1
|
Sabet NH, Wyatt TA. The alcohol exposome. Alcohol 2025; 122:81-89. [PMID: 39722409 PMCID: PMC11918757 DOI: 10.1016/j.alcohol.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/05/2024] [Accepted: 12/07/2024] [Indexed: 12/28/2024]
Abstract
Science is now in a new era of exposome research that strives to build a more all-inclusive, panoramic view in the quest for answers; this is especially true in the field of toxicology. Alcohol exposure researchers have been examining the multivariate co-exposures that may either exacerbate or initiate alcohol-related tissue/organ injuries. This manuscript presents selected key variables that represent the Alcohol Exposome. The primary variables that make up the Alcohol Exposome can include comorbidities such as cigarettes, poor diet, occupational hazards, environmental hazards, infectious agents, and aging. In addition to representing multiple factors, the Alcohol Exposome examines the various types of intercellular communications that are carried from one organ system to another and may greatly impact the types of injuries and metabolites caused by alcohol exposure. The intent of defining the Alcohol Exposome is to bring the newly expanded definition of Exposomics, meaning the study of the exposome, to the field of alcohol research and to emphasize the need for examining research results in a non-isolated environment representing a more relevant manner in which all human physiology exists.
Collapse
Affiliation(s)
- Nousha H Sabet
- Department of Internal Medicine, Division of Pulmonary, Critical Care & Sleep, University of Nebraska Medical Center, Omaha, NE, USA; Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE', USA
| | - Todd A Wyatt
- Department of Internal Medicine, Division of Pulmonary, Critical Care & Sleep, University of Nebraska Medical Center, Omaha, NE, USA; Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE', USA; Department of Veterans Affairs, Nebraska-Western Iowa Health Care System, Omaha, NE, USA.
| |
Collapse
|
2
|
Cui Y, Du X, Li Y, Wang D, Lv Z, Yuan H, Chen Y, Liu J, Sun Y, Wang W. Imbalanced and Unchecked: The Role of Metal Dyshomeostasis in Driving COPD Progression. COPD 2024; 21:2322605. [PMID: 38591165 DOI: 10.1080/15412555.2024.2322605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/19/2024] [Indexed: 04/10/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic respiratory condition characterized by persistent inflammation and oxidative stress, which ultimately leads to progressive restriction of airflow. Extensive research findings have cogently suggested that the dysregulation of essential transition metal ions, notably iron, copper, and zinc, stands as a critical nexus in the perpetuation of inflammatory processes and oxidative damage within the lungs of COPD patients. Unraveling the intricate interplay between metal homeostasis, oxidative stress, and inflammatory signaling is of paramount importance in unraveling the intricacies of COPD pathogenesis. This comprehensive review aims to examine the current literature on the sources, regulation, and mechanisms by which metal dyshomeostasis contributes to COPD progression. We specifically focus on iron, copper, and zinc, given their well-characterized roles in orchestrating cytokine production, immune cell function, antioxidant depletion, and matrix remodeling. Despite the limited number of clinical trials investigating metal modulation in COPD, the advent of emerging methodologies tailored to monitor metal fluxes and gauge responses to chelation and supplementation hold great promise in unlocking the potential of metal-based interventions. We conclude that targeted restoration of metal homeostasis represents a promising frontier for ameliorating pathological processes driving COPD progression.
Collapse
Affiliation(s)
- Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Xinqian Du
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yunqi Li
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Dan Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Huihui Yuan
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yan Chen
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Jie Liu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Ying Sun
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
3
|
Zhang C, Xi Y, Zhang Y, He P, Su X, Fan F, Wu M, Kong X, Shi Y. Genetic association analysis of dietary intake and idiopathic pulmonary fibrosis: a two-sample mendelian randomization study. BMC Pulm Med 2024; 24:15. [PMID: 38178024 PMCID: PMC10768076 DOI: 10.1186/s12890-023-02831-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND IPF is a complex lung disease whose aetiology is not fully understood, but diet may have an impact on its development and progression. Therefore, we investigated the potential causal connection between dietary intake and IPF through TSMR to offer insights for early disease prevention recommendations. METHODS The study incorporated 29 dietary exposure factors, oily fish intake, bacon intake, processed meat intake, poultry intake, beef intake, pork intake, lamb/mutton intake, non-oily fish intake, fresh fruit intake, cooked vegetable intake, baked bean intake, fresh tomato intake, tinned tomato intake, salad/raw vegetable intake, Fresh fruit intake, coffee intake, tea intake, water intake, red wine intake, average weekly beer plus cider intake, alcoholic drinks per week, cereal intake, bread intake, whole-wheat intake, whole-wheat cereal intake, cheese intake, yogurt intake, salt added to food and whole egg intake. The study explored the causal link between diet and IPF using TSMR analysis, predominantly the IVW method, and performed sensitivity analyses to validate the results. RESULT The study revealed that consuming oily fish, yogurt, and dried fruits had a protective effect against IPF, whereas the consumption of alcoholic beverages and beef was linked to an increased risk of IPF. CONCLUSION In this MR study, it was discovered that the consumption of oily fish, yogurt, and dried fruits exhibited a protective effect against IPF, whereas the intake of alcoholic beverages and beef was associated with an elevated risk of IPF. These findings underscore the significance of making informed and timely dietary decisions in IPF prevention.
Collapse
Affiliation(s)
- Chenwei Zhang
- Department of Respiratory and Critical Care Medicine, Shanxi Medical University Affiliated First Hospital, Taiyuan, 030000, China
| | - Yujia Xi
- Department of Urology, The Second Hospital of Shanxi Medical University, Taiyuan, 030000, China
| | - Yukai Zhang
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan, 030000, China
| | - Peiyun He
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan, 030000, China
| | - Xuesen Su
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan, 030000, China
| | - Fangfang Fan
- Department of Respiratory and Critical Care Medicine, Shanxi Medical University Affiliated First Hospital, Taiyuan, 030000, China
| | - Min Wu
- Department of Respiratory and Critical Care Medicine, Shanxi Medical University Affiliated First Hospital, Taiyuan, 030000, China
| | - Xiaomei Kong
- Department of Respiratory and Critical Care Medicine, Shanxi Medical University Affiliated First Hospital, Taiyuan, 030000, China.
- NHC Key Laboratory of Pneumoconiosis, Taiyuan, 030000, China.
| | - Yiwei Shi
- Department of Respiratory and Critical Care Medicine, Shanxi Medical University Affiliated First Hospital, Taiyuan, 030000, China.
- NHC Key Laboratory of Pneumoconiosis, Taiyuan, 030000, China.
| |
Collapse
|
4
|
Sueblinvong V, Fan X, Hart C, Molina S, Koval M, Guidot DM. Ethanol-exposed lung fibroblasts cause airway epithelial barrier dysfunction. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:1839-1849. [PMID: 37864530 DOI: 10.1111/acer.15174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/24/2023] [Accepted: 08/11/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Chronic alcohol ingestion predisposes to lung injury and disrepair during sepsis. Our previous studies outlined roles for transforming growth factor-beta 1 (TGFβ1) and granulocyte-macrophage colony-stimulating factor (GM-CSF) in epithelial barrier homeostasis and how alcohol perturbs their expression and signaling. Here we hypothesize that ethanol-exposed lung fibroblasts (LF) are a source of dysregulated TGFβ1 and GM-CSF and thereby alter airway epithelial barrier function. METHODS Human or rat LF were cultured ± ethanol for 2 weeks and then co-cultured with human or rat airway epithelial cells (AEC) seeded on Transwell permeable supports. In selected groups, a TGFβ1 receptor type 1 (TGFβR1) inhibitor (SB431542) or a TGFβ1 neutralizing antibody was applied. Transepithelial electrical resistance (TER) was measured prior to co-culture and on day 5 of co-culture. AEC were then analyzed for the expression of selected tight junction and mesenchymal proteins, and transwell membranes were analyzed by immunofluorescence microscopy for ZO-1 expression and localization. TGFβ1 and GM-CSF levels in conditioned media from the co-cultures were quantified by ELISA. RESULTS AEC co-cultured with ethanol-exposed LF (ELF) showed a significant reduction in TER and corresponding decreases in ZO-1 expression, whereas collagen type 1A1 and α-smooth muscle actin protein expression were increased. In parallel, in conditioned media from the ELF + AEC co-cultures, activated TGFβ1 levels increased and GM-CSF levels decreased. Notably, all the effects of ELF on the AEC were prevented by blocking TGFβ1 activity. CONCLUSIONS Prior ethanol exposure to LF induces barrier dysfunction in naive AEC in a paracrine fashion through activation of TGFβ1 signaling and suppression of GM-CSF. These experimental findings provide a potential mechanism by which chronic alcohol ingestion impairs airway epithelial integrity and renders individuals susceptible to lung injury.
Collapse
Affiliation(s)
- Viranuj Sueblinvong
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Xian Fan
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Craishun Hart
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Samuel Molina
- FUJIFILM Irvine Scientific, Warminster, Pennsylvania, USA
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David M Guidot
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
5
|
Gbotosho OT, Li W, Joiner CH, Brown LAS, Hyacinth HI. The inflammatory profiles of pulmonary alveolar macrophages and alveolar type 2 cells in SCD. Exp Biol Med (Maywood) 2023; 248:1013-1023. [PMID: 37012678 PMCID: PMC10581160 DOI: 10.1177/15353702231157940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 01/15/2023] [Indexed: 04/05/2023] Open
Abstract
The lung microenvironment plays a crucial role in maintaining lung homeostasis as well as the initiation and resolution of both acute and chronic lung injury. Acute chest syndrome (ACS) is a complication of sickle cell disease (SCD) like acute lung injury. Both the endothelial cells and peripheral blood mononuclear cells are known to secrete proinflammatory cytokines elevated during ACS episodes. However, in SCD, the lung microenvironment that may favor excessive production of proinflammatory cytokines and the contribution of other lung resident cells, such as alveolar macrophages and alveolar type 2 epithelial (AT-2) cells, to ACS pathogenesis is not completely understood. Here, we sought to understand the pulmonary microenvironment and the proinflammatory profile of lung alveolar macrophages (LAMs) and AT-2 cells at steady state in Townes sickle cell (SS) mice compared to control mice (AA). In addition, we examined lung function and micromechanics molecules essential for pulmonary epithelial barrier function in these mice. Our results showed that bronchoalveolar lavage (BAL) fluid in SS mice had elevated protein levels of pro-inflammatory cytokines interleukin (IL)-1β and IL-12 (p ⩽ 0.05) compared to AA controls. We showed for the first time, significantly increased protein levels of inflammatory mediators (Human antigen R (HuR), Toll-like receptor 4 (TLR4), MyD88, and PU.1) in AT-2 cells (1.4 to 2.2-fold) and LAM (17-21%) isolated from SS mice compared to AA control mice at steady state. There were also low levels of anti-inflammatory transcription factors (Nrf2 and PPARy) in SS mice compared to AA controls (p ⩽ 0.05). Finally, we found impaired lung function and a dysregulated composition of surfactant proteins (B and C). Our results demonstrate that SS mice at steady state had a compromised lung microenvironment with elevated expression of proinflammatory cytokines by AT-2 cells and LAM, as well as dysregulated expression of surfactant proteins necessary for maintaining the alveolar barrier integrity and lung function.
Collapse
Affiliation(s)
- Oluwabukola T Gbotosho
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Wei Li
- Aflac Cancer & Blood Disorders Center of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Clinton H Joiner
- Aflac Cancer & Blood Disorders Center of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lou Ann S Brown
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hyacinth I Hyacinth
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
6
|
Chen Y, Li F, Hua M, Liang M, Song C. Role of GM-CSF in lung balance and disease. Front Immunol 2023; 14:1158859. [PMID: 37081870 PMCID: PMC10111008 DOI: 10.3389/fimmu.2023.1158859] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 03/24/2023] [Indexed: 04/07/2023] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a hematopoietic growth factor originally identified as a stimulus that induces the differentiation of bone marrow progenitor cells into granulocytes and macrophages. GM-CSF is now considered to be a multi-origin and pleiotropic cytokine. GM-CSF receptor signals activate JAK2 and induce nuclear signals through the JAK-STAT, MAPK, PI3K, and other pathways. In addition to promoting the metabolism of pulmonary surfactant and the maturation and differentiation of alveolar macrophages, GM-CSF plays a key role in interstitial lung disease, allergic lung disease, alcoholic lung disease, and pulmonary bacterial, fungal, and viral infections. This article reviews the latest knowledge on the relationship between GM-CSF and lung balance and lung disease, and indicates that there is much more to GM-CSF than its name suggests.
