1
|
Burns E, Harner R, Kodali V, Afshari A, Antonini J, Leonard S. Comparative in vitro toxicity of compositionally distinct thermal spray particulates in human bronchial cells. Toxicol Rep 2024; 13:101851. [PMID: 39717857 PMCID: PMC11665665 DOI: 10.1016/j.toxrep.2024.101851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/29/2024] [Accepted: 12/03/2024] [Indexed: 12/25/2024] Open
Abstract
Thermal spray, in general, is a process that involves forcing a melted substance, such as metal or ceramic in the form of wire or powder, onto the surface of a targeted object to enhance its desired surface properties. In this paper, the melted substance is metal wire generated by an electric arc and forcibly coated on a rotary iron substrate using compressed air. This thermal process is referred to as double-wire arc thermal spray. The particles generated through these methods fall within the nanometer to micrometer agglomerate size range. There is concern regarding potential human health outcomes as these particles exhibit a similarity in particle morphology to welding fumes. Thermal spray wires with zinc (PMET540), iron and chromium (PMET731), and nickel (PMET885) as primary metal compositions were used to generate particulate via an electric arc wire thermal spray generator for exposure to human bronchial cells (BEAS-2B) to examine comparative toxicity ranging from 0 to 200 µg/mL. Resulting cellular viability was assessed through live cell counts, and percent cytotoxicity was measured as a function of LDH release. Oxidative stress, genotoxicity, and alteration in total antioxidant capacity were evaluated through DNA damage (COMET analysis) and antioxidant concentration at 0, 3.125, 25, and 100 µg/mL. Protein markers for endothelin-1 (ET-1), interleukin-6 (IL-6), and interleukin-8 (IL-8) were also assessed to determine inflammation and endothelial alteration. Results indicate modulation of oxidative stress response in a material and dose dependent manner. PMET540 exhibited the greatest cytotoxic effect between wires and across doses. DNA damage and antioxidant concentration induced by PMET540 were significantly higher than other wires at higher doses (DNA damage increased at 25 and 100 µg/mL; Antioxidant concentration increased at 100 µg/mL). However, ET-1 concentration significantly increased only after application of 100 µg/mL PMET885. IL-6 and IL-8 were most highly expressed in BEAS2B culture after 25 µg/mL exposure to PMET540 (99.4 % Zn). This data suggests that metal composition of thermal spray wires dictates the diverse response in human bronchial cells.
Collapse
Affiliation(s)
- E.S. Burns
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, United States
| | - R.E. Harner
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, United States
- West Virginia University, School of Medicine, Morgantown, WV, United States
| | - V. Kodali
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, United States
| | - A.A. Afshari
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, United States
| | - J.M. Antonini
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, United States
| | - S.S. Leonard
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, United States
| |
Collapse
|
2
|
Mattke J, Darden CM, Vasu S, Lawrence MC, Kirkland J, Kane RR, Naziruddin B. Inhibition of Toll-like Receptor 4 Using Small Molecule, TAK-242, Protects Islets from Innate Immune Responses. Cells 2024; 13:416. [PMID: 38474380 PMCID: PMC10931053 DOI: 10.3390/cells13050416] [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: 01/26/2024] [Revised: 02/23/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
Islet transplantation is a therapeutic option to replace β-cell mass lost during type 1 or type 3c diabetes. Innate immune responses, particularly the instant blood-mediated inflammatory reaction and activation of monocytes, play a major role in the loss of transplanted islet tissue. In this study, we aimed to investigate the inhibition of toll-like receptor 4 (TLR4) on innate inflammatory responses. We first demonstrate a significant loss of graft function shortly after transplant through the assessment of miR-375 and miR-200c in plasma as biomarkers. Using in vitro models, we investigate how targeting TLR4 mitigates islet damage and immune cell activation during the peritransplant period. The results of this study support the application of TAK-242 as a therapeutic agent to reduce inflammatory and innate immune responses to islets immediately following transplantation into the hepatic portal vein. Therefore, TLR4 may serve as a target to improve islet transplant outcomes in the future.
Collapse
Affiliation(s)
- Jordan Mattke
- Institute of Biomedical Studies, Baylor University, Waco, TX 76706, USA; (J.M.); (R.R.K.)
| | - Carly M. Darden
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX 75204, USA; (C.M.D.); (J.K.)
| | - Srividya Vasu
- Islet Cell Laboratory, Baylor Scott and White Research Institute, Dallas, TX 75204, USA; (S.V.); (M.C.L.)
| | - Michael C. Lawrence
- Islet Cell Laboratory, Baylor Scott and White Research Institute, Dallas, TX 75204, USA; (S.V.); (M.C.L.)
| | - Jeffrey Kirkland
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX 75204, USA; (C.M.D.); (J.K.)
| | - Robert R. Kane
- Institute of Biomedical Studies, Baylor University, Waco, TX 76706, USA; (J.M.); (R.R.K.)
| | - Bashoo Naziruddin
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX 75204, USA; (C.M.D.); (J.K.)
| |
Collapse
|
3
|
Shih LJ, Yang CC, Liao MT, Lu KC, Hu WC, Lin CP. An important call: Suggestion of using IL-10 as therapeutic agent for COVID-19 with ARDS and other complications. Virulence 2023; 14:2190650. [PMID: 36914565 PMCID: PMC10026935 DOI: 10.1080/21505594.2023.2190650] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023] Open
Abstract
The global coronavirus disease 2019 (COVID-19) pandemic has a detrimental impact on public health. COVID-19 usually manifests as pneumonia, which can progress into acute respiratory distress syndrome (ARDS) related to uncontrolled TH17 immune reaction. Currently, there is no effective therapeutic agent to manage COVID-19 with complications. The currently available anti-viral drug remdesivir has an effectiveness of 30% in SARS-CoV-2-induced severe complications. Thus, there is a need to identify effective agents to treat COVID-19 and the associated acute lung injury and other complications. The host immunological pathway against this virus typically involves the THαβ immune response. THαβ immunity is triggered by type 1 interferon and interleukin-27 (IL-27), and the main effector cells of the THαβ immune response are IL10-CD4 T cells, CD8 T cells, NK cells, and IgG1-producing B cells. In particular, IL-10 exerts a potent immunomodulatory or anti-inflammatory effect and is an anti-fibrotic agent for pulmonary fibrosis. Concurrently, IL-10 can ameliorate acute lung injury or ARDS, especially those caused by viruses. Owing to its anti-viral activity and anti-pro-inflammatory effects, in this review, IL-10 is suggested as a possible treatment agent for COVID-19.
Collapse
Affiliation(s)
- Li-Jane Shih
- Department of Medical Laboratory, Taoyuan Armed Forces General Hospital, Taoyuan City, Taiwan
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei City, Taiwan
| | - Chun-Chun Yang
- Department of Laboratory Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
- National Defense Medical Center, Department of Pediatrics, Tri-Service General Hospital, Taipei, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Wan-Chung Hu
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Chih-Pei Lin
- Department of Laboratory Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- h Department of Biotechnology, Ming Chuan University, Taoyuan, Taiwan
| |
Collapse
|
4
|
Tomos I, Roussis I, Matthaiou AM, Dimakou K. Molecular and Genetic Biomarkers in Idiopathic Pulmonary Fibrosis: Where Are We Now? Biomedicines 2023; 11:2796. [PMID: 37893169 PMCID: PMC10604739 DOI: 10.3390/biomedicines11102796] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) represents a chronic progressive fibrotic interstitial lung disease of unknown cause with an ominous prognosis. It remains an unprecedent clinical challenge due to its delayed diagnosis and unpredictable clinical course. The need for accurate diagnostic, prognostic and predisposition biomarkers in everyday clinical practice becomes more necessary than ever to ensure prompt diagnoses and early treatment. The identification of such blood biomarkers may also unravel novel drug targets against IPF development and progression. So far, the role of diverse blood biomarkers, implicated in various pathogenetic pathways, such as in fibrogenesis (S100A4), extracellular matrix remodelling (YKL-40, MMP-7, ICAM-1, LOXL2, periostin), chemotaxis (CCL-18, IL-8), epithelial cell injury (KL-6, SP-A, SP-D), autophagy and unfolded protein response has been investigated in IPF with various results. Moreover, the recent progress in genetics in IPF allows for a better understanding of the underlying disease mechanisms. So far, the causative mutations in pulmonary fibrosis include mutations in telomere-related genes and in surfactant-related genes, markers that could act as predisposition biomarkers in IPF. The aim of this review is to provide a comprehensive overview from the bench to bedside of current knowledge and recent insights on biomarkers in IPF, and to suggest future directions for research. Large-scale studies are still needed to confirm the exact role of these biomarkers.
Collapse
Affiliation(s)
- Ioannis Tomos
- 5th Department of Respiratory Medicine, ‘SOTIRIA’ Chest Diseases Hospital of Athens, 11527 Athens, Greece; (I.R.); (A.M.M.); (K.D.)
| | - Ioannis Roussis
- 5th Department of Respiratory Medicine, ‘SOTIRIA’ Chest Diseases Hospital of Athens, 11527 Athens, Greece; (I.R.); (A.M.M.); (K.D.)
| | - Andreas M. Matthaiou
- 5th Department of Respiratory Medicine, ‘SOTIRIA’ Chest Diseases Hospital of Athens, 11527 Athens, Greece; (I.R.); (A.M.M.); (K.D.)
- Laboratory of Molecular and Cellular Pneumonology, Medical School, University of Crete, 714 09 Heraklion, Greece
- Respiratory Physiology Laboratory, Medical School, University of Cyprus, Nicosia 2029, Cyprus
| | - Katerina Dimakou
- 5th Department of Respiratory Medicine, ‘SOTIRIA’ Chest Diseases Hospital of Athens, 11527 Athens, Greece; (I.R.); (A.M.M.); (K.D.)
| |
Collapse
|
5
|
Brigham E, Hashimoto A, Alexis NE. Air Pollution and Diet: Potential Interacting Exposures in Asthma. Curr Allergy Asthma Rep 2023; 23:541-553. [PMID: 37440094 DOI: 10.1007/s11882-023-01101-1] [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] [Accepted: 05/17/2023] [Indexed: 07/14/2023]
Abstract
PURPOSE OF REVIEW To provide a review of emerging literature describing the impact of diet on the respiratory response to air pollution in asthma. RECENT FINDINGS Asthma phenotyping (observable characteristics) and endotyping (mechanistic pathways) have increased the specificity of diagnostic and treatment pathways and opened the doors to the identification of subphenotypes with enhanced susceptibility to exposures and interventions. Mechanisms underlying the airway immune response to air pollution are still being defined but include oxidative stress, inflammation, and activation of adaptive and innate immune responses, with genetic susceptibility highlighted. Of these, neutrophil recruitment and activation appear prominent; however, understanding neutrophil function in response to pollutant exposures is a research gap. Diet may play a role in asthma pathogenesis and morbidity; therefore, diet modification is a potential target opportunity to protect against pollutant-induced lung injury. In particular, in vivo and in vitro data suggest the potential for diet to modify the inflammatory response in the airways, including impacts on neutrophil recruitment and function. Murine models provide compelling results in regard to the potential for dietary components (including fiber, antioxidants, and omega-3 fatty acids) to buffer against the inflammatory response to air pollution in the lung. Precision lifestyle approaches to asthma management and respiratory protection in the context of air pollution exposures may evolve to include diet, pending the results of further epidemiologic and causal investigation and with neutrophil recruitment and activation as a candidate mechanism.
Collapse
Affiliation(s)
- Emily Brigham
- Division of Respirology, University of British Columbia, Vancouver, BC, Canada.
- Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.
| | - Alisa Hashimoto
- Faculty of Science, University of British Columbia, BC, Vancouver, Canada
| | - Neil E Alexis
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Pediatrics, Division of Allergy, Immunology, Rheumatology and Infectious Disease, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
6
|
Maus KD, Stephenson DJ, Macknight HP, Vu NT, Hoeferlin LA, Kim M, Diegelmann RF, Xie X, Chalfant CE. Skewing cPLA 2α activity toward oxoeicosanoid production promotes neutrophil N2 polarization, wound healing, and the response to sepsis. Sci Signal 2023; 16:eadd6527. [PMID: 37433004 PMCID: PMC10565596 DOI: 10.1126/scisignal.add6527] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 06/16/2023] [Indexed: 07/13/2023]
Abstract
Uncontrolled inflammation is linked to poor outcomes in sepsis and wound healing, both of which proceed through distinct inflammatory and resolution phases. Eicosanoids are a class of bioactive lipids that recruit neutrophils and other innate immune cells. The interaction of ceramide 1-phosphate (C1P) with the eicosanoid biosynthetic enzyme cytosolic phospholipase A2 (cPLA2) reduces the production of a subtype of eicosanoids called oxoeicosanoids. We investigated the effect of shifting the balance in eicosanoid biosynthesis on neutrophil polarization and function. Knockin mice expressing a cPLA2 mutant lacking the C1P binding site (cPLA2αKI/KI mice) showed enhanced and sustained neutrophil infiltration into wounds and the peritoneum during the inflammatory phase of wound healing and sepsis, respectively. The mice exhibited improved wound healing and reduced susceptibility to sepsis, which was associated with an increase in anti-inflammatory N2-type neutrophils demonstrating proresolution behaviors and a decrease in proinflammatory N1-type neutrophils. The N2 polarization of cPLA2αKI/KI neutrophils resulted from increased oxoeicosanoid biosynthesis and autocrine signaling through the oxoeicosanoid receptor OXER1 and partially depended on OXER1-dependent inhibition of the pentose phosphate pathway (PPP). Thus, C1P binding to cPLA2α suppresses neutrophil N2 polarization, thereby impairing wound healing and the response to sepsis.
