1
|
Akutsu T, Tone K, Gochi M, Tsukamoto Y, Araya J. Wound Dehiscence and Chest Pain After Lobectomy. Clin Infect Dis 2025; 80:653-656. [PMID: 40095959 DOI: 10.1093/cid/ciae466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Affiliation(s)
- Takuya Akutsu
- Department of Respiratory Medicine, The Jikei University School of Medicine Kashiwa Hospital, Kashiwa, Chiba, Japan
| | - Kazuya Tone
- Department of Respiratory Medicine, The Jikei University School of Medicine Kashiwa Hospital, Kashiwa, Chiba, Japan
| | - Mina Gochi
- Department of Respiratory Medicine, The Jikei University School of Medicine Kashiwa Hospital, Kashiwa, Chiba, Japan
| | - Yo Tsukamoto
- Department of Surgery, The Jikei University School of Medicine Kashiwa Hospital, Kashiwa, Chiba, Japan
| | - Jun Araya
- Division of Respiratory Diseases, Department of Internal Medicine, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| |
Collapse
|
2
|
Su H, Xie H. Associations between lifestyle habits, environmental factors and respiratory diseases: a cross-sectional study from southwest China. Front Public Health 2025; 13:1513926. [PMID: 40109413 PMCID: PMC11919831 DOI: 10.3389/fpubh.2025.1513926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/07/2025] [Indexed: 03/22/2025] Open
Abstract
Background Numerous studies have demonstrated that lifestyle habits and environmental factors influence the incidence and progression of respiratory diseases. However, there is a paucity of similar research conducted in southwest China. Objective This study aims to investigate the prevalence and primary influencing factors of respiratory diseases among residents in a specific region of southwest China, and to identify vulnerable populations. Method From February 2024 to May 2024, a multi-stage stratified random sampling method was employed in a specific region of southwest China. Three monitoring points were randomly selected from six jurisdictions within this region, resulting in the collection of relevant information from a total of 4,507 residents through offline interviews. Lasso-logistic regression was conducted using R version 4.3.0 to develop a nomogram for estimating disease probabilities. Interaction analysis was performed with gender and age group serving as grouping variables, while other dimensional factors were utilized as analysis variables. Result A total of 4,507 respondents participated in this study, of whom 956 (21.21%) were identified as sick. The older adult group (>65 years) exhibited the highest prevalence (30.3%). Results from the Lasso-logistic model indicated that current smoking, alcohol abuse, passive smoking, coupled with poor indoor and outdoor environments were significant risk factors. Additionally, a history of respiratory disease, a family history of respiratory issues, negative emotions, and high stress levels may also contribute to the risk of the disease. Protective factors identified include regular exercise, adequate indoor lighting, frequent ventilation, and regular disinfection practices. The nomogram developed in this study demonstrated good discrimination, calibration, and clinical efficacy. Multiplicative interaction analysis indicated that gender and age group exhibited varying degrees of interaction with factors such as smoking, passive smoking, alcohol abuse, regular exercise, household smoke, house disinfection, dust mites, history of respiratory allergies, use of velvet products, and family history of respiratory conditions. Notably, females, adolescents, and the older adult were identified as particularly susceptible and at-risk groups for these interactions. Conclusion The prevalence of respiratory diseases is notably higher among the permanent population in southwest China. High-risk lifestyles, coupled with poor indoor and outdoor environments, pose particularly significant threats to women, adolescents, and the older adult. Consequently, improving living habits, renovating aging communities, enhancing the quality of the living environment, and prioritizing vulnerable populations remain central to the objectives of primary health services.
Collapse
Affiliation(s)
- Hengyu Su
- School of Public Health, Xinjiang Medical University, Ürümqi, China
| | - Huifang Xie
- School of Public Health, Xinjiang Medical University, Ürümqi, China
| |
Collapse
|
3
|
Ji D, Li H, Jin S, Tian C, Wu L. Examination of factors causing postoperative pneumonia in elderly hip fracture patients: A narrative review. Medicine (Baltimore) 2025; 104:e41700. [PMID: 40020120 PMCID: PMC11875577 DOI: 10.1097/md.0000000000041700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
With the increasing aging problem, the number of fractures in the elderly is also increasing, of which hip fractures are more common, known as "the last fracture of life." Postoperative pneumonia (POP) is a common complication of hip fracture, which greatly increases the mortality of patients. It is particularly important to clarify the factors of perioperative pneumonia for the prevention and treatment process. In this paper, the factors causing POP mainly include demographic factors, pre-injury comorbidities, blood index parameters, major clinical interventions and related mechanisms were reviewed, and the risk degree of the factors causing postoperative pneumonia was mainly discussed, and they were divided into independent factors and risk factors. The objective is to make the most accurate POP prevention measures for hip fracture patients according to the classification of independent factors and risk factors, and reduce the incidence of postoperative pneumonia.
Collapse
Affiliation(s)
- Dongqi Ji
- Beijing Xiaotangshan Hospital, Beijing, China
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin, China
| | - Huanhuan Li
- Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise & Health, Tianjin University of Sport, Tianjin, China
| | - Shasha Jin
- Beijing Xiaotangshan Hospital, Beijing, China
| | | | - Liang Wu
- Beijing Xiaotangshan Hospital, Beijing, China
| |
Collapse
|
4
|
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
|
5
|
Cui J, Xu Z, Yu Z, Zhang Q, Liu S, Du B, Gan L, Yan C, Xue G, Feng J, Fan Z, Fu T, Feng Y, Zhao H, Ding Z, Li X, Zhang R, Cui X, Tian Z, Huang K, Wang W, Bai Y, Zhou H, Sun Y, Yang X, Wan M, Ke Y, Yuan J. High-alcohol-producing Klebsiella pneumoniae aggravates lung injury by affecting neutrophils and the airway epithelium. Cell Rep Med 2025; 6:101886. [PMID: 39753141 DOI: 10.1016/j.xcrm.2024.101886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/27/2024] [Accepted: 12/05/2024] [Indexed: 01/24/2025]
Abstract
We have previously reported that high-alcohol-producing Klebsiella pneumoniae (HiAlc Kpn) in the gut can cause endo-alcoholic fatty liver disease. Here, we discover that 91.2% of Kpn isolates from pulmonary disease samples also produce excess ethanol, which may be associated with respiratory disease severity. To further explore the potential mechanism, a murine model is established with high-dose bacteria. Kpn stimulates granular neutrophils (G0), subsequently transforming them into phagocytic neutrophils (G1). HiAlc Kpn also causes dysfunction of pyrimidine metabolism, leading to neutrophil apoptosis. These changes inhibit phagocytosis of neutrophils and possibly suppress inflammasome-dependent innate immunity. In a persistent infective murine model, HiAlc Kpn induces lung fibrosis and production of reactive oxygen species (ROS), possibly affecting epithelial cell apoptosis and lung function. The results suggest that the subtype of neutrophil is a potential biomarker for the severity of lung injury caused by HiAlc Kpn.
Collapse
Affiliation(s)
- Jinghua Cui
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Ziying Xu
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Zihui Yu
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Qun Zhang
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Shiyu Liu
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Bing Du
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Lin Gan
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Chao Yan
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Guanhua Xue
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Junxia Feng
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Zheng Fan
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Tongtong Fu
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Yanling Feng
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Hanqing Zhao
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Zanbo Ding
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Xiaoran Li
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Rui Zhang
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Xiaohu Cui
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Ziyan Tian
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Kewu Huang
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Wenjun Wang
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yu Bai
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Haijian Zhou
- National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ying Sun
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xiaopeng Yang
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Meng Wan
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuehua Ke
- Capital Institute of Pediatrics, Beijing 100020, China.
| | - Jing Yuan
- Capital Institute of Pediatrics, Beijing 100020, China.
