1
|
Zhong X, Lyu C, Lai D, Shu Q. [Advances on physiology and pathology of subpopulations of macrophages in the lung tissue]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:650-658. [PMID: 39343742 DOI: 10.3724/zdxbyxb-2024-0129] [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] [Indexed: 10/01/2024]
Abstract
Macrophages are vital in maintaining tissue homeostasis in the lungs by modulating and regulating immune responses. Based on different origins and anatomical locations, macrophages in the lungs are categorized into alveolar macrophages, interstitial macrophages, perivascular macrophages, and inflammatory macrophages. Alveolar macrophages are located in the alveolar spaces and are primarily responsible for maintaining alveolar surfactant homeostasis, defending against pathogens and regulating immune responses. Interstitial macrophages can maintain homeostasis, regulate immunity and anti-inflammation in the lung tissue. Perivascular macrophages play a crucial role in inhibiting lung inflammation, improving pulmonary fibrosis, and regulating lung tumor progression due to antigen-presenting and immunomodulatory effects. Inflammatory macrophages, which are differentiated from monocytes during inflammation, regulate the inflammatory process. This article reviews the origins of various subpopulations of macro-phages in the lung tissue and their physiological and pathological functions as well as discusses the underlying mechanisms and potential therapeutic targets.
Collapse
Affiliation(s)
- Xiaohui Zhong
- Department of Thoracic and Cardiovascular Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Regional Medical Center for Children, Hangzhou 310052, China.
| | - Chengjie Lyu
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Regional Medical Center for Children, Hangzhou 310052, China
| | - Dengming Lai
- Department of Neonatal Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Regional Medical Center for Children, Hangzhou 310052, China
- Children's Health Innovation Research Center, Binjiang Institute of Zhejiang University, Hangzhou 310053, China
| | - Qiang Shu
- Department of Thoracic and Cardiovascular Surgery, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Regional Medical Center for Children, Hangzhou 310052, China.
| |
Collapse
|
2
|
Scheffzük C, Biedziak D, Gisch N, Goldmann T, Stamme C. Surfactant protein A modulates neuroinflammation in adult mice upon pulmonary infection. Brain Res 2024; 1840:149108. [PMID: 38964703 DOI: 10.1016/j.brainres.2024.149108] [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: 05/05/2024] [Revised: 06/17/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND One of the most common entry gates for systemic infection is the lung. In humans, pulmonary infections can lead to significant neurological impairment, ranging from acute sickness behavior to long-term disorders. Surfactant proteins (SP), essential parts of the pulmonary innate immune defense, have been detected in the brain of rats and humans. Recent evidence suggests that SP-A, the major protein component of surfactant, also plays a functional role in modulating neuroinflammation. This study aimed to determine whether SP-A deficiency affects the inflammatory response in the brain of adult mice during pulmonary infection. EXPERIMENTAL PROCEDURE Adult male wild-type (WT, n = 72) and SP-A-deficient (SP-A-/-, n = 72) mice were oropharyngeally challenged with lipopolysaccharide (LPS), Pseudomonas aeruginosa (P. aeruginosa), or PBS (control). Both, behavioral assessment and subsequent brain tissue analysis, were performed 24, 48, and 72 h after challenge. The brain concentrations of the pro-inflammatory cytokines TNF-α, IL-6, and IL-1β were determined by ELISA. Quantitative rtPCR was used to detect SP-A mRNA expression in brain homogenates and immunohistochemistry was applied for the detection of SP-A protein expression in brain coronal slices. RESULTS SP-A mRNA and histological evidence of protein expression were detected in both the lungs and brains of WT mice, with significantly higher amounts in lung samples. SP-A-/- mice exhibited significantly higher baseline concentrations of brain TNF-α, IL-6, and IL-1β compared to WT mice. Oropharyngeal application of either LPS or P. aeruginosa elicited significantly higher brain levels of TNF-α and IL-1β in SP-A-/- mice compared to WT mice at all time points. In comparison, behavioral impairment as a measure of sickness behavior, was significantly stronger in WT than in SP-A-/- mice, particularly after LPS application. CONCLUSION SP-A is known for its anti-inflammatory role in the pulmonary immune response to bacterial infection. Recent evidence suggests that in an abdominal sepsis model SP-A deficiency can lead to increased cytokine levels in the brain. Our results extend this perception and provide evidence for an anti-inflammatory role of SP-A in the brain of adult WT mice after pulmonary infection.
Collapse
Affiliation(s)
- Claudia Scheffzük
- Division of Cellular Pneumology, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany; Department of Anesthesiology and Intensive Care Medicine, University Hospital of Schleswig-Holstein, Lübeck, Germany; Department of Anesthesiology and Intensive Care Medicine, University Hospital BG Bergmannsheil Bochum, Germany.
| | - Dominika Biedziak
- Division of Cellular Pneumology, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Nicolas Gisch
- Division of Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Torsten Goldmann
- Division of Histology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany
| | - Cordula Stamme
- Division of Cellular Pneumology, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany; Department of Anesthesiology and Intensive Care Medicine, University Hospital of Schleswig-Holstein, Lübeck, Germany
| |
Collapse
|
3
|
Zhang K, Yao E, Aung T, Chuang PT. The alveolus: Our current knowledge of how the gas exchange unit of the lung is constructed and repaired. Curr Top Dev Biol 2024; 159:59-129. [PMID: 38729684 DOI: 10.1016/bs.ctdb.2024.01.002] [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] [Indexed: 05/12/2024]
Abstract
The mammalian lung completes its last step of development, alveologenesis, to generate sufficient surface area for gas exchange. In this process, multiple cell types that include alveolar epithelial cells, endothelial cells, and fibroblasts undergo coordinated cell proliferation, cell migration and/or contraction, cell shape changes, and cell-cell and cell-matrix interactions to produce the gas exchange unit: the alveolus. Full functioning of alveoli also involves immune cells and the lymphatic and autonomic nervous system. With the advent of lineage tracing, conditional gene inactivation, transcriptome analysis, live imaging, and lung organoids, our molecular understanding of alveologenesis has advanced significantly. In this review, we summarize the current knowledge of the constituents of the alveolus and the molecular pathways that control alveolar formation. We also discuss how insight into alveolar formation may inform us of alveolar repair/regeneration mechanisms following lung injury and the pathogenic processes that lead to loss of alveoli or tissue fibrosis.
Collapse
Affiliation(s)
- Kuan Zhang
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Erica Yao
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Thin Aung
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States
| | - Pao-Tien Chuang
- Cardiovascular Research Institute, University of California, San Francisco, CA, United States.
| |
Collapse
|
4
|
Gopal R, Tutuncuoglu E, Bakalov V, Wasserloos K, Li H, Lemley D, DeVito LJ, Constantinesco NJ, Reed DS, McHugh KJ, Chinnappan B, Andreas AR, Maloy A, Bain D, Alcorn JF, Pitt BR, Kaynar AM. Zinc deficiency enhances sensitivity to influenza A associated bacterial pneumonia in mice. Physiol Rep 2024; 12:e15902. [PMID: 38163670 PMCID: PMC10758336 DOI: 10.14814/phy2.15902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024] Open
Abstract
Although zinc deficiency (secondary to malnutrition) has long been considered an important contributor to morbidity and mortality of infectious disease (e.g. diarrhea disorders), epidemiologic data (including randomized controlled trials with supplemental zinc) for such a role in lower respiratory tract infection are somewhat ambiguous. In the current study, we provide the first preclinical evidence demonstrating that although diet-induced acute zinc deficiency (Zn-D: ~50% decrease) did not worsen infection induced by either influenza A (H1N1) or methicillin-resistant staph aureus (MRSA), Zn-D mice were sensitive to the injurious effects of superinfection of H1N1 with MRSA. Although the mechanism underlying the sensitivity of ZnD mice to combined H1N1/MRSA infection is unclear, it was noteworthy that this combination exacerbated lung injury as shown by lung epithelial injury markers (increased BAL protein) and decreased genes related to epithelial integrity in Zn-D mice (surfactant protein C and secretoglobins family 1A member 1). As bacterial pneumonia accounts for 25%-50% of morbidity and mortality from influenza A infection, zinc deficiency may be an important pathology component of respiratory tract infections.
Collapse
Affiliation(s)
- Radha Gopal
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Egemen Tutuncuoglu
- The Clinical Research, Investigation, and Systems Modeling of Acute Illness (CRISMA) CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Veli Bakalov
- The Clinical Research, Investigation, and Systems Modeling of Acute Illness (CRISMA) CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
Medicine InstituteAllegheny Health NetworkPittsburghPennsylvaniaUSA
| | - Karla Wasserloos
- Department of Environmental and Occupational HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
R.D. 2PortersvillePennsylvaniaUSA
| | - HuiHua Li
- Department of Environmental and Occupational HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
Department of PathologyUniversity of WisconsinMadisonWisconsinUSA
| | - David Lemley
- Department of Environmental and Occupational HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
R.D. 2PortersvillePennsylvaniaUSA
| | - Louis J. DeVito
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | | | - Douglas S. Reed
- Center for Vaccine ResearchUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Kevin J. McHugh
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Baskaran Chinnappan
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Alexis R. Andreas
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Present address:
Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Abigail Maloy
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Daniel Bain
- Department of Geology and Planetary ScienceUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - John F. Alcorn
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Bruce R. Pitt
- Department of Environmental and Occupational HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Ata Murat Kaynar
- The Clinical Research, Investigation, and Systems Modeling of Acute Illness (CRISMA) CenterUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Anesthesiology and Perioperative MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
5
|
Peers de Nieuwburgh M, Wambach JA, Griese M, Danhaive O. Towards personalized therapies for genetic disorders of surfactant dysfunction. Semin Fetal Neonatal Med 2023; 28:101500. [PMID: 38036307 PMCID: PMC10753445 DOI: 10.1016/j.siny.2023.101500] [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] [Indexed: 12/02/2023]
Abstract
Genetic disorders of surfactant dysfunction are a rare cause of chronic, progressive or refractory respiratory failure in term and preterm infants. This review explores genetic mechanisms underpinning surfactant dysfunction, highlighting specific surfactant-associated genes including SFTPB, SFTPC, ABCA3, and NKX2.1. Pathogenic variants in these genes contribute to a range of clinical presentations and courses, from neonatal hypoxemic respiratory failure to childhood interstitial lung disease and even adult-onset pulmonary fibrosis. This review emphasizes the importance of early recognition, thorough phenotype assessment, and assessment of variant functionality as essential prerequisites for treatments including lung transplantation. We explore emerging treatment options, including personalized pharmacological approaches and gene therapy strategies. In conclusion, this comprehensive review offers valuable insights into the pathogenic mechanisms of genetic disorders of surfactant dysfunction, genetic fundamentals, available and emerging therapeutic options, and underscores the need for further research to develop personalized therapies for affected infants and children.
Collapse
Affiliation(s)
- Maureen Peers de Nieuwburgh
- Division of Neonatology, Department of Pediatrics, St-Luc University Hospital, Catholic University of Louvain, Brussels, Belgium.
| | - Jennifer A Wambach
- Washington University School of Medicine/St. Louis Children's Hospital, One Children's Place, St. Louis, Missouri, USA.
| | - Matthias Griese
- Pediatric Pulmonology, Dr von Hauner Children's Hospital, University-Hospital, German Center for Lung Research (DZL), Munich, Germany.
| | - Olivier Danhaive
- Division of Neonatology, Department of Pediatrics, St-Luc University Hospital, Catholic University of Louvain, Brussels, Belgium; Division of Neonatology, Benioff Children's Hospital, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
6
|
Wang W, Peng F, Ding C, Li T, Wang H. An Analysis of Targeted Serum Lipidomics in Patients with Pneumoconiosis - China, 2022. China CDC Wkly 2023; 5:849-855. [PMID: 37814648 PMCID: PMC10560374 DOI: 10.46234/ccdcw2023.161] [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: 07/02/2023] [Accepted: 09/18/2023] [Indexed: 10/11/2023] Open
Abstract
Introduction Pneumoconiosis emerges as the most critical and prevalent occupational disease in China at present, according to research. Studies indicate that pneumoconiosis may indeed impact the body's phospholipid metabolism. Methods In this study, serum samples were taken from 46 paired participants, which included patients with pneumoconiosis and dust-exposed workers. We employed ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) technology in targeted lipidomics to investigate serum target phospholipids. Initially, a pilot study was conducted with a selection of 24 pneumoconiosis patients and 24 dust-exposed workers, using both univariate and multivariate statistical analyses to preliminarily identify significant differences in phospholipids. Subsequent to this, the remaining subjects were engaged in a validation study, wherein receiver operating characteristic (ROC) analysis was performed to further substantiate the screening potency of potential lipid biomarkers for pneumoconiosis. Results The pilot study revealed significantly reduced serum levels of 16∶0 lysophosphatidylcholines (Lyso PC), 18∶0-18∶1 phosphatidylglycerol (PG), 18∶0-18∶1 phosphatidylethanolamine (PE), 18∶0 PE, and 18∶1 lysophosphatidylethanolamine(Lyso PE) in the case group in comparison to the control group. Additionally, 18∶0 PE, 18∶0-18∶1 PE, and 18∶1 Lyso PE emerged as significant phospholipids with superior diagnostic values [area under the curve (AUC)>0.7]. A diagnostic model was established, built on 16∶0 PC and 18∶0 PE (AUC>0.8). In the ROC analyses of validation studies, the 18∶0-18∶1 PE and this diagnostic model demonstrated excellent screening efficiency (AUC>0.7). Discussion A significant divergence in phospholipid metabolism has been observed between pneumoconiosis patients and dust-exposed workers. The 18∶0-18∶1 PE present in serum could potentially function as a lipid biomarker for pneumoconiosis. Additionally, diagnostic models were developed relying on 16∶0 PC and 18∶0 PE, proving to have superior screening efficiency.