Collapse
Affiliation(s)
- Kenneth D Maus
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Daniel J Stephenson
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA 22903, USA
| | - H Patrick Macknight
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA 22903, USA
| | - Ngoc T Vu
- Department of Applied Biochemistry, School of Biotechnology, International University-VNU HCM, Ho Chi Minh City, Vietnam
| | - L Alexis Hoeferlin
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University-School of Medicine, Richmond VA 23298, USA
| | - Minjung Kim
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Robert F Diegelmann
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University-School of Medicine, Richmond VA 23298, USA
| | - Xiujie Xie
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA 22903, USA
| | - Charles E Chalfant
- Department of Medicine, Division of Hematology and Oncology, University of Virginia, Charlottesville, VA 22903, USA
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22903, USA
- Program in Cancer Biology, University of Virginia Cancer Center, Charlottesville, VA 22903, USA
- Research Service, Richmond Veterans Administration Medical Center, Richmond VA, 23298, USA
| |
Collapse
|
7
|
Calvert BA, Quiroz EJ, Lorenzana Z, Doan N, Kim S, Senger CN, Anders JJ, Wallace WD, Salomon MP, Henley J, Ryan AL. Neutrophilic inflammation promotes SARS-CoV-2 infectivity and augments the inflammatory responses in airway epithelial cells. Front Immunol 2023; 14:1112870. [PMID: 37006263 PMCID: PMC10061003 DOI: 10.3389/fimmu.2023.1112870] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction In response to viral infection, neutrophils release inflammatory mediators as part of the innate immune response, contributing to pathogen clearance through virus internalization and killing. Pre- existing co-morbidities correlating to incidence to severe COVID-19 are associated with chronic airway neutrophilia. Furthermore, examination of COVID-19 explanted lung tissue revealed a series of epithelial pathologies associated with the infiltration and activation of neutrophils, indicating neutrophil activity in response to SARS-CoV-2 infection. Methods To determine the impact of neutrophil-epithelial interactions on the infectivity and inflammatory responses to SARS-CoV-2 infection, we developed a co-culture model of airway neutrophilia. This model was infected with live SARS-CoV-2 virus the epithelial response to infection was evaluated. Results SARS-CoV-2 infection of airway epithelium alone does not result in a notable pro-inflammatory response from the epithelium. The addition of neutrophils induces the release of proinflammatory cytokines and stimulates a significantly augmented proinflammatory response subsequent SARS-CoV-2 infection. The resulting inflammatory responses are polarized with differential release from the apical and basolateral side of the epithelium. Additionally, the integrity of the \epithelial barrier is impaired with notable epithelial damage and infection of basal stem cells. Conclusions This study reveals a key role for neutrophil-epithelial interactions in determining inflammation and infectivity.
Collapse
Affiliation(s)
- Ben A. Calvert
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa, IA, United States
| | - Erik J. Quiroz
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa, IA, United States
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Zareeb Lorenzana
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ngan Doan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Seongjae Kim
- The Salk Institute of Biological Studies, La Jolla, CA, United States
| | - Christiana N. Senger
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jeffrey J. Anders
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa, IA, United States
| | - Wiliam D. Wallace
- Department of Pathology, University of Southern California, Los Angeles, CA, United States
| | - Matthew P. Salomon
- Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jill Henley
- Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa, IA, United States
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
8
|
Yee AJ, Kandasamy J, Ambalavanan N, Ren C, Halloran B, Olave N, Nicola T, Jilling T. Platelet Activating Factor Activity Modulates Hyperoxic Neonatal Lung Injury Severity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532697. [PMID: 36993203 PMCID: PMC10055044 DOI: 10.1101/2023.03.14.532697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Hyperoxia-induced inflammation contributes significantly to developmental lung injury and bronchopulmonary dysplasia (BPD) in preterm infants. Platelet activating factor (PAF) is known to be a major driver of inflammation in lung diseases such as asthma and pulmonary fibrosis, but its role in BPD has not been previously investigated. Therefore, to determine whether PAF signaling independently modulates neonatal hyperoxic lung injury and BPD pathogenesis, lung structure was assessed in 14 day-old C57BL/6 wild-type (WT) and PAF receptor knockout (PTAFR KO) mice that were exposed to 21% (normoxia) or 85% O 2 (hyperoxia) from postnatal day 4. Lung morphometry showed that PTAFR KO mice had attenuated hyperoxia-induced alveolar simplification when compared to WT mice. Functional analysis of gene expression data from hyperoxia-exposed vs. normoxia-exposed lungs of WT and PTAFR KO showed that the most upregulated pathways were the hypercytokinemia/hyperchemokinemia pathway in WT mice, NAD signaling pathway in PTAFR KO mice, and agranulocyte adhesion and diapedesis as well as other pro-fibrotic pathways such as tumor microenvironment and oncostatin-M signaling in both mice strains, indicating that PAF signaling may contribute to inflammation but may not be a significant mediator of fibrotic processes during hyperoxic neonatal lung injury. Gene expression analysis also indicated increased expression of pro-inflammatory genes such as CXCL1, CCL2 and IL-6 in the lungs of hyperoxia-exposed WT mice and metabolic regulators such as HMGCS2 and SIRT3 in the lungs of PTAFR KO mice, suggesting that PAF signaling may modulate BPD risk through changes in pulmonary inflammation and/or metabolic reprogramming in preterm infants.
Collapse
|
9
|
Asl ER, Rostamzadeh D, Duijf PHG, Mafi S, Mansoori B, Barati S, Cho WC, Mansoori B. Mutant P53 in the formation and progression of the tumor microenvironment: Friend or foe. Life Sci 2023; 315:121361. [PMID: 36608871 DOI: 10.1016/j.lfs.2022.121361] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/20/2022] [Accepted: 12/29/2022] [Indexed: 01/07/2023]
Abstract
TP53 is the most frequently mutated gene in human cancer. It encodes the tumor suppressor protein p53, which suppresses tumorigenesis by acting as a critical transcription factor that can induce the expression of many genes controlling a plethora of fundamental cellular processes, including cell cycle progression, survival, apoptosis, and DNA repair. Missense mutations are the most frequent type of mutations in the TP53 gene. While these can have variable effects, they typically impair p53 function in a dominant-negative manner, thereby altering intra-cellular signaling pathways and promoting cancer development. Additionally, it is becoming increasingly apparent that p53 mutations also have non-cell autonomous effects that influence the tumor microenvironment (TME). The TME is a complex and heterogeneous milieu composed of both malignant and non-malignant cells, including cancer-associated fibroblasts (CAFs), adipocytes, pericytes, different immune cell types, such as tumor-associated macrophages (TAMs) and T and B lymphocytes, as well as lymphatic and blood vessels and extracellular matrix (ECM). Recently, a large body of evidence has demonstrated that various types of p53 mutations directly affect TME. They fine-tune the inflammatory TME and cell fate reprogramming, which affect cancer progression. Notably, re-educating the p53 signaling pathway in the TME may be an effective therapeutic strategy in combating cancer. Therefore, it is timely to here review the recent advances in our understanding of how TP53 mutations impact the fate of cancer cells by reshaping the TME.
Collapse
Affiliation(s)
- Elmira Roshani Asl
- Department of Biochemistry, Saveh University of Medical Sciences, Saveh, Iran
| | - Davoud Rostamzadeh
- Department of Clinical Biochemistry, Yasuj University of Medical Sciences, Yasuj, Iran; Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Pascal H G Duijf
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia; Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD, Australia; Centre for Data Science, Queensland University of Technology, Brisbane, QLD, Australia; Cancer and Aging Research Program, Queensland University of Technology, Brisbane, QLD, Australia; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Sahar Mafi
- Department of Clinical Biochemistry, Yasuj University of Medical Sciences, Yasuj, Iran; Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Behnaz Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, Hong Kong
| | - Behzad Mansoori
- The Wistar Institute, Molecular & Cellular Oncogenesis Program, Philadelphia, PA, United States.
| |
Collapse
|
10
|
Zhou Z, Shen D, Wang K, Liu J, Li M, Win-Shwe TT, Nagaoka K, Li C. Pulmonary microbiota intervention alleviates fine particulate matter-induced lung inflammation in broilers. J Anim Sci 2023; 101:skad207. [PMID: 37341706 PMCID: PMC10390102 DOI: 10.1093/jas/skad207] [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: 02/06/2023] [Accepted: 06/20/2023] [Indexed: 06/22/2023] Open
Abstract
Fine particulate matter (PM2.5) released during the livestock industry endangers the respiratory health of animals. Our previous findings suggested that broilers exposed to PM2.5 exhibited lung inflammation and changes in the pulmonary microbiome. Therefore, this study was to investigate whether the pulmonary microbiota plays a causal role in the pathogenesis of PM2.5-induced lung inflammation. We first used antibiotics to establish a pulmonary microbiota intervention broiler model, which showed a significantly reduced total bacterial load in the lungs without affecting the microbiota composition or structure. Based on it, 45 AA broilers of similar body weight were randomly assigned to three groups: control (CON), PM2.5 (PM), and pulmonary microbiota intervention (ABX-PM). From 21 d of age, broilers in the ABX-PM group were intratracheally instilled with antibiotics once a day for 3 d. Meanwhile, broilers in the other two groups were simultaneously instilled with sterile saline. On 24 and 26 d of age, broilers in the PM and ABX-PM groups were intratracheally instilled with PM2.5 suspension to induce lung inflammation, and broilers in the CON group were simultaneously instilled with sterile saline. The lung histomorphology, inflammatory cytokines' expression levels, lung microbiome, and microbial growth conditions were analyzed to determine the effect of the pulmonary microbiota on PM2.5-induced lung inflammation. Broilers in the PM group showed lung histological injury, while broilers in the ABX-PM group had normal lung histomorphology. Furthermore, microbiota intervention significantly reduced mRNA expression levels of interleukin-1β, tumor necrosis factor-α, interleukin-6, interleukin-8, toll-like receptor 4 and nuclear factor kappa-B. PM2.5 induced significant changes in the β diversity and structure of the pulmonary microbiota in the PM group. However, no significant changes in microbiota structure were observed in the ABX-PM group. Moreover, the relative abundance of Enterococcus cecorum in the PM group was significantly higher than that in the CON and ABX-PM groups. And sterile bronchoalveolar lavage fluid from the PM group significantly promoted the growth of E. cecorum, indicating that PM2.5 altered the microbiota's growth condition. In conclusion, pulmonary microbiota can affect PM2.5-induced lung inflammation in broilers. PM2.5 can alter the bacterial growth environment and promote dysbiosis, potentially exacerbating inflammation.
Collapse
Affiliation(s)
- Zilin Zhou
- Jiangsu Joint International Research Laboratory of Animal Gastrointestinal Genomes, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Dan Shen
- Jiangsu Joint International Research Laboratory of Animal Gastrointestinal Genomes, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Kai Wang
- Jiangsu Joint International Research Laboratory of Animal Gastrointestinal Genomes, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Junze Liu
- Jiangsu Joint International Research Laboratory of Animal Gastrointestinal Genomes, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Mingyang Li
- Jiangsu Joint International Research Laboratory of Animal Gastrointestinal Genomes, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Tin-Tin Win-Shwe
- Center for Environmental Risk Research, National Institute for Environmental Studies, Tsukuba 305-8506, Japan
| | - Kentaro Nagaoka
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Tokyo 183-8509, Japan
| | - Chunmei Li
- Jiangsu Joint International Research Laboratory of Animal Gastrointestinal Genomes, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, PR China
| |
Collapse
|
11
|
Pantzke J, Offer S, Zimmermann EJ, Kuhn E, Streibel T, Oeder S, Di Bucchianico S, Zimmermann R. An alternative in vitro model considering cell-cell interactions in fiber-induced pulmonary fibrosis. Toxicol Mech Methods 2022:1-16. [DOI: 10.1080/15376516.2022.2156008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Jana Pantzke
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Rostock, Germany
| | - Svenja Offer
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Rostock, Germany
| | - Elias J. Zimmermann
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Rostock, Germany
| | - Evelyn Kuhn
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Neuherberg, Germany
| | - Thorsten Streibel
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Rostock, Germany
| | - Sebastian Oeder
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Neuherberg, Germany
| | - Sebastiano Di Bucchianico
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Rostock, Germany
| | - Ralf Zimmermann
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Rostock, Germany
| |
Collapse
|
12
|
Lacticaseibacillus rhamnosus attenuates acute lung inflammation in a murine model of acute respiratory distress syndrome: Relevance to cytokines associated to STAT4/T-bet and STAT3/RORɣt”. Microb Pathog 2022; 173:105831. [DOI: 10.1016/j.micpath.2022.105831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
|
13
|
An in vitro alveolar model allows for the rapid assessment of chemical respiratory sensitization with modifiable biomarker endpoints. Chem Biol Interact 2022; 368:110232. [DOI: 10.1016/j.cbi.2022.110232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/07/2022] [Accepted: 10/21/2022] [Indexed: 11/23/2022]
|
14
|
Matsuda M, Inaba M, Hamaguchi J, Tomita H, Omori M, Shimora H, Sakae H, Kitatani K, Nabe T. Local IL-10 replacement therapy was effective for steroid-insensitive asthma in mice. Int Immunopharmacol 2022; 110:109037. [PMID: 35810490 DOI: 10.1016/j.intimp.2022.109037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/27/2022] [Accepted: 07/04/2022] [Indexed: 01/24/2023]
Abstract
Subgroups of patients with severe asthma showing marked increases in sputum eosinophils and/or neutrophils are insensitive to corticosteroids. Previous reports have shown that exogenous administration of an anti-inflammatory cytokine, interleukin (IL)-10 negatively regulated both eosinophilic and neutrophilic migration into tissues. The objective of this study was to elucidate whether intratracheal IL-10 administration suppresses asthmatic responses in a steroid-insensitive model of mice. Ovalbumin (OVA)-sensitized BALB/c mice were intratracheally challenged with OVA at 500 µg/animal four times. Dexamethasone (1 mg/kg, intraperitoneal) or IL-10 (25 ng/mouse, intratracheal) was administered during the multiple challenges. The number of leukocytes, expression of intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and IL-10 receptor in the lung, and the development of airway remodeling and hyperresponsiveness were evaluated after the fourth challenge. Consistent with our previous study, dexamethasone hardly suppressed the development of airway remodeling and hyperresponsiveness. Although intratracheal IL-10 administration did not affect the development of airway remodeling, the infiltration of eosinophils and neutrophils, and the development of airway hyperresponsiveness were significantly inhibited. Moreover, IL-10 administration significantly decreased the numbers of ICAM-1+ and VCAM-1+ pulmonary vascular endothelial cells, which express IL-10 receptor 1, even though neither production of eosinophilic nor neutrophilic cytokines in the lung was inhibited. Therefore, IL-10 can suppress eosinophil and neutrophil infiltration by inhibiting the proliferation of ICAM-1+ and VCAM-1+ pulmonary vascular endothelial cells, resulting in inhibition of airway hyperresponsiveness in steroid-insensitive asthmatic mice. IL-10 replacement therapy may be clinically useful for the treatment of steroid-insensitive asthma.
