1
|
Mazzio E, Barnes A, Badisa R, Council S, Soliman KFA. Plants against cancer: the immune-boosting herbal microbiome: not of the plant, but in the plant. Basic concepts, introduction, and future resource for vaccine adjuvant discovery. Front Oncol 2023; 13:1180084. [PMID: 37588095 PMCID: PMC10426289 DOI: 10.3389/fonc.2023.1180084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 05/30/2023] [Indexed: 08/18/2023] Open
Abstract
The presence of microorganism communities (MOCs) comprised of bacteria, fungi, archaea, algae, protozoa, viruses, and the like, are ubiquitous in all living tissue, including plant and animal. MOCs play a significant role in establishing innate and acquired immunity, thereby influencing susceptibility and resistance to disease. This understanding has fostered substantial advancements in several fields such as agriculture, food science/safety, and the development of vaccines/adjuvants, which rely on administering inactivated-attenuated MOC pathogens. Historical evidence dating back to the 1800s, including reports by Drs Busch, Coley, and Fehleisen, suggested that acute febrile infection in response to "specific microbes" could trigger spontaneous tumor remission in humans. This discovery led to the purposeful administration of the same attenuated strains, known as "Coley's toxin," marking the onset of the first microbial (pathogen) associated molecular pattern (MAMPs or PAMPs)-based tumor immunotherapy, used clinically for over four decades. Today, these same MAMPS are consumed orally by billions of consumers around the globe, through "specific" mediums (immune boosting "herbal supplements") as carriers of highly concentrated MOCs accrued in roots, barks, hulls, sea algae, and seeds. The American Herbal Products Association (AHPA) mandates microbial reduction in botanical product processing but does not necessitate the removal of dead MAMP laden microbial debris, which we ingest. Moreover, while existing research has focused on the immune-modulating role of plant phytochemicals, the actual immune-boosting properties might instead reside solely in the plant's MOC MAMP laden biomass. This assertion is logical, considering that antigenic immune-provoking epitopes, not phytochemicals, are known to stimulate immune response. This review explores a neglected area of research regarding the immune-boosting effects of the herbal microbiome - a presence which is indirectly corroborated by various peripheral fields of study and poses a fundamental question: Given that food safety focuses on the elimination of harmful pathogens and crop science acknowledges the existence of plant microbiomes, what precisely are the immune effects of ingesting MAMPs of diverse structural composition and concentration, and where are these distributed in our botanicals? We will discuss the topic of concentrated edible MAMPs as acid and thermally stable motifs found in specific herbs and how these would activate cognate pattern recognition receptors (PPRs) in the upper gut-associated lymphoid tissue (GALT), including Peyer's patches and the lamina propria, to boost antibody titers, CD8+ and CD4+ T cells, NK activity, hematopoiesis, and facilitating M2 to M1 macrophage phenotype transition in a similar manner as vaccines. This new knowledge could pave the way for developing bioreactor-grown/heat-inactivated MOC therapies to boost human immunity against infections and improve tumor surveillance.
Collapse
Affiliation(s)
- Elizabeth Mazzio
- Divison of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A & M University, Tallahassee, FL, United States
| | - Andrew Barnes
- Divison of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A & M University, Tallahassee, FL, United States
| | - Ramesh Badisa
- Divison of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A & M University, Tallahassee, FL, United States
| | - Stevie Council
- John Gnabre Science Research Institute, Baltimore, MD, United States
| | - Karam F. A. Soliman
- Divison of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A & M University, Tallahassee, FL, United States
| |
Collapse
|
2
|
The Tobacco Smoke Component, Acrolein, as a Major Culprit in Lung Diseases and Respiratory Cancers: Molecular Mechanisms of Acrolein Cytotoxic Activity. Cells 2023; 12:cells12060879. [PMID: 36980220 PMCID: PMC10047238 DOI: 10.3390/cells12060879] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/05/2023] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
Acrolein, a highly reactive unsaturated aldehyde, is a ubiquitous environmental pollutant that seriously threatens human health and life. Due to its high reactivity, cytotoxicity and genotoxicity, acrolein is involved in the development of several diseases, including multiple sclerosis, neurodegenerative diseases such as Alzheimer’s disease, cardiovascular and respiratory diseases, diabetes mellitus and even the development of cancer. Traditional tobacco smokers and e-cigarette users are particularly exposed to the harmful effects of acrolein. High concentrations of acrolein have been found in both mainstream and side-stream tobacco smoke. Acrolein is considered one of cigarette smoke’s most toxic and harmful components. Chronic exposure to acrolein through cigarette smoke has been linked to the development of asthma, acute lung injury, chronic obstructive pulmonary disease (COPD) and even respiratory cancers. This review addresses the current state of knowledge on the pathological molecular mechanisms of acrolein in the induction, course and development of lung diseases and cancers in smokers.
Collapse
|
3
|
Singh T, Bello B, Jeebhay MF. Characterizing Inflammatory Cell Asthma Associated Phenotypes in Dental Health Workers Using Cytokine Profiling. FRONTIERS IN ALLERGY 2021; 2:747591. [PMID: 35387066 PMCID: PMC8974759 DOI: 10.3389/falgy.2021.747591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/11/2021] [Indexed: 11/24/2022] Open
Abstract
Cytokines elicit a pro-inflammatory response by modifying the airway microenvironment in patients with acute or chronic asthma. The expression pattern of several distinct cytokines could be a useful discriminator in asthma. This study aimed to identify asthma subject groupings based on common inflammatory patterns and to determine the relationship between these identified patterns and asthma-associated clinical indices. A sub-group of 76 dental healthcare workers (HCWs) identified from a larger cross-sectional study of 454 dental HCWs in five dental institutions were evaluated further. A self-administered questionnaire elicited the health and employment history of subjects. Sera were analyzed for atopic status, latex sensitization, and 12 cytokines (IL-1β, 3, 4, 5, 6, 7, 8, 10, 12p70, eotaxin, GM-CSF, TNF-α). Pre and post-bronchodilator spirometry was performed on all HCWs. Data clustering and factor analysis were used to identify inflammatory cluster patterns of cytokines. Associations between the cytokine cluster groupings and relevant asthma-associated clinical indices were determined using multivariate logistic regression. The classification of asthma subtype based on cytokine patterns demonstrated both eosinophilic and neutrophilic inflammatory responses. Four phenotypically distinct subgroups relating to the severity of inflammation (acute or chronic) of the cell types were identified. Cytokine determinants for the neutrophilic subtype included IL-1β, 6, 8, 10, 12p70, and TNF-α whereas for the eosinophilic subtype these included IL-3, 4, 5, 7, eotaxin, and GM-CSF. The multivariate models showed a significant association between work-related chest symptoms and all four inflammatory patterns. However, stronger associations were observed for the acute neutrophilic (OR = 6.00, p < 0.05) compared to acute and chronic eosinophilic responses (OR = 4.30, p < 0.05; OR = 4.93, p < 0.05), respectively. Subjects with airway obstruction were more likely to have a mixed cellular infiltrate. The odds of work-exacerbated asthma were increased in acute or chronic eosinophilia (OR = 7.75 and 8.12; p < 0.05), respectively as well as with acute neutrophilia (OR = 6) sub-type. This study demonstrated that neutrophilic inflammatory cell asthma phenotypes coexist with eosinophilic inflammatory phenotypes suggesting a possible dual pathway for asthma in dental health workers, probably due to mixed exposures to high molecular weight (e.g., latex) and low molecular weight (e.g., acrylates) agents.
Collapse
Affiliation(s)
- Tanusha Singh
- Immunology & Microbiology, National Institute for Occupational Health, National Health Laboratory Service, Johannesburg, South Africa
- Department of Environmental Health, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
- Department of Clinical Microbiology and Infectious Diseases, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- *Correspondence: Tanusha Singh
| | - Braimoh Bello
- Immunology & Microbiology Department, Centre for Statistical Analysis and Research, Johannesburg, South Africa
| | - Mohamed F. Jeebhay
- Division of Occupational Medicine and Centre for Environmental & Occupational Health Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
4
|
Schiffers C, van de Wetering C, Bauer RA, Habibovic A, Hristova M, Dustin CM, Lambrichts S, Vacek PM, Wouters EF, Reynaert NL, van der Vliet A. Downregulation of epithelial DUOX1 in chronic obstructive pulmonary disease. JCI Insight 2021; 6:142189. [PMID: 33301419 PMCID: PMC7934842 DOI: 10.1172/jci.insight.142189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic respiratory disease characterized by small airway remodeling and alveolar emphysema due to environmental stresses such as cigarette smoking (CS). Oxidative stress is commonly implicated in COPD pathology, but recent findings suggest that one oxidant-producing NADPH oxidase homolog, dual oxidase 1 (DUOX1), is downregulated in the airways of patients with COPD. We evaluated lung tissue sections from patients with COPD for small airway epithelial DUOX1 protein expression, in association with measures of lung function and small airway and alveolar remodeling. We also addressed the impact of DUOX1 for lung tissue remodeling in mouse models of COPD. Small airway DUOX1 levels were decreased in advanced COPD and correlated with loss of lung function and markers of emphysema and remodeling. Similarly, DUOX1 downregulation in correlation with extracellular matrix remodeling was observed in a genetic model of COPD, transgenic SPC-TNF-α mice. Finally, development of subepithelial airway fibrosis in mice due to exposure to the CS-component acrolein, or alveolar emphysema induced by administration of elastase, were in both cases exacerbated in Duox1-deficient mice. Collectively, our studies highlight that downregulation of DUOX1 may be a contributing feature of COPD pathogenesis, likely related to impaired DUOX1-mediated innate injury responses involved in epithelial homeostasis.