Collapse
Affiliation(s)
- Wenrong Wang
- National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Fangda Peng
- National Center for Occupational Safety and Health, Beijing, China
- National Key Laboratory for Engineering Control of Dust Hazard, Beijing, China
| | - Chunguang Ding
- National Center for Occupational Safety and Health, Beijing, China
- National Key Laboratory for Engineering Control of Dust Hazard, Beijing, China
| | - Tao Li
- National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Huanqiang Wang
- National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
7
|
Khudadah K, Ramadan A, Othman A, Refaey N, Elrosasy A, Rezkallah A, Heseba T, Moawad M, Mektebi A, Elejla S, Abouzid M, Abdelazeem B. Surfactant replacement therapy as promising treatment for COVID-19: an updated narrative review. Biosci Rep 2023; 43:BSR20230504. [PMID: 37497603 PMCID: PMC10412525 DOI: 10.1042/bsr20230504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/11/2023] [Accepted: 07/25/2023] [Indexed: 07/28/2023] Open
Abstract
Patients with COVID-19 exhibit similar symptoms to neonatal respiratory distress syndrome. SARS-CoV-2 spike protein has been shown to target alveolar type 2 lung cells which synthesize and secrete endogenous surfactants leading to acute respiratory distress syndrome in some patients. This was proven by post-mortem histopathological findings revealing desquamated alveolar type 2 cells. Surfactant use in patients with COVID-19 respiratory distress syndrome results in marked improvement in respiratory parameters but not mortality which needs further clinical trials comparing surfactant formulas and modes of administration to decrease the mortality. In addition, surfactants could be a promising vehicle for specific drug delivery as a liposomal carrier, which requires more and more challenging efforts. In this review, we highlight the current reviews and two clinical trials on exogenous surfactant therapy in COVID-19-associated respiratory distress in adults, and how surfactant could be a promising drug to help fight the COVID-19 infection.
Collapse
Affiliation(s)
| | - Alaa Ramadan
- Faculty of Medicine, South Valley University, Qena, Egypt
| | - Ahmed Othman
- Kuwait Oil Company Ahmadi Hospital, Al Ahmadi, Kuwait
| | - Neveen Refaey
- Women’s Health department, Faculty of Physical Therapy, Cairo University, Cairo, Egypt
| | - Amr Elrosasy
- Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ayoub Rezkallah
- Faculty of Medicine, University of Algeirs, Algeirs, Algeria
- Department of Hematology Laboratory and Blood Transfusion, Hospital Center University Lamine Debaghine, Algeirs, Algeria
| | - Toka Heseba
- Faculty of Medicine, Assuit University, Assuit, Egypt
| | - Mostafa Hossam El Din Moawad
- Faculty of Pharmacy, Clinical Department, Alexandria University, Egypt
- Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ammar Mektebi
- Faculty of Medicine, Kutahya Health Sciences University, Kutahya, Turkey
| | - Sewar A Elejla
- Faculty of Medicine, Alquds University, Jerusalem, Palestine
| | - Mohamed Abouzid
- Department of Physical Pharmacy and Pharmacokinetics, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Basel Abdelazeem
- McLaren Health Care, Flint, Michigan, U.S.A
- Michigan State University, East Lansing, Michigan, U.S.A
| |
Collapse
|
8
|
Yu Z, Wu T, Liu X, Chen H, Ren C, Zhu L. Resveratrol-Loaded Dipalmitoylphosphatidylcholine Liposomal Large Porous Microparticle Inhalations for the Treatment of Bacterial Pneumonia Caused by Acinetobacter baumannii. J Aerosol Med Pulm Drug Deliv 2023; 36:2-11. [PMID: 36695669 DOI: 10.1089/jamp.2021.0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Background: Acinetobacter baumannii-mediated bacterial pneumonia is a common disease that is harmful to human health. Dipalmitoylphosphatidylcholine (DPPC) is the major lipid component of the pulmonary surfactant (PS) found in the alveolar space; the PS helps to keep surface tension low, which allows for improved oxygen delivery. Resveratrol (RE) is a phytoalexin found in plants that is released in response to injury or infection. The therapeutic effect of Re is limited due to its low solubility and bioavailability. In this study, we report pulmonary delivery of Re-loaded DPPC liposomal large porous microparticles (RDLPMs) for treatment of A. baumannii-induced pneumonia. Methods: Novel RDLPMs were prepared by rotary evaporation and a freeze-drying method in this study. RDLPMs were evaluated by the particle size, electric potential, in vitro release, and particle size distribution. A rat model of A. baumannii-mediated pneumonia was established and used for pharmacodynamic evaluations. Results: The Re-loaded DPPC liposomes (RDLs) consisted of Re/DPPC (1:3, mol/mol) and DPPC/cholesterol (3:1, w/w), with a hydration time of 15 minutes. The RDLs had a high encapsulation efficiency of 69.8% ± 1.6%, a mean size of 191.5 ± 4.5 nm, and a high zeta potential of 12.4 ± 1.5 mV. The RDLPMs were composed of mannitol/large porous microparticles/RDLs (1:4:2, w/w/w) and had a loading efficiency of 2.20% ± 0.24%. The RDLPMs had an aerodynamic diameter (2.73 ± 0.65 μm), a good fluidity (28.30° ± 6.13°), and demonstrated high lung deposition (fine particle fraction = 43.33%). Surprisingly, while penicillin showed better microbial inhibition than the RDLPMs and Re groups in vitro, the RDLPMs were more effective in vivo. Conclusion: The RDLPMs showed good powder properties for pulmonary delivery. The RDLPMs may inhibit the nuclear factor kappa-B pathway and downregulate the expression of cytokines downstream of tumor necrosis factor-α and interleukin-1β. As well as, RDLPMs demonstrated some antibacterial properties against A. baumannii bacteria. Re, when delivered in RDLPMs as a dry powder inhaler, is a promising substitute for antibiotics in the treatment of A. baumannii pneumonia.
Collapse
Affiliation(s)
- Zicheng Yu
- Department of Laboratory, Institute of Clinical Pharmacy and Pharmacology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tingting Wu
- Department of Laboratory, Institute of Clinical Pharmacy and Pharmacology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoyan Liu
- Department of Pharmacy, Shanghai United Family Pudong Hospital, Shanghai, China
| | - Hongjun Chen
- Department of Laboratory, Institute of Clinical Pharmacy and Pharmacology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunxia Ren
- Department of Laboratory, Institute of Clinical Pharmacy and Pharmacology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lifei Zhu
- Department of Laboratory, Institute of Clinical Pharmacy and Pharmacology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Budhraja A, Basu A, Gheware A, Abhilash D, Rajagopala S, Pakala S, Sumit M, Ray A, Subramaniam A, Mathur P, Nambirajan A, Kumar S, Gupta R, Wig N, Trikha A, Guleria R, Sarkar C, Gupta I, Jain D. Molecular signature of postmortem lung tissue from COVID-19 patients suggests distinct trajectories driving mortality. Dis Model Mech 2022; 15:dmm049572. [PMID: 35438176 PMCID: PMC9194484 DOI: 10.1242/dmm.049572] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/07/2022] [Indexed: 12/19/2022] Open
Abstract
To elucidate the molecular mechanisms that manifest lung abnormalities during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, we performed whole-transcriptome sequencing of lung autopsies from 31 patients with severe COVID-19 and ten uninfected controls. Using metatranscriptomics, we identified the existence of two distinct molecular signatures of lethal COVID-19. The dominant 'classical' signature (n=23) showed upregulation of the unfolded protein response, steroid biosynthesis and complement activation, supported by massive metabolic reprogramming leading to characteristic lung damage. The rarer signature (n=8) that potentially represents 'cytokine release syndrome' (CRS) showed upregulation of cytokines such as IL1 and CCL19, but absence of complement activation. We found that a majority of patients cleared SARS-CoV-2 infection, but they suffered from acute dysbiosis with characteristic enrichment of opportunistic pathogens such as Staphylococcus cohnii in 'classical' patients and Pasteurella multocida in CRS patients. Our results suggest two distinct models of lung pathology in severe COVID-19 patients, which can be identified through complement activation, presence of specific cytokines and characteristic microbiome. These findings can be used to design personalized therapy using in silico identified drug molecules or in mitigating specific secondary infections.
Collapse
Affiliation(s)
- Anshul Budhraja
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi 110016, India
| | - Anubhav Basu
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi 110016, India
| | - Atish Gheware
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Dasari Abhilash
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi 110016, India
| | - Seesandra Rajagopala
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Suman Pakala
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Madhuresh Sumit
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi 110016, India
| | - Animesh Ray
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Arulselvi Subramaniam
- Department of Laboratory Medicine, JPNATC, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Purva Mathur
- Department of Laboratory Medicine, JPNATC, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Aruna Nambirajan
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sachin Kumar
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Ritu Gupta
- Laboratory Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital (IRCH), All India Institute of Medical Sciences, New Delhi 110029, India
| | - Naveet Wig
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Anjan Trikha
- Department of Anaesthesiology, Critical Care and Pain Medicine, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Randeep Guleria
- Department of Pulmonary Medicine and Sleep Disorders, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Chitra Sarkar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Ishaan Gupta
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi 110016, India
| | - Deepali Jain
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| |
Collapse
|
10
|
Nof E, Zidan H, Artzy-Schnirman A, Mouhadeb O, Beckerman M, Bhardwaj S, Elias-Kirma S, Gur D, Beth-Din A, Levenberg S, Korin N, Ordentlich A, Sznitman J. Human Multi-Compartment Airways-on-Chip Platform for Emulating Respiratory Airborne Transmission: From Nose to Pulmonary Acini. Front Physiol 2022; 13:853317. [PMID: 35350687 PMCID: PMC8957966 DOI: 10.3389/fphys.2022.853317] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/15/2022] [Indexed: 12/15/2022] Open
Abstract
The past decade has witnessed tremendous endeavors to deliver novel preclinical in vitro lung models for pulmonary research endpoints, including foremost with the advent of organ- and lung-on-chips. With growing interest in aerosol transmission and infection of respiratory viruses within a host, most notably the SARS-CoV-2 virus amidst the global COVID-19 pandemic, the importance of crosstalk between the different lung regions (i.e., extra-thoracic, conductive and respiratory), with distinct cellular makeups and physiology, are acknowledged to play an important role in the progression of the disease from the initial onset of infection. In the present Methods article, we designed and fabricated to the best of our knowledge the first multi-compartment human airway-on-chip platform to serve as a preclinical in vitro benchmark underlining regional lung crosstalk for viral infection pathways. Combining microfabrication and 3D printing techniques, our platform mimics key elements of the respiratory system spanning (i) nasal passages that serve as the alleged origin of infections, (ii) the mid-bronchial airway region and (iii) the deep acinar region, distinct with alveolated airways. Crosstalk between the three components was exemplified in various assays. First, viral-load (including SARS-CoV-2) injected into the apical partition of the nasal compartment was detected in distal bronchial and acinar components upon applying physiological airflow across the connected compartment models. Secondly, nebulized viral-like dsRNA, poly I:C aerosols were administered to the nasal apical compartment, transmitted to downstream compartments via respiratory airflows and leading to an elevation in inflammatory cytokine levels secreted by distinct epithelial cells in each respective compartment. Overall, our assays establish an in vitro methodology that supports the hypothesis for viral-laden airflow mediated transmission through the respiratory system cellular landscape. With a keen eye for broader end user applications, we share detailed methodologies for fabricating, assembling, calibrating, and using our multi-compartment platform, including open-source fabrication files. Our platform serves as an early proof-of-concept that can be readily designed and adapted to specific preclinical pulmonary research endpoints.
Collapse
Affiliation(s)
- Eliram Nof
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Hikaia Zidan
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Arbel Artzy-Schnirman
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Odelia Mouhadeb
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Margarita Beckerman
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Saurabh Bhardwaj
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Shani Elias-Kirma
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Didi Gur
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Adi Beth-Din
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Netanel Korin
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Arie Ordentlich
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Josué Sznitman
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
11
|
Milad N, Morissette MC. Revisiting the role of pulmonary surfactant in chronic inflammatory lung diseases and environmental exposure. Eur Respir Rev 2021; 30:30/162/210077. [PMID: 34911693 DOI: 10.1183/16000617.0077-2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
Pulmonary surfactant is a crucial and dynamic lung structure whose primary functions are to reduce alveolar surface tension and facilitate breathing. Though disruptions in surfactant homeostasis are typically thought of in the context of respiratory distress and premature infants, many lung diseases have been noted to have significant surfactant abnormalities. Nevertheless, preclinical and clinical studies of pulmonary disease too often overlook the potential contribution of surfactant alterations - whether in quantity, quality or composition - to disease pathogenesis and symptoms. In inflammatory lung diseases, whether these changes are cause or consequence remains a subject of debate. This review will outline 1) the importance of pulmonary surfactant in the maintenance of respiratory health, 2) the diseases associated with primary surfactant dysregulation, 3) the surfactant abnormalities observed in inflammatory pulmonary diseases and, finally, 4) the available research on the interplay between surfactant homeostasis and smoking-associated lung disease. From these published studies, we posit that changes in surfactant integrity and composition contribute more considerably to chronic inflammatory pulmonary diseases and that more work is required to determine the mechanisms underlying these alterations and their potential treatability.