Collapse
Affiliation(s)
- Masaya Matsuda
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Miki Inaba
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Junpei Hamaguchi
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Hiro Tomita
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Miyu Omori
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Hayato Shimora
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Harumi Sakae
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Kazuyuki Kitatani
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Takeshi Nabe
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan.
| |
Collapse
|
15
|
Identification of bronchoalveolar and blood immune-inflammatory biomarker signature associated with poor 28-day outcome in critically ill COVID-19 patients. Sci Rep 2022; 12:9502. [PMID: 35681070 PMCID: PMC9178326 DOI: 10.1038/s41598-022-13179-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/20/2022] [Indexed: 11/09/2022] Open
Abstract
The local immune-inflammatory response elicited by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is still poorly described, as well as the extent to which its characteristics may be associated with the outcome of critical Coronavirus disease 2019 (COVID-19). In this prospective monocenter study, all consecutive COVID-19 critically ill patients admitted from February to December 2020 and explored by fiberoptic bronchoscopy with bronchoalveolar lavage (BAL) were included. Biological assays, including digital ELISA cytokine profiling and targeted eicosanoid metabolomic analysis, were performed on paired blood and BAL fluid (BALF). Clinical outcome was assessed through the World Health Organization 10-point Clinical Progression Scale (WHO-CPS) at the 28th day (D28) following the admission to intensive care unit. A D28-WHO-CPS value higher than 5 defined a poor outcome. Seventy-six patients were included, 45 (59%) had a poor day-28 outcome. As compared to their counterparts, patients with D28-WHO-CPS > 5 exhibited a neutrophil-predominant bronchoalveolar phenotype, with a higher BALF neutrophil/lymphocyte ratio, a blunted local type I interferon response, a decompartimentalized immune-inflammatory response illustrated by lower BALF/blood ratio of concentrations of IL-6 (1.68 [0.30-4.41] vs. 9.53 [2.56-19.1]; p = 0.001), IL-10, IL-5, IL-22 and IFN-γ, and a biological profile of vascular endothelial injury illustrated by a higher blood concentration of VEGF and higher blood and/or BALF concentrations of several vasoactive eicosanoids. In critically ill COVID-19 patients, we identified bronchoalveolar and blood immune-inflammatory biomarker signature associated with poor 28-day outcome.
Collapse
|
16
|
Calvert BA, Quiroz EJ, Lorenzana Z, Doan N, Kim S, Senger CN, Wallace WD, Salomon MP, Henley J, Ryan AL. Neutrophilic inflammation promotes SARS-CoV-2 infectivity and augments the inflammatory responses in airway epithelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2021.08.09.455472. [PMID: 34401877 PMCID: PMC8366793 DOI: 10.1101/2021.08.09.455472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In response to viral infection, neutrophils release inflammatory mediators as part of the innate immune response, contributing to pathogen clearance through virus internalization and killing. Pre-existing co- morbidities correlating to incidence of severe COVID-19 are associated with chronic airway neutrophilia. Furthermore, examination of COVID-19 explanted lung tissue revealed a series of epithelial pathologies associated with the infiltration and activation of neutrophils, indicating neutrophil activity in response to SARS- CoV-2 infection. To determine the impact of neutrophil-epithelial interactions on the infectivity and inflammatory responses to SARS-CoV-2 infection, we developed a co-culture model of airway neutrophilia. SARS-CoV-2 infection of the airway epithelium alone does not result in a notable pro-inflammatory response from the epithelium. The addition of neutrophils induces the release of proinflammatory cytokines and stimulates a significantly augmented pro-inflammatory response subsequent SARS-CoV-2 infection. The resulting inflammatory response is polarized with differential release from the apical and basolateral side of the epithelium. Additionally, the integrity of the epithelial barrier is impaired with notable epithelial damage and infection of basal stem cells. This study reveals a key role for neutrophil-epithelial interactions in determining inflammation and infectivity in response to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- BA Calvert
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - EJ Quiroz
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Z Lorenzana
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - N Doan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - S Kim
- The Salk Institute of Biological Studies, 10010 North Torey Pines Road, La Jolla, Ca, USA
| | - CN Senger
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - WD Wallace
- Department of Pathology, University of Southern California, Los Angeles, CA, USA
| | - MP Salomon
- Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - J Henley
- Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - AL Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
17
|
Lin Q, Ni H, Zheng Z, Zhong J, Nie H. Cross-talk of four types of RNA modification writers defines the immune microenvironment in severe asthma. Ann N Y Acad Sci 2022; 1514:93-103. [PMID: 35506887 DOI: 10.1111/nyas.14782] [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: 12/20/2022]
Abstract
Adenine modifications, including m6 A, m1 A, APA, and A-to-I modifications, are the most impactful RNA modifications. These modifications are primarily produced by enzymes called writers. The main purpose of this study was to explore the cross-talk and potential roles of these writers in severe asthma. We found 13 RNA writers potentially related to severe asthma and three RNA modification patterns. Cluster 3 showed predominant neutrophil infiltration and C-type lectin receptor signaling; cluster 1 showed predominant innate immune cell infiltration and ubiquitin-proteasome system activation; and cluster 2 did not show obvious immune infiltration characteristics. We found that RNA modification writers modified immune cell-related genes and led to both accumulation of different immune cells in the airways and activation of a series of biological processes, which ultimately leads to severe asthma. TRMT6, WTAP, and TRMT6A were included in a random forest model as predictors. Cromoglicic acid, thioperamide, and fluvastatin were potential drugs for clusters 1, 2, and 3, respectively. We found that cross-talk of RNA modifications is significant in severe asthma, which provides insight into severe asthma pathogenesis and possible treatment avenues.
Collapse
Affiliation(s)
- Qibin Lin
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haiyang Ni
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhishui Zheng
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jieying Zhong
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hanxiang Nie
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
18
|
Lung Hyaluronasome: Involvement of Low Molecular Weight Ha (Lmw-Ha) in Innate Immunity. Biomolecules 2022; 12:biom12050658. [PMID: 35625586 PMCID: PMC9138743 DOI: 10.3390/biom12050658] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/01/2023] Open
Abstract
Hyaluronic acid (HA) is a major component of the extracellular matrix. It is synthesized by hyaluronan synthases (HAS) into high-molecular-weight chains (HMW-HA) that exhibit anti-inflammatory and immunomodulatory functions. In damaged, infected, and/or inflamed tissues, HMW-HA are degraded by hyaluronidases (HYAL) or reactive oxygen species (ROS) to give rise to low-molecular-weight HAs (LMW-HAs) that are potent pro-inflammatory molecules. Therefore, the size of HA regulates the balance of anti- or pro-inflammatory functions. The activities of HA depend also on its interactions with hyaladherins. HA synthesis, degradation, and activities through HA/receptors interactions define the hyaluronasome. In this review, a short overview of the role of high and low-molecular-weight HA polymers in the lungs is provided. The involvement of LMW-HA in pulmonary innate immunity via the activation of neutrophils, macrophages, dendritic cells, and epithelial cells is described to highlight LMW-HA as a therapeutic target in inflammatory respiratory diseases. Finally, the possibilities to counter LMW-HA’s deleterious effects in the lungs are discussed.
Collapse
|
19
|
Scheithauer L, Thiem S, Ünal CM, Dellmann A, Steinert M. Zinc Metalloprotease ProA from Legionella pneumophila Inhibits the Pro-Inflammatory Host Response by Degradation of Bacterial Flagellin. Biomolecules 2022; 12:624. [PMID: 35625552 PMCID: PMC9138289 DOI: 10.3390/biom12050624] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/08/2022] [Accepted: 04/09/2022] [Indexed: 01/27/2023] Open
Abstract
The environmental bacterium Legionella pneumophila is an intracellular pathogen of various protozoan hosts and able to cause Legionnaires' disease, a severe pneumonia in humans. By encoding a wide selection of virulence factors, the infectious agent possesses several strategies to manipulate its host cells and evade immune detection. In the present study, we demonstrate that the L. pneumophila zinc metalloprotease ProA functions as a modulator of flagellin-mediated TLR5 stimulation and subsequent activation of the pro-inflammatory NF-κB pathway. We found ProA to be capable of directly degrading immunogenic FlaA monomers but not the polymeric form of bacterial flagella. These results indicate a role of the protease in antagonizing immune stimulation, which was further substantiated in HEK-BlueTM hTLR5 Detection assays. Addition of purified proteins, bacterial suspensions of L. pneumophila mutant strains as well as supernatants of human lung tissue explant infection to this reporter cell line demonstrated that ProA specifically decreases the TLR5 response via FlaA degradation. Conclusively, the zinc metalloprotease ProA serves as a powerful regulator of exogenous flagellin and presumably creates an important advantage for L. pneumophila proliferation in mammalian hosts by promoting immune evasion.
Collapse
Affiliation(s)
- Lina Scheithauer
- Institut für Mikrobiologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany; (L.S.); (S.T.); (C.M.Ü.)
| | - Stefanie Thiem
- Institut für Mikrobiologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany; (L.S.); (S.T.); (C.M.Ü.)
| | - Can M. Ünal
- Institut für Mikrobiologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany; (L.S.); (S.T.); (C.M.Ü.)
| | - Ansgar Dellmann
- Institut für Pathologie, Städtisches Klinikum Braunschweig, Celler Straße 38, 38114 Braunschweig, Germany;
| | - Michael Steinert
- Institut für Mikrobiologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany; (L.S.); (S.T.); (C.M.Ü.)
- Helmholtz Center for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| |
Collapse
|
20
|
Tajbakhsh A, Gheibihayat SM, Mortazavi D, Medhati P, Rostami B, Savardashtaki A, Momtazi-Borojeni AA. The Effect of Cigarette Smoke Exposure on Efferocytosis in Chronic Obstructive Pulmonary Disease; Molecular Mechanisms and Treatment Opportunities. COPD 2021; 18:723-736. [PMID: 34865568 DOI: 10.1080/15412555.2021.1978419] [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: 01/04/2023]
Abstract
Cigarette smoking-related inflammation, cellular stresses, and tissue destruction play a key role in lung disease, such as chronic obstructive pulmonary disease (COPD). Notably, augmented apoptosis and impaired clearance of apoptotic cells, efferocytosis, contribute to the chronic inflammatory response and tissue destruction in patients with COPD. Of note, exposure to cigarette smoke can impair alveolar macrophages efferocytosis activity, which leads to secondary necrosis formation and tissue inflammation. A better understanding of the processes behind the effect of cigarette smoke on efferocytosis concerning lung disorders can help to design more efficient treatment approaches and also delay the development of lung disease, such as COPD. To this end, we aimed to seek mechanisms underlying the impairing effect of cigarette smoke on macrophages-mediated efferocytosis in COPD. Further, available therapeutic opportunities for restoring efferocytosis activity and ameliorating respiratory tract inflammation in smokers with COPD were also discussed.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Deniz Mortazavi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Pourya Medhati
- Student research committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behrouz Rostami
- Health & Treatment Center of Rostam, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Abbas Momtazi-Borojeni
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Iran's National Elites Foundation, Tehran, Iran
| |
Collapse
|
21
|
Souza TN, Valdez AF, Rizzo J, Zamith-Miranda D, Guimarães AJ, Nosanchuk JD, Nimrichter L. Host cell membrane microdomains and fungal infection. Cell Microbiol 2021; 23:e13385. [PMID: 34392593 PMCID: PMC8664998 DOI: 10.1111/cmi.13385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/14/2021] [Accepted: 07/24/2021] [Indexed: 01/13/2023]
Abstract
Lipid microdomains or lipid rafts are dynamic and tightly ordered regions of the plasma membrane. In mammalian cells, they are enriched in cholesterol, glycosphingolipids, Glycosylphosphatidylinositol-anchored and signalling-related proteins. Several studies have suggested that mammalian pattern recognition receptors are concentrated or recruited to lipid domains during host-pathogen association to enhance the effectiveness of host effector processes. However, pathogens have also evolved strategies to exploit these domains to invade cells and survive. In fungal organisms, a complex cell wall network usually mediates the first contact with the host cells. This cell wall may contain virulence factors that interfere with the host membrane microdomains dynamics, potentially impacting the infection outcome. Indeed, the microdomain disruption can dampen fungus-host cell adhesion, phagocytosis and cellular immune responses. Here, we provide an overview of regulatory strategies employed by pathogenic fungi to engage with and potentially subvert the lipid microdomains of host cells. TAKE AWAY: Lipid microdomains are ordered regions of the plasma membrane enriched in cholesterol, glycosphingolipids (GSL), GPI-anchored and signalling-related proteins. Pathogen recognition by host immune cells can involve lipid microdomain participation. During this process, these domains can coalesce in larger complexes recruiting receptors and signalling proteins, significantly increasing their signalling abilities. The antifungal innate immune response is mediated by the engagement of pathogen-associated molecular patterns to pattern recognition receptors (PRRs) at the plasma membrane of innate immune cells. Lipid microdomains can concentrate or recruit PRRs during host cell-fungi association through a multi-interactive mechanism. This association can enhance the effectiveness of host effector processes. However, virulence factors at the fungal cell surface and extracellular vesicles can re-assembly these domains, compromising the downstream signalling and favouring the disease development. Lipid microdomains are therefore very attractive targets for novel drugs to combat fungal infections.