Collapse
Affiliation(s)
- Yingzi Chen
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Anhui, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Anhui, China
| | - Fan Li
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Anhui, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Anhui, China
| | - Mengqing Hua
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Anhui, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Anhui, China
| | - Meng Liang
- Department of Biotechnology, School of Life Science, Bengbu Medical College, Anhui, China
- *Correspondence: Chuanwang Song, ; Meng Liang,
| | - Chuanwang Song
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Anhui, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Anhui, China
- *Correspondence: Chuanwang Song, ; Meng Liang,
| |
Collapse
|
7
|
Butts M, Sundaram VL, Murughiyan U, Borthakur A, Singh S. The Influence of Alcohol Consumption on Intestinal Nutrient Absorption: A Comprehensive Review. Nutrients 2023; 15:nu15071571. [PMID: 37049411 PMCID: PMC10096942 DOI: 10.3390/nu15071571] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/18/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Chronic alcohol use has been attributed to the development of malnutrition. This is in part due to the inhibitory effect of ethanol on the absorption of vital nutrients, including glucose, amino acids, lipids, water, vitamins, and minerals within the small intestine. Recent advances in research, along with new cutting-edge technologies, have advanced our understanding of the mechanism of ethanol's effect on intestinal nutrient absorption at the brush border membrane (BBM) of the small intestine. However, further studies are needed to delineate how ethanol consumption could have an impact on altered nutrient absorption under various disease conditions. Current research has elucidated the relationship of alcohol consumption on glucose, glutamine, vitamins B1 (thiamine), B2 (riboflavin), B9 (folate), C (ascorbic acid), selenium, iron, and zinc absorption within the small intestine. We conducted systematic computerized searches in PubMed using the following keywords: (1) "Alcohol effects on nutrient transport"; (2) "Alcohol mediated malabsorption of nutrients"; (3) "Alcohol effects on small intestinal nutrient transport"; and (4) "Alcohol mediated malabsorption of nutrients in small intestine". We included the relevant studies in this review. The main objective of this review is to marshal and analyze previously published research articles and discuss, in-depth, the understanding of ethanol's effect in modulating absorption of vital macro and micronutrients in health and disease conditions. This could ultimately provide great insights in the development of new therapeutic strategies to combat malnutrition associated with alcohol consumption.
Collapse
Affiliation(s)
- Molly Butts
- Department of Clinical and Translational Sciences, Marshall University, Huntington, WV 25755, USA
| | - Vijaya Lakshmi Sundaram
- Department of Clinical and Translational Sciences, Marshall University, Huntington, WV 25755, USA
| | - Usha Murughiyan
- Department of Clinical and Translational Sciences, Marshall University, Huntington, WV 25755, USA
| | - Alip Borthakur
- Department of Clinical and Translational Sciences, Marshall University, Huntington, WV 25755, USA
| | - Soudamani Singh
- Department of Clinical and Translational Sciences, Marshall University, Huntington, WV 25755, USA
| |
Collapse
|
8
|
Causal Associations Between Tobacco, Alcohol Use and Risk of Infectious Diseases: A Mendelian Randomization Study. Infect Dis Ther 2023; 12:965-977. [PMID: 36862322 PMCID: PMC10017901 DOI: 10.1007/s40121-023-00775-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/08/2023] [Indexed: 03/03/2023] Open
Abstract
INTRODUCTION The causal effects of smoking and alcohol use on the risk of infectious diseases are unclear, and it is hard investigate them in an observational study due to the potential confounding factors. The aim of this study was to use Mendelian randomization (MR) techniques to assess the causalities between smoking, alcohol use and risk of infectious diseases. METHODS Univariable and multivariable MR analyses were performed using genome-wide association data for the age of initiation of regular smoking (AgeSmk, N = 341,427), smoking initiation (SmkInit, N = 1,232,091), cigarettes per day (CigDay, N = 337,334), lifetime smoking (LifSmk, N = 462,690), drinks per week (DrnkWk, N = 941,280), sepsis (N = 486,484), pneumonia (N = 486,484), upper respiratory tract infection (URTI, N = 486,484) and urinary tract infection (UTI, N = 486,214) among individuals of European ancestry. Independent genetic variants that were significantly (P < 5 × 10-8) associated with each exposure were considered as instruments. The inverse-variance-weighted method was used in the primary analysis, which was followed by a series of sensitivity analyses. RESULTS Genetically predicted SmkInit was associated with an increased risk of sepsis (OR 1.353, 95% CI 1.079-1.696, P = 0.009), pneumonia (OR 1.770, 95% CI 1.464-2.141, P = 3.8 × 10-9) and UTI (OR 1.445, 95% CI 1.184-1.764, P = 3 × 10-4). Moreover, genetically predicted CigDay was associated with a higher risk of sepsis (OR 1.403, 95% CI 1.037-1.898, P = 0.028) and pneumonia (OR 1.501, 95% CI 1.167-1.930, P = 0.00156). Furthermore, genetically predicted LifSmk was associated with an increased risk of sepsis (OR 2.200, 95% CI 1.583-3.057, P = 2.63 × 10-6), pneumonia (OR 3.462, 95% CI 2.798-4.285, P = 3.28 × 10-30), URTI (OR 2.523, 95% CI 1.315-4.841, P = 0.005) and UTI (OR 2.036, 95% CI 1.585-2.616, P = 3.0 × 10-8). However, there was no significant causal evidence for genetically predicted DrnkWk in sepsis, pneumonia, URTI or UTI. Multivariable MR analyses and sensitivity analyses showed that the above results for causal association estimations were robust. CONCLUSION In this MR study, we demonstrated the causal association between tobacco smoking and risk of infectious diseases. However, no evidence was found to support causality between alcohol use and the risk of infectious diseases.
Collapse
|
9
|
Li Z, Liu Y, Wei R, Yong VW, Xue M. The Important Role of Zinc in Neurological Diseases. Biomolecules 2022; 13:28. [PMID: 36671413 PMCID: PMC9855948 DOI: 10.3390/biom13010028] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Zinc is one of the most abundant metal ions in the central nervous system (CNS), where it plays a crucial role in both physiological and pathological brain functions. Zinc promotes antioxidant effects, neurogenesis, and immune system responses. From neonatal brain development to the preservation and control of adult brain function, zinc is a vital homeostatic component of the CNS. Molecularly, zinc regulates gene expression with transcription factors and activates dozens of enzymes involved in neuronal metabolism. During development and in adulthood, zinc acts as a regulator of synaptic activity and neuronal plasticity at the cellular level. There are several neurological diseases that may be affected by changes in zinc status, and these include stroke, neurodegenerative diseases, traumatic brain injuries, and depression. Accordingly, zinc deficiency may result in declines in cognition and learning and an increase in oxidative stress, while zinc accumulation may lead to neurotoxicity and neuronal cell death. In this review, we explore the mechanisms of brain zinc balance, the role of zinc in neurological diseases, and strategies affecting zinc for the prevention and treatment of these diseases.
Collapse
Affiliation(s)
- Zhe Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou 450001, China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou 450001, China
| | - Ruixue Wei
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou 450001, China
| | - V. Wee Yong
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou 450001, China
| |
Collapse
|
10
|
Godbole NM, Chowdhury AA, Chataut N, Awasthi S. Tight Junctions, the Epithelial Barrier, and Toll-like Receptor-4 During Lung Injury. Inflammation 2022; 45:2142-2162. [PMID: 35779195 PMCID: PMC9649847 DOI: 10.1007/s10753-022-01708-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/31/2022] [Accepted: 06/13/2022] [Indexed: 11/25/2022]
Abstract
Lung epithelium is constantly exposed to the environment and is critically important for the orchestration of initial responses to infectious organisms, toxins, and allergic stimuli, and maintenance of normal gaseous exchange and pulmonary function. The integrity of lung epithelium, fluid balance, and transport of molecules is dictated by the tight junctions (TJs). The TJs are formed between adjacent cells. We have focused on the topic of the TJ structure and function in lung epithelial cells. This review includes a summary of the last twenty years of literature reports published on the disrupted TJs and epithelial barrier in various lung conditions and expression and regulation of specific TJ proteins against pathogenic stimuli. We discuss the molecular signaling and crosstalk among signaling pathways that control the TJ structure and function. The Toll-like receptor-4 (TLR4) recognizes the pathogen- and damage-associated molecular patterns released during lung injury and inflammation and coordinates cellular responses. The molecular aspects of TLR4 signaling in the context of TJs or the epithelial barrier are not fully known. We describe the current knowledge and possible networking of the TLR4-signaling with cellular and molecular mechanisms of TJs, lung epithelial barrier function, and resistance to treatment strategies.
Collapse
Affiliation(s)
- Nachiket M Godbole
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Asif Alam Chowdhury
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Neha Chataut
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Shanjana Awasthi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA.
| |
Collapse
|
11
|
Wigger GW, Bouton TC, Jacobson KR, Auld SC, Yeligar SM, Staitieh BS. The Impact of Alcohol Use Disorder on Tuberculosis: A Review of the Epidemiology and Potential Immunologic Mechanisms. Front Immunol 2022; 13:864817. [PMID: 35432348 PMCID: PMC9009367 DOI: 10.3389/fimmu.2022.864817] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
Globally, an estimated 107 million people have an alcohol use disorder (AUD) leading to 2.8 million premature deaths each year. Tuberculosis (TB) is one of the leading causes of death globally and over 8% of global TB cases are estimated to be attributable to AUD. Social determinants of health such as poverty and undernutrition are often shared among those with AUD and TB and could explain the epidemiologic association between them. However, recent studies suggest that these shared risk factors do not fully account for the increased risk of TB in people with AUD. In fact, AUD has been shown to be an independent risk factor for TB, with a linear increase in the risk for TB with increasing alcohol consumption. While few studies have focused on potential biological mechanisms underlying the link between AUD and TB, substantial overlap exists between the effects of alcohol on lung immunity and the mechanisms exploited by Mycobacterium tuberculosis (Mtb) to establish infection. Alcohol misuse impairs the immune functions of the alveolar macrophage, the resident innate immune effector in the lung and the first line of defense against Mtb in the lower respiratory tract. Chronic alcohol ingestion also increases oxidative stress in the alveolar space, which could in turn facilitate Mtb growth. In this manuscript, we review the epidemiologic data that links AUD to TB. We discuss the existing literature on the potential mechanisms by which alcohol increases the risk of TB and review the known effects of alcohol ingestion on lung immunity to elucidate other mechanisms that Mtb may exploit. A more in-depth understanding of the link between AUD and TB will facilitate the development of dual-disease interventions and host-directed therapies to improve lung health and long-term outcomes of TB.
Collapse
Affiliation(s)
- Gregory W Wigger
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Tara C Bouton
- Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Karen R Jacobson
- Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Sara C Auld
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States.,Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Samantha M Yeligar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States.,Atlanta VA Medical Center, Atlanta, GA, United States
| | - Bashar S Staitieh
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
12
|
Micronutrient Improvement of Epithelial Barrier Function in Various Disease States: A Case for Adjuvant Therapy. Int J Mol Sci 2022; 23:ijms23062995. [PMID: 35328419 PMCID: PMC8951934 DOI: 10.3390/ijms23062995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
The published literature makes a very strong case that a wide range of disease morbidity associates with and may in part be due to epithelial barrier leak. An equally large body of published literature substantiates that a diverse group of micronutrients can reduce barrier leak across a wide array of epithelial tissue types, stemming from both cell culture as well as animal and human tissue models. Conversely, micronutrient deficiencies can exacerbate both barrier leak and morbidity. Focusing on zinc, Vitamin A and Vitamin D, this review shows that at concentrations above RDA levels but well below toxicity limits, these micronutrients can induce cell- and tissue-specific molecular-level changes in tight junctional complexes (and by other mechanisms) that reduce barrier leak. An opportunity now exists in critical care—but also medical prophylactic and therapeutic care in general—to consider implementation of select micronutrients at elevated dosages as adjuvant therapeutics in a variety of disease management. This consideration is particularly pointed amidst the COVID-19 pandemic.
