementation were investigated in 50 voluntary participants. Participants were required to take 300 mg of TMM twice daily for three months. Level of interleukin 1β (IL-1β), interleukin 8 (IL-8), immunoglobulin A (IgA), total antioxidant capacity, malondialdehyde (MDA), 3-nitrotyrosine (3-NT), 8-hydroxydeoxyguanosine (8-OHdG), pulmonary function and respiratory symptoms were assessed during baseline and monthly follow-up visits. Results demonstrated that supplementation of TMM significantly (p < 0.05) suppressed the level of IL-1β, IL-8, MDA, as well as respiratory symptoms. In additional to that, TMM also significantly (p < 0.05) induced the level of IgA, total antioxidant capacity, as well as pulmonary function. Analyses of data indicated that gender and BMI were factors influencing the outcomes of antioxidant status. Collectively, our findings suggested that TMM supplementation effectively improves respiratory health, immunity and antioxidant status.
Collapse
|
46
|
Soriano L, Khalid T, O’Brien FJ, O’Leary C, Cryan SA. A Tissue-Engineered Tracheobronchial In Vitro Co-Culture Model for Determining Epithelial Toxicological and Inflammatory Responses. Biomedicines 2021; 9:631. [PMID: 34199462 PMCID: PMC8226664 DOI: 10.3390/biomedicines9060631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/23/2021] [Accepted: 05/29/2021] [Indexed: 11/16/2022] Open
Abstract
Translation of novel inhalable therapies for respiratory diseases is hampered due to the lack of in vitro cell models that reflect the complexity of native tissue, resulting in many novel drugs and formulations failing to progress beyond preclinical assessments. The development of physiologically-representative tracheobronchial tissue analogues has the potential to improve the translation of new treatments by more accurately reflecting in vivo respiratory pharmacological and toxicological responses. Herein, advanced tissue-engineered collagen hyaluronic acid bilayered scaffolds (CHyA-B) previously developed within our group were used to evaluate bacterial and drug-induced toxicity and inflammation for the first time. Calu-3 bronchial epithelial cells and Wi38 lung fibroblasts were grown on either CHyA-B scaffolds (3D) or Transwell® inserts (2D) under air liquid interface (ALI) conditions. Toxicological and inflammatory responses from epithelial monocultures and co-cultures grown in 2D or 3D were compared, using lipopolysaccharide (LPS) and bleomycin challenges to induce bacterial and drug responses in vitro. The 3D in vitro model exhibited significant epithelial barrier formation that was maintained upon introduction of co-culture conditions. Barrier integrity showed differential recovery in CHyA-B and Transwell® epithelial cultures. Basolateral secretion of pro-inflammatory cytokines to bacterial challenge was found to be higher from cells grown in 3D compared to 2D. In addition, higher cytotoxicity and increased basolateral levels of cytokines were detected when epithelial cultures grown in 3D were challenged with bleomycin. CHyA-B scaffolds support the growth and differentiation of bronchial epithelial cells in a 3D co-culture model with different transepithelial resistance in comparison to the same co-cultures grown on Transwell® inserts. Epithelial cultures in an extracellular matrix like environment show distinct responses in cytokine release and metabolic activity compared to 2D polarised models, which better mimic in vivo response to toxic and inflammatory stimuli offering an innovative in vitro platform for respiratory drug development.
Collapse
Affiliation(s)
- Luis Soriano
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland; (L.S.); (T.K.); (C.O.)
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- SFI Centre for Research in Medical Devices (CÚRAM), RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Tehreem Khalid
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland; (L.S.); (T.K.); (C.O.)
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI University of Medicine and Health Sciences and Trinity College Dublin, D02 YN77 Dublin, Ireland
| | - Fergal J. O’Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- SFI Centre for Research in Medical Devices (CÚRAM), RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI University of Medicine and Health Sciences and Trinity College Dublin, D02 YN77 Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Cian O’Leary
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland; (L.S.); (T.K.); (C.O.)
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- SFI Centre for Research in Medical Devices (CÚRAM), RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI University of Medicine and Health Sciences and Trinity College Dublin, D02 YN77 Dublin, Ireland
| | - Sally-Ann Cryan
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland; (L.S.); (T.K.); (C.O.)