Collapse
Affiliation(s)
- Nadia Milad
- Faculty of Medicine, Université Laval, Quebec City, QC, Canada.,Quebec Heart and Lung Institute - Université Laval, Quebec City, QC, Canada
| | - Mathieu C Morissette
- Quebec Heart and Lung Institute - Université Laval, Quebec City, QC, Canada .,Dept of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
12
|
Sriram K, Insel MB, Insel PA. Inhaled β2 Adrenergic Agonists and Other cAMP-Elevating Agents: Therapeutics for Alveolar Injury and Acute Respiratory Disease Syndrome? Pharmacol Rev 2021; 73:488-526. [PMID: 34795026 DOI: 10.1124/pharmrev.121.000356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/15/2021] [Indexed: 12/15/2022] Open
Abstract
Inhaled long-acting β-adrenergic agonists (LABAs) and short-acting β-adrenergic agonists are approved for the treatment of obstructive lung disease via actions mediated by β2 adrenergic receptors (β2-ARs) that increase cellular cAMP synthesis. This review discusses the potential of β2-AR agonists, in particular LABAs, for the treatment of acute respiratory distress syndrome (ARDS). We emphasize ARDS induced by pneumonia and focus on the pathobiology of ARDS and actions of LABAs and cAMP on pulmonary and immune cell types. β2-AR agonists/cAMP have beneficial actions that include protection of epithelial and endothelial cells from injury, restoration of alveolar fluid clearance, and reduction of fibrotic remodeling. β2-AR agonists/cAMP also exert anti-inflammatory effects on the immune system by actions on several types of immune cells. Early administration is likely critical for optimizing efficacy of LABAs or other cAMP-elevating agents, such as agonists of other Gs-coupled G protein-coupled receptors or cyclic nucleotide phosphodiesterase inhibitors. Clinical studies that target lung injury early, prior to development of ARDS, are thus needed to further assess the use of inhaled LABAs, perhaps combined with inhaled corticosteroids and/or long-acting muscarinic cholinergic antagonists. Such agents may provide a multipronged, repurposing, and efficacious therapeutic approach while minimizing systemic toxicity. SIGNIFICANCE STATEMENT: Acute respiratory distress syndrome (ARDS) after pulmonary alveolar injury (e.g., certain viral infections) is associated with ∼40% mortality and in need of new therapeutic approaches. This review summarizes the pathobiology of ARDS, focusing on contributions of pulmonary and immune cell types and potentially beneficial actions of β2 adrenergic receptors and cAMP. Early administration of inhaled β2 adrenergic agonists and perhaps other cAMP-elevating agents after alveolar injury may be a prophylactic approach to prevent development of ARDS.
Collapse
Affiliation(s)
- Krishna Sriram
- Departments of Pharmacology (K.S., P.A.I.) and Medicine (P.A.I.), University of California San Diego, La Jolla, California; Department of Medicine (M.B.I.) University of Arizona, Tucson, Arizona
| | - Michael B Insel
- Departments of Pharmacology (K.S., P.A.I.) and Medicine (P.A.I.), University of California San Diego, La Jolla, California; Department of Medicine (M.B.I.) University of Arizona, Tucson, Arizona
| | - Paul A Insel
- Departments of Pharmacology (K.S., P.A.I.) and Medicine (P.A.I.), University of California San Diego, La Jolla, California; Department of Medicine (M.B.I.) University of Arizona, Tucson, Arizona
| |
Collapse
|
13
|
Alketbi EH, Hamdy R, El‐Kabalawy A, Juric V, Pignitter M, A. Mosa K, Almehdi AM, El‐Keblawy AA, Soliman SSM. Lipid-based therapies against SARS-CoV-2 infection. Rev Med Virol 2021; 31:1-13. [PMID: 34546604 PMCID: PMC8013851 DOI: 10.1002/rmv.2214] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022]
Abstract
Viruses have evolved to manipulate host lipid metabolism to benefit their replication cycle. Enveloped viruses, including coronaviruses, use host lipids in various stages of the viral life cycle, particularly in the formation of replication compartments and envelopes. Host lipids are utilised by the virus in receptor binding, viral fusion and entry, as well as viral replication. Association of dyslipidaemia with the pathological development of Covid-19 raises the possibility that exploitation of host lipid metabolism might have therapeutic benefit against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In this review, promising host lipid targets are discussed along with potential inhibitors. In addition, specific host lipids are involved in the inflammatory responses due to viral infection, so lipid supplementation represents another potential strategy to counteract the severity of viral infection. Furthermore, switching the lipid metabolism through a ketogenic diet is another potential way of limiting the effects of viral infection. Taken together, restricting the access of host lipids to the virus, either by using lipid inhibitors or supplementation with exogenous lipids, might significantly limit SARS-CoV-2 infection and/or severity.
Collapse
Affiliation(s)
- Eman Humaid Alketbi
- Department of Applied BiologyCollege of SciencesUniversity of SharjahSharjahUnited Arab Emirates
| | - Rania Hamdy
- Research Institute for Medical and Health SciencesUniversity of SharjahSharjahUnited Arab Emirates
- Faculty of PharmacyZagazig UniversityZagazigEgypt
| | | | - Viktorija Juric
- Department of Physiological ChemistryFaculty of ChemistryUniversity of ViennaViennaAustria
| | - Marc Pignitter
- Department of Physiological ChemistryFaculty of ChemistryUniversity of ViennaViennaAustria
| | - Kareem A. Mosa
- Department of Applied BiologyCollege of SciencesUniversity of SharjahSharjahUnited Arab Emirates
- Research Institute of Science and EngineeringUniversity of SharjahSharjahUnited Arab Emirates
- Department of BiotechnologyFaculty of AgricultureAl‐Azhar UniversityCairoEgypt
| | - Ahmed M. Almehdi
- Department of ChemistryCollege of SciencesUniversity of SharjahSharjahUnited Arab Emirates
| | - Ali A. El‐Keblawy
- Department of Applied BiologyCollege of SciencesUniversity of SharjahSharjahUnited Arab Emirates
- Research Institute of Science and EngineeringUniversity of SharjahSharjahUnited Arab Emirates
| | - Sameh S. M. Soliman
- Research Institute for Medical and Health SciencesUniversity of SharjahSharjahUnited Arab Emirates
- Faculty of PharmacyZagazig UniversityZagazigEgypt
- Department of Medicinal ChemistryCollege of PharmacyUniversity of SharjahSharjahUnited Arab Emirates
| |
Collapse
|
14
|
Bocking T, Johnson L, Singh A, Desai A, Aulakh GK, Singh B. Research article expression of surfactant protein-A and D, and CD9 in lungs of 1 and 30 day old foals. BMC Vet Res 2021; 17:236. [PMID: 34225699 PMCID: PMC8256609 DOI: 10.1186/s12917-021-02943-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/23/2021] [Indexed: 11/15/2022] Open
Abstract
Background Respiratory diseases are a major cause of morbidity and mortality in the horses of all ages including foals. There is limited understanding of the expression of immune molecules such as tetraspanins and surfactant proteins (SP) and the regulation of the immune responses in the lungs of the foals. Therefore, the expression of CD9, SP-A and SP-D in foal lungs was examined. Results Lungs from one day old (n = 6) and 30 days old (n = 5) foals were examined for the expression of CD9, SP-A, and SP-D with immunohistology and Western blots. Western blot data showed significant increase in the amount of CD9 protein (p = 0.0397) but not of SP-A and SP-D at 30 days of age compared to one day. Immunohistology detected CD9 in the alveolar septa and vascular endothelium but not the bronchiolar epithelium in the lungs of the foals in both age groups. SP-A and SP-D expression was localized throughout the alveolar septa including type II alveolar epithelial cells and the vascular endothelium of the lungs in all the foals. Compared to one day old foals, the expression of SP-A and SP-D appeared to be increased in the bronchiolar epithelium of 30 day old foals. Pulmonary intravascular macrophages were also positive for SP-A and SP-D in 30 days old foals and these cells are not developed in the day old foals. Conclusions This is the first data on the expression of CD9, SP-A and SP-D in the lungs of foals. Supplementary Information The online version contains supplementary material available at 10.1186/s12917-021-02943-5.
Collapse
Affiliation(s)
- Tara Bocking
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Laura Johnson
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Amitoj Singh
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Atul Desai
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Gurpreet Kaur Aulakh
- Department of Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Baljit Singh
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada.
| |
Collapse
|
15
|
Bocking T, Singh B. Light and electron-microscopic localization of CD9 and surfactant protein A and D in normal lungs of the horse. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2021; 85:170-176. [PMID: 34248260 PMCID: PMC8243805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 02/05/2021] [Indexed: 06/13/2023]
Abstract
The lung is a complex organ, and its physiology and immunology are regulated by various immune molecules and cells. Lung surfactant, a mixture of phospholipids and proteins produced by the bronchiolar and type II alveolar epithelial cells, is one such important player in lung physiology. Compared to knowledge about the biology of the surfactant in rodents and humans, only limited data are available on the surfactant in the horse. Although there are data linking levels of surfactant proteins with respiratory disease in the horse, there are no data on the cellular localization of surfactant protein A (SP-A) and surfactant protein D (SP-D). A member of the tetraspanin family of proteins, CD9 is a cell-signaling and adhesion protein and its expression has been detected in both normal and cancer cells, including those in the lung. Because there are no immunolocalization data on SP-A, SP-D, and CD9 in the normal lungs of the horse, our objective was to conduct a light and electron microscopic immunocytochemical study on normal lungs of the horse. The data showed SP-A and SP-D in bronchiolar epithelial and type II alveolar epithelial cells. These proteins were also localized in type I alveolar epithelial cells, pulmonary intravascular macrophages, and neutrophils, which is likely an outcome of endocytosis of the proteins by these cells. CD9 was present in the airway and vascular smooth muscle cells, endothelium, and blood cells, but not in the airway epithelium. These new data provide a baseline to further examine the expression and functions of SP-A, SP-D, and CD9 proteins in inflammation associated with respiratory diseases in the horse.
Collapse
Affiliation(s)
- Tara Bocking
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan S7N 5B4
| | - Baljit Singh
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan S7N 5B4
| |
Collapse
|
16
|
Seyfoori A, Shokrollahi Barough M, Mokarram P, Ahmadi M, Mehrbod P, Sheidary A, Madrakian T, Kiumarsi M, Walsh T, McAlinden KD, Ghosh CC, Sharma P, Zeki AA, Ghavami S, Akbari M. Emerging Advances of Nanotechnology in Drug and Vaccine Delivery against Viral Associated Respiratory Infectious Diseases (VARID). Int J Mol Sci 2021; 22:6937. [PMID: 34203268 PMCID: PMC8269337 DOI: 10.3390/ijms22136937] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/19/2021] [Accepted: 06/19/2021] [Indexed: 12/12/2022] Open
Abstract
Viral-associated respiratory infectious diseases are one of the most prominent subsets of respiratory failures, known as viral respiratory infections (VRI). VRIs are proceeded by an infection caused by viruses infecting the respiratory system. For the past 100 years, viral associated respiratory epidemics have been the most common cause of infectious disease worldwide. Due to several drawbacks of the current anti-viral treatments, such as drug resistance generation and non-targeting of viral proteins, the development of novel nanotherapeutic or nano-vaccine strategies can be considered essential. Due to their specific physical and biological properties, nanoparticles hold promising opportunities for both anti-viral treatments and vaccines against viral infections. Besides the specific physiological properties of the respiratory system, there is a significant demand for utilizing nano-designs in the production of vaccines or antiviral agents for airway-localized administration. SARS-CoV-2, as an immediate example of respiratory viruses, is an enveloped, positive-sense, single-stranded RNA virus belonging to the coronaviridae family. COVID-19 can lead to acute respiratory distress syndrome, similarly to other members of the coronaviridae. Hence, reviewing the current and past emerging nanotechnology-based medications on similar respiratory viral diseases can identify pathways towards generating novel SARS-CoV-2 nanotherapeutics and/or nano-vaccines.
Collapse
Affiliation(s)
- Amir Seyfoori
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
- Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran
| | - Mahdieh Shokrollahi Barough
- Department of Immunology, Iran University of Medical Sciences, Tehran 1449614535, Iran;
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran
| | - Pooneh Mokarram
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran;
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (M.A.); (T.M.)
| | - Parvaneh Mehrbod
- Influenza and Respiratory Viruses Department, Pasteur Institute of IRAN, Tehran 1316943551, Iran;
| | - Alireza Sheidary
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14155-6451, Iran;
| | - Tayyebeh Madrakian
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (M.A.); (T.M.)
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14155-6451, Iran;
| | - Mohammad Kiumarsi
- Department of Human Anatomy and Cell Science, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
| | - Tavia Walsh
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
| | - Kielan D. McAlinden
- Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS 7248, Australia;
| | - Chandra C. Ghosh
- Roger Williams Medical Center, Immuno-Oncology Institute (Ix2), Providence, RI 02908, USA;
| | - Pawan Sharma
- Center for Translational Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Amir A. Zeki
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, U.C. Davis Lung Center, Davis School of Medicine, University of California, Davis, CA 95817, USA;
- Veterans Affairs Medical Center, Mather, CA 95817, USA
| | - Saeid Ghavami
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Department of Human Anatomy and Cell Science, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
- Biotechnology Center, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
- Center for Advanced Materials and Related Technologies, University of Victoria, Victoria, BC V8P 5C2, Canada
| |
Collapse
|
17
|
Suresh V, Mohanty V, Avula K, Ghosh A, Singh B, Reddy RK, Parida D, Suryawanshi AR, Raghav SK, Chattopadhyay S, Prasad P, Swain RK, Dash R, Parida A, Syed GH, Senapati S. Quantitative proteomics of hamster lung tissues infected with SARS-CoV-2 reveal host factors having implication in the disease pathogenesis and severity. FASEB J 2021; 35:e21713. [PMID: 34105201 PMCID: PMC8206718 DOI: 10.1096/fj.202100431r] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/07/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022]
Abstract
Syrian golden hamsters (Mesocricetus auratus) infected by severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) manifests lung pathology. In this study, efforts were made to check the infectivity of a local SARS‐CoV‐2 isolate in a self‐limiting and non‐lethal hamster model and evaluate the differential expression of lung proteins during acute infection and convalescence. The findings of this study confirm the infectivity of this isolate in vivo. Analysis of clinical parameters and tissue samples show the pathophysiological manifestation of SARS‐CoV‐2 infection similar to that reported earlier in COVID‐19 patients and hamsters infected with other isolates. However, diffuse alveolar damage (DAD), a common histopathological feature of human COVID‐19 was only occasionally noticed. The lung‐associated pathological changes were very prominent on the 4th day post‐infection (dpi), mostly resolved by 14 dpi. Here, we carried out the quantitative proteomic analysis of the lung tissues from SARS‐CoV‐2‐infected hamsters on day 4 and day 14 post‐infection. This resulted in the identification of 1585 proteins of which 68 proteins were significantly altered between both the infected groups. Pathway analysis revealed complement and coagulation cascade, platelet activation, ferroptosis, and focal adhesion as the top enriched pathways. In addition, we also identified altered expression of two pulmonary surfactant‐associated proteins (Sftpd and Sftpb), known for their protective role in lung function. Together, these findings will aid in understanding the mechanism(s) involved in SARS‐CoV‐2 pathogenesis and progression of the disease.