Collapse
|
13
|
Tajbakhsh A, Gheibihayat SM, Mortazavi D, Medhati P, Rostami B, Savardashtaki A, Momtazi-Borojeni AA. The Effect of Cigarette Smoke Exposure on Efferocytosis in Chronic Obstructive Pulmonary Disease; Molecular Mechanisms and Treatment Opportunities. COPD 2021; 18:723-736. [PMID: 34865568 DOI: 10.1080/15412555.2021.1978419] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Cigarette smoking-related inflammation, cellular stresses, and tissue destruction play a key role in lung disease, such as chronic obstructive pulmonary disease (COPD). Notably, augmented apoptosis and impaired clearance of apoptotic cells, efferocytosis, contribute to the chronic inflammatory response and tissue destruction in patients with COPD. Of note, exposure to cigarette smoke can impair alveolar macrophages efferocytosis activity, which leads to secondary necrosis formation and tissue inflammation. A better understanding of the processes behind the effect of cigarette smoke on efferocytosis concerning lung disorders can help to design more efficient treatment approaches and also delay the development of lung disease, such as COPD. To this end, we aimed to seek mechanisms underlying the impairing effect of cigarette smoke on macrophages-mediated efferocytosis in COPD. Further, available therapeutic opportunities for restoring efferocytosis activity and ameliorating respiratory tract inflammation in smokers with COPD were also discussed.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Deniz Mortazavi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Pourya Medhati
- Student research committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behrouz Rostami
- Health & Treatment Center of Rostam, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Abbas Momtazi-Borojeni
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Iran's National Elites Foundation, Tehran, Iran
| |
Collapse
|
14
|
Wetzel TJ, Wyatt TA. Dual Substance Use of Electronic Cigarettes and Alcohol. Front Physiol 2020; 11:593803. [PMID: 33224040 PMCID: PMC7667127 DOI: 10.3389/fphys.2020.593803] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 09/29/2020] [Indexed: 11/25/2022] Open
Abstract
Electronic cigarettes (ECs) are a modern nicotine delivery system that rapidly grew in widespread use, particularly in younger populations. Given the long history of the comorbidity of alcohol and nicotine use, the rising prevalence of ECs raises the question as to their role in the consumption of alcohol. Of the numerous models of ECs available, JUUL is the most popular. This narrative review aims to determine current trends in literature regarding the relationship between EC and alcohol dual use, as well as hypothesize potential pathogenic tissue damage and summarize areas for future study, including second-hand vapor exposure and calling for standardization among studies. In summary, EC users are more likely to participate in hazardous drinking and are at higher risk for alcohol use disorder (AUD). We surmise the pathogenic damage of dual use may exhibit an additive effect, particularly in pathogen clearance from the lungs, increased inflammation and decreased immune response, physical damage to epithelial cells, and exacerbation of chronic obstructive pulmonary disease (COPD)-like illnesses. A better understanding of pathogenic damages is critical to understand the risks placed on dual users when exposed to respiratory pathogens, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
Collapse
Affiliation(s)
- Tanner J Wetzel
- Department of Environmental, Agricultural and Occupational Health, University of Nebraska Medical Center, Omaha, NE, United States
| | - Todd A Wyatt
- Department of Environmental, Agricultural and Occupational Health, University of Nebraska Medical Center, Omaha, NE, United States.,Pulmonary, Critical Care, and Sleep, University of Nebraska Medical Center, Omaha, NE, United States.,VA Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| |
Collapse
|
15
|
Wessels I, Pupke JT, von Trotha KT, Gombert A, Himmelsbach A, Fischer HJ, Jacobs MJ, Rink L, Grommes J. Zinc supplementation ameliorates lung injury by reducing neutrophil recruitment and activity. Thorax 2020; 75:253-261. [PMID: 31915307 DOI: 10.1136/thoraxjnl-2019-213357] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 11/25/2019] [Accepted: 12/06/2019] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Zinc is well known for its anti-inflammatory effects, including regulation of migration and activity of polymorphonuclear neutrophils (PMN). Zinc deficiency is associated with inflammatory diseases such as acute lung injury (ALI). As deregulated neutrophil recruitment and their hyper-activation are hallmarks of ALI, benefits of zinc supplementation on the development of lipopolysaccharides (LPS)-induced ALI were tested. METHODS 64 C57Bl/6 mice, split into eight groups, were injected with 30 µg zinc 24 hours before exposure to aerosolised LPS for 4 hours. Zinc homoeostasis was characterised measuring serum and lung zinc concentrations as well as metallothionein-1 expression. Recruitment of neutrophils to alveolar, interstitial and intravascular space was assessed using flow cytometry. To determine the extent of lung damage, permeability and histological changes and the influx of protein into the bronchoalveolar lavage fluid were measured. Inflammatory status and PMN activity were evaluated via tumour necrosis factor α levels and formation of neutrophil extracellular traps. The effects of zinc supplementation prior to LPS stimulation on activation of primary human granulocytes and integrity of human lung cell monolayers were assessed as well. RESULTS Injecting zinc 24 hours prior to LPS-induced ALI indeed significantly decreased the recruitment of neutrophils to the lungs and prevented their hyperactivity and thus lung damage was decreased. Results from in vitro investigations using human cells suggest the transferability of the finding to human disease, which remains to be tested in more detail. CONCLUSION Zinc supplementation attenuated LPS-induced lung injury in a murine ALI model. Thus, the usage of zinc-based strategies should be considered to prevent detrimental consequences of respiratory infection and lung damage in risk groups.
Collapse
Affiliation(s)
- Inga Wessels
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Johanna Theresa Pupke
- Department of Vascular Surgery, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Klaus-Thilo von Trotha
- Department of Vascular Surgery, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany.,Department of Vascular Surgery, Marienhospital Aachen, Aachen, Nordrhein-Westfalen, Germany
| | - Alexander Gombert
- Department of Vascular Surgery, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Anika Himmelsbach
- Department of Cardiology, Medical Clinic I, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Henrike Josephine Fischer
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Michael J Jacobs
- Department of Vascular Surgery, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Lothar Rink
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Jochen Grommes
- Department of Vascular Surgery, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany .,Department of Vascular Surgery, Rhein-Maas Klinikum GmbH, Wurselen, Nordrhein-Westfalen, Germany
| |
Collapse
|
16
|
Saribal D, Hocaoglu-Emre FS, Karaman F, Mırsal H, Akyolcu MC. Trace Element Levels and Oxidant/Antioxidant Status in Patients with Alcohol Abuse. Biol Trace Elem Res 2020; 193:7-13. [PMID: 30805875 DOI: 10.1007/s12011-019-01681-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/17/2019] [Indexed: 12/18/2022]
Abstract
Alcohol abuse is a well-known cause of imbalance in trace element levels and oxidant/antioxidant status of individuals with long time consumption. However, the levels of these parameters in the patients on the early stages of alcohol dependence without liver damage differ on various studies. The aim of our study was to measure the levels of trace elements in the serum and oxidative/antioxidative system members in the red blood cells (RBC) of early-stage alcoholic individuals and compare with control subjects. Our study included 21 male patients recently hospitalized for alcohol abuse and 25 healthy non-abusing male controls. Levels of Fe, Zn, and Cu in the serum and MDA, SOD, CAT, and GSH in the red blood cells (RBC) of the subjects were measured. Fe, Zn, and Cu levels were lower in the study group when compared to the controls. Levels of lipid peroxidation marker MDA was high, whereas the activities of antioxidant enzymes SOD and CAT were decreased in our study group. However, levels of GSH, an antioxidant compound were higher in the alcohol abuse group. RBC SOD levels were positively correlated with Fe, Cu, Zn, and CAT. There was a positive correlation between Fe-Cu, Zn-Fe, Zn-Cu, CAT-Zn, and CAT-SOD. MDA was negatively correlated with Fe, Zn, SOD, and CAT. The results obtained from present study indicate that high levels of alcohol intake are related with increased oxidative damage and decreased levels of antioxidant enzymes and trace elements. Additionally, antioxidant compensation mechanisms are still on process in the early stages of chronic alcohol exposure.
Collapse
Affiliation(s)
- Devrim Saribal
- Department of Biophysics, Istanbul University Cerrahpaşa Medical Faculty, Istanbul, Turkey
| | | | - Fulya Karaman
- Department of Physiology, Çanakkale Onsekiz Mart University Medical Faculty, Çanakkale, Turkey
| | - Hasan Mırsal
- Center for Treatment of Substance Abuse, Balikli Rum Hospital, Istanbul, Turkey
| | - Mehmet Can Akyolcu
- Department of Biophysics, Girne American University Medical School, Kyrenia, Cyprus
| |
Collapse
|
17
|
Johnson JK, Harris FL, Ping XD, Gauthier TW, Brown LAS. Role of zinc insufficiency in fetal alveolar macrophage dysfunction and RSV exacerbation associated with fetal ethanol exposure. Alcohol 2019; 80:5-16. [PMID: 30580016 DOI: 10.1016/j.alcohol.2018.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/19/2018] [Accepted: 11/30/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND We previously reported that maternal alcohol use significantly increases the risk of sepsis in premature and term newborns. In the mouse, fetal ethanol exposure results in an immunosuppressed phenotype for the alveolar macrophage (AM) and decreases bacterial phagocytosis. In pregnant mice, ethanol decreased AM zinc homeostasis, which contributed to immunosuppression and impaired AM phagocytosis. In this study, we explored whether ethanol-induced zinc insufficiency extended to the pup AMs and contributed to immunosuppression and exacerbated viral lung infections. METHODS C57BL/6 female mice were fed a liquid diet with 25% ethanol-derived calories or pair-fed a control diet with 25% of calories as maltose-dextrin. Some pup AMs were treated in vitro with zinc acetate before measuring zinc pools or transporter expression and bacteria phagocytosis. Some dams were fed additional zinc supplements in the ethanol or control diets, and then we assessed pup AM zinc pools, zinc transporters, and the immunosuppressant TGFβ1. On postnatal day 10, some pups were given intranasal saline or respiratory syncytial virus (RSV), and then AM RSV phagocytosis and the RSV burden in the airway lining fluid were assessed. RESULTS Fetal ethanol exposure decreased pup AM zinc pools, zinc transporter expression, and bacterial clearance, but in vitro zinc treatments reversed these alterations. In addition, the expected ethanol-induced increase in TGFβ1 and immunosuppression were associated with decreased RSV phagocytosis and exacerbated RSV infections. However, additional maternal zinc supplements blocked the ethanol-induced perturbations in the pup AM zinc homeostasis and TGFβ1 immunosuppression, thereby improving RSV phagocytosis and attenuating the RSV burden in the lung. CONCLUSION These studies suggest that, despite normal maternal dietary zinc intake, in utero alcohol exposure results in zinc insufficiency, which contributes to compromised neonatal AM immune functions, thereby increasing the risk of bacterial and viral infections.
Collapse
|
18
|
Slovinsky WS, Romero F, Sales D, Shaghaghi H, Summer R. The involvement of GM-CSF deficiencies in parallel pathways of pulmonary alveolar proteinosis and the alcoholic lung. Alcohol 2019; 80:73-79. [PMID: 31229291 PMCID: PMC6592783 DOI: 10.1016/j.alcohol.2018.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/09/2018] [Accepted: 07/12/2018] [Indexed: 12/24/2022]
Abstract
Chronic alcohol consumption renders the lung more susceptible to infections by disrupting essential alveolar macrophage functions. Emerging evidence suggests that these functional deficits are due, in part, to a suppression of GM-CSF signaling, which is believed to compromise monocyte growth and maturation in the lung. However, in addition to controlling monocyte behaviors, GM-CSF also regulates surfactant homeostasis. For example, mice with targeted deletion of the gene for GM-CSF accumulate large amounts of surfactant phospholipids in their lungs. Moreover, decreased GM-CSF signaling in humans has been linked to the development of pulmonary alveolar proteinosis (PAP), a rare disorder in which surfactant lipids and proteins accumulate in alveolar macrophages and the lung exhibits enhanced susceptibility to infection. Consistent with parallel mechanisms in the PAP and alcoholic lung, we have recently reported that levels of intrapulmonary lipids, specifically triglycerides and free fatty acids, are increased in BAL fluid, whole lung digests and alveolar macrophages of chronically alcohol exposed rats. Additionally, we showed that uptake of saturated fatty acids alone could induce phenotypic and functional changes in alveolar macrophages that mimicked those in the alcohol-exposed rat and human lung. Herein, we discuss the role of GM-CSF in surfactant homeostasis and highlight the evidence that links decreased GM-CSF signaling to alveolar macrophage dysfunction in both the PAP and alcohol-exposed lung. Moreover, we discuss how lipid accumulation itself might contribute to altering alveolar macrophage function and propose how targeting these mechanisms could be employed for reducing the susceptibility to pulmonary infections in alcoholics.