Collapse
Affiliation(s)
- Taiane N Souza
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro F Valdez
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Rizzo
- Unité Biologie des ARN des Pathogènes Fongiques, Département de Mycologie, Institut Pasteur, Paris, France
| | - Daniel Zamith-Miranda
- Departments of Medicine (Division of Infectious Diseases) and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Allan Jefferson Guimarães
- Departamento de Microbiologia e Parasitologia-MIP, Instituto Biomédico, Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Joshua D Nosanchuk
- Departments of Medicine (Division of Infectious Diseases) and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Leonardo Nimrichter
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Rizk MA, El-Sayed SAES, Salman D, Marghani BH, Gadalla HE, Sayed-Ahmed MZ. Immunomodulatory Effect of Vitamin C on Proinflammatory Cytokines Production in Ossimi Lambs (Ovis aries) with Pneumonic Pasteurellosis. Animals (Basel) 2021; 11:ani11123374. [PMID: 34944151 PMCID: PMC8697947 DOI: 10.3390/ani11123374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 11/16/2022] Open
Abstract
In this study, we have investigated the impact of vitamin C on the production of pro-inflammatory cytokines (interleukin 1 β (IL-1 β), interleukin 6 (IL-6), interleukin 12p40 (IL-12p40), interferon gamma (IFNγ), and tumor necrosis factor alpha (TNF-α)) in lambs naturally infected by pneumonic pasteurellosis. Of 37 lambs, 18 lambs were identified to have pneumonic pasteurellosis and randomly allocated into two equal groups. Single subcutaneous dose of tulathromycine alone (2.5 mg kg−1) or tulathromycine combined with vitamin C (3 gm kg−1) were administrated to the diseased lambs. The serum levels of IL-1β, IL-6, IFN-γ, and TNF-α were returned to the normal levels in pneumonic lambs treated with the combination therapy. The obtained results indicate the selective influences of vitamin C on pro-inflammatory cytokines production in sera of lambs with pneumonic pasteurellosis and highlights the value of vitamin C as a potential anti-inflammatory drug and ideal immunomodulatory agent.
Collapse
Affiliation(s)
- Mohamed Abdo Rizk
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
- Correspondence:
| | - Shimaa Abd El-Salam El-Sayed
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Doaa Salman
- Department of Animal Medicine, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt;
| | - Basma H. Marghani
- Department of Physiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Hossam Elshahat Gadalla
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Mohamed Z. Sayed-Ahmed
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jizan 82722, Saudi Arabia
| |
Collapse
|
23
|
Líbalová H, Závodná T, Vrbová K, Sikorová J, Vojtíšek-Lom M, Beránek V, Pechout M, Kléma J, Ciganek M, Machala M, Neča J, Rössner P, Topinka J. Transcription profiles in BEAS-2B cells exposed to organic extracts from particulate emissions produced by a port-fuel injection vehicle, fueled with conventional fossil gasoline and gasoline-ethanol blend. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2021; 872:503414. [PMID: 34798934 DOI: 10.1016/j.mrgentox.2021.503414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 09/08/2021] [Accepted: 09/28/2021] [Indexed: 12/21/2022]
Abstract
Emissions from road traffic are among the major contributors to air pollution worldwide and represent a serious environmental health risk. Although traffic-related pollution has been most commonly associated with diesel engines, increasing evidence suggests that gasoline engines also produce a considerable amount of potentially hazardous particulate matter (PM). The primary objective of this study was to compare the intrinsic toxic properties of the organic components of PM, generated by a conventional gasoline engine fueled with neat gasoline (E0), or gasoline-ethanol blend (15 % ethanol, v/v, E15). Our results showed that while E15 has produced, compared to gasoline and per kg of fuel, comparable particle mass (μg PM/kg fuel) and slightly more particles by number, the organic extract from the particulate matter produced by E15 contained a larger amount of harmful polycyclic aromatic hydrocarbons (PAHs), as determined by the chemical analysis. To examine the toxicity, we monitored genome-wide gene expression changes in human lung BEAS-2B cells, exposed for 4 h and 24 h to a subtoxic dose of each PM extract. After 4 h exposure, numerous dysregulated genes and processes such as oxidative stress, lipid and steroid metabolism, PPARα signaling and immune response, were found to be common for both extract treatments. On the other hand, 24 h exposure resulted in more distinctive gene expression patterns. Although we identified several common modulated processes indicating the metabolism of PAHs and activation of aryl hydrocarbon receptor (AhR), E15 specifically dysregulated a variety of other genes and pathways related to cancer promotion and progression. Overall, our findings suggest that the ethanol addition to gasoline changed the intrinsic properties of PM emissions and increased the PAH content in PM organic extract, thus contributing to a more extensive toxic response particularly after 24 h exposure in BEAS-2B cells.
Collapse
Affiliation(s)
- Helena Líbalová
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine AS CR, Videnska 1083, 142 20, Prague, Czech Republic.
| | - Táňa Závodná
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine AS CR, Videnska 1083, 142 20, Prague, Czech Republic.
| | - Kristýna Vrbová
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine AS CR, Videnska 1083, 142 20, Prague, Czech Republic.
| | - Jitka Sikorová
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine AS CR, Videnska 1083, 142 20, Prague, Czech Republic.
| | - Michal Vojtíšek-Lom
- Center of Vehicles for Sustainable Mobility, Faculty of Mechanical Engineering, Czech Technical University in Prague, Technicka 4, 166 07, Prague, Czech Republic.
| | - Vít Beránek
- Center of Vehicles for Sustainable Mobility, Faculty of Mechanical Engineering, Czech Technical University in Prague, Technicka 4, 166 07, Prague, Czech Republic.
| | - Martin Pechout
- Department of Vehicles and Ground Transport, Faculty of Engineering, Czech University of Life Sciences, Kamycka 127, 165 21, Prague, Czech Republic.
| | - Jiří Kléma
- Department of Cybernetics, Faculty of Electrical Engineering, Czech Technical University in Prague, Karlovo namesti 13, 121 35, Prague, Czech Republic.
| | - Miroslav Ciganek
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 621 00, Brno, Czech Republic.
| | - Miroslav Machala
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 621 00, Brno, Czech Republic.
| | - Jiří Neča
- Department of Chemistry and Toxicology, Veterinary Research Institute, Hudcova 296/70, 621 00, Brno, Czech Republic.
| | - Pavel Rössner
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine AS CR, Videnska 1083, 142 20, Prague, Czech Republic.
| | - Jan Topinka
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine AS CR, Videnska 1083, 142 20, Prague, Czech Republic.
| |
Collapse
|
24
|
Lim S, Kim TJ, Kim YJ, Kim C, Ko SB, Kim BS. Senolytic Therapy for Cerebral Ischemia-Reperfusion Injury. Int J Mol Sci 2021; 22:ijms222111967. [PMID: 34769397 PMCID: PMC8584561 DOI: 10.3390/ijms222111967] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 12/13/2022] Open
Abstract
Ischemic stroke is one of the leading causes of death, and even timely treatment can result in severe disabilities. Reperfusion of the ischemic stroke region and restoration of the blood supply often lead to a series of cellular and biochemical consequences, including generation of reactive oxygen species (ROS), expression of inflammatory cytokines, inflammation, and cerebral cell damage, which is collectively called cerebral ischemia-reperfusion (IR) injury. Since ROS and inflammatory cytokines are involved in cerebral IR injury, injury could involve cellular senescence. Thus, we investigated whether senolytic therapy that eliminates senescent cells could be an effective treatment for cerebral IR injury. To determine whether IR induces neural cell senescence in vitro, astrocytes were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R). OGD/R induced astrocyte senescence and senescent cells in OGD/R-injured astrocytes were effectively eliminated in vitro by ABT263, a senolytic agent. IR in rats with intraluminal middle cerebral artery occlusion induced cellular senescence in the ischemic region. The senescent cells in IR-injured rats were effectively eliminated by intravenous injections of ABT263. Importantly, ABT263 treatment significantly reduced the infarct volume and improved neurological function in behavioral tests. This study demonstrated, for the first time, that senolytic therapy has therapeutic potential for cerebral IR injury.
Collapse
Affiliation(s)
- Songhyun Lim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea; (S.L.); (C.K.)
| | - Tae Jung Kim
- Department of Neurology, Seoul National University Hospital, Seoul 03080, Korea; (T.J.K.); (Y.-J.K.)
- Department of Critical Care Medicine, Seoul National University Hospital, Seoul 03080, Korea
| | - Young-Ju Kim
- Department of Neurology, Seoul National University Hospital, Seoul 03080, Korea; (T.J.K.); (Y.-J.K.)
| | - Cheesue Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea; (S.L.); (C.K.)
| | - Sang-Bae Ko
- Department of Neurology, Seoul National University Hospital, Seoul 03080, Korea; (T.J.K.); (Y.-J.K.)
- Department of Critical Care Medicine, Seoul National University Hospital, Seoul 03080, Korea
- Correspondence: (S.-B.K.); (B.-S.K.); Tel.: +82-2-2072-2278 (S.-B.K.); +82-2-880-1509 (B.-S.K.)
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea; (S.L.); (C.K.)
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Korea
- Institute of Chemical Processes, Seoul National University, Seoul 08826, Korea
- Institute of Engineering Research, Seoul National University, Seoul 08826, Korea
- Bio-MAX Institute, Seoul National University, Seoul 08826, Korea
- Correspondence: (S.-B.K.); (B.-S.K.); Tel.: +82-2-2072-2278 (S.-B.K.); +82-2-880-1509 (B.-S.K.)
| |
Collapse
|
25
|
Giacomelli C, Piccarducci R, Marchetti L, Romei C, Martini C. Pulmonary fibrosis from molecular mechanisms to therapeutic interventions: lessons from post-COVID-19 patients. Biochem Pharmacol 2021; 193:114812. [PMID: 34687672 PMCID: PMC8546906 DOI: 10.1016/j.bcp.2021.114812] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 02/07/2023]
Abstract
Pulmonary fibrosis (PF) is characterised by several grades of chronic inflammation and collagen deposition in the interalveolar space and is a hallmark of interstitial lung diseases (ILDs). Recently, infectious agents have emerged as driving causes for PF development; however, the role of viral/bacterial infections in the initiation and propagation of PF is still debated. In this context, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for the current coronavirus disease 2019 (COVID-19) pandemic, has been associated with acute respiratory distress syndrome (ARDS) and PF development. Although the infection by SARS-CoV-2 can be eradicated in most cases, the development of fibrotic lesions cannot be precluded; furthermore, whether these lesions are stable or progressive fibrotic events is still unknown. Herein, an overview of the main molecular mechanisms driving the fibrotic process together with the currently approved and newly proposed therapeutic solutions was given. Then, the most recent data that emerged from post-COVID-19 patients was discussed, in order to compare PF and COVID-19-dependent PF, highlighting shared and specific mechanisms. A better understanding of PF aetiology is certainly needed, also to develop effective therapeutic strategies and COVID-19 pathology is offering one more chance to do it. Overall, the work reported here could help to define new approaches for therapeutic intervention in the diversity of the ILD spectrum.
Collapse
Affiliation(s)
- Chiara Giacomelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Rebecca Piccarducci
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Laura Marchetti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Chiara Romei
- Multidisciplinary Team of Interstitial Lung Disease, Radiology Department, Pisa University Hospital, Via Paradisa 2, Pisa 56124, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy,Corresponding author
| |
Collapse
|
26
|
Lourenço LO, Ribeiro AM, Lopes FDTQDS, Tibério IDFLC, Tavares-de-Lima W, Prado CM. Different Phenotypes in Asthma: Clinical Findings and Experimental Animal Models. Clin Rev Allergy Immunol 2021; 62:240-263. [PMID: 34542807 DOI: 10.1007/s12016-021-08894-x] [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] [Accepted: 08/24/2021] [Indexed: 10/20/2022]
Abstract
Asthma is a respiratory allergic disease presenting a high prevalence worldwide, and it is responsible for several complications throughout life, including death. Fortunately, asthma is no longer recognized as a unique manifestation but as a very heterogenic manifestation. Its phenotypes and endotypes are known, respectively, as pathologic and molecular features that might not be directly associated with each other. The increasing number of studies covering this issue has brought significant insights and knowledge that are constantly expanding. In this review, we intended to summarize this new information obtained from clinical studies, which not only allowed for the creation of patient clusters by means of personalized medicine and a deeper molecular evaluation, but also created a connection with data obtained from experimental models, especially murine models. We gathered information regarding sensitization and trigger and emphasizing the most relevant phenotypes and endotypes, such as Th2-high asthma and Th2-low asthma, which included smoking and obesity-related asthma and mixed and paucigranulocytic asthma, not only in physiopathology and the clinic but also in how these phenotypes can be determined with relative similarity using murine models. We also further investigated how clinical studies have been treating patients using newly developed drugs focusing on specific biomarkers that are more relevant according to the patient's clinical manifestation of the disease.