Collapse
Affiliation(s)
- Caspar Schiffers
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.,Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Cheryl van de Wetering
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA.,Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Robert A Bauer
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Aida Habibovic
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Milena Hristova
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Christopher M Dustin
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Sara Lambrichts
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Pamela M Vacek
- Department of Medical Biostatistics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Emiel Fm Wouters
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands.,Ludwig Boltzman Institute for Lung Health, Vienna, Austria
| | - Niki L Reynaert
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| |
Collapse
|
5
|
Analysis of Acrolein Exposure Induced Pulmonary Response in Seven Inbred Mouse Strains and Human Primary Bronchial Epithelial Cells Cultured at Air-Liquid Interface. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3259723. [PMID: 33110918 PMCID: PMC7582059 DOI: 10.1155/2020/3259723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/07/2020] [Accepted: 09/15/2020] [Indexed: 11/23/2022]
Abstract
Background Acrolein is a major component of environmental pollutants, cigarette smoke, and is also formed by heating cooking oil. We evaluated the interstrain variability of response to subchronic inhalation exposure to acrolein among inbred mouse strains for inflammation, oxidative stress, and tissue injury responses. Furthermore, we studied the response to acrolein vapor in the lung mucosa model using human primary bronchial epithelial cells (PBEC) cultured at an air-liquid interface (ALI) to evaluate the findings of mouse studies. Methods Female 129S1/SvlmJ, A/J, BALB/cByJ, C3H/HeJ, C57BL/6J, DBA/2J, and FVB/NJ mice were exposed to 1 part per million (ppm) acrolein or filtered air for 11 weeks. Total cell counts and protein concentrations were measured in bronchoalveolar lavage (BAL) fluid to assess airway inflammation and membrane integrity. PBEC-ALI models were exposed to acrolein vapor (0.1 and 0.2 ppm) for 30 minutes. Gene expression of proinflammatory, oxidative stress, and tissue injury-repair markers was assessed (cut off: ≥2 folds; p < 0.05) in the lung models. Results Total BAL cell numbers and protein concentrations remained unchanged following acrolein exposure in all mouse strains. BALB/cByJ, C57BL/6J, and 129S1/SvlmJ strains were the most affected with an increased expression of proinflammatory, oxidative stress, and/or tissue injury markers. DBA/2J, C3H/HeJ, A/J, and FVB/NJ were affected to a lesser extent. Both matrix metalloproteinase 9 (Mmp9) and tissue inhibitor of metalloproteinase 1 (Timp1) were upregulated in the strains DBA/2J, C3H/HeJ, and FVB/NJ indicating altered protease/antiprotease balance. Upregulation of lung interleukin- (IL-) 17b transcript in the susceptible strains led us to investigate the IL-17 pathway genes in the PBEC-ALI model. Acrolein exposure resulted in an increased expression of IL-17A, C, and D; IL-1B; IL-22; and RAR-related orphan receptor A in the PBEC-ALI model. Conclusion The interstrain differences in response to subchronic acrolein exposure in mouse suggest a genetic predisposition. Altered expression of IL-17 pathway genes following acrolein exposure in the PBEC-ALI models indicates that it has a central role in chemical irritant toxicity. The findings also indicate that genetically determined differences in IL-17 signaling pathway genes in the different mouse strains may explain their susceptibility to different chemical irritants.
Collapse
|
6
|
Zirak MR, Mehri S, Karimani A, Zeinali M, Hayes AW, Karimi G. Mechanisms behind the atherothrombotic effects of acrolein, a review. Food Chem Toxicol 2019; 129:38-53. [DOI: 10.1016/j.fct.2019.04.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/18/2019] [Accepted: 04/18/2019] [Indexed: 12/31/2022]
|
7
|
Kim BG, Lee PH, Lee SH, Hong J, Jang AS. Claudins, VEGF, Nrf2, Keap1, and Nonspecific Airway Hyper-Reactivity Are Increased in Mice Co-Exposed to Allergen and Acrolein. Chem Res Toxicol 2019; 32:139-145. [PMID: 30608172 DOI: 10.1021/acs.chemrestox.8b00239] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Acrolein, an α/β-unsaturated aldehyde, is volatile at room temperature. It is a respiratory irritant found in environmental tobacco smoke, which can be generated during cooking or endogenously at sites of injury. An acute high concentration of uncontrolled irritant exposure can lead to an asthma-like syndrome known as reactive airways dysfunction syndrome (RADS). However, whether acrolein can induce RADS remains poorly understood. The aim of study is to develop a RADS model of acrolein inhalation in mice and to clarify the mechanism of RADS. Mice were treated with ovalbumin (OVA) and exposed to acrolein (5 ppm/10 min). Airway hyper-responsiveness (AHR) was measured on days 24 and 56, and samples were collected on days 25 and 57. Tight junction protein, antioxidant-associated protein, and vascular endothelial growth factor (VEGF) levels were estimated by Western blotting and immunohistochemical staining. Reactive oxygen species (ROS) was calculated using enzyme linked immunosorbent assays. Acrolein or OVA groups exhibited an increase in airway inflammatory cells and AHR compared to a sham group. These effects were further increased in mice in the OVA + acrolein exposure group than in the OVA exposure group and persisted in the acrolein exposure group for 8 weeks. CLDNs, carbonyls, VEGF, Nrf2, and Keap1 were observed in the lungs. Our data demonstrate that acrolein induces RADS and that ROS, angiogenesis, and tight junction proteins are involved in RADS in a mouse model.
Collapse
Affiliation(s)
- Byeong-Gon Kim
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine , Soonchunhyang University Bucheon Hospital , Bucheon , Gyeonggi-do Republic of Korea 14584
| | - Pureun-Haneul Lee
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine , Soonchunhyang University Bucheon Hospital , Bucheon , Gyeonggi-do Republic of Korea 14584
| | - Sun-Hye Lee
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine , Soonchunhyang University Bucheon Hospital , Bucheon , Gyeonggi-do Republic of Korea 14584
| | - Jisu Hong
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine , Soonchunhyang University Bucheon Hospital , Bucheon , Gyeonggi-do Republic of Korea 14584
| | - An-Soo Jang
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine , Soonchunhyang University Bucheon Hospital , Bucheon , Gyeonggi-do Republic of Korea 14584
| |
Collapse
|
8
|
Lu Q, Mundy M, Chambers E, Lange T, Newton J, Borgas D, Yao H, Choudhary G, Basak R, Oldham M, Rounds S. Alda-1 Protects Against Acrolein-Induced Acute Lung Injury and Endothelial Barrier Dysfunction. Am J Respir Cell Mol Biol 2017; 57:662-673. [PMID: 28763253 DOI: 10.1165/rcmb.2016-0342oc] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Inhalation of acrolein, a highly reactive aldehyde, causes lung edema. The underlying mechanism is poorly understood and there is no effective treatment. In this study, we demonstrated that acrolein not only dose-dependently induced lung edema but also promoted LPS-induced acute lung injury. Importantly, acrolein-induced lung injury was prevented and rescued by Alda-1, an activator of mitochondrial aldehyde dehydrogenase 2. Acrolein also dose-dependently increased monolayer permeability, disrupted adherens junctions and focal adhesion complexes, and caused intercellular gap formation in primary cultured lung microvascular endothelial cells (LMVECs). These effects were attenuated by Alda-1 and the antioxidant N-acetylcysteine, but not by the NADPH inhibitor apocynin. Furthermore, acrolein inhibited AMP-activated protein kinase (AMPK) and increased mitochondrial reactive oxygen species levels in LMVECs-effects that were associated with impaired mitochondrial respiration. AMPK total protein levels were also reduced in lung tissue of mice and LMVECs exposed to acrolein. Activation of AMPK with 5-aminoimidazole-4-carboxamide-1-β-4-ribofuranoside blunted an acrolein-induced increase in endothelial monolayer permeability, but not mitochondrial oxidative stress or inhibition of mitochondrial respiration. Our results suggest that acrolein-induced mitochondrial dysfunction may not contribute to endothelial barrier dysfunction. We speculate that detoxification of acrolein by Alda-1 and activation of AMPK may be novel approaches to prevent and treat acrolein-associated acute lung injury, which may occur after smoke inhalation.