Collapse
Affiliation(s)
- William S Slovinsky
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Freddy Romero
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Dominic Sales
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Hoora Shaghaghi
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Ross Summer
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| |
Collapse
|
19
|
Macrophages exposed to HIV viral protein disrupt lung epithelial cell integrity and mitochondrial bioenergetics via exosomal microRNA shuttling. Cell Death Dis 2019; 10:580. [PMID: 31371699 PMCID: PMC6675785 DOI: 10.1038/s41419-019-1803-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/01/2019] [Accepted: 07/03/2019] [Indexed: 12/14/2022]
Abstract
Antiretroviral therapy extends survival but does not eliminate HIV from its cellular reservoirs. Between immune and stromal cells in the tissue microenvironment, a dynamic intercellular communication might influence host viral immune responses via intercellular transfer of extracellular vehicles (EVs) (microvesicles, exosome, or apoptotic bodies). It is increasingly recognized that HIV-infected macrophage-secreted nucleotide-rich exosomes might play a critical role in mediating communication between macrophages and other structural cells; however, molecular mechanisms underlying cell–cell crosstalk remain unknown. Here we show that HIV-1-infected macrophages and HIV-1 proteins Tat or gp120-treated macrophages express high levels of microRNAs, including miR-23a and miR-27a. Identical miRNAs expression patterns were detected in macrophage-secreted exosomes isolated from bronchoalveolar lavage fluid of HIV transgenic rats. Tat-treated macrophage-derived exosomal miR-23a attenuated posttranscriptional modulation of key tight junction protein zonula occludens (ZO-1) 3′-UTR in epithelial cells. In parallel, exosomal miR-27a released from Tat-treated macrophages altered the mitochondrial bioenergetics of recipient lung epithelial cells by targeting peroxisome proliferator-activated receptor gamma (PPARγ), while simultaneously stimulating glycolysis. Together, exosomal miRNAs shuttle from macrophages to epithelial cells and thereby explain in part HIV-mediated lung epithelial barrier dysfunction. These studies suggest that targeting miRNAs may be of therapeutic value to enhance lung health in HIV.
Collapse
|
20
|
Zalewski PD, Beltrame JF, Wawer AA, Abdo AI, Murgia C. Roles for endothelial zinc homeostasis in vascular physiology and coronary artery disease. Crit Rev Food Sci Nutr 2018; 59:3511-3525. [PMID: 29999409 DOI: 10.1080/10408398.2018.1495614] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The discovery of the roles of nitric oxide (NO) in cardiovascular signaling has led to a revolution in the understanding of cardiovascular disease. A new perspective to this story involving zinc (Zn) is emerging. Zn and its associated Zn transporter proteins are important for the integrity and functions of both the large conduit vessels and the microvascular resistance vessels. The Zn and NO pathways are tightly coordinated. Zn ions are required for the dimerization of endothelial nitric oxide synthase and subsequent generation of NO while generation of NO leads to a rapid mobilization of endothelial Zn stores. Labile Zn may mediate important downstream actions of NO including vascular cytoprotection and vasodilation. Several vascular disease risk factors (including aging, smoking and diabetes) interfere with Zn homeostatic mechanisms and both hypozincaemia and Zn transporter protein abnormalities are linked to atherosclerosis and microvascular disease. Some vegetarian diets and long-term use of certain anti-hypertensives may also impact on Zn status. The available evidence supports the existence of a Zn regulatory pathway in the vascular wall that is coupled to the generation and actions of NO and which is compromised in Zn deficiency with consequent implications for the pathogenesis and therapy of vascular disease.
Collapse
Affiliation(s)
- P D Zalewski
- Discipline of Medicine, University of Adelaide, The Queen Elizabeth Hospital and the Basil Hetzel Institute for Translational Health Research, Woodville, South Australia
| | - J F Beltrame
- Discipline of Medicine, University of Adelaide, The Queen Elizabeth Hospital and the Basil Hetzel Institute for Translational Health Research, Woodville, South Australia
| | - A A Wawer
- Discipline of Medicine, University of Adelaide, The Queen Elizabeth Hospital and the Basil Hetzel Institute for Translational Health Research, Woodville, South Australia
| | - A I Abdo
- Discipline of Medicine, University of Adelaide, The Queen Elizabeth Hospital and the Basil Hetzel Institute for Translational Health Research, Woodville, South Australia
| | - C Murgia
- Department of Nutrition, Dietetics and Food, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
21
|
Simou E, Leonardi-Bee J, Britton J. The Effect of Alcohol Consumption on the Risk of ARDS: A Systematic Review and Meta-Analysis. Chest 2018; 154:58-68. [PMID: 29288645 PMCID: PMC6045784 DOI: 10.1016/j.chest.2017.11.041] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/13/2017] [Accepted: 11/29/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND To conduct a systematic review and meta-analysis evaluating the association between alcohol consumption and the risk of ARDS in adults. METHODS Medline, EMBASE and Web of Science were searched to identify observational studies evaluating the association between prior alcohol intake and the occurrence of ARDS among adults, published between 1985 and 2015 and with no language restriction. Reference lists were also screened. Demographic baseline data were extracted independently by two reviewers and random-effects meta-analyses were used to estimate pooled effect sizes with 95% confidence intervals. Subgroup analyses were used to explore heterogeneity. RESULTS Seventeen observational studies (177,674 people) met the inclusion criteria. Meta-analysis of 13 studies showed that any measure of high relative to low alcohol consumption was associated with a significantly increased risk of ARDS (OR, 1.89; 95% CI, 1.45-2.48; I2 = 48%; 13 studies); no evidence of publication bias was seen (P = .150). Sensitivity analyses indicated that this association was attributable primarily to an effect of a history of alcohol abuse (OR, 1.90; 95% CI, 1.40-2.60; 10 studies). Also, subgroup analyses identified that heterogeneity was explained by predisposing condition (trauma, sepsis/septic shock, pneumonia; P = .003). CONCLUSIONS Chronic high alcohol consumption significantly increases the risk of ARDS. This finding suggests that patients admitted to hospital should be screened for chronic alcohol use.
Collapse
Affiliation(s)
- Evangelia Simou
- UK Centre for Tobacco and Alcohol Studies, Division of Epidemiology and Public Health, University of Nottingham, Nottingham, UK.
| | - Jo Leonardi-Bee
- UK Centre for Tobacco and Alcohol Studies, Division of Epidemiology and Public Health, University of Nottingham, Nottingham, UK
| | - John Britton
- UK Centre for Tobacco and Alcohol Studies, Division of Epidemiology and Public Health, University of Nottingham, Nottingham, UK
| |
Collapse
|
22
|
Pulmonary Alveolar Proteinosis in Setting of Inhaled Toxin Exposure and Chronic Substance Abuse. Case Rep Pulmonol 2018; 2018:5202173. [PMID: 29607238 PMCID: PMC5828087 DOI: 10.1155/2018/5202173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/15/2017] [Accepted: 09/20/2017] [Indexed: 11/17/2022] Open
Abstract
Pulmonary alveolar proteinosis (PAP) is a rare lung disorder in which defects in alveolar macrophage maturation or function lead to the accumulation of proteinaceous surfactant in alveolar space, resulting in impaired gas exchange and hypoxemia. PAP is categorized into three types: hereditary, autoimmune, and secondary. We report a case of secondary PAP in a 47-year-old man, whose risk factors include occupational exposure to inhaled toxins, especially aluminum dust, the use of anabolic steroids, and alcohol abuse, which in mice leads to alveolar macrophage dysfunction through a zinc-dependent mechanism that inhibits granulocyte macrophage-colony stimulating factor (GM-CSF) receptor signalling. Although the rarity and vague clinical presentation of PAP can pose diagnostic challenges, clinician awareness of PAP risk factors may facilitate the diagnostic process and lead to more prompt treatment.
Collapse
|
23
|
Zinc deficiency as a mediator of toxic effects of alcohol abuse. Eur J Nutr 2017; 57:2313-2322. [PMID: 29177978 DOI: 10.1007/s00394-017-1584-y] [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] [Received: 08/08/2017] [Accepted: 11/20/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To review data on the role of ethanol-induced alteration of Zn homeostasis in mediation of adverse effects of alcohol abuse. METHODS The scholarly published articles on the association between Zn metabolism and alcohol-associated disorders (liver, brain, lung, gut dysfunction, and fetal alcohol syndrome) have been reviewed. RESULTS It is demonstrated that alcohol-induced modulation of zinc transporters results in decreased Zn levels in lungs, liver, gut, and brain. Zn deficiency in the gut results in increased gut permeability, ultimately leading to endotoxemia and systemic inflammation. Similarly, Zn deficiency in lung epithelia and alveolar macrophages decreases lung barrier function resulting in respiratory distress syndrome. In turn, increased endotoxemia significantly contributes to proinflammatory state in alcoholic liver disease. Finally, impaired gut and liver functions may play a significant role in alcoholic brain damage, being associated with both increased proinflammatory signaling and accumulation of neurotoxic metabolites. It is also hypothesized that ethanol-induced Zn deficiency may interfere with neurotransmission. Similar changes may take place in the fetus as a result of impaired placental zinc transfer, maternal zinc deficiency, or maternal Zn sequestration, resulting in fetal alcoholic syndrome. Therefore, alcoholic Zn deficiency not only mediates the adverse effects of ethanol exposure, but also provides an additional link between different alcohol-induced disorders. CONCLUSIONS Generally, current findings suggest that assessment of Zn status could be used as a diagnostic marker of metabolic disturbances in alcohol abuse, whereas modulation of Zn metabolism may be a potential tool in the treatment of alcohol-associated disorders.
Collapse
|
24
|
Gal Y, Mazor O, Falach R, Sapoznikov A, Kronman C, Sabo T. Treatments for Pulmonary Ricin Intoxication: Current Aspects and Future Prospects. Toxins (Basel) 2017; 9:E311. [PMID: 28972558 PMCID: PMC5666358 DOI: 10.3390/toxins9100311] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 09/26/2017] [Accepted: 09/29/2017] [Indexed: 12/13/2022] Open
Abstract
Ricin, a plant-derived toxin originating from the seeds of Ricinus communis (castor beans), is one of the most lethal toxins known, particularly if inhaled. Ricin is considered a potential biological threat agent due to its high availability and ease of production. The clinical manifestation of pulmonary ricin intoxication in animal models is closely related to acute respiratory distress syndrome (ARDS), which involves pulmonary proinflammatory cytokine upregulation, massive neutrophil infiltration and severe edema. Currently, the only post-exposure measure that is effective against pulmonary ricinosis at clinically relevant time-points following intoxication in pre-clinical studies is passive immunization with anti-ricin neutralizing antibodies. The efficacy of this antitoxin treatment depends on antibody affinity and the time of treatment initiation within a limited therapeutic time window. Small-molecule compounds that interfere directly with the toxin or inhibit its intracellular trafficking may also be beneficial against ricinosis. Another approach relies on the co-administration of antitoxin antibodies with immunomodulatory drugs, thereby neutralizing the toxin while attenuating lung injury. Immunomodulators and other pharmacological-based treatment options should be tailored according to the particular pathogenesis pathways of pulmonary ricinosis. This review focuses on the current treatment options for pulmonary ricin intoxication using anti-ricin antibodies, disease-modifying countermeasures, anti-ricin small molecules and their various combinations.