Collapse
Affiliation(s)
- Luiz Otávio Lourenço
- Department of Biosciences, Federal University of São Paulo, Campus Baixada Santista, Santos, SP, Brazil
| | - Alessandra Mussi Ribeiro
- Department of Biosciences, Federal University of São Paulo, Campus Baixada Santista, Santos, SP, Brazil
| | | | | | - Wothan Tavares-de-Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Carla Máximo Prado
- Department of Biosciences, Federal University of São Paulo, Campus Baixada Santista, Santos, SP, Brazil. .,Department of Medicine, School of Medicine, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
27
|
de Cevins C, Luka M, Smith N, Meynier S, Magérus A, Carbone F, García-Paredes V, Barnabei L, Batignes M, Boullé A, Stolzenberg MC, Pérot BP, Charbit B, Fali T, Pirabakaran V, Sorin B, Riller Q, Abdessalem G, Beretta M, Grzelak L, Goncalves P, Di Santo JP, Mouquet H, Schwartz O, Zarhrate M, Parisot M, Bole-Feysot C, Masson C, Cagnard N, Corneau A, Brunaud C, Zhang SY, Casanova JL, Bader-Meunier B, Haroche J, Melki I, Lorrot M, Oualha M, Moulin F, Bonnet D, Belhadjer Z, Leruez M, Allali S, Gras-Leguen C, de Pontual L, Fischer A, Duffy D, Rieux-Laucat F, Toubiana J, Ménager MM. A monocyte/dendritic cell molecular signature of SARS-CoV-2-related multisystem inflammatory syndrome in children with severe myocarditis. MED 2021; 2:1072-1092.e7. [PMID: 34414385 PMCID: PMC8363470 DOI: 10.1016/j.medj.2021.08.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/12/2021] [Accepted: 08/09/2021] [Indexed: 12/28/2022]
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in children is generally milder than in adults, but a proportion of cases result in hyperinflammatory conditions often including myocarditis. Methods To better understand these cases, we applied a multiparametric approach to the study of blood cells of 56 children hospitalized with suspicion of SARS-CoV-2 infection. Plasma cytokine and chemokine levels and blood cellular composition were measured, alongside gene expression at the bulk and single-cell levels. Findings The most severe forms of multisystem inflammatory syndrome in children (MIS-C) related to SARS-CoV-2 that resulted in myocarditis were characterized by elevated levels of pro-angiogenesis cytokines and several chemokines. Single-cell transcriptomics analyses identified a unique monocyte/dendritic cell gene signature that correlated with the occurrence of severe myocarditis characterized by sustained nuclear factor κB (NF-κB) activity and tumor necrosis factor alpha (TNF-α) signaling and associated with decreased gene expression of NF-κB inhibitors. We also found a weak response to type I and type II interferons, hyperinflammation, and response to oxidative stress related to increased HIF-1α and Vascular endothelial growth factor (VEGF) signaling. Conclusions These results provide potential for a better understanding of disease pathophysiology. Funding Agence National de la Recherche (Institut Hospitalo-Universitaire Imagine, grant ANR-10-IAHU-01; Recherche Hospitalo-Universitaire, grant ANR-18-RHUS-0010; Laboratoire d’Excellence ‘‘Milieu Intérieur,” grant ANR-10-LABX-69-01; ANR-flash Covid19 “AIROCovid” and “CoVarImm”), Institut National de la Santé et de la Recherche Médicale (INSERM), and the “URGENCE COVID-19” fundraising campaign of Institut Pasteur. Children with SARS-CoV-2 infection were initially thought to have only mild COVID-19 symptoms. However, several weeks into the first wave of SARS-CoV-2 infections, there was a surge of a postacute pathology called multisystem inflammatory syndrome in children (MIS-C). The authors recruited a cohort of children with suspicion of SARS-CoV-2 infection and uncovered hyperinflammation, hypoxic conditions, exacerbation of TNF-α signaling via NF-κB, and absence of responses to type I and type II IFN secretion in the most severe forms of MIS-C with severe myocarditis. This work led the authors to identify in monocytes and validate in peripheral blood mononuclear cells a molecular signature of 25 genes that allows discrimination of the most severe forms of MIS-C with myocarditis.
Collapse
Affiliation(s)
- Camille de Cevins
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Molecular Biology and Genomics, Translational Sciences, Sanofi R&D, Chilly-Mazarin, France
| | - Marine Luka
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Nikaïa Smith
- Translational Immunology Lab, Department of Immunology, Institut Pasteur, 75015 Paris, France
| | - Sonia Meynier
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Aude Magérus
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Francesco Carbone
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Víctor García-Paredes
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Laura Barnabei
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Maxime Batignes
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Alexandre Boullé
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Marie-Claude Stolzenberg
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Brieuc P Pérot
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Bruno Charbit
- Cytometry and Biomarkers UTechS, CRT, Institut Pasteur, 75015, Paris, France
| | - Tinhinane Fali
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Vithura Pirabakaran
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Boris Sorin
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Quentin Riller
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Ghaith Abdessalem
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
| | - Maxime Beretta
- Humoral Immunology Laboratory, Department of Immunology, Institut Pasteur, 75015, Paris, France
- INSERM U1222, Institut Pasteur, 75015, Paris, France
| | - Ludivine Grzelak
- Virus and Immunity Unit, Department of Virology, Institut Pasteur, 75015, Paris, France
| | - Pedro Goncalves
- INSERM U1223, Institut Pasteur, 75015, Paris, France
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, 75015, Paris, France
| | - James P Di Santo
- INSERM U1223, Institut Pasteur, 75015, Paris, France
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, 75015, Paris, France
| | - Hugo Mouquet
- Humoral Immunology Laboratory, Department of Immunology, Institut Pasteur, 75015, Paris, France
- INSERM U1222, Institut Pasteur, 75015, Paris, France
| | - Olivier Schwartz
- Virus and Immunity Unit, Department of Virology, Institut Pasteur, 75015, Paris, France
| | - Mohammed Zarhrate
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UMS3633, Paris Descartes Sorbonne Paris Cite University, Paris, France
| | - Mélanie Parisot
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UMS3633, Paris Descartes Sorbonne Paris Cite University, Paris, France
| | - Christine Bole-Feysot
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UMS3633, Paris Descartes Sorbonne Paris Cite University, Paris, France
| | - Cécile Masson
- Bioinformatics Platform, Structure Fédérative de Recherche Necker, INSERM UMR1163, Université de Paris, Imagine Institute, Paris, France
| | - Nicolas Cagnard
- Bioinformatics Platform, Structure Fédérative de Recherche Necker, INSERM UMR1163, Université de Paris, Imagine Institute, Paris, France
| | - Aurélien Corneau
- Sorbonne Université, UMS037, PASS, Plateforme de Cytométrie de la Pitié-Salpêtrière CyPS, 75013 Paris, France
| | - Camille Brunaud
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Shen-Ying Zhang
- Université de Paris, Imagine Institute, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Jean-Laurent Casanova
- Université de Paris, Imagine Institute, Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, 75015 Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Department of Paediatric Immuno-Haematology and Rheumatology, Reference Center for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), Hôpital Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), 75015 Paris, France
| | - Brigitte Bader-Meunier
- Department of Paediatric Immuno-Haematology and Rheumatology, Reference Center for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), Hôpital Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), 75015 Paris, France
| | - Julien Haroche
- Department of Immunology and Infectious Disease (CIMI-Paris), Pitié-Salpêtrière University Hospital, Sorbonne Université, AP-HP, 75013 Paris, France
| | - Isabelle Melki
- Department of Paediatric Immuno-Haematology and Rheumatology, Reference Center for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), Hôpital Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), 75015 Paris, France
- Department of Pediatrics, Robert-Debré University Hospital, AP-HP, Université de Paris, Paris, France
| | - Mathie Lorrot
- Department of Pediatrics, Armand-Trousseau University Hospital, AP-HP, 75012 Paris, France
| | - Mehdi Oualha
- Pediatric Intensive Care Unit, Necker-Enfants Malades University Hospital, AP-HP, Université de Paris, 75015 Paris, France
| | - Florence Moulin
- Pediatric Intensive Care Unit, Necker-Enfants Malades University Hospital, AP-HP, Université de Paris, 75015 Paris, France
| | | | | | - Marianne Leruez
- Virology Laboratory, Necker-Enfants Malades University Hospital, AP-HP, Université de Paris, 75015 Paris, France
| | - Slimane Allali
- Department of General Paediatrics and Paediatric Infectious Diseases, Necker-Enfants Malades University Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Université de Paris, 75015 Paris, France
| | - Christèle Gras-Leguen
- Pediatric Department, Nantes University Hospital, CIC 1413, INSERM, 44000 Nantes, France
| | - Loïc de Pontual
- Department of Pediatrics, Jean Verdier Hospital, Assistance Publique-Hôpitaux de Paris, Paris 13 University, Bondy, France
| | - Alain Fischer
- Department of Paediatric Immuno-Haematology and Rheumatology, Reference Center for Rheumatic, AutoImmune and Systemic Diseases in Children (RAISE), Hôpital Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris (AP-HP), 75015 Paris, France
- Université de Paris, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
- Collège de France, Paris, France
| | - Darragh Duffy
- Translational Immunology Lab, Department of Immunology, Institut Pasteur, 75015 Paris, France
- Cytometry and Biomarkers UTechS, CRT, Institut Pasteur, 75015, Paris, France
| | - Fredéric Rieux-Laucat
- Université de Paris, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, 75015 Paris, France
| | - Julie Toubiana
- Department of General Paediatrics and Paediatric Infectious Diseases, Necker-Enfants Malades University Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Université de Paris, 75015 Paris, France
- Institut Pasteur, Biodiversity and Epidemiology of Bacterial Pathogens, Paris, France
| | - Mickaël M Ménager
- Université de Paris, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| |
Collapse
|
28
|
A novel miRNA-762/NFIX pathway modulates LPS-induced acute lung injury. Int Immunopharmacol 2021; 100:108066. [PMID: 34492536 DOI: 10.1016/j.intimp.2021.108066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/03/2021] [Accepted: 08/09/2021] [Indexed: 11/23/2022]
Abstract
Severe acute lung injury (ALI) cause significant morbidity and mortality worldwide. MicroRNAs (miRNAs) are possible biomarkers and therapeutic targets for ALI. We aimed to explore the role of miR-762, a known oncogenic factor, in the pathogenesis of ALI. Levels of miR-762 in lung tissues of LPS-treated ALI mice and blood cells of patients with lung injury were measured. Injury of human lung epithelial cell line A549 was induced by LPS stimulation. A downstream target of miR-762, NFIX, was predicted using online tools. Their interactions were validated by luciferase reporter assay. Effects of targeted regulation of the miR-762/NFIX axis on cell proliferation, apoptosis, and inflammatory responses were tested in vitro in A549 cells in vivo with an ALI mouse model. We found that upregulation of miR-762 expression and downregulation of NFIX expression were associated with lung injury. Either miR-762 inhibition or NFIX overexpression in A549 lung cells significantly attenuated LPS-mediated impairment of cell proliferation and viability. Notably, increasing expressions of miR-762 inhibitor or NFIX in vivo via airway lentivirus infection alleviated the LPS-induced ALI in mice. Further, targeted downregulation of miR-762 expression or upregulation of NFIX expression in A549 cells markedly down-regulates NF-κB/IRF3 activation, and substantially reduces the production of inflammatory factors, including TNF-α, IL-6, and IL-8. This study reveals a novel role for the miR-762/NFIX pathway in ALI pathogenesis and sheds new light on targeting this pathway for diagnosis, prevention, and therapy.
Collapse
|
29
|
Recent trends of NFκB decoy oligodeoxynucleotide-based nanotherapeutics in lung diseases. J Control Release 2021; 337:629-644. [PMID: 34375688 DOI: 10.1016/j.jconrel.2021.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023]
Abstract
Nuclear factor κB (NFκB) is a unique protein complex that plays a major role in lung inflammation and respiratory dysfunction. The NFκB signaling pathway, therefore becomes an avenue for the development of potential pharmacological interventions, especially in situations where chronic inflammation is often constitutively active and plays a key role in the pathogenesis and progression of the disease. NFκB decoy oligodeoxynucleotides (ODNs) are double-stranded and carry NFκB binding sequences. They prevent the formation of NFκB-mediated inflammatory cytokines and thus have been employed in the treatment of a variety of chronic inflammatory diseases. However, the systemic administration of naked decoy ODNs restricts their therapeutic effectiveness because of their poor pharmacokinetic profile, instability, degradation by cellular enzymes and their low cellular uptake. Both structural modification and nanotechnology have shown promising results in enhancing the pharmacokinetic profiles of potent therapeutic substances and have also shown great potential in the treatment of respiratory diseases such as asthma, chronic obstructive pulmonary disease and cystic fibrosis. In this review, we examine the contribution of NFκB activation in respiratory diseases and recent advancements in the therapeutic use of decoy ODNs. In addition, we also highlight the limitations and challenges in use of decoy ODNs as therapeutic molecules, cellular uptake of decoy ODNs, and the current need for novel delivery systems to provide efficient delivery of decoy ODNs. Furthermore, this review provides a common platform for discussion on the existence of decoy ODNs, as well as outlining perspectives on the latest generation of delivery systems that encapsulate decoy ODNs and target NFκB in respiratory diseases.
Collapse
|
30
|
Rawat S, Vrati S, Banerjee A. Neutrophils at the crossroads of acute viral infections and severity. Mol Aspects Med 2021; 81:100996. [PMID: 34284874 PMCID: PMC8286244 DOI: 10.1016/j.mam.2021.100996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/22/2022]
Abstract
Neutrophils are versatile immune effector cells essential for mounting a first-line defense against invading pathogens. However, uncontrolled activation can lead to severe life-threatening complications. Neutrophils exist as a heterogeneous population, and their interaction with pathogens and other immune cells may shape the outcome of the host immune response. Diverse classes of viruses, including the recently identified novel SARS-CoV-2, have shown to alter the various aspects of neutrophil biology, offering possibilities for selective intervention. Here, we review heterogeneity within the neutrophil population, highlighting the functional consequences of circulating phenotypes and their critical involvement in exaggerating protective and pathological immune responses against the viruses. We discuss the recent findings of neutrophil extracellular traps (NETs) in COVID-19 pathology and cover other viruses, where neutrophil biology and NETs are crucial for developing disease severity. In the end, we have also pointed out the areas where neutrophil-mediated responses can be finely tuned to outline opportunities for therapeutic manipulation in controlling inflammation against viral infection.