Collapse
Affiliation(s)
- Qing Lu
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Miles Mundy
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Eboni Chambers
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Thilo Lange
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Julie Newton
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Diana Borgas
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Hongwei Yao
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Gaurav Choudhary
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Rajshekhar Basak
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Mahogany Oldham
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Sharon Rounds
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
9
|
Zhang R, Ai X, Duan Y, Xue M, He W, Wang C, Xu T, Xu M, Liu B, Li C, Wang Z, Zhang R, Wang G, Tian S, Liu H. Kaempferol ameliorates H9N2 swine influenza virus-induced acute lung injury by inactivation of TLR4/MyD88-mediated NF-κB and MAPK signaling pathways. Biomed Pharmacother 2017; 89:660-672. [DOI: 10.1016/j.biopha.2017.02.081] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 02/21/2017] [Accepted: 02/21/2017] [Indexed: 01/23/2023] Open
|
10
|
Roth-Walter F, Bergmayr C, Meitz S, Buchleitner S, Stremnitzer C, Fazekas J, Moskovskich A, Müller MA, Roth GA, Manzano-Szalai K, Dvorak Z, Neunkirchner A, Jensen-Jarolim E. Janus-faced Acrolein prevents allergy but accelerates tumor growth by promoting immunoregulatory Foxp3+ cells: Mouse model for passive respiratory exposure. Sci Rep 2017; 7:45067. [PMID: 28332605 PMCID: PMC5362909 DOI: 10.1038/srep45067] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/20/2017] [Indexed: 12/30/2022] Open
Abstract
Acrolein, a highly reactive unsaturated aldehyde, is generated in large amounts during smoking and is best known for its genotoxic capacity. Here, we aimed to assess whether acrolein at concentrations relevant for smokers may also exert immunomodulatory effects that could be relevant in allergy or cancer. In a BALB/c allergy model repeated nasal exposure to acrolein abrogated allergen-specific antibody and cytokine formation, and led to a relative accumulation of regulatory T cells in the lungs. Only the acrolein-treated mice were protected from bronchial hyperreactivity as well as from anaphylactic reactions upon challenge with the specific allergen. Moreover, grafted D2F2 tumor cells grew faster and intratumoral Foxp3+ cell accumulation was observed in these mice compared to sham-treated controls. Results from reporter cell lines suggested that acrolein acts via the aryl-hydrocarbon receptor which could be inhibited by resveratrol and 3′-methoxy-4′-nitroflavone Acrolein- stimulation of human PBMCs increased Foxp3+ expression by T cells which could be antagonized by resveratrol. Our mouse and human data thus revealed that acrolein exerts systemic immunosuppression by promoting Foxp3+ regulatory cells. This provides a novel explanation why smokers have a lower allergy, but higher cancer risk.
Collapse
Affiliation(s)
- Franziska Roth-Walter
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Cornelia Bergmayr
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sarah Meitz
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Stefan Buchleitner
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Caroline Stremnitzer
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Judit Fazekas
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Anna Moskovskich
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Mario A Müller
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Georg A Roth
- Department of Anesthesiology, General Intensive Care and Pain Medicine, Medical University of Vienna, Austria
| | - Krisztina Manzano-Szalai
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Zdenek Dvorak
- Department of Cell Biology and Genetics, Palacky University, Olomouc, Czech Republic
| | - Alina Neunkirchner
- Christian Doppler Laboratory for Immunomodulation, Medical University of Vienna, Vienna, Austria
| | - Erika Jensen-Jarolim
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria.,Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
11
|
Liu X, Zheng W, Sivasankar MP. Acute Acrolein Exposure Induces Impairment of Vocal Fold Epithelial Barrier Function. PLoS One 2016; 11:e0163237. [PMID: 27643990 PMCID: PMC5028054 DOI: 10.1371/journal.pone.0163237] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 09/06/2016] [Indexed: 01/07/2023] Open
Abstract
Acrolein is a ubiquitous pollutant abundant in cigarette smoke, mobile exhaust, and industrial waste. There is limited literature on the effects of acrolein on vocal fold tissue, although there are clinical reports of voice changes after pollutant exposures. Vocal folds are responsible for voice production. The overall objective of this study was to investigate the effects of acrolein exposure on viable, excised vocal fold epithelial tissue and to characterize the mechanism underlying acrolein toxicity. Vocal fold epithelia were studied because they form the outermost layer of the vocal folds and are a primary recipient of inhaled pollutants. Porcine vocal fold epithelia were exposed to 0, 50, 100, 500, 900 or 1300 μM of acrolein for 3 hours; the metabolic activity, epithelial resistance, epithelial permeability, tight junction protein (occludin and claudin 3) expression, cell membrane integrity and lipid peroxidation were investigated. The data demonstrated that acrolein exposure at 500 μM significantly reduced vocal fold epithelial metabolic activity by 27.2% (p≤0.001). Incubation with 100 μM acrolein caused a marked increase in epithelial permeability by 130.5% (p<0.05) and a reduction in transepithelial electrical resistance (TEER) by 180.0% (p<0.001). While the expression of tight junctional protein did not change in acrolein-treated samples, the cell membrane integrity was significantly damaged with a 45.6% increase of lipid peroxidation as compared to controls (p<0.05). Taken together, these data provide evidence that acute acrolein exposure impairs vocal fold epithelial barrier integrity. Lipid peroxidation-induced cell membrane damage may play an important role in reducing the barrier function of the epithelium.
Collapse
Affiliation(s)
- Xinxin Liu
- School of Health Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - M. Preeti Sivasankar
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- Department of Speech, Language, and Hearing Sciences, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail:
| |
Collapse
|
12
|
Danyal K, de Jong W, O'Brien E, Bauer RA, Heppner DE, Little AC, Hristova M, Habibovic A, van der Vliet A. Acrolein and thiol-reactive electrophiles suppress allergen-induced innate airway epithelial responses by inhibition of DUOX1 and EGFR. Am J Physiol Lung Cell Mol Physiol 2016; 311:L913-L923. [PMID: 27612966 DOI: 10.1152/ajplung.00276.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/05/2016] [Indexed: 01/27/2023] Open
Abstract
Acrolein is a major thiol-reactive component of cigarette smoke (CS) that is thought to contribute to increased asthma incidence associated with smoking. Here, we explored the effects of acute acrolein exposure on innate airway responses to two common airborne allergens, house dust mite and Alternaria alternata, and observed that acrolein exposure of C57BL/6 mice (5 ppm, 4 h) dramatically inhibited innate airway responses to subsequent allergen challenge, demonstrated by attenuated release of the epithelial-derived cytokines IL-33, IL-25, and IL-1α. Acrolein and other anti-inflammatory thiol-reactive electrophiles, cinnamaldehyde, curcumin, and sulforaphane, similarly inhibited allergen-induced production of these cytokines from human or murine airway epithelial cells in vitro. Based on our previous observations indicating the importance of Ca2+-dependent signaling, activation of the NADPH oxidase DUOX1, and Src/EGFR-dependent signaling in allergen-induced epithelial secretion of these cytokines, we explored the impact of acrolein on these pathways. Acrolein and other thiol-reactive electrophiles were found to dramatically prevent allergen-induced activation of DUOX1 as well as EGFR, and acrolein was capable of inhibiting EGFR tyrosine kinase activity via modification of C797. Biotin-labeling strategies indicated increased cysteine modification and carbonylation of Src, EGFR, as well as DUOX1, in response to acrolein exposure in vitro and in vivo, suggesting that direct alkylation of these proteins on accessible cysteine residues may be responsible for their inhibition. Collectively, our findings indicate a novel anti-inflammatory mechanism of CS-derived acrolein and other thiol-reactive electrophiles, by directly inhibiting DUOX1- and EGFR-mediated airway epithelial responses to airborne allergens.