Collapse
Affiliation(s)
- Voddu Suresh
- Institute of Life Sciences, Bhubaneswar, India.,Regional Centre for Biotechnology, Faridabad, India
| | | | - Kiran Avula
- Institute of Life Sciences, Bhubaneswar, India.,Regional Centre for Biotechnology, Faridabad, India
| | - Arup Ghosh
- Institute of Life Sciences, Bhubaneswar, India.,Kalinga Institute of Industrial Technology, Bhubaneswar, India
| | - Bharati Singh
- Institute of Life Sciences, Bhubaneswar, India.,Kalinga Institute of Industrial Technology, Bhubaneswar, India
| | | | - Deepti Parida
- Institute of Life Sciences, Bhubaneswar, India.,Regional Centre for Biotechnology, Faridabad, India
| | | | | | | | | | | | - Rupesh Dash
- Institute of Life Sciences, Bhubaneswar, India
| | - Ajay Parida
- Institute of Life Sciences, Bhubaneswar, India
| | | | | |
Collapse
|
18
|
Milad N, Pineault M, Lechasseur A, Routhier J, Beaulieu MJ, Aubin S, Morissette MC. Neutrophils and IL-1α Regulate Surfactant Homeostasis during Cigarette Smoking. THE JOURNAL OF IMMUNOLOGY 2021; 206:1923-1931. [PMID: 33722877 DOI: 10.4049/jimmunol.2001182] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 02/08/2021] [Indexed: 11/19/2022]
Abstract
Cigarette smoke exposure induces inflammation marked by rapid and sustained neutrophil infiltration, IL-1α, release and altered surfactant homeostasis. However, the extent to which neutrophils and IL-1α contribute to the maintenance of pulmonary surfactant homeostasis is not well understood. We sought to investigate whether neutrophils play a role in surfactant clearance as well as the effect of neutrophil depletion and IL-1α blockade on the response to cigarette smoke exposure. In vitro and in vivo administration of fluorescently labeled surfactant phosphatidylcholine was used to assess internalization of surfactant by lung neutrophils and macrophages during or following cigarette smoke exposure in mice. We also depleted neutrophils using anti-Ly-6G or anti-Gr-1 Abs, or we neutralized IL-1α using a blocking Ab to determine their respective roles in regulating surfactant homeostasis during cigarette smoke exposure. We observed that neutrophils actively internalize labeled surfactant both in vitro and in vivo and that IL-1α is required for smoke-induced elevation of surfactant protein (SP)-A and SP-D levels. Neutrophil depletion during cigarette smoke exposure led to a further increase in SP-A levels in the bronchoalveolar lavage and increased IL-1α, CCL2, GM-CSF, and G-CSF release. Finally, macrophage expression of Mmp12, a protease linked to emphysema, was increased in neutrophil-depleted groups and decreased following IL-1α blockade. Taken together, our results indicate that neutrophils and IL-1α signaling are actively involved in surfactant homeostasis and that the absence of neutrophils in the lungs during cigarette smoke exposure leads to an IL-1α-dependent exacerbation of the inflammatory response.
Collapse
Affiliation(s)
- Nadia Milad
- Québec Heart and Lung Institute-Université Laval, Quebec City, Quebec G1V 4G5, Canada.,Faculty of Medicine, Université Laval, Quebec City, Quebec G1V 0A6, Canada; and
| | - Marie Pineault
- Québec Heart and Lung Institute-Université Laval, Quebec City, Quebec G1V 4G5, Canada.,Faculty of Medicine, Université Laval, Quebec City, Quebec G1V 0A6, Canada; and
| | - Ariane Lechasseur
- Québec Heart and Lung Institute-Université Laval, Quebec City, Quebec G1V 4G5, Canada.,Faculty of Medicine, Université Laval, Quebec City, Quebec G1V 0A6, Canada; and
| | - Joanie Routhier
- Québec Heart and Lung Institute-Université Laval, Quebec City, Quebec G1V 4G5, Canada
| | - Marie-Josée Beaulieu
- Québec Heart and Lung Institute-Université Laval, Quebec City, Quebec G1V 4G5, Canada
| | - Sophie Aubin
- Québec Heart and Lung Institute-Université Laval, Quebec City, Quebec G1V 4G5, Canada
| | - Mathieu C Morissette
- Québec Heart and Lung Institute-Université Laval, Quebec City, Quebec G1V 4G5, Canada; .,Department of Medicine, Université Laval, Quebec City, Quebec G1V 0A6, Canada
| |
Collapse
|
19
|
Bastola R, Young PM, Das SC. Simulation of respiratory tract lining fluid for in vitro dissolution study. Expert Opin Drug Deliv 2021; 18:1091-1100. [PMID: 33504235 DOI: 10.1080/17425247.2021.1882991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Drug particles inhaled via the respiratory system must first dissolve in the respiratory tract lining fluid (RTLF) that lies on the surfaces of airways and alveoli, so that they are absorbed and have therapeutic action. Artificial simulated RTLFs are often used for in vitro dissolution studies to determine the solubility and dissolution of inhaled drug particles. Such studies can be used to predict bioavailability minimizing the requirement for in vivo studies. Numerous studies have been conducted to develop bio-relevant simulated RTLFs; however, to date, there is no singular simulated RTLF that closely resembles human RTLF.Areas covered: This review focuses on the composition of natural and simulated RTLFs and their use in in vitro dissolution studies.Expert opinion: There is variation in the composition and thickness of RTLF along the respiratory tract. Identification of the actual concentration of components of endogenous RTLF present in different areas of the respiratory tract helps in the development of region-specific simulated RTLFs. It is recommended that region-specific simulated RTLFs can be prepared by varying concentration of major RTLF components like mucus/gel simulants, lipids/surfactants, peptides/proteins, and inorganic/organic salts.
Collapse
Affiliation(s)
- Rakesh Bastola
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Paul M Young
- Woolcock Institute of Medical Research and Discipline of Pharmacology, Sydney Medical School, Glebe, Australia
| | - Shyamal C Das
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
20
|
Vago JP, Tavares LP, Riccardi C, Teixeira MM, Sousa LP. Exploiting the pro-resolving actions of glucocorticoid-induced proteins Annexin A1 and GILZ in infectious diseases. Biomed Pharmacother 2020; 133:111033. [PMID: 33378946 DOI: 10.1016/j.biopha.2020.111033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/10/2020] [Accepted: 11/15/2020] [Indexed: 02/08/2023] Open
Abstract
For decades, glucocorticoids (GC) have been used to treat several inflammatory conditions, including chronic and autoimmune diseases, due to their potent anti-inflammatory properties. In the context of infectious diseases, the use of GCs may be effective as adjuvant to antibiotic therapy by controlling excessive inflammatory responses resulting in better outcome in some cases. However, the use of GCs has been associated with a vast number of side effects, including increased probability of immunosuppression and consequent risk of opportunistic infection. Glucocorticoid-induced leucine zipper (GILZ) and Annexin A1 (AnxA1) are GC-induced proteins intrinsically involved with the anti-inflammatory functions of GCs without the associated adverse metabolic effects. Recent studies have shown that these GC-proteins exhibit pro-resolving effects. An essential characteristic of pro-resolving molecules is their ability to coordinate the resolution of inflammation and promote host defense in most experimental models of infection. Although the role of GILZ and AnxA1 in the context of infectious diseases remain to be better explored, herein we provide an overview of the emerging functions of these GC-proteins obtained from pre-clinical models of infectious diseases.
Collapse
Affiliation(s)
- Juliana P Vago
- Signaling in Inflammation Laboratory, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Luciana P Tavares
- Signaling in Inflammation Laboratory, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Carlo Riccardi
- Departament of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Signaling in Inflammation Laboratory, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
21
|
Yu X, Xu J, Liu W, Zhang Z, He C, Xu W. Protective effects of pulmonary surfactant on decompression sickness in rats. J Appl Physiol (1985) 2020; 130:400-407. [PMID: 33270509 DOI: 10.1152/japplphysiol.00807.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Decompression sickness (DCS) is a systemic pathophysiological process featured by bubble load. Lung dysfunction plays a harmful effect on off-gassing, which contributes to bubble load and subsequent DCS occurrence. This study aimed to investigate the effects of pulmonary surfactant on DCS as it possesses multiple advantages on the lung. Rats were divided into three groups: the normal (n = 10), the surfactant (n = 36), and the saline (n = 36) group. Animals in surfactant or saline group were administered aerosol surfactant or saline 12 h before a stimulated diving, respectively. Signs of DCS were recorded and bubble load was detected. The contents of phospholipid and surfactant protein A (SPA), protein, IL-1 and IL-6 in bronchoalveolar lavage fluid (BALF), and lung wet/dry (W/D) ratio were determined. Serum levels of IL-6, ICAM-1, E-selectin, GSH, and GSSG were detected. In surfactant-treated rats, the morbidity and mortality of DCS markedly decreased (P < 0.01 and P < 0.05, respectively). Survival time prolonged and the latency to DCS dramatically delayed (P < 0.01). More importantly, bubble load markedly decreased (P < 0.01). The increases of protein, IL-1 and IL-6 in BALF, and lung W/D ratio were alleviated. Restoration of total phospholipid and SPA in BALF and ICAM-1 and E-selectin in serum was observed. The inflammation and oxidation were attenuated (P < 0.01). In conclusion, prediving administrating exogenous surfactant by aerosolization is an efficient, simple, and safe method for DCS prevention in rats.NEW & NOTEWORTHY This is the first study exploring the effects of aerosol surfactant on DCS prevention and it was proven to be an efficient and simple method. The role of surfactant in facilitating off-gassing was thought to be the critical mechanism in bubble degrading and subsequent DCS prevention.
Collapse
Affiliation(s)
- Xuhua Yu
- Department of Diving and Hyperbaric Medicine, Naval Special Medical Center, Naval Medical University, Shanghai, China
| | - Jiajun Xu
- Department of Diving and Hyperbaric Medicine, Naval Special Medical Center, Naval Medical University, Shanghai, China
| | - Wenwu Liu
- Department of Diving and Hyperbaric Medicine, Naval Special Medical Center, Naval Medical University, Shanghai, China
| | - Ze Zhang
- The 17th detachment of the frigate, Jiangmen, China
| | - Chunyang He
- Department of Hyperbaric Oxygen, General Hospital in Western Theater of Operations, Chengdu, China
| | - Weigang Xu
- Department of Diving and Hyperbaric Medicine, Naval Special Medical Center, Naval Medical University, Shanghai, China
| |
Collapse
|
22
|
Islam ABMMK, Khan MAAK. Lung transcriptome of a COVID-19 patient and systems biology predictions suggest impaired surfactant production which may be druggable by surfactant therapy. Sci Rep 2020; 10:19395. [PMID: 33173052 PMCID: PMC7656460 DOI: 10.1038/s41598-020-76404-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
An incomplete understanding of the molecular mechanisms behind impairment of lung pathobiology by COVID-19 complicates its clinical management. In this study, we analyzed the gene expression pattern of cells obtained from biopsies of COVID-19-affected patient and compared to the effects observed in typical SARS-CoV-2 and SARS-CoV-infected cell-lines. We then compared gene expression patterns of COVID-19-affected lung tissues and SARS-CoV-2-infected cell-lines and mapped those to known lung-related molecular networks, including hypoxia induced responses, lung development, respiratory processes, cholesterol biosynthesis and surfactant metabolism; all of which are suspected to be downregulated following SARS-CoV-2 infection based on the observed symptomatic impairments. Network analyses suggest that SARS-CoV-2 infection might lead to acute lung injury in COVID-19 by affecting surfactant proteins and their regulators SPD, SPC, and TTF1 through NSP5 and NSP12; thrombosis regulators PLAT, and EGR1 by ORF8 and NSP12; and mitochondrial NDUFA10, NDUFAF5, and SAMM50 through NSP12. Furthermore, hypoxia response through HIF-1 signaling might also be targeted by SARS-CoV-2 proteins. Drug enrichment analysis of dysregulated genes has allowed us to propose novel therapies, including lung surfactants, respiratory stimulants, sargramostim, and oseltamivir. Our study presents a distinct mechanism of probable virus induced lung damage apart from cytokine storm.
Collapse
|
23
|
Bollag WB, Gonzales JN. Phosphatidylglycerol and surfactant: A potential treatment for COVID-19? Med Hypotheses 2020; 144:110277. [PMID: 33254581 PMCID: PMC7493731 DOI: 10.1016/j.mehy.2020.110277] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/11/2020] [Accepted: 09/12/2020] [Indexed: 01/08/2023]
Abstract
A hypothesis concerning the potential utility of surfactant supplementation for the treatment of critically ill patients with COVID-19 is proposed, along with a brief summary of the data in the literature supporting this idea. It is thought that surfactant, which is already approved by the Food and Drug Administration for intratracheal administration to treat neonatal respiratory distress syndrome in pre-term infants, could benefit COVID-19-infected individuals by: (1) restoring surfactant damaged by lung infection and/or decreased due to the virus-induced death of the type II pneumocytes that produce it and (2) reducing surface tension to decrease the work of breathing and limit pulmonary edema. In addition, a constituent of surfactant, phosphatidylglycerol, could mitigate COVID-19-induced lung pathology by: (3) decreasing excessive innate immune system stimulation via its inhibition of toll-like receptor-2 and -4 activation by microbial components and cellular proteins released by damaged cells, thereby limiting inflammation and the resultant pulmonary edema, and (4) possibly blocking spread of the viral infection to non-infected cells in the lung. Therefore, it is suggested that surfactant preparations containing phosphatidylglycerol be tested for their ability to improve lung function in critically ill patients with COVID-19.