| |
Collapse
|
6
|
Wang Z, Meng S, Fan Y, Liu J, Zhao L, Cui Y, Xie K. Long-term trends and comparison of the burden of lower respiratory tract infections in China and globally from 1990 to 2021: an analysis based on the Global Burden of Disease study 2021. Front Public Health 2024; 12:1507672. [PMID: 39720797 PMCID: PMC11666531 DOI: 10.3389/fpubh.2024.1507672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 11/20/2024] [Indexed: 12/26/2024] Open
Abstract
Background This study aimed to describe the temporal trends in the age and sex burdens of lower respiratory infections (LRIs) in China and globally from 1990 to 2021 and to analyze their epidemiological characteristics to formulate corresponding strategies to control LRIs. Methods This study utilized open data from the Global Burden of Disease (GBD) database from 1990 to 2021 to assess the burden of disease based on the prevalence, incidence, mortality, years lost (YLLs), years lived with disability (YLDs), and disability-adjusted life-years (DALYs) of LRIs in China and globally. Moreover, a comprehensive comparative analysis of the epidemiological characteristics of LRIs in China and globally was conducted via the Joinpoint regression model, age-period-cohort model (APC model), and stratified analysis of the study method from multiple dimensions, such as age, sex, and period. Finally, we used an autoregressive integrated moving average (ARIMA) model to predict the disease burden in LRIs over the next 15 years. Results From 1990 to 2021, China's age-standardized incidence, deaths, and disability-adjusted life year (DALY) rates per 100,000 people decreased from 5,481.13 (95% CI: 5,149.05, 5,836.35) to 2,853.81 (95% CI: 2,663.94, 3,067.55), from 60.65 (95% CI. 52.96, 66.66) to 14.03 (95% CI: 11.68, 17) and from 3,128.39 (95% CI: 2,724.11, 3,579.57) to 347.67 (95% CI: 301.28, 402.94). The global age-standardized incidence, deaths, and DALY rates per 100,000 people, on the other hand, decreased from 6,373.17 (95% CI: 5,993.51, 6,746.04) to 4,283.61 (95% CI: 4,057.03, 4,524.89) and from 61.81 (95% CI: 56.66, 66.74) to 28.67 (95% CI: 25.92, 31.07) and from 3,472.9 (95% CI: 3,090.71, 3,872.11) to 1,168.8 (95% CI: 1,016.96, 1,336.95). The decline in the aforementioned indicators is greater in the female population than in the male population, and the decrease in China is more pronounced than the global trend. In China, the age-standardized incidence and mortality rates of LRIs showed an annual average percentage change (AAPC) of -2.12 (95% CI: -2.20, -2.03) and -4.77 (95% CI: -5.14, -4.39), respectively. Globally, the age-standardized incidence and mortality rates for LRIs decreased by -1.28 (95% CI: -1.37, -1.18) and -2.47 (95% CI: -2.61, -2.32). By 2036, the incidence of lower respiratory infections (LRI) among men and women in China is projected to decrease by 36.55 and 46.87%, respectively, while the mortality rates are expected to decline to 12.67% for men and increase by 71.85% for women. In comparison, the global decline in LRI incidence is lower than that observed in China, yet the reduction in mortality rates is greater globally than in China. Conclusions Age-standardized incidence, mortality and disability-adjusted life years (DALYs) decreased more in China than at the global level between 1990 and 2021. Compared with the previous period, the COVID-19 pandemic has led to a significant decrease in the disease burden of LRIs. As the population continues to age, the disease burden of LRIs in the old adult population will become a major new public health challenge.
Collapse
Affiliation(s)
- Zhiwei Wang
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shuqi Meng
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Fan
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianfeng Liu
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lina Zhao
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Cui
- Department of Pathogen Biology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
7
|
Rojas DA, Coronado K, Pérez-Reytor D, Karahanian E. Reduction of Alcohol-Dependent Lung Pathological Features in Rats Treated with Fenofibrate. Int J Mol Sci 2024; 25:12814. [PMID: 39684525 DOI: 10.3390/ijms252312814] [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: 10/21/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Alcohol use disorder (AUD) is a public health problem characterized by a marked increment in systemic inflammation. In the last few years, it has been described as the role of alcohol in neuroinflammation affecting some aspects of neuronal function. Interestingly, inflammation is reduced with fenofibrate treatment, a PPARα agonist used to treat dyslipidemia. On the other hand, alcohol has been associated with chronic inflammation and fibrosis in the lungs, affecting their normal function and increasing respiratory infections. However, a deep characterization of the role of alcohol in the worsening of chronic respiratory diseases has not been described completely. In this work, we present a novel study using rats treated with alcohol and fenofibrate to evaluate the relevant features of chronic respiratory disease: inflammation, mucus hypersecretion, and fibrosis. The analysis of extracted lungs showed an increment in the inflammatory infiltrates and pro-inflammatory cytokine levels associated with alcohol. Interestingly, the treatment with fenofibrate decreased the expression of these markers and the infiltrates observed in the lungs. The levels of mucin Muc5ac showed an increment in animals treated with alcohol. However, this increment was markedly reduced if animals were subsequently treated with fenofibrate. Finally, we documented an increment of collagen deposition around airways in the animals treated with alcohol compared with control animals. However, fenofibrate treatment reduced this deposition to a level similar to the control animals. These results showed the role of alcohol in the increment of pathological features in the lungs. Moreover, these features were attenuated due to the fibrate treatment, which allows us to glimpse this drug's promising role as lung anti-inflammatory therapy.
Collapse
Affiliation(s)
- Diego A Rojas
- Instituto de Ciencias Biomédicas (ICB), Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910132, Chile
| | - Krishna Coronado
- Instituto de Ciencias Biomédicas (ICB), Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910132, Chile
| | - Diliana Pérez-Reytor
- Instituto de Ciencias Biomédicas (ICB), Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910132, Chile
| | - Eduardo Karahanian
- Instituto de Ciencias Biomédicas (ICB), Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910132, Chile
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago 8910132, Chile
| |
Collapse
|
8
|
Pommerolle L, Arif M, Behee M, Appolonia CN, Basu A, Wolf KM, Zawatsky CN, Johnson N, Rivellini O, Park JK, Cinar R. Chronic Alcohol Intake Compromises Lung Immunity by Altering Immunometabolism in Humans and Mouse Models. Am J Respir Cell Mol Biol 2024; 71:559-576. [PMID: 39024537 PMCID: PMC11568473 DOI: 10.1165/rcmb.2024-0086oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/02/2024] [Indexed: 07/20/2024] Open
Abstract
Chronic alcohol consumption disrupts lung immunity and host defense mechanisms, rendering individuals with alcohol use disorder more susceptible to developing inflammatory lung conditions with poor prognoses. Here, we focused on investigating the molecular and cellular effects of alcohol ingestion on lung immunity in male and female subjects using population-based human lung transcriptomics analysis and an experimental mouse model of chronic alcohol drinking using the National Institute on Alcohol Abuse and Alcoholism alcohol feeding model. Flow cytometry and transcriptomics analyses in lungs revealed a sexually dimorphic effect of chronic alcohol drinking on lung immunity in both human and mouse. Male lungs were more sensitive to chronic alcohol drinking-induced dysregulation of lung immunity compared with female lungs. Furthermore, comparative transcriptomics analysis using lungs and liver samples from matched human and mouse subjects demonstrated that lungs were more sensitive than liver to the effects of alcohol in downregulating immune-related genes and pathways. Furthermore, the transcriptomics analysis provided evidence that immunometabolic change is a central driver in lung alteration by downregulating the immune pathways and upregulating metabolic pathways. Chronic alcohol consumption resulted in reduced mTOR signaling and decreased immune cell populations. The mTOR signaling axis may serve as an upstream regulator of alcohol-induced dysregulation in lung immunity.
Collapse
Affiliation(s)
| | - Muhammad Arif
- Section on Fibrotic Disorders
- Laboratory of Cardiovascular Physiology and Tissue Injury, and
| | | | | | | | | | | | | | - Olivia Rivellini
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland
| | - Joshua K. Park
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland
| | | |
Collapse
|
9
|
Martins FRB, Beltrami VA, Zenóbio IC, Martins DG, da Silva Gurgel IL, de Assis Rabelo Ribeiro N, Queiroz-Junior CM, Bonaventura D, Rezende BM, Teixeira MM, Pinho V, Oliveira NL, Soriani FM. Chronic ethanol exposure decreases H3K27me3 in the Il6 promoter region of macrophages and generates persistent dysfunction on neutrophils during fungal infection. Inflamm Res 2024; 73:1747-1763. [PMID: 39127870 DOI: 10.1007/s00011-024-01928-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/17/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
OBJECTIVE AND DESIGN The aim of this study was to investigate the effects of ethanol exposure on epigenetic markers in bone marrow (BM) and their impact on inflammatory response during Aspergillus fumigatus infection. RESULTS Chronic ethanol exposure decreased H3K27me3 enrichment in the Il6 promoter region while increased H3K4me3 enrichment in Tnf. Chimeric mice were generated by transplanting BM from mice exposed to ethanol or water. Infection of ethanol-chimeric mice culminated in higher clinical scores, although there was no effect on mortality. However, previous chronic exposure to ethanol affects persistently the inflammatory response in lung tissue, demonstrated by increased lung damage, neutrophil accumulation and IL-6, TNF and CXCL2 production in ethanol-chimeric mice, resulting in a decreased neutrophil infiltration into the alveolar space. Neutrophil killing and phagocytosis were also significantly lower. Moreover, BM derived macrophages (BMDM) from ethanol-chimeric mice stimulated with A. fumigatus conidia exhibited higher levels of TNF, CXCL2 and IL-6 release and a higher killing activity. The Il6 promoter of BMDM from ethanol-chimeric mice exhibited a reduction in H3K27me3 enrichment, a finding also observed in BM donors exposed to ethanol. CONCLUSIONS These evidences demonstrate that prior chronic alcohol exposure of bone-marrow modify immune effector cells functions impairing the inflammatory response during A. fumigatus infection. These findings highlight the persistent impact of chronic ethanol exposure on infectious disease outcomes.
Collapse
Affiliation(s)
- Flávia Rayssa Braga Martins
- Department of Genetics, Ecology, and Evolution, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Vinicius Amorim Beltrami
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Isabelle Cruz Zenóbio
- Department of Genetics, Ecology, and Evolution, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Débora Gonzaga Martins
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Isabella Luísa da Silva Gurgel
- Department of Genetics, Ecology, and Evolution, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Daniella Bonaventura
- Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Barbara Maximino Rezende
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Pinho
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Nathalia Luisa Oliveira
- Department of Genetics, Ecology, and Evolution, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Microbiology and Immunology, McGill University, Montreal, Canada
| | - Frederico Marianetti Soriani
- Department of Genetics, Ecology, and Evolution, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
10
|
McGee MY, Enten GA, Boshra SN, Ogunsina O, Gaponenko V, Gao X, Majetschak M. Ethanol promotes protease activated receptor 1: Chemokine (C-X-C motif) receptor 4 heteromerization and enhances thrombin-induced impairment of human lung endothelial cell barrier function. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167335. [PMID: 38969148 PMCID: PMC11330351 DOI: 10.1016/j.bbadis.2024.167335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
Ethanol enhances the propensity of PAR1 and CXCR4 to form heteromers. Ethanol increases PAR1:CXCR4 heteromer expression in human lung microvascular endothelial cells (HULEC-5a). Ethanol enhances the efficacy of PAR1 to activate Gα12 upon thrombin stimulation in cells co-expressing CXCR4. Ethanol dose-dependently increases the efficacy of thrombin to impair HULEC-5a barrier function at clinically relevant concentrations. Interference with PAR1:CXCR4 heteromerization mitigates effects of ethanol on thrombin-induced impairment of HULEC-5a barrier function. Our findings provide a molecular mechanism that is likely to contribute to the increased risk of acute respiratory distress syndrome with alcohol abuse.