Collapse
Affiliation(s)
- Karamatullah Danyal
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Willem de Jong
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Edmund O'Brien
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Robert A Bauer
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - David E Heppner
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Andrew C Little
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Milena Hristova
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Aida Habibovic
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
13
|
Yeager RP, Kushman M, Chemerynski S, Weil R, Fu X, White M, Callahan-Lyon P, Rosenfeldt H. Proposed Mode of Action for Acrolein Respiratory Toxicity Associated with Inhaled Tobacco Smoke. Toxicol Sci 2016; 151:347-64. [DOI: 10.1093/toxsci/kfw051] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
14
|
Randall MJ, Haenen GRMM, Bouwman FG, van der Vliet A, Bast A. The tobacco smoke component acrolein induces glucocorticoid resistant gene expression via inhibition of histone deacetylase. Toxicol Lett 2015; 240:43-9. [PMID: 26481333 DOI: 10.1016/j.toxlet.2015.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 09/15/2015] [Accepted: 10/11/2015] [Indexed: 01/24/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is the leading cause of cigarette smoke-related death worldwide. Acrolein, a crucial reactive electrophile found in cigarette smoke mimics many of the toxic effects of cigarette smoke-exposure in the lung. In macrophages, cigarette smoke is known to hinder histone deacetylases (HDACs), glucocorticoid-regulated enzymes that play an important role in the pathogenesis of glucocorticoid resistant inflammation, a common feature of COPD. Thus, we hypothesize that acrolein plays a role in COPD-associated glucocorticoid resistance. To examine the role of acrolein on glucocorticoid resistance, U937 monocytes, differentiated with PMA to macrophage-like cells were treated with acrolein for 0.5h followed by stimulation with hydrocortisone for 8h, or treated simultaneously with LPS and hydrocortisone for 8h without acrolein. GSH and nuclear HDAC activity were measured, or gene expression was analyzed by qPCR. Acrolein-mediated TNFα gene expression was not suppressed by hydrocortisone whereas LPS-induced TNFα expression was suppressed. Acrolein also significantly inhibited nuclear HDAC activity in macrophage-like cells. Incubation of recombinant HDAC2 with acrolein led to the formation of an HDAC2-acrolein adduct identified by mass spectrometry. Therefore, these results suggest that acrolein-induced inflammatory gene expression is resistant to suppression by the endogenous glucocorticoid, hydrocortisone.
Collapse
Affiliation(s)
- Matthew J Randall
- Department of Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands; Department of Pathology, College of Medicine, University of Vermont, 89 Beaumont Avenue, Burlington, VT 05405, USA.
| | - Guido R M M Haenen
- Department of Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Freek G Bouwman
- Department of Human Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Albert van der Vliet
- Department of Pathology, College of Medicine, University of Vermont, 89 Beaumont Avenue, Burlington, VT 05405, USA
| | - Aalt Bast
- Department of Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
15
|
Moghe A, Ghare S, Lamoreau B, Mohammad M, Barve S, McClain C, Joshi-Barve S. Molecular mechanisms of acrolein toxicity: relevance to human disease. Toxicol Sci 2015; 143:242-55. [PMID: 25628402 DOI: 10.1093/toxsci/kfu233] [Citation(s) in RCA: 341] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Acrolein, a highly reactive unsaturated aldehyde, is a ubiquitous environmental pollutant and its potential as a serious environmental health threat is beginning to be recognized. Humans are exposed to acrolein per oral (food and water), respiratory (cigarette smoke, automobile exhaust, and biocide use) and dermal routes, in addition to endogenous generation (metabolism and lipid peroxidation). Acrolein has been suggested to play a role in several disease states including spinal cord injury, multiple sclerosis, Alzheimer's disease, cardiovascular disease, diabetes mellitus, and neuro-, hepato-, and nephro-toxicity. On the cellular level, acrolein exposure has diverse toxic effects, including DNA and protein adduction, oxidative stress, mitochondrial disruption, membrane damage, endoplasmic reticulum stress, and immune dysfunction. This review addresses our current understanding of each pathogenic mechanism of acrolein toxicity, with emphasis on the known and anticipated contribution to clinical disease, and potential therapies.
Collapse
Affiliation(s)
- Akshata Moghe
- *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202
| | - Smita Ghare
- *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202
| | - Bryan Lamoreau
- *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202
| | - Mohammad Mohammad
- *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202
| | - Shirish Barve
- *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202 *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202
| | - Craig McClain
- *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202 *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202 *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202
| | - Swati Joshi-Barve
- *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202 *Department of Pharmacology and Toxicology, Department of Medicine and Robley Rex VAMC, Louisville, Kentucky 40202
| |
Collapse
|
16
|
O'Brien E, Spiess PC, Habibovic A, Hristova M, Bauer RA, Randall MJ, Poynter ME, van der Vliet A. Inhalation of the reactive aldehyde acrolein promotes antigen sensitization to ovalbumin and enhances neutrophilic inflammation. J Immunotoxicol 2015; 13:191-7. [PMID: 25875327 DOI: 10.3109/1547691x.2015.1033571] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Acrolein (ACR), an α,β-unsaturated aldehyde and a major component of tobacco smoke, is a highly reactive electrophilic respiratory irritant implicated in asthma pathogenesis and severity. However, few studies have directly investigated the influence of ACR exposure on allergen sensitization and pulmonary inflammation. The present study was designed to examine the impact of ACR inhalation on allergic sensitization to the inhaled antigen ovalbumin (OVA), as well as pulmonary inflammation during subsequent OVA challenge. Adult male C57BL/6 mice were exposed to inhaled OVA (1%, 30 min/day, 4 days/week) and/or ACR (5 ppm, 4 h/day, 4 days/week) over 2 weeks and subsequently challenged with aerosolized OVA (1%, 30 min/day) over three consecutive days. Serum anti-OVA IgG1 levels were increased significantly in animals exposed to both OVA and ACR, compared to animals exposed to either OVA or ACR alone. In addition, differential cell counts and histological analysis revealed an increase in BAL neutrophils in animals exposed to both OVA and ACR. However, exposure to both OVA and ACR did not influence mRNA expression of the cytokines il5, il10, il13 or tnfa, but significantly increased mRNA expression of ccl20. Moreover, ACR exposure enhanced lung mRNA levels of il17f and tgfb1, suggesting development of enhanced inhalation tolerance to OVA. Overall, the findings indicate that ACR inhalation can promote airway-mediated sensitization to otherwise innocuous inhaled antigens, such as OVA, but also enhances immune tolerance, thereby favoring neutrophilic airway inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Matthew E Poynter
- b Department of Medicine , College of Medicine, University of Vermont , Burlington , VT , USA
| | | |
Collapse
|
17
|
Factors affecting susceptibility to Mycobacterium tuberculosis: a close view of immunological defence mechanism. Appl Biochem Biotechnol 2014; 174:2663-73. [PMID: 25296626 DOI: 10.1007/s12010-014-1217-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 09/01/2014] [Indexed: 10/24/2022]
Abstract
Tuberculosis is the most deadly infectious disease. In particular, pulmonary tuberculosis, being the predominant one, is highly contagious. In past the 200 years, one billion tuberculosis (TB) deaths had occurred, and it is anticipated that in the next 25 years, more than 40 million people may be killed by TB unless control measures are implemented. There are various causes which increase the susceptibility to Mycobacterium tuberculosis infection; these include weakened immune system which occurs through various diseases and medications like human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome (AIDS), type II diabetes, end-stage kidney disease, alcoholism and intravenous drug use, certain cancers, cancer treatment such as chemotherapy, malnutrition and very young or advanced age. Some other factors include tobacco use, which increases the risk of getting TB and dying from it. In this manuscript, the authors tried to summarize all the alterations occurring in immune system at cellular and molecular level which occur due to infection, metabolic changes and chemical exposure, which increase susceptibility to mycobacterial infection.
Collapse
|
18
|
Vroman H, van den Blink B, Kool M. Mode of dendritic cell activation: the decisive hand in Th2/Th17 cell differentiation. Implications in asthma severity? Immunobiology 2014; 220:254-61. [PMID: 25245013 DOI: 10.1016/j.imbio.2014.09.016] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/12/2014] [Accepted: 09/05/2014] [Indexed: 11/16/2022]
Abstract
Asthma is a heterogeneous chronic inflammatory disease of the airways, with reversible airflow limitations and airway remodeling. The classification of asthma phenotypes was initially based on different combinations of clinical symptoms, but they are now unfolding to link biology to phenotype. As such, patients can suffer from a predominant eosinophilic, neutrophilic or even mixed eosinophilic/neutrophilic inflammatory response. In adult asthma patients, eosinophilic inflammation is usually seen in mild-to-moderate disease and neutrophilic inflammation in more severe disease. The underlying T cell response is predominated by T helper (Th) 2, Th17, or a mixed Th2/Th17 cell immune response. Dendritic cells (DCs) are "professional" antigen presenting cells (APCs), since their principal function is to present antigens and induce a primary immune response in resting naive T cells. DCs also drive the differentiation into distinctive Th subsets. The expression of co-stimulatory molecules and cytokines by DCs and surrounding cells determines the outcome of Th cell differentiation. The nature of DC activation will determine the expression of specific co-stimulatory molecules and cytokines, specifically needed for induction of the different Th cell programs. Thus DC activation is crucial for the subsequent effector Th immune responses. In this review, we will discuss underlying mechanisms that initiate DC activation in favor of Th2 differentiation versus Th1/Th17 and Th17 differentiation in the development of mild versus moderate to severe asthma.