Collapse
Affiliation(s)
- Wendy B Bollag
- Charlie Norwood VA Medical Center, Augusta, GA 30904, United States; Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA 30912, United States; Department of Dermatology, Medical College of Georgia at Augusta University, Augusta, GA 30912, United States; Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, United States.
| | - Joyce N Gonzales
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, United States
| |
Collapse
|
24
|
Pramod K, Kotta S, Jijith US, Aravind A, Abu Tahir M, Manju CS, Gangadharappa HV. Surfactant-based prophylaxis and therapy against COVID-19: A possibility. Med Hypotheses 2020; 143:110081. [PMID: 32653736 PMCID: PMC7340033 DOI: 10.1016/j.mehy.2020.110081] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/06/2020] [Accepted: 07/03/2020] [Indexed: 12/17/2022]
Abstract
Hand hygiene by washing with soap and water is recommended for the prevention of COVID-19 spread. Soaps and detergents are explained to act by damaging viral spike glycoproteins (peplomers) or by washing out the virus through entrapment in the micelles. Technically, soaps come under a functional category of molecules known as surfactants. Surfactants are widely used in pharmaceutical formulations as excipients. We wonder why surfactants are still not tried for prophylaxis or therapy against COVID-19? That too when many of them have proven antiviral properties. Moreover, lung surfactants have already shown benefits in respiratory viral infections. Therefore, we postulate that surfactant-based prophylaxis and therapy would be promising. We believe that our hypothesis would stimulate debate or new research exploring the possibility of surfactant-based prophylaxis and therapy against COVID-19. The success of a surfactant-based technique would save the world from any such pandemic in the future too.
Collapse
Affiliation(s)
- K Pramod
- College of Pharmaceutical Sciences, Govt. Medical College, Kozhikode, Kerala, India.
| | - Sabna Kotta
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Saudi Arabia
| | - U S Jijith
- College of Pharmaceutical Sciences, Govt. Medical College, Kozhikode, Kerala, India
| | - A Aravind
- College of Pharmaceutical Sciences, Govt. Medical College, Thiruvananthapuram, Kerala, India
| | - M Abu Tahir
- Formulation & Development, Steril-gene Life Sciences, Puducherry, India
| | - C S Manju
- College of Pharmaceutical Sciences, Govt. Medical College, Kozhikode, Kerala, India
| | - H V Gangadharappa
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Sri Shivarathreshwara Nagar, Bannimantap, Mysuru 570015, Karnataka, India
| |
Collapse
|
25
|
Kamga Gninzeko FJ, Valentine MS, Tho CK, Chindal SR, Boc S, Dhapare S, Momin MAM, Hassan A, Hindle M, Farkas DR, Longest PW, Heise RL. Excipient Enhanced Growth Aerosol Surfactant Replacement Therapy in an In Vivo Rat Lung Injury Model. J Aerosol Med Pulm Drug Deliv 2020; 33:314-322. [PMID: 32453638 DOI: 10.1089/jamp.2020.1593] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: In neonatal respiratory distress syndrome, breathing support and surfactant therapy are commonly used to enable the alveoli to expand. Surfactants are typically delivered through liquid instillation. However, liquid instillation does not specifically target the small airways. We have developed an excipient enhanced growth (EEG) powder aerosol formulation using Survanta®. Methods: EEG Survanta powder aerosol was delivered using a novel dry powder inhaler via tracheal insufflation to surfactant depleted rats at nominal doses of 3, 5, 10, and 20 mg of powder containing 0.61, 0.97, 1.73, and 3.46 mg of phospholipids (PL), whereas liquid Survanta was delivered via syringe instillation at doses of 2 and 4 mL/kg containing 18.6 and 34 mg of PL. Ventilation mechanics were measured before and after depletion, and after treatment. We hypothesized that EEG Survanta powder aerosol would improve lung mechanics compared with instilled liquid Survanta in surfactant depleted rats. Results and Conclusion: EEG Survanta powder aerosol at a dose of 0.61 mg PL significantly improved lung compliance and elastance compared with the liquid Survanta at a dose of 18.6 mg, which represents improved primary efficacy of the aerosol at a 30-fold lower dose of PL. There was no significant difference in white blood cell count of the lavage from the EEG Survanta group compared with liquid Survanta. These results provide an in vivo proof-of-concept for EEG Survanta powder aerosol as a promising method of surfactant replacement therapy.
Collapse
Affiliation(s)
- Franck J Kamga Gninzeko
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Michael S Valentine
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Cindy K Tho
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Sahil R Chindal
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Susan Boc
- Department of Pharmaceutics, and Virginia Commonwealth University, Richmond, Virginia, USA
| | - Sneha Dhapare
- Department of Pharmaceutics, and Virginia Commonwealth University, Richmond, Virginia, USA
| | | | - Amr Hassan
- Department of Pharmaceutics, and Virginia Commonwealth University, Richmond, Virginia, USA
| | - Michael Hindle
- Department of Pharmaceutics, and Virginia Commonwealth University, Richmond, Virginia, USA
| | - Dale R Farkas
- Department of Mechanical and Nuclear Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - P Worth Longest
- Department of Mechanical and Nuclear Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Rebecca L Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
26
|
Deinhardt-Emmer S, Rennert K, Schicke E, Cseresnyés Z, Windolph M, Nietzsche S, Heller R, Siwczak F, Haupt KF, Carlstedt S, Schacke M, Figge MT, Ehrhardt C, Löffler B, Mosig AS. Co-infection with Staphylococcus aureus after primary influenza virus infection leads to damage of the endothelium in a human alveolus-on-a-chip model. Biofabrication 2020; 12:025012. [PMID: 31994489 DOI: 10.1088/1758-5090/ab7073] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pneumonia is one of the most common infectious diseases worldwide. The influenza virus can cause severe epidemics, which results in significant morbidity and mortality. Beyond the virulence of the virus itself, epidemiological data suggest that bacterial co-infections are the major cause of increased mortality. In this context, Staphylococcus aureus represents a frequent causative bacterial pathogen. Currently available models have several limitations in the analysis of the pathogenesis of infections, e.g. some bacterial toxins strongly act in a species-specific manner. Human 2D mono-cell culture models often fail to maintain the differentiation of alveolus-specific functions. A detailed investigation of the underlying pathogenesis mechanisms requires a physiological interaction of alveolus-specific cell types. The aim of the present work was to establish a human in vitro alveolus model system composed of vascular and epithelial cell structures with cocultured macrophages resembling the human alveolus architecture and functions. We demonstrate that high barrier integrity maintained for up to 14 d in our model containing functional tissue-resident macrophages. We show that flow conditions and the presence of macrophages increased the barrier function. The infection of epithelial cells induced a high inflammatory response that spread to the endothelium. Although the integrity of the epithelium was not compromised by a single infection or co-infection, we demonstrated significant endothelial cell damage associated with loss of barrier function. We established a novel immune-responsive model that reflects the complex crosstalk between pathogens and host. The in vitro model allows for the monitoring of spatiotemporal spreading of the pathogens and the characterization of morphological and functional alterations attributed to infection. The alveolus-on-a-chip represents a promising platform for mechanistic studies of host-pathogen interactions and the identification of molecular and cellular targets of novel treatment strategies in pneumonia.
Collapse
Affiliation(s)
- Stefanie Deinhardt-Emmer
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, D-07747 Jena, Germany. Center for Sepsis Control and Care, Jena University Hospital, D-07747 Jena, Germany. Section of Experimental Virology, Institute of Medical Microbiology, Jena University Hospital, Hans-Knöll-Str. 2, D-07745, Jena, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Agudelo CW, Kumley BK, Area-Gomez E, Xu Y, Dabo AJ, Geraghty P, Campos M, Foronjy R, Garcia-Arcos I. Decreased surfactant lipids correlate with lung function in chronic obstructive pulmonary disease (COPD). PLoS One 2020; 15:e0228279. [PMID: 32027677 PMCID: PMC7004328 DOI: 10.1371/journal.pone.0228279] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/12/2020] [Indexed: 01/10/2023] Open
Abstract
Smoke exposure is known to decrease total pulmonary surfactant and alter its composition, but the role of surfactant in chronic obstructive pulmonary disease (COPD) remains unknown. We aimed to analyze the compositional changes in the surfactant lipidome in COPD and identify specific lipids associated with pulmonary function decline. Bronchoalveolar lavage (BAL) fluid was obtained from 12 former smokers with COPD and 5 non-smoking, non-asthmatic healthy control volunteers. Lipids were extracted and analyzed by liquid chromatography and mass spectrometry. Pulmonary function data were obtained by spirometry, and correlations of lung function with lipid species were determined. Wild-type C57BL/6 mice were exposed to 6 months of second-hand smoke in a full-body chamber. Surfactant lipids were decreased by 60% in subjects with COPD. All phospholipid classes were dramatically decreased, including ether phospholipids, which have not been studied in pulmonary surfactant. Availability of phospholipid, cholesterol, and sphingomyelin in BAL strongly correlated with pulmonary function and this was attributable to specific lipid species of phosphatidylcholine with surface tension reducing properties, and of phosphatidylglycerol with antimicrobial roles, as well as to other less studied lipid species. Mice exposed to smoke for six months recapitulated surfactant lipidomic changes observed in human subjects with COPD. In summary, we show that the surfactant lipidome is substantially altered in subjects with COPD, and decreased availability of phospholipids correlated with decreased pulmonary function. Further investigation of surfactant alterations in COPD would improve our understanding of its physiopathology and reveal new potential therapeutic targets.
Collapse
Affiliation(s)
- Christina W. Agudelo
- Department of Medicine, SUNY Downstate Medical Center, New York, New York, United States of America
| | - Britta K. Kumley
- Department of Medicine, SUNY Downstate Medical Center, New York, New York, United States of America
| | - Estela Area-Gomez
- Department of Neurology, Columbia University, New York, New York, United States of America
| | - Yimeng Xu
- Department of Neurology, Columbia University, New York, New York, United States of America
| | - Abdoulaye J. Dabo
- Department of Medicine, SUNY Downstate Medical Center, New York, New York, United States of America
| | - Patrick Geraghty
- Department of Medicine, SUNY Downstate Medical Center, New York, New York, United States of America
- Department of Cell Biology, SUNY Downstate Medical Center, New York, New York, United States of America
| | - Michael Campos
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Robert Foronjy
- Department of Medicine, SUNY Downstate Medical Center, New York, New York, United States of America
- Department of Cell Biology, SUNY Downstate Medical Center, New York, New York, United States of America
| | - Itsaso Garcia-Arcos
- Department of Medicine, SUNY Downstate Medical Center, New York, New York, United States of America
- Department of Cell Biology, SUNY Downstate Medical Center, New York, New York, United States of America
| |
Collapse
|
28
|
Methylisothiazolinone induces apoptotic cell death via matrix metalloproteinase activation in human bronchial epithelial cells. Toxicol In Vitro 2020; 62:104661. [DOI: 10.1016/j.tiv.2019.104661] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/16/2019] [Accepted: 09/19/2019] [Indexed: 12/16/2022]
|
29
|
Ohgoda O, Robinson IN. Toxicological evaluation of DSPC (1,2-distearoyl-sn-glycero-3-phosphocholine). ACTA ACUST UNITED AC 2020. [DOI: 10.2131/fts.7.55] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Osamu Ohgoda
- Clinical Pharmacology & Drug Safety and Metabolism Department, Science and Data Technology Division, R&D, AstraZeneca K.K
| | - Ian N. Robinson
- Regulatory Safety Centre of Excellence, Clinical Pharmacology and Safety Sciences, R&D BioPharmaceuticals, AstraZeneca UK Ltd, United Kingdom
| |
Collapse
|
30
|
Atia T, Abdel-Gawad S. Pulmonary toxicity induced by exposure to phthalates, an experimental study. Inhal Toxicol 2019; 31:376-383. [PMID: 31777295 DOI: 10.1080/08958378.2019.1695025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
Background: A plasticizer product, di-(2-ethylhexyl) phthalate (DEHP), is widely used in many consumer products, such as food packages, personal care products, children` toys, and medical devices. Phthalates are known to be released into the biological fluids and redistributed into various tissues linked with multiple health problems.Aim: We aimed to study the possible toxic effect of phthalate exposure on the lung tissues.Methods: Thirty male Wister rats were randomly divided into three groups equally, received the following for two weeks once daily via gastric intubation: control group; received normal saline. The DEHP treated group received 2,85mg/kg per BW of DEHP dissolved in normal saline. The DEHP recovery group, received the same as the treated group, followed by two weeks without any treatment. For light microscopic study; the lung tissues were dissected, cut into small pieces, processed, embedded in paraffin wax, sectioned and stained with Hematoxylin and Eosin as well as Masson trichrome stains. For electron microscopic study; the lung tissues were fixed in glutaraldehyde, processed, embedded in epoxy, cut into ultrathin sections, and stained with uranyl acetate and lead citrate.Results: Compared to the control group, the alveolar tissues in the treated group showed a significant increase in collagen deposition and inflammatory cellular infiltration. The number of type-II pneumocytes and alveolar macrophages/field were also significantly increased. However, these pathological changes improved slightly after stopping exposure to DEHP.Conclusion: DEHP has a toxic effect on the lung tissues, which after its withdrawal did not improve completely.