Collapse
Affiliation(s)
- Michelle Y McGee
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Garrett A Enten
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Sadia N Boshra
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA; Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Ololade Ogunsina
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL, USA
| | - Xianlong Gao
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Matthias Majetschak
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA; Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|
11
|
Song HW, Tian JH, Song HP, Guo SJ, Lin YH, Pan JS. Tracking multidrug resistant tuberculosis: a 30-year analysis of global, regional, and national trends. Front Public Health 2024; 12:1408316. [PMID: 39319291 PMCID: PMC11421170 DOI: 10.3389/fpubh.2024.1408316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/20/2024] [Indexed: 09/26/2024] Open
Abstract
Objectives To provide valuable insights for targeted interventions and resource allocation, our analysis delved into the multifaceted burden, trends, risks, and projections of multi drug resistant tuberculosis (MDR-TB). Methods This research employed data from the Global Burden of Disease (GBD) 2019 dataset, which used a comparative risk assessment to quantify the disease burden resulting from risk factors. Initially, this database was utilized to extract details concerning the disability-adjusted life years (DALYs), mortality, incidence, and the number of individuals afflicted by MDR-TB. Subsequently, regression analyses were conducted using the Joinpoint program to figure average annual percent change (AAPC) to ascertain the trend. Thirdly, the age-period-cohort model (APCM) was adopted to analyze evolutions in incidence and mortality. Finally, utilizing the Nordpred model within R software, we projected the incidence and mortality of MDR-TB from 2020 to 2030. Results MDR-TB remained a pressing global health concern in regions with lower socio-demographic indexes (SDI), where the AAPC in DALYs topped 7% from 1990 to 2019. In 2019, the cumulative DALYs attributed to MDR-TB tallied up to 4.2 million, with India, the Russian Federation, and China bearing the brunt. Notably, the incidence rates have shown a steadfast presence over the past decade, and a troubling forecast predicts an uptick in these areas from 2020 to 2030. Additionally, the risk of contracting MDR-TB grew with advancing age, manifesting most acutely among men aged 40+ in lower SDI regions. Strikingly, alcohol consumption had been identified as a significant contributor, surpassing the impacts of smoking and high fasting plasma glucose, leading to 0.7 million DALYs in 2019. Conclusions A robust strategy is needed to end tuberculosis (TB) by 2030, especially in lower SDI areas.
Collapse
Affiliation(s)
- Hui-Wen Song
- Department of Infectious Diseases, Sanming First Hospital Affiliated to Fujian Medical University, Sanming, Fujian, China
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Department of Hepatology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jian-Hua Tian
- Department of Infectious Diseases, Sanming First Hospital Affiliated to Fujian Medical University, Sanming, Fujian, China
| | - Hui-Ping Song
- Department of Infectious Diseases, Sanming First Hospital Affiliated to Fujian Medical University, Sanming, Fujian, China
| | - Si-Jie Guo
- Department of Infectious Diseases, Sanming First Hospital Affiliated to Fujian Medical University, Sanming, Fujian, China
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
| | - Ye-Hong Lin
- Department of Infectious Diseases, Sanming First Hospital Affiliated to Fujian Medical University, Sanming, Fujian, China
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
| | - Jin-Shui Pan
- Department of Hepatology, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Hepatology Research Institute, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Liver and Intestinal Diseases, Fuzhou, Fujian, China
| |
Collapse
|
12
|
Narasimha VL, Nath S, Alam B, Kumari B, Kumari P, Kumari S, Kaur J, Varshney S. Prevalence and association between alcohol, tobacco, and COVID-19: a study from a tribal predominant district in eastern India. Front Public Health 2024; 12:1415178. [PMID: 39220445 PMCID: PMC11361974 DOI: 10.3389/fpubh.2024.1415178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Alcohol and tobacco use has been proposed to significantly affect COVID-19 outcomes. The study aimed to estimate the prevalence of alcohol and tobacco use among COVID-19-positive patients and compare it with the general population prevalence rates. It also aimed to assess and determine the association between the severity of COVID-19 illness and the complications with alcohol and tobacco use. Method For this, a cross-sectional, retrospective, telephone-based study was conducted using a structured questionnaire among COVID-19 diagnosed patients in the district of Deoghar of the Indian state of Jharkhand. A multinomial logistic regression is done to determine the association. Results Among 1,425 patients interviewed, tobacco and alcohol were used by 22.31 and 9.96%, significantly more than the prevalence of tobacco (Z = 4.9485, p < 0.00001) and alcohol use (Z = 7.118, p < 0.00001), respectively, in the district (tobacco-11.7% and alcohol-4.8%).In a regression model, patients with co-morbidity had higher odds of severe [3.34 (1.99-5.62)] and moderate [2.95 (1.97-4.41)] COVID-19. Young [0.12 (0.04-0.38)] and middle-aged individuals [0.23 (0.13-0.4)], people below the poverty line 0.28 (0.11-0.69) are at lower odds of severe COVID-19. Tobacco users [1.58 (1.16-2.14)], alcohol users [1.53 (1.03-2.28)], incomplete vaccination [3.24 (1.49-7.01)], and patients with comorbidity [3.6 (2.79-4.68)] were found to have higher odds of post-COVID-19 complications. Discussion People with COVID-19 in our study population had significantly higher tobacco and alcohol use compared to the general population. Tobacco and alcohol use significantly increases the risk of post-COVID-19 complications. The study highlights the need for addiction treatment services to prevent complications during future pandemics.
Collapse
Affiliation(s)
| | - Santanu Nath
- All India Institute of Medical Sciences, Deoghar, India
| | - Benazir Alam
- All India Institute of Medical Sciences, Deoghar, India
| | - Bipasa Kumari
- All India Institute of Medical Sciences, Deoghar, India
| | - Pooja Kumari
- All India Institute of Medical Sciences, Deoghar, India
| | | | - Jagdish Kaur
- World Health Organization-South-East Asia Regional Office, New Delhi, India
| | | |
Collapse
|
13
|
Ma Z, Wu S, Guo Y, Ouyang S, Wang N. Association of advanced lung cancer inflammation index with all-cause and cardiovascular mortality in US patients with rheumatoid arthritis. Front Nutr 2024; 11:1397326. [PMID: 38873560 PMCID: PMC11169605 DOI: 10.3389/fnut.2024.1397326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/16/2024] [Indexed: 06/15/2024] Open
Abstract
Introduction As a systemic autoimmune disorder, the prognosis of rheumatoid arthritis (RA) is intricately linked to inflammation. This study aimed to investigate the association between the advanced lung cancer inflammation index (ALI), a comprehensive indicator of inflammation combined with nutritional status, and all-cause and cardiovascular mortality among patients diagnosed with RA. Methods The 2,305 RA patients from NHANES (2001-2018) included in the analysis were categorized into three groups according to ALI tertiles. Weighted Kaplan-Meier and multivariate COX regression analyses evaluated the relationship between ALI and mortality. The time-dependent characteristic curve (ROC) was used to assess the prediction accuracy of ALI. Results During a median follow-up of 7.92 years, 591 participants died from all causes, including 197 from cardiovascular diseases. Increased ALI was associated with a decreased probability of death. The full COX model revealed lower all-cause mortality hazard risks in the T2 (HR: 0.67, 95%CI: 0.54-0.83) and T3 (HR: 0.47 95%CI: 0.33-0.67, p for tend <0.001) groups compared to T1, and the risk of cardiovascular mortality was also lower in the groups of T2 (HR: 0.47, 95%CI: 0.31-0.70) and T3 (HR: 0.34, 95%CI: 0.19-0.62, p for trend <0.001). Furthermore, the ROC analysis underscored the strong predictive capability of ALI (AUC for 1-year all-cause and cardiovascular mortality were 0.73 and 0.79, respectively). Conclusion This cohort study demonstrated the higher accuracy of ALI in predicting mortality in RA patients, highlighting the important clinical value of ALI in risk assessment and prognosis evaluation.