Collapse
Affiliation(s)
- Heleen Vroman
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | | - Mirjam Kool
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
19
|
DeJarnett N, Conklin DJ, Riggs DW, Myers JA, O'Toole TE, Hamzeh I, Wagner S, Chugh A, Ramos KS, Srivastava S, Higdon D, Tollerud DJ, DeFilippis A, Becher C, Wyatt B, McCracken J, Abplanalp W, Rai SN, Ciszewski T, Xie Z, Yeager R, Prabhu SD, Bhatnagar A. Acrolein exposure is associated with increased cardiovascular disease risk. J Am Heart Assoc 2014; 3:jah3635. [PMID: 25099132 PMCID: PMC4310380 DOI: 10.1161/jaha.114.000934] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Acrolein is a reactive aldehyde present in high amounts in coal, wood, paper, and tobacco smoke. It is also generated endogenously by lipid peroxidation and the oxidation of amino acids by myeloperoxidase. In animals, acrolein exposure is associated with the suppression of circulating progenitor cells and increases in thrombosis and atherogenesis. The purpose of this study was to determine whether acrolein exposure in humans is also associated with increased cardiovascular disease (CVD) risk. Methods and Results Acrolein exposure was assessed in 211 participants of the Louisville Healthy Heart Study with moderate to high (CVD) risk by measuring the urinary levels of the major acrolein metabolite—3‐hydroxypropylmercapturic acid (3‐HPMA). Generalized linear models were used to assess the association between acrolein exposure and parameters of CVD risk, and adjusted for potential demographic confounders. Urinary 3‐HPMA levels were higher in smokers than nonsmokers and were positively correlated with urinary cotinine levels. Urinary 3‐HPMA levels were inversely related to levels of both early (AC133+) and late (AC133−) circulating angiogenic cells. In smokers as well as nonsmokers, 3‐HPMA levels were positively associated with both increased levels of platelet–leukocyte aggregates and the Framingham Risk Score. No association was observed between 3‐HPMA and plasma fibrinogen. Levels of C‐reactive protein were associated with 3‐HPMA levels in nonsmokers only. Conclusions Regardless of its source, acrolein exposure is associated with platelet activation and suppression of circulating angiogenic cell levels, as well as increased CVD risk.
Collapse
Affiliation(s)
- Natasha DeJarnett
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Department of Environmental and Occupational Health Sciences, University of Louisville, Louisville, KY (N.D.J., D.J.T., R.Y.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Daniel J Conklin
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Daniel W Riggs
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - John A Myers
- Department of Pediatrics, University of Louisville, Louisville, KY (J.A.M.)
| | - Timothy E O'Toole
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Ihab Hamzeh
- Baylor College of Medicine, Houston, TX (I.H.)
| | - Stephen Wagner
- Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Atul Chugh
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Kenneth S Ramos
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY (K.S.R., A.B.)
| | - Sanjay Srivastava
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Deirdre Higdon
- Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - David J Tollerud
- Department of Environmental and Occupational Health Sciences, University of Louisville, Louisville, KY (N.D.J., D.J.T., R.Y.)
| | - Andrew DeFilippis
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.) Department of Medicine, Johns Hopkins University, Baltimore, MD (A.D.F.)
| | - Carrie Becher
- Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Brad Wyatt
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - James McCracken
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Wes Abplanalp
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Shesh N Rai
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Department of Bioinformatics and Biostatics, University of Louisville, Louisville, KY (S.N.R.) Biostatistics Shared Facility, JG Brown Cancer Center, University of Louisville, Louisville, KY (S.N.R.)
| | - Tiffany Ciszewski
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Zhengzhi Xie
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| | - Ray Yeager
- Department of Environmental and Occupational Health Sciences, University of Louisville, Louisville, KY (N.D.J., D.J.T., R.Y.)
| | - Sumanth D Prabhu
- Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.) Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL (S.D.P.)
| | - Aruni Bhatnagar
- Diabetes and Obesity Center, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., A.C., S.S., A.D.F., B.W., J.M.C., W.A., S.N.R., T.C., Z.X., A.B.) Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY (K.S.R., A.B.) Institue of Molecular Cardiology, University of Louisville, Louisville, KY (N.D.J., D.J.C., D.W.R., T.E.T., S.W., A.C., S.S., D.H., A.D.F., C.B., B.W., J.M.C., W.A., T.C., Z.X., S.D.P., A.B.)
| |
Collapse
|
20
|
Zhang RH, Li CH, Wang CL, Xu MJ, Xu T, Wei D, Liu BJ, Wang GH, Tian SF. N-acetyl-l-cystine (NAC) protects against H9N2 swine influenza virus-induced acute lung injury. Int Immunopharmacol 2014; 22:1-8. [PMID: 24968347 PMCID: PMC7106131 DOI: 10.1016/j.intimp.2014.06.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/06/2014] [Accepted: 06/08/2014] [Indexed: 12/16/2022]
Abstract
The antioxidant N-acetyl-l-cysteine (NAC) had been shown to inhibit replication of seasonal human influenza A viruses. Here, the effects of NAC on H9N2 swine influenza virus-induced acute lung injury (ALI) were investigated in mice. BALB/c mice were inoculated intranasally with 107 50% tissue culture infective doses (TCID50) of A/swine/HeBei/012/2008/(H9N2) viruses with or without NAC treatments to induce ALI model. The result showed that pulmonary inflammation, pulmonary edema, MPO activity, total cells, neutrophils, macrophages, TNF-α, IL-6, IL-1β and CXCL-10 in BALF were attenuated by NAC. Moreover, our data showed that NAC significantly inhibited the levels of TLR4 protein and TLR4 mRNA in the lungs. Pharmacological inhibitors of TLR4 (E5564) exerted similar effects like those determined for NAC in H9N2 swine influenza virus-infected mice. These results suggest that antioxidants like NAC represent a potential additional treatment option that could be considered in the case of an influenza A virus pandemic. NAC protects against H9N2 swine influenza virus-induced acute lung injury (ALI). NAC protects against acute lung injury by inactivation of TLR4. Eritoran (E5564), a TLR4 antagonist, also protects against acute lung injury.
Collapse
Affiliation(s)
- Rui-Hua Zhang
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, PR China
| | - Chun-Hong Li
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, PR China
| | - Cun-Lian Wang
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, PR China
| | - Ming-Ju Xu
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, PR China
| | - Tong Xu
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, PR China.
| | - Dong Wei
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, PR China
| | - Bao-Jian Liu
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, PR China
| | - Guo-Hua Wang
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, PR China
| | - Shu-Fei Tian
- Key Laboratory of Preventive Veterinary Medicine, Department of Veterinary Medicine, Animal Science College, Hebei North University, Zhangjiakou 075131, PR China
| |
Collapse
|
21
|
Spiess PC, Kasahara D, Habibovic A, Hristova M, Randall MJ, Poynter ME, van der Vliet A. Acrolein exposure suppresses antigen-induced pulmonary inflammation. Respir Res 2013; 14:107. [PMID: 24131734 PMCID: PMC3852782 DOI: 10.1186/1465-9921-14-107] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 10/08/2013] [Indexed: 12/20/2022] Open
Abstract
Background Adverse health effects of tobacco smoke arise partly from its influence on innate and adaptive immune responses, leading to impaired innate immunity and host defense. The impact of smoking on allergic asthma remains unclear, with various reports demonstrating that cigarette smoke enhances asthma development but can also suppress allergic airway inflammation. Based on our previous findings that immunosuppressive effects of smoking may be largely attributed to one of its main reactive electrophiles, acrolein, we explored the impact of acrolein exposure in a mouse model of ovalbumin (OVA)-induced allergic asthma. Methods C57BL/6 mice were sensitized to ovalbumin (OVA) by intraperitoneal injection with the adjuvant aluminum hydroxide on days 0 and 7, and challenged with aerosolized OVA on days 14–16. In some cases, mice were also exposed to 5 ppm acrolein vapor for 6 hrs/day on days 14–17. Lung tissues or brochoalveolar lavage fluids (BALF) were collected either 6 hrs after a single initial OVA challenge and/or acrolein exposure on day 14 or 48 hrs after the last OVA challenge, on day 18. Inflammatory cells and Th1/Th2 cytokine levels were measured in BALF, and lung tissue samples were collected for analysis of mucus and Th1/Th2 cytokine expression, determination of protein alkylation, cellular thiol status and transcription factor activity. Results Exposure to acrolein following OVA challenge of OVA-sensitized mice resulted in markedly attenuated allergic airway inflammation, demonstrated by decreased inflammatory cell infiltrates, mucus hyperplasia and Th2 cytokines. Acrolein exposure rapidly depleted lung tissue glutathione (GSH) levels, and induced activation of the Nrf2 pathway, indicated by accumulation of Nrf2, increased alkylation of Keap1, and induction of Nrf2-target genes such as HO-1. Additionally, analysis of inflammatory signaling pathways showed suppressed activation of NF-κB and marginally reduced activation of JNK in acrolein-exposed lungs, associated with increased carbonylation of RelA and JNK. Conclusion Acrolein inhalation suppresses Th2-driven allergic inflammation in sensitized animals, due to direct protein alkylation resulting in activation of Nrf2 and anti-inflammatory gene expression, and inhibition of NF-κB or JNK signaling. Our findings help explain the paradoxical anti-inflammatory effects of cigarette smoke exposure in allergic airways disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Albert van der Vliet
- Department of Pathology, College of Medicine, D205 Given Building, 89 Beaumont Ave, Burlington, VT 05405, USA.