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- SFI Centre for Research in Medical Devices (CÚRAM), RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI University of Medicine and Health Sciences and Trinity College Dublin, D02 YN77 Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, D02 PN40 Dublin, Ireland
| |
Collapse
|
47
|
No association between pyrite content and lung cell responses to coal particles. Sci Rep 2021; 11:8193. [PMID: 33854135 PMCID: PMC8046986 DOI: 10.1038/s41598-021-87517-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 03/30/2021] [Indexed: 11/29/2022] Open
Abstract
There has been an increase in the identification of cases of coal workers’ pneumoconiosis (CWP) in recent years around the world. While there are a range of possible explanations for this, studies have implicated the pyrite content of coal as a key determinant of CWP risk. However, experimental studies to support this link are limited. The aim of this study was to assess the association between the pyrite content, and subsequent release of bioavailable iron, in coal particles and the response of lung cells involved in the pathogenesis of CWP (epithelial cells, macrophages and fibroblasts). Using real-world Australian coal samples, we found no evidence of an association between the pyrite content of the coal and the magnitude of the detrimental cell response. We did find evidence of an increase in IL-8 production by epithelial cells with increasing bioavailable iron (p = 0.01), however, this was not linked to the pyrite content of the coal (p = 0.75) and we did not see any evidence of a positive association in the other cell types. Given the lack of association between the pyrite content of real-world coal particles and lung cell cytotoxicity (epithelial cells and macrophages), inflammatory cytokine production (epithelial cells, macrophages and fibroblasts), and cell proliferation (fibroblasts) our data do not support the use of coal pyrite content as a predictor of CWP risk.
Collapse
|
48
|
Gusarova GA, Das SR, Islam MN, Westphalen K, Jin G, Shmarakov IO, Li L, Bhattacharya S, Bhattacharya J. Actin fence therapy with exogenous V12Rac1 protects against acute lung injury. JCI Insight 2021; 6:135753. [PMID: 33749665 PMCID: PMC8026177 DOI: 10.1172/jci.insight.135753] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/11/2021] [Indexed: 02/05/2023] Open
Abstract
High mortality in acute lung injury (ALI) results from sustained proinflammatory signaling by alveolar receptors, such as TNF-α receptor type 1 (TNFR1). Factors that determine the sustained signaling are not known. Unexpectedly, optical imaging of live alveoli revealed a major TNF-α–induced surge of alveolar TNFR1 due to a Ca2+-dependent mechanism that decreased the cortical actin fence. Mouse mortality due to inhaled LPS was associated with cofilin activation, actin loss, and the TNFR1 surge. The constitutively active form of the GTPase, Rac1 (V12Rac1), given intranasally (i.n.) as a noncovalent construct with a cell-permeable peptide, enhanced alveolar filamentous actin (F-actin) and blocked the TNFR1 surge. V12Rac1 also protected against ALI-induced mortality resulting from i.n. instillation of LPS or of Pseudomonas aeruginosa. We propose a potentially new therapeutic paradigm in which actin enhancement by exogenous Rac1 strengthens the alveolar actin fence, protecting against proinflammatory receptor hyperexpression, and therefore blocking ALI.
Collapse
Affiliation(s)
- Galina A Gusarova
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Shonit R Das
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Mohammad N Islam
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Kristin Westphalen
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Guangchun Jin
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | | | - Li Li
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Sunita Bhattacharya
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.,Department of Pediatrics, and
| | - Jahar Bhattacharya
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.,Department of Physiology and Cellular Biophysics, Vagelos College of Physicians and Surgeons of Columbia University, New York, New York, USA
| |
Collapse
|
49
|
Pereira ABM, de Oliveira JR, Teixeira MM, da Silva PR, Rodrigues Junior V, Rogerio ADP. IL-27 regulates IL-4-induced chemokine production in human bronchial epithelial cells. Immunobiology 2020; 226:152029. [PMID: 33278712 DOI: 10.1016/j.imbio.2020.152029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 10/29/2020] [Accepted: 11/12/2020] [Indexed: 11/25/2022]
Abstract
IL-4 coordinates the Th2-type immune response in inflammatory diseases such as asthma. IL-27 can inhibit the development of both Th2 and Th1 cells. However, IL-27 can also drive naïve T cells to differentiate toward the Th1 phenotype. In this study, we investigated the effects of IL-27 on the activation of IL-4-induced human bronchial epithelial cells (BEAS-2B). Compared to controls, both IL-4 and IL-27 (25-100 ng/mL) increased the concentrations of CCL2 and IL-8 in a dose-dependent manner. However, compared to cells stimulated individually with IL-4 or IL-27, treatment with a combination of both cytokines reduced CCL2 and IL-8 concentrations in a dose- and time-dependent manner. IL-4 increased the activation of p38 MAPK, ERK1/2, STAT6 and NF-κB, while IL-27 increased the activation of p38 MAPK and ERK1/2 but not STAT6 and NF-κB. Compared to IL-4-stimulated cells, cells treated with both IL-27 and IL-4 displayed decreased activation of STAT6 and NF-κB but not ERK1/2 and p38 MAPK. Taken together, these results suggest that IL-27 plays a pro-inflammatory role when administered alone but downregulates bronchial epithelial cell activation when combined with IL-4. Therefore, IL-27 may be an interesting target for the treatment of Th2 inflammatory diseases.