Collapse
Affiliation(s)
- Zhuang Ma
- School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Shixin Wu
- School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Yitong Guo
- School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Shiyi Ouyang
- School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Ningning Wang
- School of Public Health, Guangzhou Medical University, Guangzhou, China
- Department of Health Statistics, School of Public Health, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
14
|
Lu H. Inflammatory liver diseases and susceptibility to sepsis. Clin Sci (Lond) 2024; 138:435-487. [PMID: 38571396 DOI: 10.1042/cs20230522] [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: 09/03/2023] [Revised: 01/09/2024] [Accepted: 03/12/2024] [Indexed: 04/05/2024]
Abstract
Patients with inflammatory liver diseases, particularly alcohol-associated liver disease and metabolic dysfunction-associated fatty liver disease (MAFLD), have higher incidence of infections and mortality rate due to sepsis. The current focus in the development of drugs for MAFLD is the resolution of non-alcoholic steatohepatitis and prevention of progression to cirrhosis. In patients with cirrhosis or alcoholic hepatitis, sepsis is a major cause of death. As the metabolic center and a key immune tissue, liver is the guardian, modifier, and target of sepsis. Septic patients with liver dysfunction have the highest mortality rate compared with other organ dysfunctions. In addition to maintaining metabolic homeostasis, the liver produces and secretes hepatokines and acute phase proteins (APPs) essential in tissue protection, immunomodulation, and coagulation. Inflammatory liver diseases cause profound metabolic disorder and impairment of energy metabolism, liver regeneration, and production/secretion of APPs and hepatokines. Herein, the author reviews the roles of (1) disorders in the metabolism of glucose, fatty acids, ketone bodies, and amino acids as well as the clearance of ammonia and lactate in the pathogenesis of inflammatory liver diseases and sepsis; (2) cytokines/chemokines in inflammatory liver diseases and sepsis; (3) APPs and hepatokines in the protection against tissue injury and infections; and (4) major nuclear receptors/signaling pathways underlying the metabolic disorders and tissue injuries as well as the major drug targets for inflammatory liver diseases and sepsis. Approaches that focus on the liver dysfunction and regeneration will not only treat inflammatory liver diseases but also prevent the development of severe infections and sepsis.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, U.S.A
| |
Collapse
|
15
|
Winkler R, Lu H. Cell-Specific Regulation of Inflammatory Cytokines and Acute-Phase Proteins by the Glucocorticoid Receptor. Mediators Inflamm 2023; 2023:4399998. [PMID: 39619227 PMCID: PMC11606692 DOI: 10.1155/2023/4399998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/08/2023] [Accepted: 10/24/2023] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Literature and data mining found abnormal induction of chemokine (C-X-C motif) ligand 1 (CXCL1) and CXCL8 and down-regulation of CXCL2 in inflammatory liver diseases. This study was performed to understand the glucocorticoid receptor's (GR's) effects on chemokine and acute-phase protein expression in human liver, in settings of bacterial infection (modeled using LPS) or inflammation (modeled using TNFα). METHODS Primary human hepatocytes (PHH) were treated with combinations of tumor necrosis factor alpha (TNFα), lipopolysaccharide (LPS), and dexamethasone (DEX) for 24 h, following which chemokine mRNA and protein expression were analyzed using qPCR and enzyme-linked immunosorbent assay assays. Dual luciferase assays were performed on transfected cell lines. Mutant CXCL2 promoters were used in dual luciferase assays to identify specific regions of the CXCL2 promoter affected by GR, TNFα, or hepatocyte nuclear factor 4α (HNF4α, a liver-enriched transcription factor). RESULTS In PHH from donor 1, GR strongly inhibited LPS-induced CXCL1 and CXCL8 translation and transcription, whereas CXCL2 transcription tended to increase with DEX treatment. In PHH from donor 2, DEX treatment inhibited protein expression and secretion of CXCL1 and CXCL8 induced by TNFα and/or LPS, whereas CXCL2 upregulation was largely unaffected by DEX treatment. In nonliver HEK293T cells GR activity inhibited CXCL2 promoter activity. However, in liver-derived HEPG2 cells, GR induced CXCL2 promoter activity. A 407-base pair region upstream of CXCL2 promoter is necessary for full GR functionality in HEPG2 cells. TNFα synergized with HNF4α in inducing CXCL2 promoter activity in HEPG2 cells. CONCLUSIONS GR's effects on chemokine expression are cell-type specific and chemokine specific. GR down-regulated CXCL1 and CXCL8 in different cell types, whereas the specific activation of CXCL2 in hepatocytes and down-regulation of CXCL2 in nonhepatocytes by GR appears due to cell-specific utilization of CXCL2 promoter. By specifically increasing GR activity in the liver, we may normalize chemokine imbalances and prevent sepsis in inflammatory liver diseases.
Collapse
Affiliation(s)
- Rebecca Winkler
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
16
|
Zhang Y, Fang XM. The pan-liver network theory: From traditional chinese medicine to western medicine. CHINESE J PHYSIOL 2023; 66:401-436. [PMID: 38149555 DOI: 10.4103/cjop.cjop-d-22-00131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023] Open
Abstract
In traditional Chinese medicine (TCM), the liver is the "general organ" that is responsible for governing/maintaining the free flow of qi over the entire body and storing blood. According to the classic five elements theory, zang-xiang theory, yin-yang theory, meridians and collaterals theory, and the five-viscera correlation theory, the liver has essential relationships with many extrahepatic organs or tissues, such as the mother-child relationships between the liver and the heart, and the yin-yang and exterior-interior relationships between the liver and the gallbladder. The influences of the liver to the extrahepatic organs or tissues have been well-established when treating the extrahepatic diseases from the perspective of modulating the liver by using the ancient classic prescriptions of TCM and the acupuncture and moxibustion. In modern medicine, as the largest solid organ in the human body, the liver has the typical functions of filtration and storage of blood; metabolism of carbohydrates, fats, proteins, hormones, and foreign chemicals; formation of bile; storage of vitamins and iron; and formation of coagulation factors. The liver also has essential endocrine function, and acts as an immunological organ due to containing the resident immune cells. In the perspective of modern human anatomy, physiology, and pathophysiology, the liver has the organ interactions with the extrahepatic organs or tissues, for example, the gut, pancreas, adipose, skeletal muscle, heart, lung, kidney, brain, spleen, eyes, skin, bone, and sexual organs, through the circulation (including hemodynamics, redox signals, hepatokines, metabolites, and the translocation of microbiota or its products, such as endotoxins), the neural signals, or other forms of pathogenic factors, under normal or diseases status. The organ interactions centered on the liver not only influence the homeostasis of these indicated organs or tissues, but also contribute to the pathogenesis of cardiometabolic diseases (including obesity, type 2 diabetes mellitus, metabolic [dysfunction]-associated fatty liver diseases, and cardio-cerebrovascular diseases), pulmonary diseases, hyperuricemia and gout, chronic kidney disease, and male and female sexual dysfunction. Therefore, based on TCM and modern medicine, the liver has the bidirectional interaction with the extrahepatic organ or tissue, and this established bidirectional interaction system may further interact with another one or more extrahepatic organs/tissues, thus depicting a complex "pan-hepatic network" model. The pan-hepatic network acts as one of the essential mechanisms of homeostasis and the pathogenesis of diseases.
Collapse
Affiliation(s)
- Yaxing Zhang
- Department of Physiology; Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong; Issue 12th of Guangxi Apprenticeship Education of Traditional Chinese Medicine (Shi-Cheng Class of Guangxi University of Chinese Medicine), College of Continuing Education, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xian-Ming Fang
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine (Guangxi Hospital of Integrated Chinese Medicine and Western Medicine, Ruikang Clinical Faculty of Guangxi University of Chinese Medicine), Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
17
|
Lewis SA, Cinco IR, Doratt BM, Blanton MB, Hoagland C, Newman N, Davies M, Grant KA, Messaoudi I. Chronic alcohol consumption dysregulates innate immune response to SARS-CoV-2 in the lung. EBioMedicine 2023; 97:104812. [PMID: 37793211 PMCID: PMC10562860 DOI: 10.1016/j.ebiom.2023.104812] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Alcohol consumption is widespread with over half of the individuals over 18 years of age in the U.S. reporting alcohol use in the last 30 days. Moreover, 9 million Americans engaged in binge or chronic heavy drinking (CHD) in 2019. CHD negatively impacts pathogen clearance and tissue repair, including in the respiratory tract, thereby increasing susceptibility to infection. Although, it has been hypothesized that chronic alcohol consumption negatively impacts COVID-19 outcomes; the interplay between chronic alcohol use and SARS-CoV-2 infection outcomes has yet to be elucidated. METHODS In this study we employed luminex, scRNA sequencing, and flow cytometry to investigate the impact of chronic alcohol consumption on SARS-CoV-2 anti-viral responses in bronchoalveolar lavage cell samples from humans with alcohol use disorder and rhesus macaques that engaged in chronic drinking. FINDINGS Our data show that in both humans (n = 6) and macaques (n = 11), the induction of key antiviral cytokines and growth factors was decreased with chronic ethanol consumption. Moreover, in macaques fewer differentially expressed genes mapped to Gene Ontology terms associated with antiviral immunity following 6 month of ethanol consumption while TLR signaling pathways were upregulated. INTERPRETATION These data are indicative of aberrant inflammation and reduced antiviral responses in the lung with chronic alcohol drinking. FUNDING This study was supported by NIH 1R01AA028735-04 (Messaoudi), U01AA013510-20 (Grant), R24AA019431-14 (Grant), R24AA019661 (Burnham), P-51OD011092 (ONPRC core grant support). The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.