| |
Collapse
|
22
|
Sultana S, Ali R, Talegaonkar S, Ahmad FJ, Mittal G, Bhatnagar A. In vivo lung deposition and sub-acute inhalation toxicity studies of nano-sized alendronate sodium as an antidote for inhaled toxic substances in Sprague Dawley rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2013; 36:636-647. [PMID: 23851119 DOI: 10.1016/j.etap.2013.05.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/17/2013] [Accepted: 05/31/2013] [Indexed: 06/02/2023]
Abstract
INTRODUCTION Alendronate sodium is a bisphosphonate agent used for the treatment of osteoporosis and other bone diseases. It has a strong chelating property to bind or, to some extent, counteract the effects of substances, such as magnesium, calcium citrate, ferrous fumarate, carbonyl iron, as well as the zinc gluconate, sulfate and acetate salts. The objective of the present study was to evaluate lung deposition and sub-acute inhalation toxicity of the alendronate sodium respiratory formulation. METHODS Particle dimension of aerosols of alendronate was measured using a particle size analyzer. Alendronate was radiolabeled using Technetium-99m for in vitro and in vivo biodistribution studies. Alendronate at doses, 0.5%, 1.0%, and 1.5% in ethanol-saline respiratory formulation was inhaled twice a day up to 5 weeks for inhalation toxicity investigations. Hematological, biochemical and lung toxicity biomarkers in bronchoalveolar lavage (BAL) fluid were determined at the end of the experiment. Histopathological analysis of lung tissues was carried out to observe any microscopic changes RESULTS Particle size analysis revealed the size within 300-500nm. Anderson cascade impactor results showed that the particles exhibited higher respirable fraction (55.52%) with MMAD of 4.66μm. Hematology, serum biochemistry and lung toxicity biomarkers in BAL fluid performed in the sub-acute toxicity studies indicated no adverse effects of alendronate sodium inhalation except for a significant increase in cholesterol levels and marginal increase in BAL fluid protein. At autopsy, no histopathological changes in major organs were observed. CONCLUSIONS The lung deposition and safety evaluation data observed from these studies suggested that aerosolized nanosized alendronate sodium by the inhalation route could be a new and promising route of administration as an antidote to radioactive substances through an increase in the bioavailability of the drug as well as a decrease in side effects on systemic delivery.
Collapse
Affiliation(s)
- Shaheen Sultana
- Jamia Hamdard, Faculty of Pharmacy, Department of Pharmaceutics, Delhi 110062, India.
| | - Rashid Ali
- Institute of Nuclear Medicine and Allied Sciences, Department of Nuclear Medicine, DRDO, Brig. S K Mazumdar Marg, Delhi 110054, India.
| | - Sushama Talegaonkar
- Jamia Hamdard, Faculty of Pharmacy, Department of Pharmaceutics, Delhi 110062, India.
| | - Farhan Jalees Ahmad
- Jamia Hamdard, Faculty of Pharmacy, Department of Pharmaceutics, Delhi 110062, India.
| | - Gaurav Mittal
- Institute of Nuclear Medicine and Allied Sciences, Department of Nuclear Medicine, DRDO, Brig. S K Mazumdar Marg, Delhi 110054, India.
| | - Aseem Bhatnagar
- Institute of Nuclear Medicine and Allied Sciences, Department of Nuclear Medicine, DRDO, Brig. S K Mazumdar Marg, Delhi 110054, India.
| |
Collapse
|
23
|
Crotonaldehyde induces apoptosis and immunosuppression in alveolar macrophages. Toxicol In Vitro 2013; 27:128-37. [DOI: 10.1016/j.tiv.2012.09.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 08/08/2012] [Accepted: 09/11/2012] [Indexed: 11/23/2022]
|
24
|
Reis Gonçalves CT, Reis Gonçalves CG, de Almeida FM, dos Santos Lopes FDTQ, dos Santos Durão ACC, dos Santos FA, da Silva LFF, Marcourakis T, Castro-Faria-Neto HC, Vieira RDP, Dolhnikoff M. Protective effects of aerobic exercise on acute lung injury induced by LPS in mice. Crit Care 2012; 16:R199. [PMID: 23078757 PMCID: PMC3682301 DOI: 10.1186/cc11807] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 08/24/2012] [Accepted: 09/18/2012] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION The regular practice of physical exercise has been associated with beneficial effects on various pulmonary conditions. We investigated the mechanisms involved in the protective effect of exercise in a model of lipopolysaccharide (LPS)-induced acute lung injury (ALI). METHODS Mice were divided into four groups: Control (CTR), Exercise (Exe), LPS, and Exercise + LPS (Exe + LPS). Exercised mice were trained using low intensity daily exercise for five weeks. LPS and Exe + LPS mice received 200 µg of LPS intratracheally 48 hours after the last physical test. We measured exhaled nitric oxide (eNO); respiratory mechanics; neutrophil density in lung tissue; protein leakage; bronchoalveolar lavage fluid (BALF) cell counts; cytokine levels in BALF, plasma and lung tissue; antioxidant activity in lung tissue; and tissue expression of glucocorticoid receptors (Gre). RESULTS LPS instillation resulted in increased eNO, neutrophils in BALF and tissue, pulmonary resistance and elastance, protein leakage, TNF-alpha in lung tissue, plasma levels of IL-6 and IL-10, and IL-1beta, IL-6 and KC levels in BALF compared to CTR (P ≤0.02). Aerobic exercise resulted in decreases in eNO levels, neutrophil density and TNF-alpha expression in lung tissue, pulmonary resistance and elastance, and increased the levels of IL-6, IL-10, superoxide dismutase (SOD-2) and Gre in lung tissue and IL-1beta in BALF compared to the LPS group (P ≤0.04). CONCLUSIONS Aerobic exercise plays important roles in protecting the lungs from the inflammatory effects of LPS-induced ALI. The effects of exercise are mainly mediated by the expression of anti-inflammatory cytokines and antioxidants, suggesting that exercise can modulate the inflammatory-anti-inflammatory and the oxidative-antioxidative balance in the early phase of ALI.
Collapse
Affiliation(s)
- Cintia Tokio Reis Gonçalves
- Departamento de Patologia (LIM05) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 sala 1155, CEP 01246-903, São Paulo, Brazil
| | - Carlos Gustavo Reis Gonçalves
- Departamento de Patologia (LIM05) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 sala 1155, CEP 01246-903, São Paulo, Brazil
| | - Francine Maria de Almeida
- Departamento de Clínica Médica (LIM20) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 sala 1226, CEP 01246-903, São Paulo, Brazil
| | | | - Ana Carolina Cardoso dos Santos Durão
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 580 Bl 13B, CEP 05503-900, São Paulo, Brazil
| | - Fabiana Almeida dos Santos
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 580 Bl 13B, CEP 05503-900, São Paulo, Brazil
| | - Luiz Fernando Ferraz da Silva
- Departamento de Patologia (LIM05) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 sala 1155, CEP 01246-903, São Paulo, Brazil
| | - Tania Marcourakis
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Av. Prof. Lineu Prestes, 580 Bl 13B, CEP 05503-900, São Paulo, Brazil
| | - Hugo C Castro-Faria-Neto
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fiocruz, Av. Brasil, 4365, CEP 21045-900, Rio de Janeiro, Brazil
| | - Rodolfo de Paula Vieira
- Departamento de Patologia (LIM05) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 sala 1155, CEP 01246-903, São Paulo, Brazil
- Universidade Nove de Julho - UNINOVE, Rua Vergueiro 239/245, Vergueiro, CEP 01504-000, São Paulo - SP, Brazil
| | - Marisa Dolhnikoff
- Departamento de Patologia (LIM05) da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Arnaldo, 455 sala 1155, CEP 01246-903, São Paulo, Brazil
| |
Collapse
|
25
|
Moretto N, Volpi G, Pastore F, Facchinetti F. Acrolein effects in pulmonary cells: relevance to chronic obstructive pulmonary disease. Ann N Y Acad Sci 2012; 1259:39-46. [PMID: 22758635 DOI: 10.1111/j.1749-6632.2012.06531.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Acrolein (2-propenal) is a highly reactive α,β-unsaturated aldehyde and a respiratory irritant that is ubiquitously present in the environment but that can also be generated endogenously at sites of inflammation. Acrolein is abundant in tobacco smoke, which is the major environmental risk factor for chronic obstructive pulmonary disease (COPD), and elevated levels of acrolein are found in the lung fluids of COPD patients. Its high electrophilicity makes acrolein notorious for its facile reaction with biological nucleophiles, leading to the modification of proteins and DNA and depletion of antioxidant defenses. As a consequence, acrolein results in oxidative stress as well as altered intracellular signaling and gene transcription/translation. In pulmonary cells, acrolein, at subtoxic concentrations, can activate intracellular stress kinases, alter the production of inflammatory mediators and proteases, modify innate immune response, induce mucus hypersecretion, and damage airway epithelium. A better comprehension of the mechanisms underlying acrolein effects in the airways may suggest novel treatment strategies in COPD.