Collapse
Affiliation(s)
- Surender Rawat
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Sudhanshu Vrati
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Arup Banerjee
- Regional Centre for Biotechnology, Faridabad, Haryana, India.
| |
Collapse
|
31
|
Zhang M, Bai X, Cao W, Ji J, Wang L, Yang Y, Yang H. The Influence of Corticosteroids, Immunosuppressants and Biologics on Patients With Inflammatory Bowel Diseases, Psoriasis and Rheumatic Diseases in the Era of COVID-19: A Review of Current Evidence. Front Immunol 2021; 12:677957. [PMID: 34335579 PMCID: PMC8317986 DOI: 10.3389/fimmu.2021.677957] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/14/2021] [Indexed: 12/15/2022] Open
Abstract
Patients with inflammatory bowel disease, psoriasis or other rheumatic diseases treated with corticosteroids, immunomodulators and biologics might face additional risk during COVID-19 epidemic due to their immunocompromised status. However, there was still no unanimous opinion on the use of these therapy during COVID-19 epidemic. Current studies suggested that systemic corticosteroids might increase the risk of hospitalization, as well as risks of ventilation, ICU, and death among patients with immune-mediated inflammatory diseases. Anti-TNF agent was associated with lower rate of hospitalization, as well as lower risks of ventilation, ICU, and death. No significant changes in rates of hospitalization, ventilation, ICU and mortality were observed in patients treated with immunomodulators or biologics apart from anti-TNF agents. The underlying mechanism of these results might be related to pathway of antiviral immune response and cytokine storm induced by SARS-COV-2 infection. Decision on the use of corticosteroids, immunomodulators and biologics should be made after weighing the benefits and potential risks based on individual patients.
Collapse
Affiliation(s)
- Mengyuan Zhang
- School of Medicine, Peking Union Medical College (PUMC), PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyin Bai
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Wei Cao
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Junyi Ji
- School of Medicine, Tsinghua University, Beijing, China
| | - Luo Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Yang Yang
- Department of Pharmacy, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Hong Yang
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| |
Collapse
|
32
|
Proboszcz M, Goryca K, Nejman-Gryz P, Przybyłowski T, Górska K, Krenke R, Paplińska-Goryca M. Phenotypic Variations of Mild-to-Moderate Obstructive Pulmonary Diseases According to Airway Inflammation and Clinical Features. J Inflamm Res 2021; 14:2793-2806. [PMID: 34234506 PMCID: PMC8254142 DOI: 10.2147/jir.s309844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 05/20/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose Asthma and chronic obstructive pulmonary disease (COPD) are complex and heterogeneous inflammatory diseases. We sought to investigate distinct disease profiles based on clinical, cellular and molecular data from patients with mild-to-moderate obstructive pulmonary diseases. Patients and Methods Patients with mild-to-moderate allergic asthma (n=30) and COPD (n=30) were prospectively recruited. Clinical characteristics and induced sputum were collected. In total, 35 mediators were assessed in induced sputum. Logistic regression analysis was conducted to identify the optimal factors that were able to discriminate between asthma and COPD. Further, the data were explored using hierarchical clustering in order to discover and compare clusters of combined samples of asthma and COPD patients. Clinical parameters, cellular composition, and sputum mediators of asthma and COPD were assessed between and within obtained clusters. Results We found five clinical and biochemical variables, namely IL-6, IL-8, CCL4, FEV1/VC ratio pre-bronchodilator (%), and sputum neutrophils (%) that differentiated asthma and COPD and were suitable for discrimination purposes. A combination of those variables yielded high sensitivity and specificity in the differentiation between asthma and COPD, although only FEV1/VC ratio pre-bronchodilator (%) proven significant in the combined model. In cluster analysis, two main clusters were identified: cluster 1, asthma predominant with evidence of eosinophilic airway inflammation and low level of Th1 and Th2 cytokines; and cluster 2, COPD predominant with elevated levels of Th1 and Th2 mediators. Conclusion The inflammatory profile of sputum samples from patients with stable mild-to-moderate asthma and COPD is not disease specific, varies within the disease and might be similar between these diseases. This study highlights the need for phenotyping the mild-to-moderate stages according to their clinical and molecular features.
Collapse
Affiliation(s)
- Małgorzata Proboszcz
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof Goryca
- Genomics Core Facility, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Patrycja Nejman-Gryz
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Warsaw, Poland
| | - Tadeusz Przybyłowski
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Górska
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Warsaw, Poland
| | - Rafał Krenke
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Paplińska-Goryca
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
33
|
Takiguchi H, Yang CX, Yang CWT, Sahin B, Whalen BA, Milne S, Akata K, Yamasaki K, Yang JSW, Cheung CY, Vander Werff R, McNagny KM, Leitao Filho FS, Shaipanich T, van Eeden SF, Obeidat M, Leung JM, Sin DD. Macrophages with reduced expressions of classical M1 and M2 surface markers in human bronchoalveolar lavage fluid exhibit pro-inflammatory gene signatures. Sci Rep 2021; 11:8282. [PMID: 33859282 PMCID: PMC8050093 DOI: 10.1038/s41598-021-87720-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 04/01/2021] [Indexed: 02/01/2023] Open
Abstract
The classical M1/M2 polarity of macrophages may not be applicable to inflammatory lung diseases including chronic obstructive pulmonary disease (COPD) due to the complex microenvironment in lungs and the plasticity of macrophages. We examined macrophage sub-phenotypes in bronchoalveolar lavage (BAL) fluid in 25 participants with CD40 (a M1 marker) and CD163 (a M2 marker). Of these, we performed RNA-sequencing on each subtype in 10 patients using the Illumina NextSeq 500. Approximately 25% of the macrophages did not harbor classical M1 or M2 surface markers (double negative, DN), and these cells were significantly enriched in COPD patients compared with non-COPD patients (46.7% vs. 14.5%, p < 0.001). 1886 genes were differentially expressed in the DN subtype compared with all other subtypes at a 10% false discovery rate. The 602 up-regulated genes included 15 mitochondrial genes and were enriched in 86 gene ontology (GO) biological processes including inflammatory responses. Modules associated with cellular functions including oxidative phosphorylation were significantly down-regulated in the DN subtype. Macrophages in the human BAL fluid, which were negative for both M1/M2 surface markers, harbored a gene signature that was pro-inflammatory and suggested dysfunction in cellular homeostasis. These macrophages may contribute to the pathogenesis and manifestations of inflammatory lung diseases such as COPD.
Collapse
Affiliation(s)
- Hiroto Takiguchi
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Chen X Yang
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada
| | - Cheng Wei Tony Yang
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada
| | - Basak Sahin
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada
| | - Beth A Whalen
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada
| | - Stephen Milne
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada
- Division of Respiratory Medicine, UBC Department of Medicine, Vancouver, BC, Canada
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Kentaro Akata
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada
| | - Kei Yamasaki
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada
| | - Julia Shun Wei Yang
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada
| | - Chung Yan Cheung
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada
| | - Ryan Vander Werff
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Kelly M McNagny
- The Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Fernando Sergio Leitao Filho
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada
| | - Tawimas Shaipanich
- Division of Respiratory Medicine, UBC Department of Medicine, Vancouver, BC, Canada
| | - Stephan F van Eeden
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada
- Division of Respiratory Medicine, UBC Department of Medicine, Vancouver, BC, Canada
| | - Ma'en Obeidat
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada
| | - Janice M Leung
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada
- Division of Respiratory Medicine, UBC Department of Medicine, Vancouver, BC, Canada
| | - Don D Sin
- St Paul's Hospital, The University of British Columbia (UBC) Centre for Heart Lung Innovation (HLI), Vancouver, BC, Canada.
- Division of Respiratory Medicine, UBC Department of Medicine, Vancouver, BC, Canada.
| |
Collapse
|
34
|
de Loyola MB, dos Reis TTA, de Oliveira GXLM, da Fonseca Palmeira J, Argañaraz GA, Argañaraz ER. Alpha-1-antitrypsin: A possible host protective factor against Covid-19. Rev Med Virol 2021; 31:e2157. [PMID: 32844538 PMCID: PMC7461031 DOI: 10.1002/rmv.2157] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 12/20/2022]
Abstract
Understanding Covid-19 pathophysiology is crucial for a better understanding of the disease and development of more effective treatments. Alpha-1-antitrypsin (A1AT) is a constitutive tissue protector with antiviral and anti-inflammatory properties. A1AT inhibits SARS-CoV-2 infection and two of the most important proteases in the pathophysiology of Covid-19: the transmembrane serine protease 2 (TMPRSS2) and the disintegrin and metalloproteinase 17 (ADAM17). It also inhibits the activity of inflammatory molecules, such as IL-8, TNF-α, and neutrophil elastase (NE). TMPRSS2 is essential for SARS-CoV-2-S protein priming and viral infection. ADAM17 mediates ACE2, IL-6R, and TNF-α shedding. ACE2 is the SARS-CoV-2 entry receptor and a key component for the balance of the renin-angiotensin system, inflammation, vascular permeability, and pulmonary homeostasis. In addition, clinical findings indicate that A1AT levels might be important in defining Covid-19 outcomes, potentially partially explaining associations with air pollution and with diabetes. In this review, we focused on the interplay between A1AT with TMPRSS2, ADAM17 and immune molecules, and the role of A1AT in the pathophysiology of Covid-19, opening new avenues for investigating effective treatments.
Collapse
Affiliation(s)
| | | | | | - Julys da Fonseca Palmeira
- Laboratory of Molecular Neurovirology, Faculty of Health ScienceUniversity of BrasíliaBrasiliaBrazil
| | - Gustavo A. Argañaraz
- Laboratory of Molecular Neurovirology, Faculty of Health ScienceUniversity of BrasíliaBrasiliaBrazil
| | - Enrique R. Argañaraz
- Laboratory of Molecular Neurovirology, Faculty of Health ScienceUniversity of BrasíliaBrasiliaBrazil
| |
Collapse
|
35
|
Chung NPY, Khan KMF, Kaner RJ, O'Beirne SL, Crystal RG. HIV induces airway basal progenitor cells to adopt an inflammatory phenotype. Sci Rep 2021; 11:3988. [PMID: 33597552 PMCID: PMC7889866 DOI: 10.1038/s41598-021-82143-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
Despite the introduction of anti-retroviral therapy, chronic HIV infection is associated with an increased incidence of other comorbidities such as COPD. Based on the knowledge that binding of HIV to human airway basal stem/progenitor cells (BC) induces a destructive phenotype by increased MMP-9 expression through MAPK signaling pathways, we hypothesized that HIV induces the BC to express inflammatory mediators that contribute to the pathogenesis of emphysema. Our data demonstrate that airway BC isolated from HAART-treated HIV+ nonsmokers spontaneously release inflammatory mediators IL-8, IL-1β, ICAM-1 and GM-CSF. Similarly, exposure of normal BC to HIV in vitro up-regulates expression of the same inflammatory mediators. These HIV-BC derived mediators induce migration of alveolar macrophages (AM) and neutrophils and stimulate AM proliferation. This HIV-induced inflammatory phenotype likely contributes to lung inflammation in HIV+ individuals and provides explanation for the increased incidence of COPD in HIV+ individuals.
Collapse
Affiliation(s)
- Nancy P Y Chung
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
| | - K M Faisal Khan
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
| | - Robert J Kaner
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Sarah L O'Beirne
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, 1300 York Avenue, Box 164, New York, NY, 10065, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
36
|
Kessie DK, Lodes N, Oberwinkler H, Goldman WE, Walles T, Steinke M, Gross R. Activity of Tracheal Cytotoxin of Bordetella pertussis in a Human Tracheobronchial 3D Tissue Model. Front Cell Infect Microbiol 2021; 10:614994. [PMID: 33585281 PMCID: PMC7873972 DOI: 10.3389/fcimb.2020.614994] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/02/2020] [Indexed: 11/13/2022] Open
Abstract
Bordetella pertussis is a highly contagious pathogen which causes whooping cough in humans. A major pathophysiology of infection is the extrusion of ciliated cells and subsequent disruption of the respiratory mucosa. Tracheal cytotoxin (TCT) is the only virulence factor produced by B. pertussis that has been able to recapitulate this pathology in animal models. This pathophysiology is well characterized in a hamster tracheal model, but human data are lacking due to scarcity of donor material. We assessed the impact of TCT and lipopolysaccharide (LPS) on the functional integrity of the human airway mucosa by using in vitro airway mucosa models developed by co-culturing human tracheobronchial epithelial cells and human tracheobronchial fibroblasts on porcine small intestinal submucosa scaffold under airlift conditions. TCT and LPS either alone and in combination induced blebbing and necrosis of the ciliated epithelia. TCT and LPS induced loss of ciliated epithelial cells and hyper-mucus production which interfered with mucociliary clearance. In addition, the toxins had a disruptive effect on the tight junction organization, significantly reduced transepithelial electrical resistance and increased FITC-Dextran permeability after toxin incubation. In summary, the results indicate that TCT collaborates with LPS to induce the disruption of the human airway mucosa as reported for the hamster tracheal model.
Collapse
Affiliation(s)
- David K. Kessie
- Biocentre, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| | - Nina Lodes
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Heike Oberwinkler
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - William E. Goldman
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Thorsten Walles
- Department of Thoracic Surgery, University of Medicine Magdeburg, Magdeburg, Germany
| | - Maria Steinke
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Roy Gross
- Biocentre, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
37
|
Maxwell AJ, Ding J, You Y, Dong Z, Chehade H, Alvero A, Mor Y, Draghici S, Mor G. Identification of key signaling pathways induced by SARS-CoV2 that underlie thrombosis and vascular injury in COVID-19 patients. J Leukoc Biol 2021; 109:35-47. [PMID: 33242368 PMCID: PMC7753679 DOI: 10.1002/jlb.4covr0920-552rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/05/2020] [Accepted: 11/05/2020] [Indexed: 12/19/2022] Open
Abstract
The SARS-CoV-2 pandemic has led to hundreds of thousands of deaths and billions of dollars in economic damage. The immune response elicited from this virus is poorly understood. An alarming number of cases have arisen where COVID-19 patients develop complications on top of the symptoms already associated with SARS, such as thrombosis, injuries of vascular system, kidney, and liver, as well as Kawasaki disease. In this review, a bioinformatics approach was used to elucidate the immune response triggered by SARS-CoV-2 infection in primary human lung epithelial and transformed human lung alveolar. Additionally, examined the potential mechanism behind several complications that have been associated with COVID-19 and determined that a specific cytokine storm is leading to excessive neutrophil recruitment. These neutrophils are directly leading to thrombosis, organ damage, and complement activation via neutrophil extracellular trap release.