Collapse
Affiliation(s)
- Sloan A Lewis
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine, USA
| | - Isaac R Cinco
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, USA
| | - Brianna M Doratt
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, USA
| | - Madison B Blanton
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, USA; Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, USA
| | - Cherise Hoagland
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, USA
| | - Natali Newman
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, USA
| | - Michael Davies
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, USA
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, USA
| | - Ilhem Messaoudi
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, USA.
| |
Collapse
|
18
|
Chaudhary MR, Chaudhary S, Sharma Y, Singh TA, Mishra AK, Sharma S, Mehdi MM. Aging, oxidative stress and degenerative diseases: mechanisms, complications and emerging therapeutic strategies. Biogerontology 2023; 24:609-662. [PMID: 37516673 DOI: 10.1007/s10522-023-10050-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/28/2023] [Indexed: 07/31/2023]
Abstract
Aging accompanied by several age-related complications, is a multifaceted inevitable biological progression involving various genetic, environmental, and lifestyle factors. The major factor in this process is oxidative stress, caused by an abundance of reactive oxygen species (ROS) generated in the mitochondria and endoplasmic reticulum (ER). ROS and RNS pose a threat by disrupting signaling mechanisms and causing oxidative damage to cellular components. This oxidative stress affects both the ER and mitochondria, causing proteopathies (abnormal protein aggregation), initiation of unfolded protein response, mitochondrial dysfunction, abnormal cellular senescence, ultimately leading to inflammaging (chronic inflammation associated with aging) and, in rare cases, metastasis. RONS during oxidative stress dysregulate multiple metabolic pathways like NF-κB, MAPK, Nrf-2/Keap-1/ARE and PI3K/Akt which may lead to inappropriate cell death through apoptosis and necrosis. Inflammaging contributes to the development of inflammatory and degenerative diseases such as neurodegenerative diseases, diabetes, cardiovascular disease, chronic kidney disease, and retinopathy. The body's antioxidant systems, sirtuins, autophagy, apoptosis, and biogenesis play a role in maintaining homeostasis, but they have limitations and cannot achieve an ideal state of balance. Certain interventions, such as calorie restriction, intermittent fasting, dietary habits, and regular exercise, have shown beneficial effects in counteracting the aging process. In addition, interventions like senotherapy (targeting senescent cells) and sirtuin-activating compounds (STACs) enhance autophagy and apoptosis for efficient removal of damaged oxidative products and organelles. Further, STACs enhance biogenesis for the regeneration of required organelles to maintain homeostasis. This review article explores the various aspects of oxidative damage, the associated complications, and potential strategies to mitigate these effects.
Collapse
Affiliation(s)
- Mani Raj Chaudhary
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Sakshi Chaudhary
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Yogita Sharma
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Thokchom Arjun Singh
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Alok Kumar Mishra
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Shweta Sharma
- Chitkara School of Health Sciences, Chitkara University, Chandigarh, Punjab, 140401, India
| | - Mohammad Murtaza Mehdi
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| |
Collapse
|
19
|
McMahan RH, Anton P, Coleman LG, Cresci GAM, Crews FT, Crotty KM, Luck ME, Molina PE, Vachharajani V, Weinberg J, Yeligar SM, Choudhry MA, McCullough RL, Kovacs EJ. Alcohol and Immunology: Mechanisms of multi-organ damage. Summary of the 2022 alcohol and Immunology research interest group (AIRIG) meeting. Alcohol 2023; 110:57-63. [PMID: 37061143 PMCID: PMC10330898 DOI: 10.1016/j.alcohol.2023.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/17/2023]
Abstract
On October 26th, 2022 the annual Alcohol and Immunology Research Interest Group (AIRIG) meeting was held as a satellite symposium at the annual meeting of the Society for Leukocyte Biology in Hawaii. The 2022 meeting focused broadly on the immunological consequences of acute, chronic, and prenatal alcohol exposure and how these contribute to damage in multiple organs and tissues. These included alcohol-induced neuroinflammation, impaired lung immunity, intestinal dysfunction, and decreased anti-microbial and anti-viral responses. In addition, research presented covered multiple pathways behind alcohol-induced cellular dysfunction, including mitochondrial metabolism, cellular bioenergetics, gene regulation, and epigenetics. Finally, the work presented highlighted potential biomarkers and novel avenues of treatment for alcohol-induced organ damage.
Collapse
Affiliation(s)
- Rachel H McMahan
- Division of GI, Trauma, and Endocrine Surgery, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Alcohol Research Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| | - Paige Anton
- Alcohol Research Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Leon G Coleman
- Department of Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Gail A M Cresci
- Departments of Pediatric Gastroenterology, Hepatology & Nutrition, Cleveland Clinic Children's Hospital and Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Fulton T Crews
- Department of Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Kathryn M Crotty
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, United States; Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Marisa E Luck
- Alcohol Research Program, Burn & Shock Trauma Research Institute, Department of Surgery, Integrative Cell Biology Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL, United States
| | - Patricia E Molina
- Department of Physiology and Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Vidula Vachharajani
- Department of Inflammation and Immunity, Critical Care Medicine, Respiratory Institute, Lerner Research Institute, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, United States
| | - Joanne Weinberg
- Department of Cellular & Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Samantha M Yeligar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, United States; Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Mashkoor A Choudhry
- Alcohol Research Program, Burn & Shock Trauma Research Institute, Department of Surgery, Integrative Cell Biology Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL, United States
| | - Rebecca L McCullough
- Alcohol Research Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Elizabeth J Kovacs
- Division of GI, Trauma, and Endocrine Surgery, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Alcohol Research Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Molecular Biology Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States; Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| |
Collapse
|
20
|
Wang C, Ramasamy A, Verduzco-Gutierrez M, Brode WM, Melamed E. Acute and post-acute sequelae of SARS-CoV-2 infection: a review of risk factors and social determinants. Virol J 2023; 20:124. [PMID: 37328773 PMCID: PMC10276420 DOI: 10.1186/s12985-023-02061-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 05/02/2023] [Indexed: 06/18/2023] Open
Abstract
SARS-CoV-2 infection leading to Coronavirus Disease 2019 (COVID-19) has caused more than 762 million infections worldwide, with 10-30% of patients suffering from post-acute sequelae of SARS-CoV-2 infections (PASC). Initially thought to primarily affect the respiratory system, it is now known that SARS-CoV-2 infection and PASC can cause dysfunction in multiple organs, both during the acute and chronic stages of infection. There are also multiple risk factors that may predispose patients to worse outcomes from acute SARS-CoV-2 infection and contribute to PASC, including genetics, sex differences, age, reactivation of chronic viruses such as Epstein Barr Virus (EBV), gut microbiome dysbiosis, and behavioral and lifestyle factors, including patients' diet, alcohol use, smoking, exercise, and sleep patterns. In addition, there are important social determinants of health, such as race and ethnicity, barriers to health equity, differential cultural perspectives and biases that influence patients' access to health services and disease outcomes from acute COVID-19 and PASC. Here, we review risk factors in acute SARS-CoV-2 infection and PASC and highlight social determinants of health and their impact on patients affected with acute and chronic sequelae of COVID-19.
Collapse
Affiliation(s)
- Chumeng Wang
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Akshara Ramasamy
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Monica Verduzco-Gutierrez
- Department of Physical Medicine and Rehabilitation, University of Texas at San Antonio, San Antonio, TX, USA
| | - W Michael Brode
- Department of Internal Medicine, Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Esther Melamed
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
21
|
Tong NH, Salvatori P. Positive Correlation Between Heavy Alcoholic Drinking and SARS-Cov-2 Non-Infection Rate. Cureus 2023; 15:e40130. [PMID: 37304380 PMCID: PMC10249648 DOI: 10.7759/cureus.40130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction During the SARS-CoV-2 pandemic, rumors claimed that alcohol drinking could someway be useful in contrasting the contagion and even the disease. It appears opportune to bring some robust data to determine whether heavy alcohol drinkers and non-drinkers experienced different infection rates. Methods A cross-sectional study through a simple survey based on the social media software Weixin and the mini survey program Wenjuanxing was carried out in China after the zero-Covid policy ended, namely from 15:00 January 1, 2023, to 12:35 January 3, 2023. The evaluation was conducted among subjects belonging to the first author's Weixin community, mostly residents in the higher populated China area. Study participants received a questionary and were asked about their virus infection status, and were classified into two groups: (a) infected, meaning he/she has been infected at least once (whether recovered or not); (b) remain uninfected, meaning the virus has not infected him/her. A total of 211 subjects adhered to the survey. Alcoholic drinking behavior about liquors with no less than 40% alcohol content in volume was retrieved from the participants. In China, such beverages are almost uniquely referred to as the Chinese Spirits or BaiJiu. The frequency of drinking quantified the drinking behavior, and it is classified into three groups: never drink or drink occasionally (group A); drink one or two times per week (group B); drink three times per week or more often (group C). The hypothesis of an existing relationship between infection status and drinking behavior was advanced before data collection. The numbers of the uninfected people in each of the three drinking groups were counted, and the rates of not-infection were calculated. The rates are compared with each other to conclude whether significant differences exist, considering the size of the samples. The conclusion is drawn from standard hypothesis testing. Results The male/female ratio was 108/103 (51.2% and 48.8%), the mean age was 38.8 years (range 21-68), and the median age of 37.4 years. The total 211 participants fell into three groups with different drinking frequencies, with counts (percentages in total 211 participants) 139 (65.9%) in group A, 28 (13.3%) in group B, and 44 (20.8%) in group C. The number (percentage within the group) of uninfected members in groups A, B, and C are 29 (20.9%), 7 (25.0%), and 17 (38.6%), respectively. The statistical analysis through the Cochran-Armitage trend test gave a significative result: p=0.0209. Conclusions Within the methodology's limitations, this study shows the significant relationship between alcohol drinking habits and the chances of avoiding SARS-CoV-2 infection. A possible hypothesis explaining these findings is advanced. However, the authors warn about misleading conclusions and advocate research that could properly guide ethanol use in the present and other possible pandemics. Limitations This study is based on self-reported data from a specific community in China. There could be recall bias and social desirability bias, and the generalizability of the findings to other populations could be limited. Other factors that could influence infection rates, such as age, occupation, and health status, are not controlled in the present study. There could be other explanations for the observed relationship between alcohol drinking habits and infection rates.