Collapse
Affiliation(s)
- Nadia Moretto
- Department of Pharmacology, Chiesi Farmaceutici SpA, Parma, Italy
| | | | | | | |
Collapse
|
26
|
Hizume DC, Toledo AC, Moriya HT, Saraiva-Romanholo BM, Almeida FM, Arantes-Costa FM, Vieira RP, Dolhnikoff M, Kasahara DI, Martins MA. Cigarette smoke dissociates inflammation and lung remodeling in OVA-sensitized and challenged mice. Respir Physiol Neurobiol 2012; 181:167-176. [PMID: 22446562 DOI: 10.1016/j.resp.2012.03.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 03/08/2012] [Accepted: 03/08/2012] [Indexed: 01/29/2023]
Abstract
We evaluated the effects of cigarette smoke (CS) on lung inflammation and remodeling in a model of ovalbumin (OVA)-sensitized and OVA-challenged mice. Male BALB/c mice were divided into 4 groups: non-sensitized and air-exposed (control); non-sensitized and exposed to cigarette smoke (CS), sensitized and air-exposed (OVA) (50 μg+OVA 1% 3 times/week for 3 weeks) and sensitized and cigarette smoke exposed mice (OVA+CS). IgE levels were not affected by CS exposure. The increases in total bronchoalveolar fluid cells in the OVA group were attenuated by co-exposure to CS, as were the changes in IL-4, IL-5, and eotaxin levels as well as tissue elastance (p<0.05). In contrast, only the OVA+CS group showed a significant increase in the protein expression of IFN-γ, VEGF, GM-CSF and collagen fiber content (p<0.05). In our study, exposure to cigarette smoke in OVA-challenged mice resulted in an attenuation of pulmonary inflammation but led to an increase in pulmonary remodeling and resulted in the dissociation of airway inflammation from lung remodeling.
Collapse
Affiliation(s)
- Deborah C Hizume
- Department of Medicine (LIM-20), School of Medicine, University of Sao Paulo, Sao Paulo, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Acrolein is a respiratory irritant that can be generated during cooking and is in environmental tobacco smoke. More plentiful in cigarette smoke than polycyclic aromatic hydrocarbons (PAH), acrolein can adduct tumor suppressor p53 (TP53) DNA and may contribute to TP53-mutations in lung cancer. Acrolein is also generated endogenously at sites of injury, and excessive breath levels (sufficient to activate metalloproteinases and increase mucin transcripts) have been detected in asthma and chronic obstructive pulmonary disease (COPD). Because of its reactivity with respiratory-lining fluid or cellular macromolecules, acrolein alters gene regulation, inflammation, mucociliary transport, and alveolar-capillary barrier integrity. In laboratory animals, acute exposures have lead to acute lung injury and pulmonary edema similar to that produced by smoke inhalation whereas lower concentrations have produced bronchial hyperreactivity, excessive mucus production, and alveolar enlargement. Susceptibility to acrolein exposure is associated with differential regulation of cell surface receptor, transcription factor, and ubiquitin-proteasome genes. Consequent to its pathophysiological impact, acrolein contributes to the morbidly and mortality associated with acute lung injury and COPD, and possibly asthma and lung cancer.
Collapse
Affiliation(s)
- Kiflai Bein
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15219-3130, USA.
| | | |
Collapse
|
28
|
Hristova M, Spiess PC, Kasahara DI, Randall MJ, Deng B, van der Vliet A. The tobacco smoke component, acrolein, suppresses innate macrophage responses by direct alkylation of c-Jun N-terminal kinase. Am J Respir Cell Mol Biol 2012; 46:23-33. [PMID: 21778411 PMCID: PMC3262655 DOI: 10.1165/rcmb.2011-0134oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 07/07/2011] [Indexed: 12/21/2022] Open
Abstract
The respiratory innate immune system is often compromised by tobacco smoke exposure, and previous studies have indicated that acrolein, a reactive electrophile in tobacco smoke, may contribute to the immunosuppressive effects of smoking. Exposure of mice to acrolein at concentrations similar to those in cigarette smoke (5 ppm, 4 h) significantly suppressed alveolar macrophage responses to bacterial LPS, indicated by reduced induction of nitric oxide synthase 2, TNF-α, and IL-12p40. Mechanistic studies with bone marrow-derived macrophages or MH-S macrophages demonstrated that acrolein (1-30 μM) attenuated these LPS-mediated innate responses in association with depletion of cellular glutathione, although glutathione depletion itself was not fully responsible for these immunosuppressive effects. Inhibitory actions of acrolein were most prominent after acute exposure (<2 h), indicating the involvement of direct and reversible interactions of acrolein with critical signaling pathways. Among the key signaling pathways involved in innate macrophage responses, acrolein marginally affected LPS-mediated activation of nuclear factor (NF)-κB, and significantly suppressed phosphorylation of c-Jun N-terminal kinase (JNK) and activation of c-Jun. Using biotin hydrazide labeling, NF-κB RelA and p50, as well as JNK2, a critical mediator of innate macrophage responses, were revealed as direct targets for alkylation by acrolein. Mass spectrometry analysis of acrolein-modified recombinant JNK2 indicated adduction to Cys(41) and Cys(177), putative important sites involved in mitogen-activated protein kinase (MAPK) kinase (MEK) binding and JNK2 phosphorylation. Our findings indicate that direct alkylation of JNK2 by electrophiles, such as acrolein, may be a prominent and hitherto unrecognized mechanism in their immunosuppressive effects, and may be a major factor in smoking-induced effects on the immune system.
Collapse
Affiliation(s)
| | | | | | | | - Bin Deng
- Department of Biology and Proteomics Core Facility, University of Vermont, Burlington, Vermont
| | | |
Collapse
|
29
|
Srivastava S, Sithu SD, Vladykovskaya E, Haberzettl P, Hoetker DJ, Siddiqui MA, Conklin DJ, D'Souza SE, Bhatnagar A. Oral exposure to acrolein exacerbates atherosclerosis in apoE-null mice. Atherosclerosis 2011; 215:301-8. [PMID: 21371710 DOI: 10.1016/j.atherosclerosis.2011.01.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 12/20/2010] [Accepted: 01/03/2011] [Indexed: 12/13/2022]
Abstract
BACKGROUND Acrolein is a dietary aldehyde that is present in high concentrations in alcoholic beverages and foods including cheese, donuts and coffee. It is also abundant in tobacco smoke, automobile exhaust and industrial waste and is generated in vivo during inflammation and oxidative stress. OBJECTIVES The goal of this study was to examine the effects of dietary acrolein on atherosclerosis. METHODS Eight-week-old male apoE-null mice were gavage-fed acrolein (2.5mg/kg/day) for 8 weeks. Atherosclerotic lesion formation and composition and plasma lipids and platelet factor 4 (PF4) levels were measured. Effects of acrolein and PF4 on endothelial cell function was measured in vitro. RESULTS Acrolein feeding increased the concentration of cholesterol in the plasma. NMR analysis of the lipoproteins showed that acrolein feeding increased the abundance of small and medium VLDL particles. Acrolein feeding also increased atherosclerotic lesion formation in the aortic valve and the aortic arch. Immunohistochemical analysis showed increased macrophage accumulation in the lesions of acrolein-fed mice. Plasma PF4 levels and accumulation of PF4 in atherosclerotic lesions was increased in the acrolein-fed mice. Incubation of endothelial cells with the plasma of acrolein-fed mice augmented transmigration of monocytic cells, which was abolished by anti-PF4 antibody treatment. CONCLUSIONS Dietary exposure to acrolein exacerbates atherosclerosis in apoE-null mice. Consumption of foods and beverages rich in unsaturated aldehydes such as acrolein may be a contributing factor to the progression of atherosclerotic lesions.