Collapse
Affiliation(s)
- Anthony J Maxwell
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Jiahui Ding
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Yuan You
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Zhong Dong
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Hussein Chehade
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Ayesha Alvero
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Yechiel Mor
- Department of Internal Medicine Wayne State University, Detroit, Michigan, USA
| | - Sorin Draghici
- Department of Computer Science, Wayne State University, Detroit, Michigan, USA
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics, Gynecology, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
38
|
Ronit A, Berg RMG, Bay JT, Haugaard AK, Ahlström MG, Burgdorf KS, Ullum H, Rørvig SB, Tjelle K, Foss NB, Benfield T, Marquart HV, Plovsing RR. Compartmental immunophenotyping in COVID-19 ARDS: A case series. J Allergy Clin Immunol 2021; 147:81-91. [PMID: 32979342 PMCID: PMC7581505 DOI: 10.1016/j.jaci.2020.09.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/27/2020] [Accepted: 09/03/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Severe immunopathology may drive the deleterious manifestations that are observed in the advanced stages of coronavirus disease 2019 (COVID-19) but are poorly understood. OBJECTIVE Our aim was to phenotype leukocyte subpopulations and the cytokine milieu in the lungs and blood of critically ill patients with COVID-19 acute respiratory distress syndrome (ARDS). METHODS We consecutively included patients less than 72 hours after intubation following informed consent from their next of kin. Bronchoalveolar lavage fluid was evaluated by microscopy; bronchoalveolar lavage fluid and blood were assessed by 10-color flow cytometry and a multiplex cytokine panel. RESULTS Four mechanically ventilated patients (aged 40-75 years) with moderate-to-severe COVID-19 ARDS were included. Immature neutrophils dominated in both blood and lungs, whereas CD4 and CD8 T-cell lymphopenia was observed in the 2 compartments. However, regulatory T cells and TH17 cells were found in higher fractions in the lung. Lung CD4 and CD8 T cells and macrophages expressed an even higher upregulation of activation markers than in blood. A wide range of cytokines were expressed at high levels both in the blood and in the lungs, most notably, IL-1RA, IL-6, IL-8, IP-10, and monocyte chemoattactant protein-1, consistent with hyperinflammation. CONCLUSION COVID-19 ARDS exhibits a distinct immunologic profile in the lungs, with a depleted and exhausted CD4 and CD8 T-cell population that resides within a heavily hyperinflammatory milieu.
Collapse
Affiliation(s)
- Andreas Ronit
- Department of Infectious Diseases, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ronan M G Berg
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine and PET and Centre for Physical Activity Research, University of Copenhagen, Copenhagen, Denmark; Neurovascular Research Laboratory, Faculty of Life Sciences and Education, University of South Wales, Pontypridd, United Kingdom
| | - Jakob T Bay
- Department of Clinical Immunology, University of Copenhagen, Copenhagen, Denmark
| | - Anna K Haugaard
- Department of Clinical Immunology, University of Copenhagen, Copenhagen, Denmark
| | - Magnus G Ahlström
- Department of Clinical Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | - Henrik Ullum
- Department of Clinical Immunology, University of Copenhagen, Copenhagen, Denmark
| | - Sara B Rørvig
- Department of Pathology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Tjelle
- Department of Anesthesiology and Intensive Care, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai B Foss
- Department of Anesthesiology and Intensive Care, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Benfield
- Department of Infectious Diseases, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | | | - Ronni R Plovsing
- Department of Anesthesiology and Intensive Care, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
39
|
Ween MP, Moshensky A, Thredgold L, Bastian NA, Hamon R, Badiei A, Nguyen PT, Herewane K, Jersmann H, Bojanowski CM, Shin J, Reynolds PN, Crotty Alexander LE, Hodge SJ. E-cigarettes and health risks: more to the flavor than just the name. Am J Physiol Lung Cell Mol Physiol 2020; 320:L600-L614. [PMID: 33295836 DOI: 10.1152/ajplung.00370.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The growing interest in regulating flavored E-liquids must incorporate understanding of the "flavoring profile" of each E-liquid-which flavorings (flavoring chemicals) are present and at what concentrations not just focusing on the flavor on the label. We investigated the flavoring profile of 10 different flavored E-liquids. We assessed bronchial epithelial cell viability and apoptosis, phagocytosis of bacteria and apoptotic cells by macrophages after exposure to E-cigarette vapor extract (EVE). We validated our data in normal human bronchial epithelial cells (NHBE) and alveolar macrophages (AM) from healthy donors. We also assessed cytokine release and validated in the saliva from E-cigarette users. Increased necrosis/apoptosis (16.1-64.5% apoptosis) in 16HBE cells was flavor dependent, and NHBEs showed an increased susceptibility to flavors. In THP-1 differentiated macrophages phagocytosis was also flavor dependent, with AM also showing increased susceptibility to flavors. Further, Banana and Chocolate were shown to reduce surface expression of phagocytic target recognition receptors on alveolar macrophages. Banana and Chocolate increased IL-8 secretion by NHBE, whereas all 4 flavors reduced AM IL-1β secretion, which was also reduced in the saliva of E-cigarette users compared with healthy controls. Flavorant profiles of E-liquids varied from simple 2 compound mixtures to complex mixtures containing over a dozen flavorants. E-liquids with high benzene content, complex flavoring profiles, high chemical concentration had the greatest impacts. The Flavorant profile of E-liquids is key to disruption of the airway status quo by increasing bronchial epithelial cell apoptosis, causing alveolar macrophage phagocytic dysfunction, and altering airway cytokines.
Collapse
Affiliation(s)
- M P Ween
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - A Moshensky
- Pulmonary Critical Care Section, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, California
| | - L Thredgold
- Department of Occupational and Environmental Health, School of Public Health, University of Adelaide, Adelaide, South Australia, Australia
| | - N A Bastian
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - R Hamon
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, South Australia, Australia
| | - A Badiei
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - P T Nguyen
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - K Herewane
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - H Jersmann
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - C M Bojanowski
- Pulmonary Critical Care Section, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, California
| | - J Shin
- Pulmonary Critical Care Section, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, California
| | - P N Reynolds
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia.,School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - L E Crotty Alexander
- Pulmonary Critical Care Section, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, California
| | - S J Hodge
- School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
40
|
Dai H, Wei G, Wang Y, Ma N, Chang G, Shen X. Sodium butyrate promotes lipopolysaccharide-induced innate immune responses by enhancing mitogen-activated protein kinase activation and histone acetylation in bovine mammary epithelial cells. J Dairy Sci 2020; 103:11636-11652. [PMID: 33010913 DOI: 10.3168/jds.2020-18198] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 07/22/2020] [Indexed: 12/29/2022]
Abstract
The innate immune response plays a crucial role in recovery from infectious diseases by promoting the clearance of pathogens. Sodium butyrate (NaB) is an energy source for cellular processes with the potential to regulate the innate immune response. The present study aimed to evaluate the effect of NaB on the innate immune response in a bovine mammary alveolar cell line (MAC-T) initiated by lipopolysaccharides (LPS). Thus, treatments were conducted as follows: treated with 1× PBS for 24 h (control), pretreated with 1 mM NaB (optimized by cell viability assays and dose-dependent experiment) for 18 h followed by treatment of 1× PBS for 6 h (NaB), pretreated with 1× PBS for 18 h followed by stimulation with LPS (1 µg/mL) for 6 h (LPS), and pretreated with 1 mM NaB for 18 h followed by stimulation with LPS (1 µg/mL) for 6 h (NaB + LPS). Different inhibitors were also used to elucidate the underlying mechanism. Furthermore, cells were treated with NaB and heat-inactivated Escherichia coli to test the effect of NaB on transcription of genes related to the innate immune response triggered by the major causative pathogen of mastitis. Each treatment had 3 replicates and was repeated 3 times. Proinflammatory cytokines, chemokines, and β-defensins are crucial secretion factors in innate immunity, and transcription of these factors was increased by NaB during challenge with LPS or heat-inactivated E. coli in MAC-T cells. Acetylation of histone H3 protein, which promotes gene expression by affecting the structure of chromatin, was also upregulated by NaB in response to LPS stimulation. P38 mitogen-activated protein kinases (MAPK), JNK, and Erk 1 and 2 are key upstream regulators of the expression of proinflammatory cytokines, chemokines, and β-defensins, and their activity was enhanced by NaB during LPS stimulation. Furthermore, inhibitors were used to assess the role of MAPK signaling in the effects of NaB. The results showed that inhibitors of p38 MAPK, Erk, and JNK attenuated the NaB-induced upregulation of TNF and β-defensin 5 (DEFB5) transcription, and that the inhibitor of Erk attenuated the NaB-induced upregulation of IL1B transcription during LPS challenge. Enhanced transcription of CXCL8 by NaB was blocked by the inhibitor of Erk and p38 MAPK during LPS stimulation. Overall, NaB boosted the LPS-induced innate immune response by promoting the expression of proinflammatory cytokines, chemokines, and β-defensins, which was associated with enhanced MAPK signaling activation and histone H3 acetylation.
Collapse
Affiliation(s)
- Hongyu Dai
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - Guozhen Wei
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - Yan Wang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - Nana Ma
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - Guangjun Chang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China
| | - Xiangzhen Shen
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, P. R. China.
| |
Collapse
|
41
|
Leija-Martínez JJ, Huang F, Del-Río-Navarro BE, Sanchéz-Muñoz F, Muñoz-Hernández O, Giacoman-Martínez A, Hall-Mondragon MS, Espinosa-Velazquez D. IL-17A and TNF-α as potential biomarkers for acute respiratory distress syndrome and mortality in patients with obesity and COVID-19. Med Hypotheses 2020; 144:109935. [PMID: 32795834 PMCID: PMC7413092 DOI: 10.1016/j.mehy.2020.109935] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/29/2020] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19) was declared a pandemic and international health emergency by the World Health Organization. Patients with obesity with COVID-19 are 7 times more likely to need invasive mechanical ventilation than are patients without obesity (OR 7.36; 95% CI: 1.63–33.14, p = 0.021). Acute respiratory distress syndrome (ARDS) is one of the main causes of death related to COVID-19 and is triggered by a cytokine storm that damages the respiratory epithelium. Interleukins that cause the chronic low-grade inflammatory state of obesity, such as interleukin (IL)-1β, IL-6, monocyte chemoattractant peptide (MCP)-1, and, in particular, IL-17A and tumour necrosis factor alpha (TNF-α), also play very important roles in lung damage in ARDS. Therefore, obesity is associated with an immune state favourable to a cytokine storm. Our hypothesis is that serum concentrations of TNF-α and IL-17A are more elevated in patients with obesity and COVID-19, and consequently, they have a greater probability of developing ARDS and death. The immunobiology of IL-17A and TNF-α opens a new fascinating field of research for COVID-19.
Collapse
Affiliation(s)
- José J Leija-Martínez
- Universidad Nacional Autónoma de México, Programa de Maestría y Doctorado en Ciencias Médicas, Odontológicas y de la Salud, Mexico City, Mexico; Hospital Infantil de Mexico Federico Gómez, Research Laboratory of Pharmacology, Mexico City, Mexico
| | - Fengyang Huang
- Universidad Nacional Autónoma de México, Programa de Maestría y Doctorado en Ciencias Médicas, Odontológicas y de la Salud, Mexico City, Mexico; Hospital Infantil de Mexico Federico Gómez, Research Laboratory of Pharmacology, Mexico City, Mexico.
| | - Blanca E Del-Río-Navarro
- Universidad Nacional Autónoma de México, Programa de Maestría y Doctorado en Ciencias Médicas, Odontológicas y de la Salud, Mexico City, Mexico; Hospital Infantil de México Federico Gómez, Department of Pediatric Allergy Clinical Immunology, Mexico City, Mexico
| | - Fausto Sanchéz-Muñoz
- Universidad Nacional Autónoma de México, Programa de Maestría y Doctorado en Ciencias Médicas, Odontológicas y de la Salud, Mexico City, Mexico; Departamento de Inmunología, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City, Mexico
| | - Onofre Muñoz-Hernández
- Universidad Nacional Autónoma de México, Programa de Maestría y Doctorado en Ciencias Médicas, Odontológicas y de la Salud, Mexico City, Mexico
| | - Abraham Giacoman-Martínez
- Hospital Infantil de Mexico Federico Gómez, Research Laboratory of Pharmacology, Mexico City, Mexico
| | - Margareth S Hall-Mondragon
- Hospital Infantil de México Federico Gómez, Department of Pediatric Allergy Clinical Immunology, Mexico City, Mexico
| | - Dario Espinosa-Velazquez
- Hospital Infantil de México Federico Gómez, Department of Pediatric Allergy Clinical Immunology, Mexico City, Mexico
| |
Collapse
|
42
|
Hwang HJ, Lee YR, Kang D, Lee HC, Seo HR, Ryu JK, Kim YN, Ko YG, Park HJ, Lee JS. Endothelial cells under therapy-induced senescence secrete CXCL11, which increases aggressiveness of breast cancer cells. Cancer Lett 2020; 490:100-110. [PMID: 32659248 DOI: 10.1016/j.canlet.2020.06.019] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022]
Abstract
The effects of senescence associated secretory phenotype (SASP) from therapy-induced senescent endothelial cells on tumor microenvironment (TME) remains to be clarified. Here, we investigated effects of ionizing radiation (IR)- and doxorubicin-induced senescent HUVEC on TME. MDA-MB-231 cancer cells treated with conditioned medium (CM) from senescent HUVEC or co-cultured with senescent HUVEC significantly increased cancer cell proliferation, migration, and invasion. We found that CXCL11 plays a principal role in the senescent CM-induced aggressive activities of MDA-MB-231 cells. When we treated HUVEC with a neutralizing anti-CXCL11 antibody or CXCL11 SiRNA, or treated MDA-MB-231 cells with CXCR3 SiRNA, we observed synergistic diminution of the ability of the HUVEC SASP to alter the migration and spheroid invasion of cancer cells. ERK activation was involved in the HUVEC SASP-induced aggressive activity of MDA-MB-231 cells. Finally, we observed the in vivo effect of CXCL11 from the senescent HUVEC in tumor-bearing mice. Together, our results demonstrate that SASP from endothelial cells experiencing therapy-induced senescence promotes the aggressive behavior of cancer cells, and that CXCL11 can potentially be targeted to prevent the adverse effects of therapy-induced senescent endothelial cells on the tumor microenvironment.