Collapse
Affiliation(s)
- Tarek Atia
- Histology and Cell Biology, Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj, KSA
| | - Sahar Abdel-Gawad
- Histology and Cell Biology, Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj, KSA
| |
Collapse
|
31
|
The Impact of Lipid Corona on Rifampicin Intramacrophagic Transport Using Inhaled Solid Lipid Nanoparticles Surface-Decorated with a Mannosylated Surfactant. Pharmaceutics 2019; 11:pharmaceutics11100508. [PMID: 31581554 PMCID: PMC6835947 DOI: 10.3390/pharmaceutics11100508] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/24/2019] [Accepted: 09/27/2019] [Indexed: 12/15/2022] Open
Abstract
The mimicking of physiological conditions is crucial for the success of accurate in vitro studies. For inhaled nanoparticles, which are designed for being deposited on alveolar epithelium and taken up by macrophages, it is relevant to investigate the interactions with pulmonary surfactant lining alveoli. As a matter of fact, the formation of a lipid corona layer around the nanoparticles could modulate the cell internalization and the fate of the transported drugs. Based on this concept, the present research focused on the interactions between pulmonary surfactant and Solid Lipid Nanoparticle assemblies (SLNas), loaded with rifampicin, an anti-tuberculosis drug. SLNas were functionalized with a synthesized mannosylated surfactant, both alone and in a blend with sodium taurocholate, to achieve an active targeting to mannose receptors present on alveolar macrophages (AM). Physico-chemical properties of the mannosylated SLNas satisfied the requirements relative to suitable respirability, drug payload, and AM active targeting. Our studies have shown that a lipid corona is formed around SLNas in the presence of Curosurf, a commercial substitute of the natural pulmonary surfactant. The lipid corona promoted an additional resistance to the drug diffusion for SLNas functionalized with the mannosylated surfactant and this improved drug retention within SLNas before AM phagocytosis takes place. Moreover, lipid corona formation did not modify the role of nanoparticle mannosylation towards the specific receptors on MH-S cell membrane.
Collapse
|
32
|
Rao M, Dodoo E, Zumla A, Maeurer M. Immunometabolism and Pulmonary Infections: Implications for Protective Immune Responses and Host-Directed Therapies. Front Microbiol 2019; 10:962. [PMID: 31134013 PMCID: PMC6514247 DOI: 10.3389/fmicb.2019.00962] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 04/16/2019] [Indexed: 12/12/2022] Open
Abstract
The biology and clinical efficacy of immune cells from patients with infectious diseases or cancer are associated with metabolic programming. Host immune- and stromal-cell genetic and epigenetic signatures in response to the invading pathogen shape disease pathophysiology and disease outcomes. Directly linked to the immunometabolic axis is the role of the host microbiome, which is also discussed here in the context of productive immune responses to lung infections. We also present host-directed therapies (HDT) as a clinically viable strategy to refocus dysregulated immunometabolism in patients with infectious diseases, which requires validation in early phase clinical trials as adjuncts to conventional antimicrobial therapy. These efforts are expected to be continuously supported by newly generated basic and translational research data to gain a better understanding of disease pathology while devising new molecularly defined platforms and therapeutic options to improve the treatment of patients with pulmonary infections, particularly in relation to multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Martin Rao
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Ernest Dodoo
- Department of Oncology and Haematology, Krankenhaus Nordwest, Frankfurt, Germany
| | - Alimuddin Zumla
- Division of Infection and Immunity, University College London, NIHR Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Markus Maeurer
- ImmunoSurgery Unit, Champalimaud Centre for the Unknown, Lisbon, Portugal.,Department of Oncology and Haematology, Krankenhaus Nordwest, Frankfurt, Germany
| |
Collapse
|
33
|
Garikapati V, Karnati S, Bhandari DR, Baumgart-Vogt E, Spengler B. High-resolution atmospheric-pressure MALDI mass spectrometry imaging workflow for lipidomic analysis of late fetal mouse lungs. Sci Rep 2019; 9:3192. [PMID: 30816198 PMCID: PMC6395778 DOI: 10.1038/s41598-019-39452-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 01/17/2019] [Indexed: 12/19/2022] Open
Abstract
Mass spectrometry imaging (MSI) provides label-free, non-targeted molecular and spatial information of the biomolecules within tissue. Lipids play important roles in lung biology, e.g. as surfactant, preventing alveolar collapse during normal and forced respiration. Lipidomic characterization of late fetal mouse lungs at day 19 of gestation (E19) has not been performed yet. In this study we employed high-resolution atmospheric pressure scanning microprobe matrix-assisted laser desorption/ionization MSI for the lipidomic analysis of E19 mouse lungs. Molecular species of different lipid classes were imaged in E19 lung sections at high spatial and mass resolution in positive- and negative-ion mode. Lipid species were characterized based on accurate mass and on-tissue tandem mass spectrometry. In addition, a dedicated sample preparation protocol, homogenous deposition of matrices on tissue surfaces and data processing parameters were optimized for the comparison of signal intensities of lipids between different tissue sections of E19 lungs of wild type and Pex11β-knockout mice. Our study provides lipid information of E19 mouse lungs, optimized experimental and data processing strategies for the direct comparison of signal intensities of metabolites (lipids) among the tissue sections from MSI experiments. To best of our knowledge, this is the first MSI and lipidomic study of E19 mouse lungs.
Collapse
Affiliation(s)
- Vannuruswamy Garikapati
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen, Germany.,Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology, Justus Liebig University Giessen, Giessen, Germany
| | - Srikanth Karnati
- Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology, Justus Liebig University Giessen, Giessen, Germany.,Institute for Anatomy and Cell Biology, Julius Maximilians University Würzburg, Würzburg, Germany
| | - Dhaka Ram Bhandari
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology, Justus Liebig University Giessen, Giessen, Germany
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen, Germany.
| |
Collapse
|
34
|
Habiel DM, Espindola MS, Kitson C, Azzara AV, Coelho AL, Stripp B, Hogaboam CM. Characterization of CD28 null T cells in idiopathic pulmonary fibrosis. Mucosal Immunol 2019; 12:212-222. [PMID: 30315241 PMCID: PMC6301115 DOI: 10.1038/s41385-018-0082-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/02/2018] [Accepted: 08/08/2018] [Indexed: 02/04/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fibrotic lung disease, with unknown etiopathogenesis and suboptimal therapeutic options. Previous reports have shown that increased T-cell numbers and CD28null phenotype is predictive of prognosis in IPF, suggesting that these cells might have a role in this disease. Flow cytometric analysis of explanted lung cellular suspensions showed a significant increase in CD8+ CD28null T cells in IPF relative to normal lung explants. Transcriptomic analysis of CD3+ T cells isolated from IPF lung explants revealed a loss of CD28-transcript expression and elevation of pro-inflammatory cytokine expression in IPF relative to normal T cells. IPF lung explant-derived T cells (enriched with CD28null T cells), but not normal donor lung CD28+ T cells induced dexamethasone-resistant lung remodeling in humanized NSG mice. Finally, CD28null T cells expressed similar CTLA4 and significantly higher levels of PD-1 proteins relative to CD28+ T cells and blockade of either proteins in humanized NSG mice, using anti-CTLA4, or anti-PD1, mAb treatment-accelerated lung fibrosis. Together, these results demonstrate that IPF CD28null T cells may promote lung fibrosis but the immune checkpoint proteins, CTLA-4 and PD-1, appears to limit this effect.
Collapse
Affiliation(s)
- David M Habiel
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
| | - Milena S Espindola
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Chris Kitson
- Bristol-Myers Squibb, Fibrosis Discovery Biology, Pennington, NJ, 08534, USA
| | - Anthony V Azzara
- Bristol-Myers Squibb, Fibrosis Discovery Biology, Pennington, NJ, 08534, USA
| | - Ana Lucia Coelho
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Barry Stripp
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Cory M Hogaboam
- Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
| |
Collapse
|
35
|
Girguis MS, Strickland MJ, Hu X, Liu Y, Chang HH, Kloog I, Belanoff C, Bartell SM, Vieira VM. Exposure to acute air pollution and risk of bronchiolitis and otitis media for preterm and term infants. JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2018; 28:348-357. [PMID: 29269754 PMCID: PMC6013343 DOI: 10.1038/s41370-017-0006-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 08/03/2017] [Accepted: 08/18/2017] [Indexed: 05/29/2023]
Abstract
Our aim is to estimate associations between acute increases in particulate matter with diameter of 2.5 µm or less (PM2.5) concentrations and risk of infant bronchiolitis and otitis media among Massachusetts births born 2001 through 2008.Our case-crossover study included 20,017 infant bronchiolitis and 42,336 otitis media clinical encounter visits. PM2.5 was modeled using satellite, remote sensing, meteorological and land use data. We applied conditional logistic regression to estimate odds ratios (ORs) and confidence intervals (CIs) per 10-µg/m3 increase in PM2.5. We assessed effect modification to determine the most susceptible subgroups. Infant bronchiolitis risk was elevated for PM2.5 exposure 1 day (OR = 1.07, 95% CI = 1.03-1.11) and 4 days (OR = 1.04, 95% CI = 0.99-1.08) prior to clinical encounter, but not 7 days. Non-significant associations with otitis media varied depending on lag. Preterm infants were at substantially increased risk of bronchiolitis 1 day prior to clinical encounter (OR = 1.17, 95% CI = 1.08-1.28) and otitis media 4 and 7 days prior to clinical encounter (OR = 1.09, 95% CI = 1.02-1.16 and OR = 1.08, 95% CI = 1.02-1.15, respectively). In conclusion, preterm infants are most susceptible to infant bronchiolitis and otitis media associated with acute PM2.5 exposures.
Collapse
Affiliation(s)
- Mariam S Girguis
- Program in Public Health, University of California, Irvine, Irvine, CA, USA
| | | | - Xuefei Hu
- Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Yang Liu
- Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Howard H Chang
- Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Itai Kloog
- The Department of Geography and Environmental Development, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Candice Belanoff
- Department of Community Health Sciences, Boston University School of Public Health, Boston, MA, USA
| | - Scott M Bartell
- Program in Public Health, University of California, Irvine, Irvine, CA, USA
- Department of Statistics, Donald Bren School of Information and Computer Sciences, University of California, Irvine, CA, USA
- Department of Epidemiology, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Verónica M Vieira
- Program in Public Health, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
36
|
Schneider-Futschik EK, Paulin OKA, Hoyer D, Roberts KD, Ziogas J, Baker MA, Karas J, Li J, Velkov T. Sputum Active Polymyxin Lipopeptides: Activity against Cystic Fibrosis Pseudomonas aeruginosa Isolates and Their Interactions with Sputum Biomolecules. ACS Infect Dis 2018; 4:646-655. [PMID: 29566483 PMCID: PMC5952261 DOI: 10.1021/acsinfecdis.7b00238] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
The
mucoid biofilm mode of growth of Pseudomonas aeruginosa (P. aeruginosa) in the lungs of cystic fibrosis
patients makes eradication of infections with antibiotic therapy very
difficult. The lipopeptide antibiotics polymyxin B and colistin are
currently the last-resort therapies for infections caused by multidrug-resistant P. aeruginosa. In the present study, we investigated
the antibacterial activity of a series of polymyxin lipopeptides (polymyxin
B, colistin, FADDI-003, octapeptin A3, and polymyxin A2) against a panel of polymyxin-susceptible and polymyxin-resistant P. aeruginosa cystic fibrosis isolates grown under
planktonic or biofilm conditions in artificial sputum and their interactions
with sputum component biomolecules. In sputum media under planktonic
conditions, the lipopeptides FADDI-003 and octapeptin A3 displayed very promising activity against the polymyxin-resistant
isolate FADDI-PA066 (polymyxin B minimum inhibitory concentration
(MIC) = 32 mg/L), while retaining their activity against the polymyxin-sensitive
strains FADDI-PA021 (polymyxin B MIC = 1 mg/L) and FADDI-PA020 (polymyxin
B MIC = 2 mg/L). Polymyxin A2 was only effective against
the polymyxin-sensitive isolates. However, under biofilm growth conditions,
the hydrophobic lipopeptide FADDI-003 was inactive compared to the
more hydrophilic lipopeptides, octapeptin A3, polymyxin
A2, polymyxin B, and colistin. Transmission electron micrographs
revealed octapeptin A3 caused reduction in the cell numbers
in biofilm as well as biofilm disruption/“antibiofilm”
activity. We therefore assessed the interactions of the lipopeptides
with the component sputum biomolecules, mucin, deoxyribonucleic acid
(DNA), surfactant, F-actin, lipopolysaccharide, and phospholipids.
We observed the general trend that sputum biomolecules reduce lipopeptide
antibacterial activity. Collectively, our data suggests that, in the
airways, lipopeptide binding to component sputum biomolecules may
reduce antibacterial efficacy and is dependent on the physicochemical
properties of the lipopeptide.
Collapse
Affiliation(s)
- Elena K. Schneider-Futschik
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Olivia K. A. Paulin
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Daniel Hoyer
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Kade D. Roberts
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - James Ziogas
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Mark A. Baker
- Priority Research Centre in Reproductive Science, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia
| | - John Karas
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jian Li
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Tony Velkov
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
37
|
Shinde A, Luo J, Bharathi SS, Shi H, Beck ME, McHugh KJ, Alcorn JF, Wang J, Goetzman ES. Increased mortality from influenza infection in long-chain acyl-CoA dehydrogenase knockout mice. Biochem Biophys Res Commun 2018; 497:700-704. [PMID: 29458021 DOI: 10.1016/j.bbrc.2018.02.135] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 02/15/2018] [Indexed: 10/18/2022]
Abstract
We previously showed that the mitochondrial fatty acid oxidation enzyme long-chain acyl-CoA dehydrogenase (LCAD) is expressed in alveolar type II pneumocytes and that LCAD-/- mice have altered breathing mechanics and surfactant defects. Here, we hypothesized that LCAD-/- mice would be susceptible to influenza infection. Indeed, LCAD-/- mice demonstrated increased mortality following infection with 2009 pandemic influenza (A/CA/07/09). However, the mortality was not due to increased lung injury, as inflammatory cell counts, viral titers, and histology scores all showed non-significant trends toward milder injury in LCAD-/- mice. To confirm this, LCAD-/- were infected with a second, mouse-adapted H1N1 virus (A/PR/8/34), to which they responded with significantly less lung injury. While both strains become increasingly hypoglycemic over the first week post-infection, LCAD-/- mice lose body weight more rapidly than wild-type mice. Surprisingly, while acutely fasted LCAD-/- mice develop hepatic steatosis, influenza-infected LCAD-/- mice do not. They do, however, become more hypothermic than wild-type mice and demonstrate increased blood lactate values. We conclude that LCAD-/- mice succumb to influenza from bioenergetic starvation, likely due to increased reliance upon glucose for energy.