Collapse
Affiliation(s)
- Sanjay Srivastava
- Diabetes and Obesity Center, University of Louisville, Louisville, KY 40202, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Shang S, Ordway D, Henao-Tamayo M, Bai X, Oberley-Deegan R, Shanley C, Orme IM, Case S, Minor M, Ackart D, Hascall-Dove L, Ovrutsky AR, Kandasamy P, Voelker DR, Lambert C, Freed BM, Iseman MD, Basaraba RJ, Chan ED. Cigarette Smoke Increases Susceptibility to Tuberculosis—Evidence From In Vivo and In Vitro Models. J Infect Dis 2011; 203:1240-8. [PMID: 21357942 DOI: 10.1093/infdis/jir009] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Shaobin Shang
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Olson N, Kasahara DI, Hristova M, Bernstein R, Janssen-Heininger Y, van der Vliet A. Modulation of NF-κB and hypoxia-inducible factor--1 by S-nitrosoglutathione does not alter allergic airway inflammation in mice. Am J Respir Cell Mol Biol 2010; 44:813-23. [PMID: 20693401 DOI: 10.1165/rcmb.2010-0035oc] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Induction of nitric oxide synthase (NOS)-2 and production of nitric oxide (NO) are common features of allergic airway disease. Conditions of severe asthma are associated with deficiency of airway S-nitrosothiols, a biological product of NO that can suppress inflammation by S-nitrosylation of the proinflammatory transcription factor, NF-κB. Therefore, restoration of airway S-nitrosothiols might have therapeutic benefit, and this was tested in a mouse model of ovalbumin (OVA)-induced allergic inflammation. Naive or OVA-sensitized animals were administered S-nitrosoglutathione (GSNO; 50 μl, 10 mM) intratracheally before OVA challenge and analyzed 48 hours later. GSNO administration enhanced lung tissue S-nitrosothiol levels and reduced NF-κB activity in OVA-challenged animals compared with control animals, but did not lead to significant changes in total bronchoalveolar lavage cell counts, differentials, or mucus metaplasia markers. Administration of GSNO also altered the activation of hypoxia-inducible factor (HIF)-1, leading to HIF-1 activation in naive mice, but suppressed HIF-1 activation in OVA-challenged mice. We assessed the contribution of endogenous NOS2 in regulating NF-κB and/or HIF-1 activation and allergic airway inflammation using NOS2(-/-) mice. Although OVA-induced NF-κB activation was slightly increased in NOS2(-/-) mice, associated with small increases in bronchoalveolar lavage neutrophils, other markers of allergic inflammation and HIF-1 activation were similar in NOS2(-/-) and wild-type mice. Collectively, our studies indicate that instillation of GSNO can suppress NF-κB activation during allergic airway inflammation, but does not significantly affect overall markers of inflammation or mucus metaplasia, thus potentially limiting its therapeutic potential due to effects on additional signaling pathways, such as HIF-1.
Collapse
Affiliation(s)
- Nels Olson
- Department of Pathology, College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | | | | | | | | | | |
Collapse
|
32
|
Sithu SD, Srivastava S, Siddiqui MA, Vladykovskaya E, Riggs DW, Conklin DJ, Haberzettl P, O'Toole TE, Bhatnagar A, D'Souza SE. Exposure to acrolein by inhalation causes platelet activation. Toxicol Appl Pharmacol 2010; 248:100-10. [PMID: 20678513 DOI: 10.1016/j.taap.2010.07.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 07/16/2010] [Accepted: 07/16/2010] [Indexed: 12/16/2022]
Abstract
Acrolein is a common air pollutant that is present in high concentrations in wood, cotton, and tobacco smoke, automobile exhaust and industrial waste and emissions. Exposure to acrolein containing environmental pollutants such as tobacco smoke and automobile exhaust has been linked to the activation of the coagulation and hemostasis pathways and thereby to the predisposition of thrombotic events in human. To examine the effects of acrolein on platelets, adult male C57Bl/6 mice were subjected acute (5ppm for 6h) or sub-chronic (1ppm, 6h/day for 4days) acrolein inhalation exposures. The acute exposure to acrolein did not cause pulmonary inflammation and oxidative stress, dyslipidemia or induce liver damage or muscle injury. Platelet GSH levels in acrolein-exposed mice were comparable to controls, but acrolein-exposure increased the abundance of protein-acrolein adducts in platelets. Platelets isolated from mice, exposed to both acute and sub-chronic acrolein levels, showed increased ADP-induced platelet aggregation. Exposure to acrolein also led to an increase in the indices of platelet activation such as the formation of platelet-leukocyte aggregates in the blood, plasma PF4 levels, and increased platelet-fibrinogen binding. The bleeding time was decreased in acrolein exposed mice. Plasma levels of PF4 were also increased in mice exposed to environmental tobacco smoke. Similar to inhalation exposure, acrolein feeding to mice also increased platelet activation and established a pro-thrombotic state in mice. Together, our data suggest that acrolein is an important contributing factor to the pro-thrombotic risk in human exposure to pollutants such as tobacco smoke or automobile exhaust, or through dietary consumption.
Collapse
Affiliation(s)
- Srinivas D Sithu
- Department of Physiology and Biophysics, University of Louisville, Louisville, KY 40202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Deshmukh HS, McLachlan A, Atkinson JJ, Hardie WD, Korfhagen TR, Dietsch M, Liu Y, Di PYP, Wesselkamper SC, Borchers MT, Leikauf GD. Matrix metalloproteinase-14 mediates a phenotypic shift in the airways to increase mucin production. Am J Respir Crit Care Med 2009; 180:834-45. [PMID: 19661247 DOI: 10.1164/rccm.200903-0328oc] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Induced mainly by cigarette smoking, chronic obstructive pulmonary disease (COPD) is a global public health problem characterized by progressive difficulty in breathing and increased mucin production. Previously, we reported that acrolein levels found in COPD sputum could activate matrix metalloproteinase-9 (MMP9). OBJECTIVES To determine whether acrolein increases expression and activity of MMP14, a critical membrane-bound endopeptidase that can initial a MMP-activation cascade. METHODS MMP14 activity and adduct formation were measured following direct acrolein treatment. MMP14 expression and activity was measured in human airway epithelial cells. MMP14 immunohistochemistry was performed with COPD tissue, and in acrolein- or tobacco-exposed mice. MEASUREMENTS AND MAIN RESULTS In a cell-free system, acrolein, in concentrations equal to those found in COPD sputum, directly adducted cysteine 319 in the MMP14 hemopexin-like domain and activated MMP14. In cells, acrolein increased MMP14 activity, which was inhibited by a proprotein convertase inhibitor, hexa-d-arginine. In the airway epithelium of COPD subjects, immunoreactive MMP14 protein increased. In mouse lung, acrolein or tobacco smoke increased lung MMP14 activity and protein. In cells, acrolein-induced MMP14 transcripts were inhibited by an epidermal growth factor receptor (EGFR) neutralizing antibody, EGFR kinase inhibitor, metalloproteinase inhibitor, or mitogen-activated protein kinase (MAPK) 3/2 or MAPK8 inhibitors, but not a MAPK14 inhibitor. Decreasing the MMP14 protein and activity in vitro by small interfering (si)RNA to MMP14 diminished the acrolein-induced MUC5AC transcripts. In acrolein-exposed mice or transgenic mice with lung-specific transforming growth factor-alpha (an EGFR ligand) expression, lung MMP14 and MUC5AC levels increased and these effects were inhibited by a EGFR inhibitor, erlotinib. CONCLUSIONS Taken together, these findings implicate acrolein-induced MMP14 expression and activity in mucin production in COPD.
Collapse
Affiliation(s)
- Hitesh S Deshmukh
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Gaschler GJ, Skrtic M, Zavitz CCJ, Lindahl M, Onnervik PO, Murphy TF, Sethi S, Stämpfli MR. Bacteria challenge in smoke-exposed mice exacerbates inflammation and skews the inflammatory profile. Am J Respir Crit Care Med 2009; 179:666-75. [PMID: 19179487 DOI: 10.1164/rccm.200808-1306oc] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE The pathogenesis of chronic obstructive pulmonary disease is associated with acute episodes of bacterial exacerbations. The most commonly isolated bacteria during episodes of exacerbation is nontypeable Haemophilus influenzae (NTHI). OBJECTIVES In this study, we investigated the in vivo consequences of cigarette smoke exposure on the inflammatory response to an NTHI challenge. METHODS C57BL/6 and BALB/c mice were exposed to cigarette smoke for 8 weeks and subsequently challenged intranasally with NTHI. MEASUREMENTS AND MAIN RESULTS We observed increased pulmonary inflammation and lung damage in cigarette smoke-exposed NTHI-challenged mice as compared with control NTHI-challenged mice. Furthermore, although NTHI challenge in control mice was marked by increases in tumor necrosis factor-alpha, IL-6, MIP-2, and KC/GROalpha, NTHI challenge in cigarette smoke-exposed mice led to a prominent up-regulation of a different subset of inflammatory mediators, most notably MCP-1, -3, and -5, IP-10, and MIP-1gamma. This skewed inflammatory mediator expression was also observed after ex vivo NTHI stimulation of alveolar macrophages, signifying their importance to this altered response. Importantly, corticosteroids attenuated inflammation after NTHI challenge in both cigarette smoke-exposed and control mice; however, this was associated with significantly increased bacterial burden. CONCLUSIONS Collectively, these data suggest that cigarette smoke exacerbates the inflammatory response to a bacterial challenge via skewed inflammatory mediator expression.
Collapse
Affiliation(s)
- Gordon J Gaschler
- Medical Sciences Graduate Program, McMaster University, 1200 Main St. West, Hamilton, ON, L8N 3Z5, Canada
| | | | | | | | | | | | | | | |
Collapse
|