Collapse
Affiliation(s)
- Aline Beatriz Mahler Pereira
- Institute of Health Sciences, Department of Clinical Medicine, Laboratory of Experimental Immunopharmacology, Federal University of Triangulo Mineiro, Uberaba, MG 38025-350, Brazil
| | - Jhony Robison de Oliveira
- Institute of Health Sciences, Department of Clinical Medicine, Laboratory of Experimental Immunopharmacology, Federal University of Triangulo Mineiro, Uberaba, MG 38025-350, Brazil
| | - Maxelle Martins Teixeira
- Institute of Health Sciences, Department of Clinical Medicine, Laboratory of Experimental Immunopharmacology, Federal University of Triangulo Mineiro, Uberaba, MG 38025-350, Brazil
| | - Paulo Roberto da Silva
- Institute of Health Sciences, Department of Clinical Medicine, Laboratory of Experimental Immunopharmacology, Federal University of Triangulo Mineiro, Uberaba, MG 38025-350, Brazil
| | - Virmondes Rodrigues Junior
- Institute of Health Sciences, Department of Clinical Medicine, Laboratory of Experimental Immunopharmacology, Federal University of Triangulo Mineiro, Uberaba, MG 38025-350, Brazil
| | - Alexandre de Paula Rogerio
- Institute of Health Sciences, Department of Clinical Medicine, Laboratory of Experimental Immunopharmacology, Federal University of Triangulo Mineiro, Uberaba, MG 38025-350, Brazil.
| |
Collapse
|
50
|
Zhang C, Wang YE, Miao H, Hou J. Efficacy and Safety of Aqueous Interleukin-8-Guided Treatment in Cytomegalovirus Retinitis after Bone Marrow Hematopoietic Stem Cell Transplantation. Ocul Immunol Inflamm 2020; 30:758-765. [PMID: 33064057 DOI: 10.1080/09273948.2020.1823422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE To explore the optimal treatment for cytomegalovirus retinitis (CMVR) in patients status-post Allogeneic bone marrow hematopoietic stem cell transplantation (Allo-HSCT), based on aqueous humor indicators. METHODS A randomized controlled study with 35 eyes. Eyes were randomized with a 1:1 ratio to standard treatment group (Group 1, with treatment endpoint as aqueous CMV-DNA load<103 copy/ml), and interleukin (IL)-8 group (Group 2, with treatment endpoint as aqueous IL-8 level <30 pg/ml or CMV-DNA load<103 copy/ml) to receive antiviral intravitreal injections. Number of injections, CMVR recurrence rate, complication rate, and vision changes were analyzed and compared. RESULTS The mean number of injections in group 2 was less than in group 1 (6 vs 8 respectively, p<0.05). There were no significant differences in CMVR recurrence, complication and vision recovery rate. CONCLUSION Incorporating aqueous humor IL-8 level into the criteria of CMVR treatment decision can safely and effectively reduce the number of intravitreal injections needed and can be used as important indicators to assess treatment endpoint.
Collapse
Affiliation(s)
- Chuan Zhang
- Department of Ophthalmology & Clinical Centre of Optometry, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroidal Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Ye Elaine Wang
- Harvard Eye Associates, Private Practice, Laguna Hills, CA, USA.,Department of Ophthalmology, Stein Eye Institute, UCLA School of Medicine, Los Angeles, CA, USA
| | - Heng Miao
- Department of Ophthalmology & Clinical Centre of Optometry, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroidal Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Jing Hou
- Department of Ophthalmology & Clinical Centre of Optometry, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroidal Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| |
Collapse
|