Collapse
Affiliation(s)
- Apurva Shinde
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, United States
| | - Jiadi Luo
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, United States
| | - Sivakama S Bharathi
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, United States
| | - Huifang Shi
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, United States
| | - Megan E Beck
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, United States
| | - Kevin J McHugh
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, United States
| | - John F Alcorn
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, United States
| | - Jieru Wang
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, United States
| | - Eric S Goetzman
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, United States.
| |
Collapse
|
38
|
Fessler MB, Summer RS. Surfactant Lipids at the Host-Environment Interface. Metabolic Sensors, Suppressors, and Effectors of Inflammatory Lung Disease. Am J Respir Cell Mol Biol 2017; 54:624-35. [PMID: 26859434 DOI: 10.1165/rcmb.2016-0011ps] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The lipid composition of pulmonary surfactant is unlike that of any other body fluid. This extracellular lipid reservoir is also uniquely susceptible by virtue of its direct and continuous exposure to environmental oxidants, inflammatory agents, and pathogens. Historically, the greatest attention has been focused on those biophysical features of surfactant that serve to reduce surface tension at the air-liquid interface. More recently, surfactant lipids have also been recognized as bioactive molecules that maintain immune quiescence in the lung but can also be remodeled by the inhaled environment into neolipids that mediate key roles in inflammation, immunity, and fibrosis. This review focuses on the roles in inflammatory and infectious lung disease of two classes of native surfactant lipids, glycerophospholipids and sterols, and their corresponding oxidized species, oxidized glycerophospholipids and oxysterols. We highlight evidence that surfactant composition is sensitive to circulating lipoproteins and that the lipid milieu of the alveolus should thus be recognized as susceptible to diet and common systemic metabolic disorders. We also discuss intriguing evidence suggesting that oxidized surfactant lipids may represent an evolutionary link between immunity and tissue homeostasis that arose in the primordial lung. Taken together, the emerging picture is one in which the unique environmental susceptibility of the lung, together with its unique extracellular lipid requirements, may have made this organ both an evolutionary hub and an engine for lipid-immune cross-talk.
Collapse
Affiliation(s)
- Michael B Fessler
- 1 Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina; and
| | - Ross S Summer
- 2 Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
39
|
Agrawal S, Srivastava R, Rahmatpanah F, Madiraju C, BenMohamed L, Agrawal A. Airway epithelial cells enhance the immunogenicity of human myeloid dendritic cells under steady state. Clin Exp Immunol 2017; 189:279-289. [PMID: 28470729 DOI: 10.1111/cei.12983] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2017] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) and airway epithelial cells (AECs) are in close proximity, and AECs secrete factors such as retinoic acid which induce tolerance in DCs at homeostasis. However, the question remains as to how DCs in the lung are able to respond to pathogens in the immunosuppressive environment. Using an in vitro human myeloid DC (mDC)-AEC co-culture system, we demonstrate that AECs induced several gene changes in the mDCs cultured with AECs compared to the mDCs not cultured with AECs. Analysis revealed that several chemokine genes were altered. These chemokine genes could serve to attract neutrophils, natural killer (NK) T as well as T helper type 1 (Th1)/Th2 cells to the airways. Genes priming lipid and major histocompatibility complex (MHC) class II antigen presentation were also up-regulated, along with certain anti-microbial protein genes. In addition, the expression and function of pathogen-sensing Toll-like receptors (TLRs) as well as Nod-like receptors (NLRs) and their downstream signalling molecules were up-regulated in mDCs cultured with AECs. Moreover, murine mucosal DCs from the lung expressed significantly higher levels of TLRs and NLRs compared to peripheral DCs from the spleen. These results indicate that AECs prime mDCs to enhance their immunogenicity, which could be one of the mechanisms that compensates for the immunosuppressive mucosal environment.
Collapse
Affiliation(s)
- S Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, Irvine, CA, USA
| | - R Srivastava
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, Irvine, CA, USA
| | - F Rahmatpanah
- Department of pathology, University of California, Irvine, Irvine, CA, USA
| | - C Madiraju
- Division of Basic and Clinical Immunology, Department of Medicine, Irvine, CA, USA
| | - L BenMohamed
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, Irvine, CA, USA
| | - A Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, Irvine, CA, USA
| |
Collapse
|
40
|
Location, function, and ontogeny of pulmonary macrophages during the steady state. Pflugers Arch 2017; 469:561-572. [DOI: 10.1007/s00424-017-1965-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 03/01/2017] [Indexed: 12/12/2022]
|
41
|
Elhosseiny NM, El-Tayeb OM, Yassin AS, Lory S, Attia AS. The secretome of Acinetobacter baumannii ATCC 17978 type II secretion system reveals a novel plasmid encoded phospholipase that could be implicated in lung colonization. Int J Med Microbiol 2016; 306:633-641. [PMID: 27713027 DOI: 10.1016/j.ijmm.2016.09.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/24/2016] [Accepted: 09/30/2016] [Indexed: 02/01/2023] Open
Abstract
Acinetobacter baumannii infections are compounded with a striking lack of treatment options. In many Gram-negative bacteria, secreted proteins play an important early role in avoiding host defences. Typically, these proteins are targeted to the external environment or into host cells using dedicated transport systems. Despite the fact that medically relevant species of Acinetobacter possess a type II secretion system (T2SS), only recently, its significance as an important pathway for delivering virulence factors has gained attention. Using in silico analysis to characterize the genetic determinants of the T2SS, which are found clustered in other organisms, in Acinetobacter species, they appear to have a unique genetic organization and are distributed throughout the genome. When compared to other T2SS orthologs, individual components of the T2SS apparatus showed the highest similarity to those of Pseudomonas aeruginosa. A mutant of Acinetobacter baumannii strain ATCC 17978 lacking the secretin component of the T2SS (ΔgspD), together with a trans-complemented mutant, were tested in a series of in vitro and in vivo assays to determine the role of T2SS in pathogenicity. The ΔgspD mutant displayed decreased lipolytic activity, associated with attenuated colonization ability in a murine pneumonia model. These phenotypes are linked to LipAN, a novel plasmid-encoded phospholipase, identified through mass spectroscopy as a T2SS substrate. Recombinant LipAN showed specific phospholipase activity in vitro. Proteomics on the T2-dependent secretome of ATCC 17978 strain revealed its potential dedication to the secretion of a number of lipolytic enzymes, among others which could contribute to its virulence. This study highlights the role of T2SS as an active contributor to the virulence of A. baumannii potentially through secretion of a newly identified phospholipase.
Collapse
Affiliation(s)
- Noha M Elhosseiny
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Ossama M El-Tayeb
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Aymen S Yassin
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Stephen Lory
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Ahmed S Attia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
42
|
Wang J, Mei H, Liu CZ, Zhang YY, Liu CL, Song D, Zhang YH. [Relationship between R236C site in exon 7 of SP-B gene and respiratory distress syndrome in Han newborns in western Inner Mongolia]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2016; 18:802-805. [PMID: 27655533 PMCID: PMC7389981 DOI: 10.7499/j.issn.1008-8830.2016.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/07/2016] [Indexed: 06/06/2023]
Abstract
OBJECTIVE To detect and analyze the genetic variation in exon 7 of lung surfactant protein B (SP-B), and to investigate the relationship between the genetic variation and the incidence of neonatal respiratory distress syndrome (NRDS) in Han populations in western Inner Mongolia. METHODS In the case-control study, 47 Han infants with NRDS were assigned to case group. All the 47 patients had the last three generations of their ancestors reside in western Inner Mongolia. Forty-seven Han newborns without NRDS were assigned to control group. PCR-based gene analysis was used to determine the mutation in exon 7 of SP-B gene and genotype and allele frequencies of the R236C site in exon 7 of SP-B gene. RESULTS In Han newborns in western Inner Mongolia, there was no mutation in exon 7 of SP-B gene; two genotypes, CC and CT, were identified in the R236C site in exon 7 of SP-B gene. No TT genotype was found in the two groups. There were no significant differences in the genotype frequency of CC or CT as well as the allele frequency of C or T between the case and control groups (CC: 72% vs 85%, P>0.05; CT: 28% vs 15%, P>0.05; C: 85% vs 93%, P>0.05; T: 15% vs 7%, P>0.05). CONCLUSIONS There is no mutation in exon 7 of SP-B gene in Han infants with NRDS in western Inner Mongolia. There is no significant association between the gene polymorphism of the R236C site in exon 7 of SP-B gene and the incidence of NRDS in Han populations in that region.
Collapse
Affiliation(s)
- Jing Wang
- Department of Neonatology, Affiliated Hospital, Inner Mongolia Medical University, Hohhot 010059, China.
| | | | | | | | | | | | | |
Collapse
|
43
|
Sweeney S, Leo BF, Chen S, Abraham-Thomas N, Thorley AJ, Gow A, Schwander S, Zhang JJ, Shaffer MSP, Chung KF, Ryan MP, Porter AE, Tetley TD. Pulmonary surfactant mitigates silver nanoparticle toxicity in human alveolar type-I-like epithelial cells. Colloids Surf B Biointerfaces 2016; 145:167-175. [PMID: 27182651 DOI: 10.1016/j.colsurfb.2016.04.040] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 04/04/2016] [Accepted: 04/19/2016] [Indexed: 01/01/2023]
Abstract
Accompanying increased commercial applications and production of silver nanomaterials is an increased probability of human exposure, with inhalation a key route. Nanomaterials that deposit in the pulmonary alveolar region following inhalation will interact firstly with pulmonary surfactant before they interact with the alveolar epithelium. It is therefore critical to understand the effects of human pulmonary surfactant when evaluating the inhalation toxicity of silver nanoparticles. In this study, we evaluated the toxicity of AgNPs on human alveolar type-I-like epithelial (TT1) cells in the absence and presence of Curosurf(®) (a natural pulmonary surfactant substitute), hypothesising that the pulmonary surfactant would act to modify toxicity. We demonstrated that 20nm citrate-capped AgNPs induce toxicity in human alveolar type I-like epithelial cells and, in agreement with our hypothesis, that pulmonary surfactant acts to mitigate this toxicity, possibly through reducing AgNP dissolution into cytotoxic Ag(+) ions. For example, IL-6 and IL-8 release by TT1 cells significantly increased 10.7- and 35-fold, respectively (P<0.01), 24h after treatment with 25μg/ml AgNPs. In contrast, following pre-incubation of AgNPs with Curosurf(®), this effect was almost completely abolished. We further determined that the mechanism of this toxicity is likely associated with Ag(+) ion release and lysosomal disruption, but not with increased reactive oxygen species generation. This study provides a critical understanding of the toxicity of AgNPs in target human alveolar type-I-like epithelial cells and the role of pulmonary surfactant in mitigating this toxicity. The observations reported have important implications for the manufacture and application of AgNPs, in particular for applications involving use of aerosolised AgNPs.
Collapse
Affiliation(s)
- Sinbad Sweeney
- Lung Cell Biology, Airways Disease, National Heart & Lung Institute, Imperial College London, London, UK
| | - Bey Fen Leo
- Department of Materials and London Centre for Nanotechnology, Imperial College London, London, UK; Department of Mechanical Engineering, University of Malaya, Kuala Lumpur, Malaysia
| | - Shu Chen
- Department of Chemistry and London Centre for Nanotechnology, Imperial College London, London, UK
| | - Nisha Abraham-Thomas
- Lung Cell Biology, Airways Disease, National Heart & Lung Institute, Imperial College London, London, UK
| | - Andrew J Thorley
- Lung Cell Biology, Airways Disease, National Heart & Lung Institute, Imperial College London, London, UK
| | - Andrew Gow
- Department of Toxicology, Ernst Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Stephan Schwander
- Department of Environmental and Occupational Health, School of Public Health, Rutgers University, Piscataway, NJ, USA
| | - Junfeng Jim Zhang
- Division of Environmental Sciences & Policy, Nicholas School of the Environment and Duke Global Health Institute,, Duke University, Durham, USA
| | - Milo S P Shaffer
- Department of Chemistry and London Centre for Nanotechnology, Imperial College London, London, UK
| | - Kian Fan Chung
- Respiratory Medicine and Experimental Studies Unit, Airways Disease, National Heart & Lung Institute, Imperial College London, London, UK
| | - Mary P Ryan
- Department of Materials and London Centre for Nanotechnology, Imperial College London, London, UK
| | - Alexandra E Porter
- Department of Materials and London Centre for Nanotechnology, Imperial College London, London, UK
| | - Teresa D Tetley
- Lung Cell Biology, Airways Disease, National Heart & Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
44
|
Metabolism and Pathogenicity of Pseudomonas aeruginosa Infections in the Lungs of Individuals with Cystic Fibrosis. Microbiol Spectr 2016; 3. [PMID: 26350318 DOI: 10.1128/microbiolspec.mbp-0003-2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Individuals with the genetic disease cystic fibrosis (CF) accumulate mucus or sputum in their lungs. This sputum is a potent growth substrate for a range of potential pathogens, and the opportunistic bacterium Pseudomonas aeruginosa is generally most difficult of these to eradicate. As a result, P. aeruginosa infections are frequently maintained in the CF lung throughout life, and are the leading cause of death for these individuals. While great effort has been expended to better understand and treat these devastating infections, only recently have researchers begun to rigorously examine the roles played by specific nutrients in CF sputum to cue P. aeruginosa pathogenicity. This chapter summarizes the current state of knowledge regarding how P. aeruginosa metabolism in CF sputum affects initiation and maintenance of these infections. It contains an overview of CF lung disease and the mechanisms of P. aeruginosa pathogenicity. Several model systems used to study these infections are described with emphasis on the challenge of replicating the chronic infections observed in humans with CF. Nutrients present in CF sputum are surveyed, and the impacts of these nutrients on the infection are discussed. The chapter concludes by addressing the future of this line of research including the use of next-generation technologies and the potential for metabolism-based therapeutics.