Collapse
Affiliation(s)
- Ning-Hua Tong
- Department of Physics, Renmin University of China, Beijing, CHN
| | | |
Collapse
|
22
|
Martins FRB, de Oliveira MD, Souza JAM, Queiroz-Junior CM, Lobo FP, Teixeira MM, Malacco NL, Soriani FM. Chronic ethanol exposure impairs alveolar leukocyte infiltration during pneumococcal pneumonia, leading to an increased bacterial burden despite increased CXCL1 and nitric oxide levels. Front Immunol 2023; 14:1175275. [PMID: 37275853 PMCID: PMC10235596 DOI: 10.3389/fimmu.2023.1175275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/05/2023] [Indexed: 06/07/2023] Open
Abstract
Ethanol abuse is a risk factor for the development of pneumonia caused by Streptococcus pneumoniae, a critical pathogen for public health. The aim of this article was to investigate the inflammatory mechanisms involved in pneumococcal pneumonia that may be associated with chronic ethanol exposure. Male C57BL6/J-Unib mice were exposed to 20% (v/v) ethanol for twelve weeks and intranasally infected with 5x104 CFU of S. pneumoniae. Twenty-four hours after infection, lungs, bronchoalveolar lavage and blood samples were obtained to assess the consequences of chronic ethanol exposure during infection. Alcohol-fed mice showed increased production of nitric oxide and CXCL1 in alveoli and plasma during pneumococcal pneumonia. Beside this, ethanol-treated mice exhibited a decrease in leukocyte infiltration into the alveoli and reduced frequency of severe lung inflammation, which was associated with an increase in bacterial load. Curiously, no changes were observed in survival after infection. Taken together, these results demonstrate that chronic ethanol exposure alters the inflammatory response during S. pneumoniae lung infection in mice with a reduction in the inflammatory infiltrate even in the presence of higher levels of the chemoattractant CXCL1.
Collapse
Affiliation(s)
- Flávia Rayssa Braga Martins
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maycon Douglas de Oliveira
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jéssica Amanda Marques Souza
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Francisco Pereira Lobo
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Frederico Marianetti Soriani
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
23
|
Lewis SA, Cinco IR, Doratt BM, Blanton MB, Hoagland C, Davies M, Grant KA, Messaoudi I. Chronic alcohol consumption dysregulates innate immune response to SARS-CoV-2 in the lung. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.02.539139. [PMID: 37205543 PMCID: PMC10187161 DOI: 10.1101/2023.05.02.539139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Alcohol consumption is widespread with over half of the individuals over 18 years of age in the U.S. reporting alcohol use in the last 30 days. Moreover, 9 million Americans engaged in binge or chronic heavy drinking (CHD) in 2019. CHD negatively impacts pathogen clearance and tissue repair, including in the respiratory tract, thereby increasing susceptibility to infection. Although, it has been hypothesized that chronic alcohol consumption negatively impacts COVID-19 outcomes; the interplay between chronic alcohol use and SARS-CoV-2 infection outcomes has yet to be elucidated. Therefore, in this study we investigated the impact of chronic alcohol consumption on SARS-CoV-2 anti-viral responses in bronchoalveolar lavage cell samples from humans with alcohol use disorder and rhesus macaques that engaged in chronic drinking. Our data show that in both humans and macaques, the induction of key antiviral cytokines and growth factors was decreased with chronic ethanol consumption. Moreover, in macaques fewer differentially expressed genes mapped to Gene Ontology terms associated with antiviral immunity following 6 month of ethanol consumption while TLR signaling pathways were upregulated. These data are indicative of aberrant inflammation and reduced antiviral responses in the lung with chronic alcohol drinking.
Collapse
Affiliation(s)
- Sloan A. Lewis
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California Irvine
| | - Isaac R. Cinco
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky
| | - Brianna M. Doratt
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky
| | - Madison B. Blanton
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky
| | - Cherise Hoagland
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University
| | - Michael Davies
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University
| | - Kathleen A. Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University
| | - Ilhem Messaoudi
- Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky
| |
Collapse
|
24
|
Pontes Pereira TT, Fideles Duarte-Andrade F, Gardone Vitório J, do Espírito Santo Pereira T, Braga Martins FR, Marques Souza JA, Malacco NL, Mathias Melo E, Costa Picossi CR, Pinto E, Santiago Gomez R, Martins Teixeira M, Nori de Macedo A, André Baptista Canuto G, Soriani FM. Chronic alcohol administration alters metabolomic profile of murine bone marrow. Front Immunol 2023; 14:1128352. [PMID: 37090737 PMCID: PMC10113543 DOI: 10.3389/fimmu.2023.1128352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/06/2023] [Indexed: 04/25/2023] Open
Abstract
Introduction People with hazardous alcohol use are more susceptible to viral, bacterial, and fungal infections due to the effect of alcohol on immune system cell function. Metabolized ethanol reduces NAD+ to NADH, affecting critical metabolic pathways. Here, our aim was to investigate whether alcohol is metabolized by bone marrow cells and if it impacts the metabolic pathways of leukocyte progenitor cells. This is said to lead to a qualitative and quantitative alteration of key metabolites which may be related to the immune response. Methods We addressed this aim by using C57BL/6 mice under chronic ethanol administration and evaluating the metabolomic profile of bone marrow total cells by gas chromatography-coupled mass spectrometry (GC-MS). Results We identified 19 metabolites. Our data demonstrated that chronic ethanol administration alters the metabolomic profile in the bone marrow, resulting in a statistically diminished abundance of five metabolites in ethanol-treated animals: uracil, succinate, proline, nicotinamide, and tyrosine. Discussion Our results demonstrate for the first time in the literature the effects of alcohol consumption on the metabolome content of hematopoietic tissue and open a wide range of further studies to investigate mechanisms by which alcohol compromises the cellular function of the immune system.
Collapse
Affiliation(s)
| | | | - Jéssica Gardone Vitório
- Department of Clinic, Pathology and Dental Surgery, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | | - Eliza Mathias Melo
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Ernani Pinto
- Nuclear Energy Center in Agriculture, Escola Superior de Agricultura Luiz de Queiroz, University of São Paulo, Piracicaba, Brazil
| | - Ricardo Santiago Gomez
- Department of Clinic, Pathology and Dental Surgery, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Gisele André Baptista Canuto
- Department of Analytical Chemistry of the Institute of Chemistry, Universidade Federal da Bahia, Salvador, Brazil
| | | |
Collapse
|
25
|
Lai T, Zhu Y, Lu N, He W. Alcohol Consumption within 48 hours before Onset Is Associated with Adverse Clinical Outcomes in Hypertriglyceridemic Pancreatitis. J Clin Med 2023; 12:jcm12072566. [PMID: 37048649 PMCID: PMC10095359 DOI: 10.3390/jcm12072566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/26/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
(1) Background: Some patients with hypertriglyceridemic pancreatitis (HTGP) drink occasionally or moderately, but do not meet the diagnostic criteria for alcoholic pancreatitis. This study aims to investigate whether occasional or moderate alcohol consumption affects the clinical outcomes of patients with HTGP. (2) Methods: This retrospective study included 373 patients with HTGP from January 2007 to December 2021. HTGP patients with occasional or moderate alcohol (OMA) consumption before onset were divided into the OMA group, and HTGP patients without alcohol (WA) consumption were divided into the WA group. The OMA group was further divided into two groups: the drinking within 48 h before onset (DW) group, and the without drinking within 48 h before onset (WDW) group. The clinical data of the two groups were compared and multivariable logistic regression was used to analyze independent risk factors for the primary outcomes. (3) Results: The proportion of men (95.7% vs. 67.6%, p < 0.001) and smoking history (61.7% vs. 15.1%, p < 0.001) in the OMA group were higher than those in the WA group. Occasional or moderate alcohol consumption was independently associated with a high incidence of SAP (adjusted odds ratio (AdjOR), 1.57; 95% CI, 1.02–2.41; p = 0.041), and necrotizing pancreatitis (AdjOR, 1.60; 95% CI, 1.04–2.48; p = 0.034). After dividing the OMA group into two subgroups, we found that drinking within 48 h before onset was independently associated with a high incidence of SAP (AdjOR, 3.09; 95% CI, 1.66–5.77; p < 0.001), and necrotizing pancreatitis (AdjOR, 2.71; 95% CI, 1.46–5.05; p = 0.002). (4) Conclusion: Occasional or moderate alcohol consumption is associated with poor clinical outcomes in patients with HTGP, particularly if they drank alcohol within 48 h before the onset of the disease.