Collapse
Affiliation(s)
- Yoav Gal
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Ohad Mazor
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Reut Falach
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Anita Sapoznikov
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Chanoch Kronman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Tamar Sabo
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| |
Collapse
|
25
|
Hoyt LR, Randall MJ, Ather JL, DePuccio DP, Landry CC, Qian X, Janssen-Heininger YM, van der Vliet A, Dixon AE, Amiel E, Poynter ME. Mitochondrial ROS induced by chronic ethanol exposure promote hyper-activation of the NLRP3 inflammasome. Redox Biol 2017; 12:883-896. [PMID: 28463821 PMCID: PMC5413213 DOI: 10.1016/j.redox.2017.04.020] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/10/2017] [Accepted: 04/10/2017] [Indexed: 02/07/2023] Open
Abstract
Alcohol use disorders are common both in the United States and globally, and are associated with a variety of co-morbid, inflammation-linked diseases. The pathogenesis of many of these ailments are driven by the activation of the NLRP3 inflammasome, a multi-protein intracellular pattern recognition receptor complex that facilitates the cleavage and secretion of the pro-inflammatory cytokines IL-1β and IL-18. We hypothesized that protracted exposure of leukocytes to ethanol would amplify inflammasome activation, which would help to implicate mechanisms involved in diseases associated with both alcoholism and aberrant NLRP3 inflammasome activation. Here we show that long-term ethanol exposure of human peripheral blood mononuclear cells and a mouse macrophage cell line (J774) amplifies IL-1β secretion following stimulation with NLRP3 agonists, but not with AIM2 or NLRP1b agonists. The augmented NRLP3 activation was mediated by increases in iNOS expression and NO production, in conjunction with increases in mitochondrial membrane depolarization, oxygen consumption rate, and ROS generation in J774 cells chronically exposed to ethanol (CE cells), effects that could be inhibited by the iNOS inhibitor SEITU, the NO scavenger carboxy-PTIO, and the mitochondrial ROS scavenger MitoQ. Chronic ethanol exposure did not alter K+ efflux or Zn2+ homeostasis in CE cells, although it did result in a lower intracellular concentration of NAD+. Prolonged administration of acetaldehyde, the product of alcohol dehydrogenase (ADH) mediated metabolism of ethanol, mimicked chronic ethanol exposure, whereas ADH inhibition prevented ethanol-induced IL-1β hypersecretion. Together, these results indicate that increases in iNOS and mitochondrial ROS production are critical for chronic ethanol-induced IL-1β hypersecretion, and that protracted exposure to the products of ethanol metabolism are probable mediators of NLRP3 inflammasome hyperactivation.
Chronic ethanol exposure amplifies NLRP3 inflammasome-induced IL-1β secretion. NO and mitochondrial ROS mediate chronic ethanol-augmented IL-1β secretion. Alcohol dehydrogenase-generated metabolites cause NLRP3 inflammasome over-activation.
Collapse
Affiliation(s)
- Laura R Hoyt
- Vermont Lung Center, University of Vermont, Burlington, VT 05405, USA; Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Matthew J Randall
- Vermont Lung Center, University of Vermont, Burlington, VT 05405, USA; Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Jennifer L Ather
- Vermont Lung Center, University of Vermont, Burlington, VT 05405, USA; Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Daniel P DePuccio
- Department of Chemistry, University of Vermont, Burlington, VT 05405, USA
| | - Christopher C Landry
- Department of Chemistry, University of Vermont, Burlington, VT 05405, USA; Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, USA
| | - Xi Qian
- Vermont Lung Center, University of Vermont, Burlington, VT 05405, USA; Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Yvonne M Janssen-Heininger
- Vermont Lung Center, University of Vermont, Burlington, VT 05405, USA; Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT 05405, USA; Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, USA
| | - Albert van der Vliet
- Vermont Lung Center, University of Vermont, Burlington, VT 05405, USA; Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT 05405, USA; Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, USA
| | - Anne E Dixon
- Vermont Lung Center, University of Vermont, Burlington, VT 05405, USA; Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Eyal Amiel
- Department of Medical Laboratory and Radiation Sciences, University of Vermont, Burlington, VT 05405, USA; Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, USA
| | - Matthew E Poynter
- Vermont Lung Center, University of Vermont, Burlington, VT 05405, USA; Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405, USA; Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, USA.
| |
Collapse
|
26
|
Mehta AJ, Guidot DM. Alcohol and the Lung. Alcohol Res 2017; 38:243-254. [PMID: 28988576 PMCID: PMC5513688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Among the many organ systems affected by harmful alcohol use, the lungs are particularly susceptible to infections and injury. The mechanisms responsible for rendering people with alcohol use disorder (AUD) vulnerable to lung damage include alterations in host defenses of the upper and lower airways, disruption of alveolar epithelial barrier integrity, and alveolar macrophage immune dysfunction. Collectively, these derangements encompass what has been termed the "alcoholic lung" phenotype. Alcohol-related reductions in antioxidant levels also may contribute to lung disease in people with underlying AUD. In addition, researchers have identified several regulatory molecules that may play crucial roles in the alcohol-induced disease processes. Although there currently are no approved therapies to combat the detrimental effects of chronic alcohol consumption on the respiratory system, these molecules may be potential therapeutic targets to guide future investigation.
Collapse
Affiliation(s)
- Ashish J Mehta
- Ashish J. Mehta, M.D., is an Assistant Professor of Medicine in the Department of Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Emory University, Atlanta, Georgia, and a Staff Physician at the Atlanta VA Medical Center, Decatur, Georgia. David M. Guidot, M.D., is a Professor of Medicine in the Department of Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Emory University, Atlanta, Georgia, and a Staff Physician at the Atlanta VA Medical Center, Decatur, Georgia
| | - David M Guidot
- Ashish J. Mehta, M.D., is an Assistant Professor of Medicine in the Department of Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Emory University, Atlanta, Georgia, and a Staff Physician at the Atlanta VA Medical Center, Decatur, Georgia. David M. Guidot, M.D., is a Professor of Medicine in the Department of Medicine, Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Emory University, Atlanta, Georgia, and a Staff Physician at the Atlanta VA Medical Center, Decatur, Georgia
| |
Collapse
|
27
|
Crowell KT, Phillips BE, Kelleher SL, Soybel DI, Lang CH. Immune and metabolic responses in early and late sepsis during mild dietary zinc restriction. J Surg Res 2016; 210:47-58. [PMID: 28457340 DOI: 10.1016/j.jss.2016.10.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/22/2016] [Accepted: 10/26/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND Mild dietary zinc (Zn) deficiency is widespread in human populations, but its influence on recovery after acute illness is poorly understood. In a mouse model of abdominal sepsis (cecal ligation puncture), systemic immune responses and liver metabolism were monitored in early (24 h) and late (5 d) phases, under control conditions and during mild dietary Zn restriction. METHODS Mice were fed diets adequate or marginally deficient (ZM) in Zn (30 versus 10 mg zinc/kg diet) for 4 wk, before undergoing laparotomy alone (nonseptic control) or cecal ligation puncture (septic). RESULTS Among nonseptic mice, the ZM state was not associated with differences in inflammation or metabolic responses. Among septic mice, mortality did not differ between the zinc adequate and ZM groups. In the early phase, the ZM state amplified increases in plasma interleukin (IL) 6, tumor necrosis factor alpha, and IL-10, while dampening the interferon gamma response. In the late phase, subtle but significant ZM-associated increases were observed in plasma IL-5 and interferon gamma levels and hepatic protein synthesis, the latter of which appeared to be mammalian target of rapamycin independent and was associated with increased hepatic tumor necrosis factor alpha messenger RNA content. CONCLUSIONS Without increasing mortality, the ZM state is associated with a more disordered acute systemic inflammatory response and persistence or enhancement of acute phase responses within the liver parenchyma.
Collapse
Affiliation(s)
- Kristen T Crowell
- Department of Surgery, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Brett E Phillips
- Department of Surgery, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Shannon L Kelleher
- Department of Surgery, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania
| | - David I Soybel
- Department of Surgery, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Department of Nutritional Sciences, Pennsylvania State University, University Park, Pennsylvania
| | - Charles H Lang
- Department of Surgery, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania.
| |
Collapse
|
28
|
Yeligar SM, Chen MM, Kovacs EJ, Sisson JH, Burnham EL, Brown LAS. Alcohol and lung injury and immunity. Alcohol 2016; 55:51-59. [PMID: 27788778 DOI: 10.1016/j.alcohol.2016.08.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 07/07/2016] [Accepted: 08/24/2016] [Indexed: 02/06/2023]
Abstract
Annually, excessive alcohol use accounts for more than $220 billion in economic costs and 80,000 deaths, making excessive alcohol use the third leading lifestyle-related cause of death in the US. Patients with an alcohol-use disorder (AUD) also have an increased susceptibility to respiratory pathogens and lung injury, including a 2-4-fold increased risk of acute respiratory distress syndrome (ARDS). This review investigates some of the potential mechanisms by which alcohol causes lung injury and impairs lung immunity. In intoxicated individuals with burn injuries, activation of the gut-liver axis drives pulmonary inflammation, thereby negatively impacting morbidity and mortality. In the lung, the upper airway is the first checkpoint to fail in microbe clearance during alcohol-induced lung immune dysfunction. Brief and prolonged alcohol exposure drive different post-translational modifications of novel proteins that control cilia function. Proteomic approaches are needed to identify novel alcohol targets and post-translational modifications in airway cilia that are involved in alcohol-dependent signal transduction pathways. When the upper airway fails to clear inhaled pathogens, they enter the alveolar space where they are primarily cleared by alveolar macrophages (AM). With chronic alcohol ingestion, oxidative stress pathways in the AMs are stimulated, thereby impairing AM immune capacity and pathogen clearance. The epidemiology of pneumococcal pneumonia and AUDs is well established, as both increased predisposition and illness severity have been reported. AUD subjects have increased susceptibility to pneumococcal pneumonia infections, which may be due to the pro-inflammatory response of AMs, leading to increased oxidative stress.
Collapse
Affiliation(s)
- Samantha M Yeligar
- Department of Medicine, Emory University and Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
| | - Michael M Chen
- Burn and Shock Trauma Research Institute, Alcohol Research Program, Integrative Cell Biology Program, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, USA
| | - Elizabeth J Kovacs
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Joseph H Sisson
- Pulmonary, Critical Care, Sleep and Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ellen L Burnham
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Lou Ann S Brown
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
29
|
Sun Z, Yang L, Chen KF, Chen GW, Peng YP, Chen JK, Suo G, Yu J, Wang WC, Lin CH. Nano zerovalent iron particles induce pulmonary and cardiovascular toxicity in an in vitro human co-culture model. Nanotoxicology 2016; 10:881-90. [DOI: 10.3109/17435390.2015.1133861] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Zhelin Sun
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China,
- School of Life Sciences, Shanghai University, Shanghai, China,
| | - Lingyan Yang
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China,
| | - Ku-Fan Chen
- Department of Civil Engineering, National Chi Nan University, Nantou, Taiwan,
| | - Guan-Wen Chen
- Department of Food and Beverage Management, Taipei College of Maritime Technology, Taipei, Taiwan,
| | - Yen-Ping Peng
- Department of Environmental Science and Engineering, Tunghai University, Taichung, Taiwan,
| | - Jen-Kun Chen
- Institute of Biomedical Engineering & Nanomedicine, National Health Research Institutes, Miaoli, Taiwan,
| | - Guangli Suo
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China,
| | - Jiantao Yu
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China,
| | - Wen-Cheng Wang
- Research Center for Environmental Changes, Academia Sinica, Taipei, Taiwan, and
| | - Chia-Hua Lin
- Department of Biotechnology, National Formosa University, Yunlin, Taiwan
| |
Collapse
|
30
|
Mehta AJ. Alcoholism and critical illness: A review. World J Crit Care Med 2016; 5:27-35. [PMID: 26855891 PMCID: PMC4733453 DOI: 10.5492/wjccm.v5.i1.27] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 10/07/2015] [Accepted: 12/04/2015] [Indexed: 02/06/2023] Open
Abstract
Alcohol is the most commonly used and abused drug in the world, and alcohol use disorders pose a tremendous burden to healthcare systems around the world. The lifetime prevalence of alcohol abuse in the United States is estimated to be around 18%, and the economic consequences of these disorders are staggering. Studies on hospitalized patients demonstrate that about one in four patients admitted to critical care units will have alcohol-related issues, and unhealthy alcohol consumption is responsible for numerous clinical problems encountered in intensive care unit (ICU) settings. Patients with alcohol use disorders are not only predisposed to developing withdrawal syndromes and other conditions that often require intensive care, they also experience a considerably higher rate of complications, longer ICU and hospital length of stay, greater resource utilization, and significantly increased mortality compared to similar critically ill patients who do not abuse alcohol. Specific disorders seen in the critical care setting that are impacted by alcohol abuse include delirium, pneumonia, acute respiratory distress syndrome, sepsis, gastrointestinal hemorrhage, trauma, and burn injuries. Despite the substantial burden of alcohol-induced disease in these settings, critical care providers often fail to identify individuals with alcohol use disorders, which can have significant implications for this vulnerable population and delay important clinical interventions.