Collapse
Affiliation(s)
- Hyun Jung Hwang
- Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, South Korea; Department of Molecular Medicine, Inha University College of Medicine, Incheon, South Korea
| | - Ye-Rim Lee
- Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, South Korea; Department of Molecular Medicine, Inha University College of Medicine, Incheon, South Korea
| | - Donghee Kang
- Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, South Korea; Department of Molecular Medicine, Inha University College of Medicine, Incheon, South Korea
| | - Hyung Chul Lee
- Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, South Korea; Department of Molecular Medicine, Inha University College of Medicine, Incheon, South Korea
| | - Haeng Ran Seo
- Cancer Biology Research Laboratory, Institute Pasteur Korea, Gyeonggi-do, South Korea
| | - Ji-Kan Ryu
- Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, South Korea; Department of Urology, Inha University College of Medicine, Incheon, South Korea
| | - Yong-Nyun Kim
- Division of Translational Research, Research Institute, National Cancer Center, Goyang, 10408, South Korea
| | - Young-Gyu Ko
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Heon Joo Park
- Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, South Korea; Department of Microbiology, Inha University College of Medicine, Incheon, South Korea
| | - Jae-Seon Lee
- Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, South Korea; Department of Molecular Medicine, Inha University College of Medicine, Incheon, South Korea.
| |
Collapse
|
43
|
Fu M, Su H, Su Z, Yin Z, Jin J, Wang L, Zhang Q, Xu X. Transcriptome analysis of Corynebacterium pseudotuberculosis-infected spleen of dairy goats. Microb Pathog 2020; 147:104370. [PMID: 32653437 DOI: 10.1016/j.micpath.2020.104370] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
Caseous lymphadenitis is a chronic disease of goats caused by Corynebacterium pseudotuberculosis (C.pseudotuberculosis) which causes great harm to the dairy goats industry. In order to obtain detailed information about the pathogenesis and host immune response in C.pseudotuberculosis-infected goats, in this study, the gene expression difference of spleen tissue after infection with C.pseudotuberculosis was analyzed by high-throughput sequencing. Transcripts obtained over 412 700 462 clean reads after reassembly were 21 343 genes detected, of which 14 720 were known genes and 7623 new genes were predicted. There were 448 up-regulated and 519 down-regulated differentially expressed genes (DEGs). Gene Ontology (GO) analysis indicated that all of the DEGs were annotated into biological process, cellular component and molecular function. Most of these unigenes are annotated in cellular processes, the cell and binding. KEGG analysis of the DEGs showed that a total of 8733 DEGs unigenes were annotated into 459 pathways classified into 6 main categories. Most of these annotated unigenes were related to immune system response to the infectious diseases pathways. In addition, 14 DEGs were verified by quantitative real-time PCR. As the first, in vivo, RNAseq analysis of dairy goats and C.pseudotuberculosis infection, this study provides knowledge about the transcriptomics of spleen in C.pseudotuberculosis-infected goats, from which a complex molecular pathways and immune response mechanism are involved in C.pseudotuberculosis infection.
Collapse
Affiliation(s)
- Mingzhe Fu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Hong Su
- College of Animal Medicine, Xinjiang Agricultural University, Urumqi, 830000, China
| | - Zhanqiang Su
- College of Animal Medicine, Xinjiang Agricultural University, Urumqi, 830000, China
| | - Zheng Yin
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jian Jin
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Lixiang Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Qi Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Xingang Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
44
|
Liu Y, Davis AS, Ma L, Bishop L, Cissé OH, Kutty G, Kovacs JA. MUC1 mediates Pneumocystis murina binding to airway epithelial cells. Cell Microbiol 2020; 22:e13182. [PMID: 32017380 DOI: 10.1111/cmi.13182] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/05/2019] [Accepted: 01/27/2020] [Indexed: 02/06/2023]
Abstract
Previous studies have shown that Pneumocystis binds to pneumocytes, but the proteins responsible for binding have not been well defined. Mucins are the major glycoproteins present in mucus, which serves as the first line of defence during airway infection. MUC1 is the best characterised membrane-tethered mucin and is expressed on the surface of most airway epithelial cells. Although by electron microscopy Pneumocystis primarily binds to type I pneumocytes, it can also bind to type II pneumocytes. We hypothesized that Pneumocystis organisms can bind to MUC1 expressed by type II pneumocytes. Overexpression of MUC1 in human embryonic kidney HEK293 cells increased Pneumocystis binding, while knockdown of MUC1 expression by siRNA in A549 cells, a human adenocarcinoma-derived alveolar type II epithelial cell line, decreased Pneumocystis binding. Immunofluorescence labelling indicated that MUC1 and Pneumocystis were co-localised in infected mouse lung tissue. Incubation of A549 cells with Pneumocystis led to phosphorylation of ERK1/2 that increased with knockdown of MUC1 expression by siRNA. Pneumocystis caused increased IL-6 and IL-8 secretion by A549 cells, and knockdown of MUC1 further increased their secretion in A549 cells. Taken together, these results suggest that binding of Pneumocystis to MUC1 expressed by airway epithelial cells may facilitate establishment of productive infection.
Collapse
Affiliation(s)
- Yueqin Liu
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - A Sally Davis
- Diagnostic Medicine/Pathobiology, Kansas State University College of Veterinary Medicine, Manhattan, Kansas, USA
| | - Liang Ma
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Lisa Bishop
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Ousmane H Cissé
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Geetha Kutty
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Joseph A Kovacs
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
45
|
Sze E, Bhalla A, Nair P. Mechanisms and therapeutic strategies for non-T2 asthma. Allergy 2020; 75:311-325. [PMID: 31309578 DOI: 10.1111/all.13985] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 06/10/2019] [Accepted: 06/26/2019] [Indexed: 12/27/2022]
Abstract
Non-T2 asthma is traditionally defined as asthma without features of T2 asthma. The definition is arbitrary and is generally based on the presence of neutrophils in sputum, or the absence (or normal levels) of eosinophils or other T2 markers in sputum (paucigranulocytic), airway biopsies or in blood. This definition may be imprecise as we gain more knowledge from applying transcriptomics and proteomics to blood and airway samples. The prevalence of non-T2 asthma is also difficult to estimate as most studies are cross-sectional and influenced by concomitant treatment with glucocorticosteroids, and by the presence of recognized or unrecognized airway infections. No specific therapies have shown any clinical benefits in patients with asthma that is associated with a non-T2 inflammatory process. It remains to be seen if such an endotype truly exists and to identify treatments to target that endotype. Meanwhile, identifying intense airway neutrophilia as an indicator of airway infection and airway hyperresponsiveness as an indicator of smooth muscle dysfunction, and treating them appropriately, and not increasing glucocorticosteroids in patients who do not have obvious T2 inflammation, seem reasonable.
Collapse
Affiliation(s)
- Eric Sze
- New Territories West Cluster Tuen Mun Hospital Tuen Mun Hong Kong
- St Joseph's Healthcare & Department of Medicine Firestone Institute for Respiratory Health, McMaster University Hamilton Ontario Canada
| | - Anurag Bhalla
- St Joseph's Healthcare & Department of Medicine Firestone Institute for Respiratory Health, McMaster University Hamilton Ontario Canada
| | - Parameswaran Nair
- St Joseph's Healthcare & Department of Medicine Firestone Institute for Respiratory Health, McMaster University Hamilton Ontario Canada
| |
Collapse
|
46
|
Leija-Martínez JJ, Huang F, Del-Río-Navarro BE, Sanchéz-Muñoz F, Romero-Nava R, Muñoz-Hernandez O, Rodríguez-Cortés O, Hall-Mondragon MS. Decreased methylation profiles in the TNFA gene promoters in type 1 macrophages and in the IL17A and RORC gene promoters in Th17 lymphocytes have a causal association with non-atopic asthma caused by obesity: A hypothesis. Med Hypotheses 2019; 134:109527. [PMID: 31877441 DOI: 10.1016/j.mehy.2019.109527] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/09/2019] [Indexed: 12/18/2022]
Abstract
Obesity is a serious public health problem worldwide and has been associated in epidemiological studies with a unique type of non-atopic asthma, although the causal association of asthma and obesity has certain criteria, such as the strength of association, consistency, specificity, temporality, biological gradient, coherence, analogy and experimentation; nevertheless, the biological plausibility of this association remains uncertain. Various mechanisms have been postulated, such as immunological, hormonal, mechanical, environmental, genetic and epigenetic mechanisms. Our hypothesis favours immunological mechanisms because some cytokines, such as tumour necrosis factor alpha (TNF-α) and interleukin (IL)-17A, are responsible for orchestrating low-grade systemic inflammation associated with obesity; however, these cytokines are regulated by epigenetic mechanisms, such as gene promoter methylation.
Collapse
Affiliation(s)
- José J Leija-Martínez
- Universidad Nacional Autónoma de México, Mexico City, Mexico; Hospital Infantil de Mexico Federico Gómez, Research Laboratory of Pharmacology, Mexico City, Mexico
| | - Fengyang Huang
- Universidad Nacional Autónoma de México, Mexico City, Mexico; Hospital Infantil de Mexico Federico Gómez, Research Laboratory of Pharmacology, Mexico City, Mexico.
| | - Blanca E Del-Río-Navarro
- Universidad Nacional Autónoma de México, Mexico City, Mexico; Hospital Infantil de México Federico Gómez, Department of Pediatric Allergy Clinical Immunology, Mexico City, Mexico
| | - Fausto Sanchéz-Muñoz
- Universidad Nacional Autónoma de México, Mexico City, Mexico; Departamento de Inmunología, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City, Mexico
| | - Rodrigo Romero-Nava
- Hospital Infantil de Mexico Federico Gómez, Research Laboratory of Pharmacology, Mexico City, Mexico; Laboratory of Pharmacology, Department of Health Sciences, Division of Health and Biological Sciences, Metropolitan Autonomous University of Iztapalapa, Mexico City, Mexico
| | | | - Octavio Rodríguez-Cortés
- Laboratorio 103, SEPI, Escuela Superior de Medicina, Instituto Politécnico Nacional, Calle Plan de San Luis y Díaz Mirón S/N, Casco de Santo Tomas, Miguel Hidalgo, 11340 Ciudad de México, Mexico
| | | |
Collapse
|
47
|
Zhang P, Mak JC, Man RY, Leung SW. Flavonoids reduces lipopolysaccharide-induced release of inflammatory mediators in human bronchial epithelial cells: Structure-activity relationship. Eur J Pharmacol 2019; 865:172731. [PMID: 31610186 DOI: 10.1016/j.ejphar.2019.172731] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 12/16/2022]
Abstract
Flavonoids are polyphenolic compounds that are widely present in food and Chinese medicine. The aim of the present study was to identify the flavonoids with anti-inflammatory effects in the airway; and to determine the role of anti-oxidant and cyclic adenosine monophosphate (cAMP) in the anti-inflammatory effect. Human bronchial epithelial BEAS-2B cells were exposed to bacterial endotoxin lipopolysaccharide (LPS) in the absence or presence of different flavonoids, which are categorized according to their chemical structures in seven subclasses [anthocyanidins, chalcones, flavanes, flavanones, flavones, flavonols, isoflavones]. Among the 17 flavonoids tested, only apigenin (flavones), luteolin (flavones), daidzein (isoflavones) and genistein (isoflavones) reduced LPS-induced release of inflammatory cytokines/chemokines interleukin (IL)-6, IL-8 and monocyte chemoattractant protein-1 in BEAS-2B cells. Quercetin caused further increase in LPS-induced IL-6 and IL-8 levels. It alone significantly increased nuclear factor-kappa B (NF-κB) p65 activity and the cellular oxidative stress marker malondialdehyde (MDA) level in BEAS-2B cells. By contrast, apigenin and genistein reduced LPS-induced increases in nuclear NF-κB activity and MDA level. Apigenin and genistein, but not quercetin, increased the cAMP level in BEAS-2B cells, and the cell-permeable cAMP analogue, 8-Br-cAMP, inhibited LPS-induced increase of IL-8 level. These findings suggest that the presence of C5-OH, C7-OH, C2=C3 and C4=O functional groups in the flavonoids is associated with greater anti-inflammatory effect, while that of C3-OH or glycosylation group at the A-ring greatly decreased the anti-inflammatory effect. The anti-inflammatory effect of these flavonoids may be related to their anti-oxidant properties, and partly to their ability in increasing cAMP level.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| | - Judith Cw Mak
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, PR China; Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| | - Ricky Yk Man
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, PR China
| | - Susan Ws Leung
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, PR China.
| |
Collapse
|
48
|
L–lysine ameliorates sepsis-induced acute lung injury in a lipopolysaccharide-induced mouse model. Biomed Pharmacother 2019; 118:109307. [DOI: 10.1016/j.biopha.2019.109307] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 01/19/2023] Open
|
49
|
Assessment of Cytokines, Biochemical Markers of Malnutrition and Frailty Syndrome Patients Considered for Lung Transplantation. Transplant Proc 2019; 51:2009-2013. [DOI: 10.1016/j.transproceed.2019.04.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 04/05/2019] [Indexed: 11/21/2022]
|
50
|
Abstract
Cellular senescence, a process of cell proliferation arrest in response to various stressors, has been considered to be important factor in age-related disease. Identification of senescent cells in tissues is limited and the role of senescent cells is poorly understood. Recently however, several studies showed the characterization of senescent cells in various pathologic conditions and the role of senescent cells in disease progression is becoming important. Senescent cells are growth-arrested cells, however, the senescence associated secretory phenotype (SASP) of senescent cells could modify the tissues’ microenvironment. Here, we discuss the progress and understanding of the role of senescent cells in tissues of pathologic conditions and discuss the development of new therapeutic paradigms, such as senescent cells-targeted therapy.
Collapse
Affiliation(s)
- Young Hwa Kim
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Tae Jun Park
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Korea
| |
Collapse
|