Collapse
Affiliation(s)
| | | | | | - Wenhua He
- Correspondence: ; Tel.: +86-13879182642
| |
Collapse
|
26
|
Muralidharan A, Bauer CD, Katafiasz DM, Strah HM, Siddique A, Reid SP, Bailey KL, Wyatt TA. Synergistic Detrimental Effects of Cigarette Smoke, Alcohol, and SARS-CoV-2 in COPD Bronchial Epithelial Cells. Pathogens 2023; 12:498. [PMID: 36986420 PMCID: PMC10056639 DOI: 10.3390/pathogens12030498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/09/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Lung conditions such as COPD, as well as risk factors such as alcohol misuse and cigarette smoking, can exacerbate COVID-19 disease severity. Synergistically, these risk factors can have a significant impact on immunity against pathogens. Here, we studied the effect of a short exposure to alcohol and/or cigarette smoke extract (CSE) in vitro on acute SARS-CoV-2 infection of ciliated human bronchial epithelial cells (HBECs) collected from healthy and COPD donors. We observed an increase in viral titer in CSE- or alcohol-treated COPD HBECs compared to untreated COPD HBECs. Furthermore, we treated healthy HBECs accompanied by enhanced lactate dehydrogenase activity, indicating exacerbated injury. Finally, IL-8 secretion was elevated due to the synergistic damage mediated by alcohol, CSE, and SARS-CoV-2 in COPD HBECs. Together, our data suggest that, with pre-existing COPD, short exposure to alcohol or CSE is sufficient to exacerbate SARS-CoV-2 infection and associated injury, impairing lung defences.
Collapse
Affiliation(s)
- Abenaya Muralidharan
- Department of Pathology and Microbiology, College of Medicine, The University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Christopher D. Bauer
- Pulmonary, Critical Care, and Sleep Medicine Division, Department of Internal Medicine, College of Medicine, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Dawn M. Katafiasz
- Pulmonary, Critical Care, and Sleep Medicine Division, Department of Internal Medicine, College of Medicine, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Heather M. Strah
- Pulmonary, Critical Care, and Sleep Medicine Division, Department of Internal Medicine, College of Medicine, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Aleem Siddique
- Department of Surgery, College of Medicine, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - St Patrick Reid
- Department of Pathology and Microbiology, College of Medicine, The University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kristina L. Bailey
- Pulmonary, Critical Care, and Sleep Medicine Division, Department of Internal Medicine, College of Medicine, the University of Nebraska Medical Center, Omaha, NE 68198, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Todd A. Wyatt
- Pulmonary, Critical Care, and Sleep Medicine Division, Department of Internal Medicine, College of Medicine, the University of Nebraska Medical Center, Omaha, NE 68198, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
- Department of Environmental, Agricultural & Occupational Health, College of Public Health, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
27
|
Yeligar SM, Harris FL, Brown LAS, Hart CM. Pharmacological reversal of post-transcriptional alterations implicated in alcohol-induced alveolar macrophage dysfunction. Alcohol 2023; 106:30-43. [PMID: 36328183 PMCID: PMC10080543 DOI: 10.1016/j.alcohol.2022.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
Alcohol use disorders (AUD) cause alveolar macrophage (AM) immune dysfunction and increase risk of lung infections. Excessive alcohol use causes AM oxidative stress, which impairs AM phagocytosis and pathogen clearance from the alveolar space. Alcohol induces expression of NADPH oxidases (Noxes), primary sources of oxidative stress in AM. In contrast, alcohol decreases AM peroxisome proliferator-activated receptor gamma (PPARγ), a critical regulator of AM immune function. To explore the underlying molecular mechanisms for these effects of alcohol, we hypothesized that ethanol promotes CCAAT/enhancer-binding protein beta (C/EBPβ)-mediated suppression of Nox-related microRNAs (miRs), in turn enhancing AM Nox expression, oxidative stress, and phagocytic dysfunction. We also hypothesized that PPARγ activation with pioglitazone (PIO) would reverse alcohol-induced C/EBPβ expression and attenuate AM oxidative stress and phagocytic dysfunction. Cells from the mouse AM cell line (MH-S) were exposed to ethanol in vitro or primary AM were isolated from mice fed ethanol in vivo. Ethanol enhanced C/EBPβ expression, decreased Nox 1-related miR-1264 and Nox 2-related miR-107 levels, and increased Nox1, Nox2, and Nox 4 expression in MH-S cells in vitro and mouse AM in vivo. These alcohol-induced AM derangements were abrogated by loss of C/EBPβ, overexpression of miRs-1264 or -107, or PIO treatment. These findings identify C/EBPβ and Nox-related miRs as novel therapeutic targets for PPARγ ligands, which could provide a translatable strategy to mitigate susceptibility to lung infections in people with a history of AUD. These studies further clarify the molecular underpinnings for a previous clinical trial using short-term PIO treatment to improve AM immunity in AUD individuals.
Collapse
Affiliation(s)
- Samantha M Yeligar
- Emory University, Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Atlanta, Georgia, United States; Atlanta Veterans Affairs Health Care System, Decatur, Georgia, United States.
| | - Frank L Harris
- Emory University, Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory + Children's Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, Georgia, United States
| | - Lou Ann S Brown
- Emory University, Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory + Children's Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, Georgia, United States
| | - C Michael Hart
- Emory University, Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Atlanta, Georgia, United States; Atlanta Veterans Affairs Health Care System, Decatur, Georgia, United States
| |
Collapse
|
28
|
COVID-19 Vaccination and Alcohol Consumption: Justification of Risks. Pathogens 2023; 12:pathogens12020163. [PMID: 36839435 PMCID: PMC9967163 DOI: 10.3390/pathogens12020163] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 01/20/2023] Open
Abstract
Since the beginning of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, pharmaceutical companies and research institutions have been actively working to develop vaccines, and the mass roll-out of vaccinations against COVID-19 began in January 2021. At the same time, during lockdowns, the consumption of alcoholic beverages increased. During the peak of vaccination, consumption remained at high levels around the world, despite the gradual relaxation of quarantine restrictions. Two of the popular queries on search engines were whether it is safe to drink alcohol after vaccination and whether this will affect the effectiveness of vaccines. Over the past two years, many studies have been published suggesting that excessive drinking not only worsens the course of an acute respiratory distress syndrome caused by the SARS-CoV-2 virus but can also exacerbate post-COVID-19 syndrome. Despite all sorts of online speculation, there is no specific scientific data on alcohol-induced complications after vaccination in the literature. Most of the published vaccine clinical trials do not include groups of patients with a history of alcohol-use disorders. This review analyzed the well-known and new mechanisms of action of COVID-19 vaccines on the immune system and the effects of alcohol and its metabolites on these mechanisms.
Collapse
|
29
|
Zsichla L, Müller V. Risk Factors of Severe COVID-19: A Review of Host, Viral and Environmental Factors. Viruses 2023; 15:175. [PMID: 36680215 PMCID: PMC9863423 DOI: 10.3390/v15010175] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
The clinical course and outcome of COVID-19 are highly variable, ranging from asymptomatic infections to severe disease and death. Understanding the risk factors of severe COVID-19 is relevant both in the clinical setting and at the epidemiological level. Here, we provide an overview of host, viral and environmental factors that have been shown or (in some cases) hypothesized to be associated with severe clinical outcomes. The factors considered in detail include the age and frailty, genetic polymorphisms, biological sex (and pregnancy), co- and superinfections, non-communicable comorbidities, immunological history, microbiota, and lifestyle of the patient; viral genetic variation and infecting dose; socioeconomic factors; and air pollution. For each category, we compile (sometimes conflicting) evidence for the association of the factor with COVID-19 outcomes (including the strength of the effect) and outline possible action mechanisms. We also discuss the complex interactions between the various risk factors.
Collapse
Affiliation(s)
- Levente Zsichla
- Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary
- National Laboratory for Health Security, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Viktor Müller
- Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary
- National Laboratory for Health Security, Eötvös Loránd University, 1117 Budapest, Hungary
| |
Collapse
|
30
|
Muralidharan A, Bauer C, Katafiasz DM, Pham D, Oyewole OO, Morwitzer MJ, Roy E, Bailey KL, Reid SP, Wyatt TA. Malondialdehyde acetaldehyde adduction of surfactant protein D attenuates SARS-CoV-2 spike protein binding and virus neutralization. Alcohol Clin Exp Res 2023; 47:95-103. [PMID: 36352814 PMCID: PMC9878066 DOI: 10.1111/acer.14974] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/01/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Over 43% of the world's population regularly consumes alcohol. Although not commonly known, alcohol can have a significant impact on the respiratory environment. Living in the time of the COVID-19 pandemic, alcohol misuse can have a particularly deleterious effect on SARS-CoV-2-infected individuals and, in turn, the overall healthcare system. Patients with alcohol use disorders have higher odds of COVID-19-associated hospitalization and mortality. Even though the detrimental role of alcohol on COVID-19 outcomes has been established, the underlying mechanisms are yet to be fully understood. Alcohol misuse has been shown to induce oxidative damage in the lungs through the production of reactive aldehydes such as malondialdehyde and acetaldehyde (MAA). MAA can then form adducts with proteins, altering their structure and function. One such protein is surfactant protein D (SPD), which plays an important role in innate immunity against pathogens. METHODS AND RESULTS In this study, we examined whether MAA adduction of SPD (SPD-MAA) attenuates the ability of SPD to bind SARS-CoV-2 spike protein, reversing SPD-mediated virus neutralization. Using ELISA, we show that SPD-MAA is unable to competitively bind spike protein and prevent ACE2 receptor binding. Similarly, SPD-MAA fails to inhibit entry of wild-type SARS-CoV-2 virus into Calu-3 cells, a lung epithelial cell line, as well as ciliated primary human bronchial epithelial cells isolated from healthy individuals. CONCLUSIONS Overall, MAA adduction of SPD, a consequence of alcohol overconsumption, represents one mechanism of compromised lung innate defense against SARS-CoV-2, highlighting a possible mechanism underlying COVID-19 severity and related mortality in patients who misuse alcohol.