Collapse
|
31
|
Phillips BE, Geletzke AK, Smith PB, Podany AB, Chacon A, Kelleher SL, Patterson AD, Soybel DI. Impaired recovery from peritoneal inflammation in a mouse model of mild dietary zinc restriction. Mol Nutr Food Res 2016; 60:672-81. [DOI: 10.1002/mnfr.201500688] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/30/2015] [Accepted: 10/31/2015] [Indexed: 12/31/2022]
Affiliation(s)
| | | | - Philip B. Smith
- Departments of Veterinary and Biomedical Sciences; University Park PA USA
- The Huck Institutes of the Life Sciences; University Park PA USA
| | | | | | - Shannon L. Kelleher
- Departments of Surgery; Hershey PA USA
- Cellular and Molecular Physiology; Hershey PA USA
| | - Andrew D. Patterson
- Departments of Veterinary and Biomedical Sciences; University Park PA USA
- Molecular Toxicology; University Park PA USA
| | - David I. Soybel
- Departments of Surgery; Hershey PA USA
- Cellular and Molecular Physiology; Hershey PA USA
| |
Collapse
|
32
|
Traphagen N, Tian Z, Allen-Gipson D. Chronic Ethanol Exposure: Pathogenesis of Pulmonary Disease and Dysfunction. Biomolecules 2015; 5:2840-53. [PMID: 26492278 PMCID: PMC4693259 DOI: 10.3390/biom5042840] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/04/2015] [Accepted: 09/28/2015] [Indexed: 12/18/2022] Open
Abstract
Ethanol (EtOH) is the world’s most commonly used drug, and has been widely recognized as a risk factor for developing lung disorders. Chronic EtOH exposure affects all of the organ systems in the body and increases the risk of developing pulmonary diseases such as acute lung injury and pneumonia, while exacerbating the symptoms and resulting in increased mortality in many other lung disorders. EtOH and its metabolites inhibit the immune response of alveolar macrophages (AMs), increase airway leakage, produce damaging reactive oxygen species (ROS), and disrupt the balance of antioxidants/oxidants within the lungs. In this article, we review the role of EtOH exposure in the pathogenesis and progression of pulmonary disease.
Collapse
Affiliation(s)
- Nicole Traphagen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida Health, Tampa, FL 33612, USA.
| | - Zhi Tian
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida Health, Tampa, FL 33612, USA.
| | - Diane Allen-Gipson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida Health, Tampa, FL 33612, USA.
- Department of Internal Medicine, Division of Allergy and Immunology, University of South Florida Health, Tampa, FL 33612, USA.
| |
Collapse
|
33
|
Potential Role of the Gut/Liver/Lung Axis in Alcohol-Induced Tissue Pathology. Biomolecules 2015; 5:2477-503. [PMID: 26437442 PMCID: PMC4693244 DOI: 10.3390/biom5042477] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/11/2015] [Accepted: 09/21/2015] [Indexed: 01/01/2023] Open
Abstract
Both Alcoholic Liver Disease (ALD) and alcohol-related susceptibility to acute lung injury are estimated to account for the highest morbidity and mortality related to chronic alcohol abuse and, thus, represent a focus of intense investigation. In general, alcohol-induced derangements to both organs are considered to be independent and are often evaluated separately. However, the liver and lung share many general responses to damage, and specific responses to alcohol exposure. For example, both organs possess resident macrophages that play key roles in mediating the immune/inflammatory response. Additionally, alcohol-induced damage to both organs appears to involve oxidative stress that favors tissue injury. Another mechanism that appears to be shared between the organs is that inflammatory injury to both organs is enhanced by alcohol exposure. Lastly, altered extracellular matrix (ECM) deposition appears to be a key step in disease progression in both organs. Indeed, recent studies suggest that early subtle changes in the ECM may predispose the target organ to an inflammatory insult. The purpose of this chapter is to review the parallel mechanisms of liver and lung injury in response to alcohol consumption. This chapter will also explore the potential that these mechanisms are interdependent, as part of a gut-liver-lung axis.
Collapse
|
34
|
Liu Y, Chen H, Sun Z, Chen X. Molecular mechanisms of ethanol-associated oro-esophageal squamous cell carcinoma. Cancer Lett 2015; 361:164-173. [PMID: 25766659 PMCID: PMC4765374 DOI: 10.1016/j.canlet.2015.03.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/04/2015] [Accepted: 03/04/2015] [Indexed: 02/06/2023]
Abstract
Alcohol drinking is a major etiological factor of oro-esophageal squamous cell carcinoma (OESCC). Both local and systemic effects of ethanol may promote carcinogenesis, especially among chronic alcoholics. However, molecular mechanisms of ethanol-associated OESCC are still not well understood. In this review, we summarize current understandings and propose three mechanisms of ethanol-associated OESCC: (1) Disturbance of systemic metabolism of nutrients: during ethanol metabolism in the liver, systemic metabolism of retinoids, zinc, iron and methyl groups is altered. These nutrients are known to be associated with the development of OESCC. (2) Disturbance of redox metabolism in squamous epithelial cells: when ethanol is metabolized in oro-esophageal squamous epithelial cells, reactive oxygen species are generated and produce oxidative damage. Meanwhile, ethanol may also disturb fatty-acid metabolism in these cells. (3) Disturbance of signaling pathways in squamous epithelial cells: due to its physico-chemical properties, ethanol changes cell membrane fluidity and shape, and may thus impact multiple signaling pathways. Advanced molecular techniques in genomics, epigenomics, metabolomics and microbiomics will help us elucidate how ethanol promotes OESCC.
Collapse
Affiliation(s)
- Yao Liu
- Department of Oral Medicine, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China; Cancer Research Program, JLC-BBRI, North Carolina Central University, Durham, NC 27707, USA
| | - Hao Chen
- Cancer Research Program, JLC-BBRI, North Carolina Central University, Durham, NC 27707, USA
| | - Zheng Sun
- Department of Oral Medicine, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China.
| | - Xiaoxin Chen
- Cancer Research Program, JLC-BBRI, North Carolina Central University, Durham, NC 27707, USA.
| |
Collapse
|
35
|
Ward C, Schlingmann B, Stecenko AA, Guidot DM, Koval M. NF-κB inhibitors impair lung epithelial tight junctions in the absence of inflammation. Tissue Barriers 2015; 3:e982424. [PMID: 25838984 DOI: 10.4161/21688370.2014.982424] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/27/2014] [Indexed: 01/11/2023] Open
Abstract
NF-κB (p50/p65) is the best characterized transcription factor known to regulate cell responses to inflammation. However, NF-κB is also constitutively expressed. We used inhibitors of the classical NF-κB signaling pathway to determine whether this transcription factor has a role in regulating alveolar epithelial tight junctions. Primary rat type II alveolar epithelial cells were isolated and cultured on Transwell permeable supports coated with collagen for 5 d to generate a model type I cell monolayer. Treatment of alveolar epithelial monolayers overnight with one of 2 different IκB kinase inhibitors (BAY 11-7082 or BMS-345541) resulted in a dose-dependent decrease in TER at concentrations that did not affect cell viability. In response to BMS-345541 treatment there was an increase in total claudin-4 and claudin-5 along with a decrease in claudin-18, as determined by immunoblot. However, there was little effect on the total amount of cell-associated claudin-7, occludin, junctional adhesion molecule A (JAM-A), zonula occludens (ZO)-1 or ZO-2. Moreover, treatment with BMS-345541 resulted in altered tight junction morphology as assessed by immunofluorescence microscopy. Cells treated with BMS-345541 had an increase in claudin-18 containing projections emanating from tight junctions ("spikes") that were less prominent in control cells. There also were several areas of cell-cell contact which lacked ZO-1 and ZO-2 localization as well as rearrangements to the actin cytoskeleton in response to BMS-345541. Consistent with an anti-inflammatory effect, BMS-345541 antagonized the deleterious effects of lipopolysaccharide (LPS) on alveolar epithelial barrier function. However, BMS-345541 also inhibited the ability of GM-CSF to increase alveolar epithelial TER. These data suggest a dual role for NF-κB in regulating alveolar barrier function and that constitutive NF-κB function is required for the integrity of alveolar epithelial tight junctions.
Collapse
Key Words
- ARDS, Acute Respiratory Distress Syndrome
- GM-CSF, Granulocyte Macrophage Colony Stimulating Factor
- IL, interleukin
- IκB, Inhibitor of κB
- JAM-A, junctional adhesion molecule A
- LPS, lipolysaccharide
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- PBS, phosphate buffered saline
- TER, transepithelial resistance
- TNF, Tumor Necrosis Factor
- ZO, zonula occludens
- alveolus
- claudin
- lung barrier
- tight junction
Collapse
Affiliation(s)
- Christina Ward
- Pulmonary Division; Department of Medicine; Emory University School of Medicine ; Atlanta, GA USA
| | - Barbara Schlingmann
- Pulmonary Division; Department of Medicine; Emory University School of Medicine ; Atlanta, GA USA
| | - Arlene A Stecenko
- Department of Pediatrics; Emory University School of Medicine ; Atlanta, GA USA
| | - David M Guidot
- Pulmonary Division; Department of Medicine; Emory University School of Medicine ; Atlanta, GA USA
| | - Michael Koval
- Pulmonary Division; Department of Medicine; Emory University School of Medicine ; Atlanta, GA USA ; Department of Cell Biology; Emory University School of Medicine ; Atlanta, GA USA
| |
Collapse
|
36
|
Trevejo-Nunez G, Kolls JK, de Wit M. Alcohol Use As a Risk Factor in Infections and Healing: A Clinician's Perspective. Alcohol Res 2015; 37:177-84. [PMID: 26695743 PMCID: PMC4590615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Physicians have recognized for more than a century that alcohol use is associated with infections and that alcoholics are especially at risk for pneumonia. Clear evidence now indicates that alcohol has a systemic effect on every organ. This review first presents a clinical case to describe a patient with immunity issues complicated by alcohol use-a setting familiar to many clinicians. This is followed by a description of the molecular mechanisms that explain the secondary immune deficiency produced by alcohol in the host, focusing mostly on the gut and lower respiratory mucosal immunity. The goal of this review is to increase awareness of the new mechanisms being investigated to understand how alcohol affects the human immune system and the development of new strategies to attenuate adverse outcomes in the affected population.
Collapse
|
37
|
Simet SM, Sisson JH. Alcohol's Effects on Lung Health and Immunity. Alcohol Res 2015; 37:199-208. [PMID: 26695745 PMCID: PMC4590617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
It has long been known that people with alcohol use disorder (AUD) not only may develop physical dependence but also may experience devastating long-term health problems. The most common and identifiable alcohol-associated health problems include liver cirrhosis, pancreatitis, cardiomyopathies, neuropathies, and dementia. However, the lung also is adversely affected by alcohol abuse, a fact often overlooked by clinicians and the public. Individuals with AUD are more likely to develop pneumonia, tuberculosis (TB), respiratory syncytial virus (RSV) infection, and acute respiratory distress syndrome (ARDS). Increased susceptibility to these and other pulmonary infections is caused by impaired immune responses in people with AUD. The key immune cells involved in combating pulmonary conditions such as pneumonia, TB, RSV infection, and ARDS are neutrophils, lymphocytes, alveolar macrophages, and the cells responsible for innate immune responses. Researchers are only now beginning to understand how alcohol affects these cells and how these effects contribute to the pathophysiology of pulmonary diseases in people with AUD.