Collapse
|
45
|
Tinglev ÅD, Ullah S, Ljungkvist G, Viklund E, Olin AC, Beck O. Characterization of exhaled breath particles collected by an electret filter technique. J Breath Res 2016; 10:026001. [DOI: 10.1088/1752-7155/10/2/026001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
46
|
Glaser K, Fehrholz M, Curstedt T, Kunzmann S, Speer CP. Effects of the New Generation Synthetic Reconstituted Surfactant CHF5633 on Pro- and Anti-Inflammatory Cytokine Expression in Native and LPS-Stimulated Adult CD14+ Monocytes. PLoS One 2016; 11:e0146898. [PMID: 26790130 PMCID: PMC4720484 DOI: 10.1371/journal.pone.0146898] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 12/24/2015] [Indexed: 02/01/2023] Open
Abstract
Background Surfactant replacement therapy is the standard of care for the prevention and treatment of neonatal respiratory distress syndrome. New generation synthetic surfactants represent a promising alternative to animal-derived surfactants. CHF5633, a new generation reconstituted synthetic surfactant containing SP-B and SP-C analogs and two synthetic phospholipids has demonstrated biophysical effectiveness in vitro and in vivo. While several surfactant preparations have previously been ascribed immunomodulatory capacities, in vitro data on immunomodulation by CHF5633 are limited, so far. Our study aimed to investigate pro- and anti-inflammatory effects of CHF5633 on native and LPS-stimulated human adult monocytes. Methods Highly purified adult CD14+ cells, either native or simultaneously stimulated with LPS, were exposed to CHF5633, its components, or poractant alfa (Curosurf®). Subsequent expression of TNF-α, IL-1β, IL-8 and IL-10 mRNA was quantified by real-time quantitative PCR, corresponding intracellular cytokine synthesis was analyzed by flow cytometry. Potential effects on TLR2 and TLR4 mRNA and protein expression were monitored by qPCR and flow cytometry. Results Neither CHF5633 nor any of its components induced inflammation or apoptosis in native adult CD14+ monocytes. Moreover, LPS-induced pro-inflammatory responses were not aggravated by simultaneous exposure of monocytes to CHF5633 or its components. In LPS-stimulated monocytes, exposure to CHF5633 led to a significant decrease in TNF-α mRNA (0.57 ± 0.23-fold, p = 0.043 at 4h; 0.56 ± 0.27-fold, p = 0.042 at 14h). Reduction of LPS-induced IL-1β mRNA expression was not significant (0.73 ± 0.16, p = 0.17 at 4h). LPS-induced IL-8 and IL-10 mRNA and protein expression were unaffected by CHF5633. For all cytokines, the observed CHF5633 effects paralleled a Curosurf®-induced modulation of cytokine response. TLR2 and TLR4 mRNA and protein expression were not affected by CHF5633 and Curosurf®, neither in native nor in LPS-stimulated adult monocytes. Conclusion The new generation reconstituted synthetic surfactant CHF5633 was tested for potential immunomodulation on native and LPS-activated adult human monocytes. Our data confirm that CHF5633 does not exert unintended pro-inflammatory effects in both settings. On the contrary, CHF5633 significantly suppressed TNF-α mRNA expression in LPS-stimulated adult monocytes, indicating potential anti-inflammatory effects.
Collapse
Affiliation(s)
- Kirsten Glaser
- University Children´s Hospital, University of Würzburg, Würzburg, Germany
- * E-mail:
| | - Markus Fehrholz
- University Children´s Hospital, University of Würzburg, Würzburg, Germany
| | - Tore Curstedt
- Department of Molecular Medicine and Surgery, Karolinska Institutet at Karolinska University Hospital, Stockholm, Sweden
| | - Steffen Kunzmann
- University Children´s Hospital, University of Würzburg, Würzburg, Germany
| | - Christian P. Speer
- University Children´s Hospital, University of Würzburg, Würzburg, Germany
| |
Collapse
|
47
|
Orgeig S, Morrison JL, Daniels CB. Evolution, Development, and Function of the Pulmonary Surfactant System in Normal and Perturbed Environments. Compr Physiol 2015; 6:363-422. [PMID: 26756637 DOI: 10.1002/cphy.c150003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Surfactant lipids and proteins form a surface active film at the air-liquid interface of internal gas exchange organs, including swim bladders and lungs. The system is uniquely positioned to meet both the physical challenges associated with a dynamically changing internal air-liquid interface, and the environmental challenges associated with the foreign pathogens and particles to which the internal surface is exposed. Lungs range from simple, transparent, bag-like units to complex, multilobed, compartmentalized structures. Despite this anatomical variability, the surfactant system is remarkably conserved. Here, we discuss the evolutionary origin of the surfactant system, which likely predates lungs. We describe the evolution of surfactant structure and function in invertebrates and vertebrates. We focus on changes in lipid and protein composition and surfactant function from its antiadhesive and innate immune to its alveolar stability and structural integrity functions. We discuss the biochemical, hormonal, autonomic, and mechanical factors that regulate normal surfactant secretion in mature animals. We present an analysis of the ontogeny of surfactant development among the vertebrates and the contribution of different regulatory mechanisms that control this development. We also discuss environmental (oxygen), hormonal and biochemical (glucocorticoids and glucose) and pollutant (maternal smoking, alcohol, and common "recreational" drugs) effects that impact surfactant development. On the adult surfactant system, we focus on environmental variables including temperature, pressure, and hypoxia that have shaped its evolution and we discuss the resultant biochemical, biophysical, and cellular adaptations. Finally, we discuss the effect of major modern gaseous and particulate pollutants on the lung and surfactant system.
Collapse
Affiliation(s)
- Sandra Orgeig
- School of Pharmacy & Medical Sciences and Sansom Institute for Health Research, University of South Australia, Adelaide, Australia
| | - Janna L Morrison
- School of Pharmacy & Medical Sciences and Sansom Institute for Health Research, University of South Australia, Adelaide, Australia
| | - Christopher B Daniels
- School of Pharmacy & Medical Sciences and Sansom Institute for Health Research, University of South Australia, Adelaide, Australia
| |
Collapse
|
48
|
Inactivation of Peroxiredoxin 6 by the Pla Protease of Yersinia pestis. Infect Immun 2015; 84:365-74. [PMID: 26553463 DOI: 10.1128/iai.01168-15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/02/2015] [Indexed: 02/03/2023] Open
Abstract
Pneumonic plague represents the most severe form of disease caused by Yersinia pestis due to its ease of transmission, rapid progression, and high mortality rate. The Y. pestis outer membrane Pla protease is essential for the development of pneumonic plague; however, the complete repertoire of substrates cleaved by Pla in the lungs is not known. In this study, we describe a proteomic screen to identify host proteins contained within the bronchoalveolar lavage fluid of mice that are cleaved and/or processed by Y. pestis in a Pla-dependent manner. We identified peroxiredoxin 6 (Prdx6), a host factor that contributes to pulmonary surfactant metabolism and lung defense against oxidative stress, as a previously unknown substrate of Pla. Pla cleaves Prdx6 at three distinct sites, and these cleavages disrupt both the peroxidase and phospholipase A2 activities of Prdx6. In addition, we found that infection with wild-type Y. pestis reduces the abundance of extracellular Prdx6 in the lungs compared to that after infection with Δpla Y. pestis, suggesting that Pla cleaves Prdx6 in the pulmonary compartment. However, following infection with either wild-type or Δpla Y. pestis, Prdx6-deficient mice exhibit no differences in bacterial burden, host immune response, or lung damage from wild-type mice. Thus, while Pla is able to disrupt Prdx6 function in vitro and reduce Prdx6 levels in vivo, the cleavage of Prdx6 has little detectable impact on the progression or outcome of pneumonic plague.
Collapse
|
49
|
Guzmán-Silva A, Vázquez de Lara LG, Torres-Jácome J, Vargaz-Guadarrama A, Flores-Flores M, Pezzat Said E, Lagunas-Martínez A, Mendoza-Milla C, Tanzi F, Moccia F, Berra-Romani R. Lung Beractant Increases Free Cytosolic Levels of Ca2+ in Human Lung Fibroblasts. PLoS One 2015; 10:e0134564. [PMID: 26230503 PMCID: PMC4521834 DOI: 10.1371/journal.pone.0134564] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 07/11/2015] [Indexed: 12/12/2022] Open
Abstract
Beractant, a natural surfactant, induces an antifibrogenic phenotype and apoptosis in normal human lung fibroblasts (NHLF). As intracellular Ca2+ signalling has been related to programmed cell death, we aimed to assess the effect of beractant on intracellular Ca2+ concentration ([Ca2+]i) in NHLF in vitro. Cultured NHLF were loaded with Fura-2 AM (3 μM) and Ca2+ signals were recorded by microfluorimetric techniques. Beractant causes a concentration-dependent increase in [Ca2+]i with a EC50 of 0.82 μg/ml. The application of beractant, at a concentration of 500 μg/ml, which has been shown to exert an apoptotic effect in human fibroblasts, elicited different patterns of Ca2+ signals in NHLF: a) a single Ca2+ spike which could be followed by b) Ca2+ oscillations, c) a sustained Ca2+ plateau or d) a sustained plateau overlapped by Ca2+ oscillations. The amplitude and pattern of Ca2+ transients evoked by beractant were dependent on the resting [Ca2+]i. Pharmacological manipulation revealed that beractant activates a Ca2+ signal through Ca2+ release from intracellular stores mediated by phospholipase Cβ (PLCβ), Ca2+ release from inositol 1,4,5-trisphosphate receptors (IP3Rs) and Ca2+ influx via a store-operated pathway. Moreover, beractant-induced Ca2+ release was abolished by preventing membrane depolarization upon removal of extracellular Na+ and Ca2+. Finally, the inhibition of store-operated channels prevented beractant-induced NHLF apoptosis and downregulation of α1(I) procollagen expression. Therefore, beractant utilizes SOCE to exert its pro-apoptotic and antifibrinogenic effect on NHLF.
Collapse
Affiliation(s)
- Alejandro Guzmán-Silva
- Department of Biomedicine, School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla, Puebla, México
| | - Luis G. Vázquez de Lara
- Experimental Medicine Laboratory, School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla, Puebla, México
| | - Julián Torres-Jácome
- Physiology Institute, Benemérita Universidad Autónoma de Puebla, Puebla, Puebla, México
| | - Ajelet Vargaz-Guadarrama
- Department of Biomedicine, School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla, Puebla, México
| | - Marycruz Flores-Flores
- Department of Biomedicine, School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla, Puebla, México
| | - Elias Pezzat Said
- Experimental Medicine Laboratory, School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla, Puebla, México
| | - Alfredo Lagunas-Martínez
- Instituto Nacional de Salud Pública, Centro de Investigación sobre Enfermedades Infecciosas, Cuernavaca, Morelos, México
| | - Criselda Mendoza-Milla
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México City, México
| | - Franco Tanzi
- Laboratory of General Physiology, Department of Biology and Biotechnology ‘‘Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology ‘‘Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Roberto Berra-Romani
- Department of Biomedicine, School of Medicine, Benemérita Universidad Autónoma de Puebla, Puebla, Puebla, México
| |
Collapse
|
50
|
Porter E, Ma DC, Alvarez S, Faull KF. Antimicrobial lipids: Emerging effector molecules of innate host defense. World J Immunol 2015; 5:51-61. [DOI: 10.5411/wji.v5.i2.51] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 06/28/2015] [Accepted: 07/17/2015] [Indexed: 02/05/2023] Open
Abstract
The antimicrobial properties of host derived lipids have become increasingly recognized and evidence is mounting that antimicrobial lipids (AMLs), like antimicrobial peptides, are effector molecules of the innate immune system and are regulated by its conserved pathways. This review, with primary focus on the human body, provides some background on the biochemistry of lipids, summarizes their biological functions, expands on their antimicrobial properties and site-specific composition, presents modes of synergism with antimicrobial peptides, and highlights the more recent reports on the regulation of AML production as well as bacterial resistance mechanisms. Based on extant data a concept of innate epithelial defense is proposed where epithelial cells, in response to microbial products and proinflammatory cytokines and through activation of conserved innate signaling pathways, increase their lipid uptake and up-regulate transcription of enzymes involved in lipid biosynthesis, and induce transcription of antimicrobial peptides as well as cytokines and chemokines. The subsequently secreted antimicrobial peptides and lipids then attack and eliminate the invader, assisted by or in synergism with other antimicrobial molecules delivered by other defense cells that have been recruited to the site of infection, in most of the cases. This review invites reconsideration of the interpretation of cholesteryl ester accumulation in macrophage lipid droplets in response to infection as a solely proinflammatory event, and proposes a direct antimicrobial role of lipid droplet- associated cholesteryl esters. Finally, for the interested, but new- to- the-field investigator some starting points for the characterization of AMLs are provided. Before it is possible to utilize AMLs for anti-infectious therapeutic and prophylactic approaches, we need to better understand pathogen responses to these lipids and their role in the pathogenesis of chronic infectious disease.
Collapse
|