Collapse
|
38
|
Konomi JV, Harris FL, Ping XD, Gauthier TW, Brown LAS. Zinc insufficiency mediates ethanol-induced alveolar macrophage dysfunction in the pregnant female mouse. Alcohol Alcohol 2015; 50:30-8. [PMID: 25371044 PMCID: PMC4318936 DOI: 10.1093/alcalc/agu073] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 06/25/2014] [Accepted: 07/18/2014] [Indexed: 11/14/2022] Open
Abstract
AIMS (a) Establish the minimum number of weeks of chronic ethanol ingestion needed to perturb zinc homeostasis, (b) Examine intracellular zinc status in the alveolar macrophages (AMs) when ethanol ingestion is combined with pregnancy, (c) Investigate whether in vitro zinc treatment reverses the effects of ethanol ingestion on the AM. METHODS C57BL/6 female mice were fed a liquid diet (±25% ethanol-derived calories) during preconception and pregnancy. The control group was pair-fed to the ethanol group. In the isolated AMs, we measured intracellular AM zinc levels, zinc transporter expression, alternative activation and phagocytic index. Zinc acetate was added to some cells prior to analysis. RESULTS Intracellular zinc levels in the AM decreased within 3 weeks of ethanol ingestion. After ethanol ingestion prior to and during pregnancy, zinc transporter expression and intracellular zinc levels were decreased in the AMs when compared with controls. Bacterial clearance was decreased because the AMs were alternatively activated. In vitro additions of zinc reversed these effects of ethanol. CONCLUSION Ethanol ingestion prior to and during pregnancy perturbed AM zinc balance resulting in impaired bacterial clearance, but these effects were ameliorated by in vitro zinc treatments.
Collapse
Affiliation(s)
- Juna V Konomi
- Nutrition and Health Sciences, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, GA, USA Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory University, Emory + Children's Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, GA 30322, USA
| | - Frank L Harris
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory University, Emory + Children's Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, GA 30322, USA
| | - Xiao-Du Ping
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory University, Emory + Children's Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, GA 30322, USA
| | - Theresa W Gauthier
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory University, Emory + Children's Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, GA 30322, USA
| | - Lou Ann S Brown
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory University, Emory + Children's Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, GA 30322, USA
| |
Collapse
|
39
|
Hamon R, Homan CC, Tran HB, Mukaro VR, Lester SE, Roscioli E, Bosco MD, Murgia CM, Ackland ML, Jersmann HP, Lang C, Zalewski PD, Hodge SJ. Zinc and zinc transporters in macrophages and their roles in efferocytosis in COPD. PLoS One 2014; 9:e110056. [PMID: 25350745 PMCID: PMC4211649 DOI: 10.1371/journal.pone.0110056] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 09/07/2014] [Indexed: 11/19/2022] Open
Abstract
Our previous studies have shown that nutritional zinc restriction exacerbates airway inflammation accompanied by an increase in caspase-3 activation and an accumulation of apoptotic epithelial cells in the bronchioles of the mice. Normally, apoptotic cells are rapidly cleared by macrophage efferocytosis, limiting any secondary necrosis and inflammation. We therefore hypothesized that zinc deficiency is not only pro-apoptotic but also impairs macrophage efferocytosis. Impaired efferocytic clearance of apoptotic epithelial cells by alveolar macrophages occurs in chronic obstructive pulmonary disease (COPD), cigarette-smoking and other lung inflammatory diseases. We now show that zinc is a factor in impaired macrophage efferocytosis in COPD. Concentrations of zinc were significantly reduced in the supernatant of bronchoalveolar lavage fluid of patients with COPD who were current smokers, compared to healthy controls, smokers or COPD patients not actively smoking. Lavage zinc was positively correlated with AM efferocytosis and there was decreased efferocytosis in macrophages depleted of Zn in vitro by treatment with the membrane-permeable zinc chelator TPEN. Organ and cell Zn homeostasis are mediated by two families of membrane ZIP and ZnT proteins. Macrophages of mice null for ZIP1 had significantly lower intracellular zinc and efferocytosis capability, suggesting ZIP1 may play an important role. We investigated further using the human THP-1 derived macrophage cell line, with and without zinc chelation by TPEN to mimic zinc deficiency. There was no change in ZIP1 mRNA levels by TPEN but a significant 3-fold increase in expression of another influx transporter ZIP2, consistent with a role for ZIP2 in maintaining macrophage Zn levels. Both ZIP1 and ZIP2 proteins were localized to the plasma membrane and cytoplasm in normal human lung alveolar macrophages. We propose that zinc homeostasis in macrophages involves the coordinated action of ZIP1 and ZIP2 transporters responding differently to zinc deficiency signals and that these play important roles in macrophage efferocytosis.
Collapse
Affiliation(s)
- Rhys Hamon
- Discipline of Medicine, University of Adelaide, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
| | - Claire C. Homan
- Discipline of Medicine, University of Adelaide, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
| | - Hai B. Tran
- Discipline of Medicine, University of Adelaide, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
- Department of Thoracic Medicine, Royal Adelaide Hospital, Lung Research Laboratory, Hanson Institute, Adelaide, South Australia, Australia
| | - Violet R. Mukaro
- Department of Thoracic Medicine, Royal Adelaide Hospital, Lung Research Laboratory, Hanson Institute, Adelaide, South Australia, Australia
| | - Susan E. Lester
- Rheumatology Unit, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
| | - Eugene Roscioli
- Discipline of Surgery, University of Adelaide, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
| | - Mariea D. Bosco
- Discipline of Medicine, University of Adelaide, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
| | | | - Margaret Leigh Ackland
- Centre for Cellular and Molecular Biology, School of Life and Environmental Sciences, Deakin University, Burwood, Melbourne, Victoria, Australia
| | - Hubertus P. Jersmann
- Department of Thoracic Medicine, Royal Adelaide Hospital, Lung Research Laboratory, Hanson Institute, Adelaide, South Australia, Australia
| | - Carol Lang
- Discipline of Medicine, University of Adelaide, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
| | - Peter D. Zalewski
- Discipline of Medicine, University of Adelaide, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
- * E-mail:
| | - Sandra J. Hodge
- Discipline of Medicine, University of Adelaide, The Queen Elizabeth Hospital, Woodville, South Australia, Australia
- Department of Thoracic Medicine, Royal Adelaide Hospital, Lung Research Laboratory, Hanson Institute, Adelaide, South Australia, Australia
| |
Collapse
|
40
|
Li B, Cui W, Tan Y, Luo P, Chen Q, Zhang C, Qu W, Miao L, Cai L. Zinc is essential for the transcription function of Nrf2 in human renal tubule cells in vitro and mouse kidney in vivo under the diabetic condition. J Cell Mol Med 2014; 18:895-906. [PMID: 24597671 PMCID: PMC4119395 DOI: 10.1111/jcmm.12239] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 12/20/2013] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence from human and laboratory studies showed the effect of zinc (Zn) on diabetic complications. Nuclear factor-erythroid 2-related factor 2 (Nrf2) plays important role in the prevention of oxidative damage. This study was to define whether Zn statues (deficiency or supplement) affect the Nrf2 expression and function, and also affect the damage severity of human renal tubular (HK11) cells exposed to high glucose (HG) with palmitate (Pal) and kidney of diabetic mice induced by multiple low-dose streptozotocins. For Zn deficiency diabetic mice were treated with Zn chelator PTEN at 5 mg/kg bw daily for 4 months. Results showed that HG/Pal significantly increased the expression of pro-fibrotic mediators, connective tissue growth factor and PAI-1, in HK11 cells, which was exacerbated by TPEN that depleted intracellular free Zn and decreased Nrf2 expression and transcription. Zn supplement prevented the effects of TPEN and also increased Akt and GSK-3β phosphorylation with a decrease in Nrf2 nuclear exporter, Fyn. All these effects of Zn were abolished by Akt inhibitor. Therefore, Zn up-regulates Nrf2 function via activating Akt-mediated inhibition of Fyn function. Treatment of diabetic mice with TPEN decreased renal Zn level and Nrf2 expression and transcription, with an exacerbation of renal oxidative damage, inflammation and fibrosis. These results suggest the essentiality of Zn for Nrf2 expression and transcription function.
Collapse
Affiliation(s)
- Bing Li
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China; Kosair Children's Hospital Research Institute, and Departments of Pediatrics and Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA; Department of Nephrology, Jilin Province People's Hospital, Changchun, China
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Yu C, Mei XT, Zheng YP, Xu DH. Zn(II)-curcumin protects against hemorheological alterations, oxidative stress and liver injury in a rat model of acute alcoholism. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 37:729-737. [PMID: 24607687 DOI: 10.1016/j.etap.2014.02.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 02/09/2014] [Accepted: 02/12/2014] [Indexed: 06/03/2023]
Abstract
Curcumin can chelate metal ions, forming metallocomplexes. We compared the effects of Zn(II)-curcumin with curcumin against hemorheological alterations, oxidative stress and liver injury in a rat model of acute alcoholism. Oral administration of Zn(II)-curcumin dose-dependently prevented the ethanol-induced elevation of serum malondialdehyde (MDA) content and reductions in glutathione level and superoxide dismutase (SOD) activity. Zn(II)-curcumin also inhibited ethanol-induced liver injury. Additionally, Zn(II)-curcumin dose-dependently inhibited hemorheological abnormalities, including the ethanol-induced elevation of whole blood viscosity, plasma viscosity, blood viscosity at corrected hematocrit (45%), erythrocyte aggregation index, erythrocyte rigidity index and hematocrit. Compared to curcumin at the same dose, Zn(II)-curcumin more effectively elevated SOD activity, ameliorated liver injury and improved hemorheological variables. These results suggest that Zn(II)-curcumin protected the rats from ethanol-induced liver injury and hemorheological abnormalities via the synergistic effect of curcumin and zinc.
Collapse
Affiliation(s)
- Chuan Yu
- Laboratory of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Xue-Ting Mei
- Laboratory of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Yan-Ping Zheng
- Laboratory of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China
| | - Dong-Hui Xu
- Laboratory of Traditional Chinese Medicine and Marine Drugs, Department of Biochemistry, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China.
| |
Collapse
|
42
|
Cribbs SK, Rimland D. Alcohol and HIV: Experimental and Clinical Evidence of Combined Impact on the Lung. ALCOHOL USE DISORDERS AND THE LUNG 2014. [PMCID: PMC7121129 DOI: 10.1007/978-1-4614-8833-0_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Despite antiretroviral therapy, lung disease is a leading cause of death in individuals infected with human immunodeficiency virus type 1 (HIV). Individuals infected with HIV are susceptible to serious bacterial and viral infections, such as pneumococcus and influenza, which are particularly problematic for lung health, resulting in lung injury. Additionally, HIV-infected individuals are susceptible to a number of pulmonary diseases for unknown reasons. Alcohol, the most commonly abused drug in the world, continues to exact an enormous toll on morbidity and mortality in individuals living with HIV. Chronic alcohol abuse has been shown to affect lung immunity, resulting in significant lung injury. There is a paucity of literature on the additive effects of HIV and alcohol, two diseases of immune senescence, in the lung. This chapter begins by discussing the latest literature evaluating the epidemiology of HIV, alcohol use, and lung health focusing on two prevalent infections, tuberculosis and pneumococcal pneumonia. In parallel, we discuss the interactions of alcohol and HIV on the risk for acute lung injury and subsequent morbidity and mortality. We then discuss the pathophysiology of how these two diseases of immune dysfunction affect the lung, with a focus on the oxidative stress, alveolar macrophage host immune capacity, and immunomodulatory role of zinc in the airway. Finally, we review the latest literature on how HIV and alcohol affect other pulmonary disorders including chronic obstructive pulmonary disease, pulmonary hypertension, and lung cancer.
Collapse
|
43
|
Mehta AJ, Yeligar SM, Elon L, Brown LA, Guidot DM. Alcoholism causes alveolar macrophage zinc deficiency and immune dysfunction. Am J Respir Crit Care Med 2013; 188:716-23. [PMID: 23805851 DOI: 10.1164/rccm.201301-0061oc] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
RATIONALE Alcohol use disorders cause oxidative stress in the lower airways and increase susceptibility to pneumonia and lung injury. Currently, no therapeutic options exist to mitigate the pulmonary consequences of alcoholism. OBJECTIVES We recently determined in an animal model that alcohol ingestion impairs pulmonary zinc metabolism and causes alveolar macrophage immune dysfunction. The objective of this research is to determine the effects of alcoholism on zinc bioavailability and alveolar macrophage function in human subjects. METHODS We recruited otherwise healthy alcoholics (n = 17) and matched control subjects (n = 17) who underwent bronchoscopy for isolation of alveolar macrophages, which were analyzed for intracellular zinc, phagocytic function, and surface expression of granulocyte-macrophage colony-stimulating factor receptor; all three of these indices are decreased in experimental models. MEASUREMENTS AND MAIN RESULTS Alcoholic subjects had normal serum zinc, but significantly decreased alveolar macrophage intracellular zinc levels (adjusted means [SE], 718 [41] vs. 948 [25] RFU/cell; P < 0.0001); bacterial phagocytosis (adjusted means [SE], 1,027 [48] vs. 1,509 [76] RFU/cell; P < 0.0001); and expression of granulocyte-macrophage colony-stimulating factor receptor β subunit (adjusted means [SE], 1,471 [42] vs. 2,114 [35] RFU/cell; P < 0.0001]. Treating alveolar macrophages with zinc acetate and glutathione in vitro increased intracellular zinc levels and improved their phagocytic function. CONCLUSIONS These novel clinical findings provide evidence that alcohol abuse is associated with significant zinc deficiency and immune dysfunction within the alveolar space and suggest that dietary supplementation with zinc and glutathione precursors could enhance airway innate immunity and decrease the risk for pneumonia or lung injury in these vulnerable individuals.
Collapse
Affiliation(s)
- Ashish J Mehta
- 1 Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | | | | | | | | |
Collapse
|
44
|
Mehta AJ. Pulmonary consequences of alcoholism: A critical review. OA ALCOHOL 2013; 1:10.13172/2053-0285-1-2-861. [PMID: 37886715 PMCID: PMC10601137 DOI: 10.13172/2053-0285-1-2-861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Alcohol use and abuse are prevailing practices in people throughout the world. Unfortunately, alcohol use disorders pose tremendous costs to both society and the individual. While alcoholism has many well-known medical consequences such as liver injury and pancreatitis, the effects of chronic alcohol exposure on the respiratory system are often overlooked. Specifically, studies have shown that alcohol abuse causes significant derangements in the lung and predisposes individuals to the development of pneumonia and acute lung injury. Several important processes are responsible for this increased susceptibility to pulmonary pathology, including alterations in nonimmunological defense systems, impairment of lung immunity, and alveolar epithelial barrier dysfunction. These crucial defects comprise what has been referred to as the "alcohol lung phenotype". Importantly, these abnormalities not only increase the risk of lung infections and injury, they cause worse morbidity and mortality in alcoholics compared to non-alcoholics. While there are no current therapies to combat these alcohol-induced pulmonary abnormalities, current research has revealed several important mechanisms that may be exploited to develop new treatment options for this vulnerable population.
Collapse
Affiliation(s)
- Ashish J Mehta
- Atlanta VA Medical Center and Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
45
|
Abstract
The distal airways are covered with a heterogeneous layer of cells known as the alveolar epithelium. Alveolar epithelial cells provide the major barrier between the airspace and fluid filled tissue compartments. As such, regulation of the alveolar epithelium is critical to maintain a healthy lung and for optimal gas exchange. In this chapter, we discuss functional roles for alveolar epithelial cells with particular emphasis on intercellular junctions and communication. As a thin layer of cells directly exposed to atmospheric oxygen, alveoli are particularly sensitive to oxidant insults. Alcohol significantly diminishes the normal antioxidant reserves of the alveolar epithelium, thereby rendering it sensitized for an exaggerated damage response to acute and chronic injuries. The effects of alcohol on alveolar epithelia are discussed along with open questions and potential therapeutic targets to prevent the pathophysiology of alcoholic lung disease.
Collapse
|
46
|
Guidot DM, Mehta AJ. Alcohol-Mediated Zinc Deficiency Within the Alveolar Space: A Potential Fundamental Mechanism Underlying Oxidative Stress and Cellular Dysfunction in the Alcoholic Lung. ALCOHOL USE DISORDERS AND THE LUNG 2013; 14. [PMCID: PMC7120872 DOI: 10.1007/978-1-4614-8833-0_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Zinc is one of the most abundant trace elements in the human body, and its presence is essential for numerous biological processes including enzymatic activity, immune function, protein synthesis, and wound healing. Given these important roles, zinc has a sophisticated transport system to regulate its homeostasis. Determination of zinc status, however, is difficult to determine as serum levels are closely maintained and are not an accurate reflection of total body zinc or metabolism at the organ level. Fortunately, the discovery of zinc-specific fluorescent dyes has allowed for a much better assessment of zinc status in the respiratory system and has revealed that alcoholism perturbs this highly developed zinc metabolism such that its distribution to the lung and alveolar space is significantly decreased. As a result, this pulmonary zinc deficiency impairs function in the alveolar macrophage, which is the primary host immune cell within the lower airway. Experimental models have demonstrated that correction of this zinc deficiency restores immune function to the alveolar macrophage as best reflected by improved bacterial clearance in response to infection. While the precise mechanisms underlying alcohol-induced zinc deficiency are still under investigation, there is experimental evidence of several important connections with granulocyte–macrophage colony-stimulating factor and oxidative stress, suggesting that alteration of zinc homeostasis may be a fundamental mechanism underlying the cellular pathology seen in the alcohol lung phenotype. This chapter reviews zinc homeostasis and offers insight into our understanding of zinc deficiency in the setting of alcoholism and the potential of zinc as a therapeutic modality in the vulnerable alcoholic host.
Collapse
Affiliation(s)
- David M. Guidot
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University School of Medicine and the Atlanta VA Medical Center, Atlanta, Georgia USA
| | - Ashish J. Mehta
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University School of Medicine and the Atlanta VA Medical Center, Decatur, Georgia USA
| |
Collapse
|
47
|
Curry-McCoy TV, Venado A, Guidot DM, Joshi PC. Alcohol ingestion disrupts alveolar epithelial barrier function by activation of macrophage-derived transforming growth factor beta1. Respir Res 2013; 14:39. [PMID: 23547562 PMCID: PMC3623812 DOI: 10.1186/1465-9921-14-39] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 03/12/2013] [Indexed: 12/13/2022] Open
Abstract
Background Chronic alcohol abuse causes oxidative stress and impairs alveolar epithelial barrier integrity, thereby rendering the lung susceptible to acute edematous injury. Experimentally, alcohol-induced oxidative stress increases the expression of transforming growth factor β1 (TGFβ1) in the lung; however, we do not know the precise contribution of various alveolar cells in this process. In the present study, we focused on cell-cell interactions between alveolar macrophages and epithelial cells and the potential mechanisms by which TGFβ1 may become activated in the alveolar space of the alcoholic lung. Methods Primary alveolar macrophages and epithelial cells were isolated from control- and alcohol-fed Sprague–Dawley rats. Expression of TGFβ1 and the epithelial integrin αvβ6 were examined by real time PCR and either immunocytochemistry or flow cytometry. Alveolar epithelial cells were cultured on transwell supports in the presence of macrophage cell lysate from control- or alcohol-fed rats or in the presence of viable macrophages ± alcohol. Epithelial barrier function was assessed by transepithelial resistance (TER) and paracellular flux of Texas Red dextran. Results TGFβ1 expression was increased in alveolar macrophages from alcohol-fed rats, and TGFβ1 protein was predominantly membrane-bound. Importantly, alveolar macrophage cellular lysate from alcohol-fed rats decreased TER and increased paracellular dextran flux in primary alveolar epithelial cell monolayers as compared to the lysates from control-fed rats. Alcohol-induced epithelial barrier dysfunction was prevented by anti-TGFβ1 antibody treatment, indicating the presence of bioactive TGFβ1 in the macrophage lysate. In addition, co-culturing macrophages and epithelial cells in the presence of alcohol decreased epithelial barrier function, which also was prevented by anti-TGFβ1 and anti-αvβ6 treatment. In parallel, chronic alcohol ingestion in vivo, or direct treatment with active TGFβ1 in vitro, increased the expression of αvβ6 integrin, which is known to activate TGFβ1, in alveolar epithelial cells. Conclusions Taken together, these data suggest that interactions between alveolar epithelial cells and macrophages contribute to the alcohol-mediated disruption of epithelial barrier function via the expression and activation of TGFβ1 at points of cell-cell contact.
Collapse
Affiliation(s)
- Tiana V Curry-McCoy
- Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University School of Medicine, Atlanta, GA 30322-1047, USA
| | | | | | | |
Collapse
|
48
|
Jensen JS, Fan X, Guidot DM. Alcohol causes alveolar epithelial oxidative stress by inhibiting the nuclear factor (erythroid-derived 2)-like 2-antioxidant response element signaling pathway. Am J Respir Cell Mol Biol 2013; 48:511-7. [PMID: 23306837 PMCID: PMC4080903 DOI: 10.1165/rcmb.2012-0334oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 01/10/2013] [Indexed: 12/28/2022] Open
Abstract
Excessive alcohol use increases the risk of acute lung injury and pneumonia. Chronic alcohol ingestion causes oxidative stress within the alveolar space, including near depletion of glutathione (GSH), which impairs alveolar epithelial and macrophage function, in experimental animals and human subjects. However, the fundamental mechanism(s) by which alcohol induces such profound lung oxidative stress is unknown. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a redox-sensitive master transcription factor that regulates activation of the antioxidant response element (ARE). As the alveolar epithelium controls GSH levels within the alveolar space, we hypothesized that alcohol also decreases Nrf2 expression and/or activation within the alveolar epithelium. In this study, we determined that alcohol ingestion in vivo or direct alcohol exposure in vitro down-regulated the Nrf2-ARE pathway in lung epithelial cells, decreased the expression of antioxidant genes, and lowered intracellular GSH levels. RNA silencing of Nrf2 gene expression in alveolar epithelial cells in vitro decreased expression of these same antioxidant genes, and likewise lowered intracellular GSH levels, findings that mirrored the effects of alcohol. In contrast, treating alcohol-exposed alveolar epithelial cells in vitro with the Nrf2 activator, sulforaphane, preserved Nrf2 expression, ARE activation, intracellular GSH levels, and epithelial barrier function. These new experimental findings implicate down-regulation of the Nrf2-ARE signaling pathway as a fundamental mechanism by which alcohol causes profound oxidative stress and alveolar epithelial dysfunction, and suggest that treatments, such as sulforaphane, that activate this pathway could mitigate the pathophysiological consequences of alcohol on the lung and other organs.
Collapse
Affiliation(s)
- J. Spencer Jensen
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia; and
- Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Xian Fan
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia; and
- Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - David M. Guidot
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia; and
- Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
49
|
Abstract
After iron, zinc is the most abundant essential trace metal. Intracellular zinc ([Zn]i) is maintained across a wide range of cells and species in a tight quota (100 to 500 μM) by a dynamic process of transport, intracellular vesicular storage, and binding to a large number of proteins (estimated at 3-10% of human proteome). As such, zinc is an integral component of numerous metalloenzymes, structural proteins, and transcription factors. It is generally assumed that a vanishingly small component of [Zn]i, referred to as free or labile zinc, and operationally defined as the pool sensitive to chelation (by agents such as N, N, N’, N’-tetrakis [2-pyridylmethyl] ethylenediamine [TPEN]) and capable of detection by a variety of chemical and genetic sensors, participates in signal transduction pathways. Zinc deficiencies, per se, can arise from acquired (malnutrition, alcoholism) or genetic (mutations in molecules affecting zinc homeostasis, the informative and first example being acrodermatitis enteropathica) factors or as a component of various diseases (e.g., sickle cell disease, cystic fibrosis, sepsis). Hypozincemia has profound effects on developing humans, and all facets of physiological function (neuronal, endocrine, immunological) are affected, although considerably less is known regarding cardiovascular pathophysiology. In this review, we provide an update on current knowledge of molecular and cellular aspects of zinc homeostasis and then focus on implications of zinc signaling in pulmonary endothelium as it relates to programmed cell death, altered contractility, and septic and aseptic injury to this segment of the lung.
Collapse
Affiliation(s)
- Kalidasan Thambiayya
- Department of Bioengineering, University of Pittsburgh and University of Pittsburgh School of Medicine and Graduate School Public Health, Pittsburgh, Pennsylvania, USA
| | | | | | | |
Collapse
|
50
|
Zhang J, Li B, Zhang Y, Li A, Yu X, Huang Q, Fan C, Cai X. Synchrotron radiation X-ray fluorescence analysis of biodistribution and pulmonary toxicity of nanoscale titanium dioxide in mice. Analyst 2013; 138:6511-6. [DOI: 10.1039/c3an01267k] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|