Collapse
Affiliation(s)
- Abenaya Muralidharan
- Department of Pathology and Microbiology, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Christopher Bauer
- Department of Internal Medicine, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Dawn M. Katafiasz
- Department of Internal Medicine, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Danielle Pham
- Department of Internal Medicine, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Opeoluwa O. Oyewole
- Department of Pathology and Microbiology, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - M. Jane Morwitzer
- Department of Pathology and Microbiology, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Enakshi Roy
- Department of Pathology and Microbiology, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Kristina L. Bailey
- Department of Internal Medicine, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Veterans Affairs Nebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| | - St Patrick Reid
- Department of Pathology and Microbiology, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Todd A. Wyatt
- Department of Internal Medicine, College of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Veterans Affairs Nebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
- Department of Environmental, Agricultural and Occupational Health, College of Public HealthUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
31
|
Crotty K, Anton P, Coleman LG, Morris NL, Lewis SA, Samuelson DR, McMahan RH, Hartmann P, Kim A, Ratna A, Mandrekar P, Wyatt TA, Choudhry MA, Kovacs EJ, McCullough R, Yeligar SM. A critical review of recent knowledge of alcohol's effects on the immunological response in different tissues. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:36-44. [PMID: 36446606 PMCID: PMC9974783 DOI: 10.1111/acer.14979] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/02/2022] [Accepted: 11/15/2022] [Indexed: 11/23/2022]
Abstract
Alcohol misuse contributes to the dysregulation of immune responses and multiorgan dysfunction across various tissues, which are associated with higher risk of morbidity and mortality in people with alcohol use disorders. Organ-specific immune cells, including microglia in the brain, alveolar macrophages in the lungs, and Kupffer cells in the liver, play vital functions in host immune defense through tissue repair and maintenance of homeostasis. However, binge drinking and chronic alcohol misuse impair these immune cells' abilities to regulate inflammatory signaling and metabolism, thus contributing to multiorgan dysfunction. Further complicating these delicate systems, immune cell dysfunction associated with alcohol misuse is exacerbated by aging and gut barrier leakage. This critical review describes recent advances in elucidating the potential mechanisms by which alcohol misuse leads to derangements in host immunity and highlights current gaps in knowledge that may be the focus of future investigations.
Collapse
Affiliation(s)
- Kathryn Crotty
- Department of Medicine, Emory University, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
| | - Paige Anton
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
- Alcohol Research Program, University of Colorado Denver, Aurora, Colorado, USA
| | - Leon G Coleman
- Department of Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Niya L Morris
- Department of Medicine, Emory University, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
| | - Sloan A Lewis
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | - Derrick R Samuelson
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Rachel H McMahan
- Alcohol Research Program, University of Colorado Denver, Aurora, Colorado, USA
- Department of Surgery, University of Colorado, Aurora, Colorado, USA
| | - Phillipp Hartmann
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Adam Kim
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Anuradha Ratna
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Pranoti Mandrekar
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Todd A Wyatt
- Department of Environmental, Agricultural and Occupational Health, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Mashkoor A Choudhry
- Alcohol Research Program, Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago, Maywood, Illinois, USA
| | - Elizabeth J Kovacs
- Alcohol Research Program, University of Colorado Denver, Aurora, Colorado, USA
- Department of Surgery, University of Colorado, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Affairs (VA) Medical Center, Aurora, Colorado, USA
| | - Rebecca McCullough
- Department of Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
- Alcohol Research Program, University of Colorado Denver, Aurora, Colorado, USA
| | - Samantha M Yeligar
- Department of Medicine, Emory University, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
| |
Collapse
|
32
|
Forsyth CB, Voigt RM, Swanson GR, Bishehsari F, Shaikh M, Zhang L, Engen P, Keshavarzian A. Alcohol use disorder as a potential risk factor for COVID-19 severity: A narrative review. Alcohol Clin Exp Res 2022; 46:1930-1943. [PMID: 36394508 PMCID: PMC9722573 DOI: 10.1111/acer.14936] [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: 06/13/2022] [Revised: 07/25/2022] [Accepted: 08/31/2022] [Indexed: 11/19/2022]
Abstract
In Dec. 2019-January 2020, a pneumonia illness originating in Wuhan, China, designated as coronavirus disease 2019 (COVID-19) was shown to be caused by a novel RNA coronavirus designated as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). People with advanced age, male sex, and/or underlying health conditions (obesity, type 2 diabetes, cardiovascular disease, hypertension, chronic kidney disease, and chronic lung disease) are especially vulnerable to severe COVID-19 symptoms and death. These risk factors impact the immune system and are also associated with poor health, chronic illness, and shortened longevity. However, a large percent of patients without these known risk factors also develops severe COVID-19 disease that can result in death. Thus, there must exist risk factors that promote exaggerated inflammatory and immune response to the SARS-CoV-2 virus leading to death. One such risk factor may be alcohol misuse and alcohol use disorder because these can exacerbate viral lung infections like SARS, influenza, and pneumonia. Thus, it is highly plausible that alcohol misuse is a risk factor for either increased infection rate when individuals are exposed to SARS-CoV-2 virus and/or more severe COVID-19 in infected patients. Alcohol use is a well-known risk factor for lung diseases and ARDS in SARS patients. We propose that alcohol has three key pathogenic elements in common with other COVID-19 severity risk factors: namely, inflammatory microbiota dysbiosis, leaky gut, and systemic activation of the NLRP3 inflammasome. We also propose that these three elements represent targets for therapy for severe COVID-19.
Collapse
Affiliation(s)
- Christopher B. Forsyth
- Department of Internal Medicine, Section of Gastroenterology, Rush University Medical Center, Chicago, IL 60612
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612
- Rush University Graduate College, Rush University Medical Center, Chicago, IL 60612
| | - Robin M. Voigt
- Department of Internal Medicine, Section of Gastroenterology, Rush University Medical Center, Chicago, IL 60612
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612
- Rush University Graduate College, Rush University Medical Center, Chicago, IL 60612
| | - Garth R. Swanson
- Department of Internal Medicine, Section of Gastroenterology, Rush University Medical Center, Chicago, IL 60612
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612
- Rush University Graduate College, Rush University Medical Center, Chicago, IL 60612
| | - Faraz Bishehsari
- Department of Internal Medicine, Section of Gastroenterology, Rush University Medical Center, Chicago, IL 60612
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612
- Rush University Graduate College, Rush University Medical Center, Chicago, IL 60612
| | - Maliha Shaikh
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
| | - Lijuan Zhang
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
| | - Phillip Engen
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
| | - Ali Keshavarzian
- Department of Internal Medicine, Section of Gastroenterology, Rush University Medical Center, Chicago, IL 60612
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL 60612
- Rush University Graduate College, Rush University Medical Center, Chicago, IL 60612
| |
Collapse
|
33
|
Narrative Review: Glucocorticoids in Alcoholic Hepatitis—Benefits, Side Effects, and Mechanisms. J Xenobiot 2022; 12:266-288. [PMID: 36278756 PMCID: PMC9589945 DOI: 10.3390/jox12040019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Alcoholic hepatitis is a major health and economic burden worldwide. Glucocorticoids (GCs) are the only first-line drugs recommended to treat severe alcoholic hepatitis (sAH), with limited short-term efficacy and significant side effects. In this review, I summarize the major benefits and side effects of GC therapy in sAH and the potential underlying mechanisms. The review of the literature and data mining clearly indicate that the hepatic signaling of glucocorticoid receptor (GR) is markedly impaired in sAH patients. The impaired GR signaling causes hepatic down-regulation of genes essential for gluconeogenesis, lipid catabolism, cytoprotection, and anti-inflammation in sAH patients. The efficacy of GCs in sAH may be compromised by GC resistance and/or GC’s extrahepatic side effects, particularly the side effects of intestinal epithelial GR on gut permeability and inflammation in AH. Prednisolone, a major GC used for sAH, activates both the GR and mineralocorticoid receptor (MR). When GC non-responsiveness occurs in sAH patients, the activation of MR by prednisolone might increase the risk of alcohol abuse, liver fibrosis, and acute kidney injury. To improve the GC therapy of sAH, the effort should be focused on developing the biomarker(s) for GC responsiveness, liver-targeting GR agonists, and strategies to overcome GC non-responsiveness and prevent alcohol relapse in sAH patients.
Collapse
|
34
|
Manna PR, Gray ZC, Sikdar M, Reddy H. COVID-19 and its genomic variants: Molecular pathogenesis and therapeutic interventions. EXCLI JOURNAL 2022; 21:1196-1221. [PMID: 36381644 PMCID: PMC9650701 DOI: 10.17179/excli2022-5315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/05/2022] [Indexed: 11/25/2022]
Abstract
Coronavirus disease-19 (COVID-19), caused by a β-coronavirus and its genomic variants, is associated with substantial morbidities and mortalities globally. The COVID-19 virus and its genomic variants enter host cells upon binding to the angiotensin converting enzyme 2 receptors that are expressed in a variety of tissues, but predominantly in the lungs, heart, and blood vessels. Patients afflicted with COVID-19 may be asymptomatic or present with critical symptoms possibly due to diverse lifestyles, immune responses, aging, and underlying medical conditions. Geriatric populations, especially men in comparison to women, with immunocompromised conditions, are most vulnerable to severe COVID-19 associated infections, complications, and mortalities. Notably, whereas immunomodulation, involving nutritional consumption, is essential to protecting an individual from COVID-19, immunosuppression is detrimental to a person with this aggressive disease. As such, immune health is inversely correlated to COVID-19 severity and resulting consequences. Advances in genomic and proteomic technologies have helped us to understand the molecular events underlying symptomatology, transmission and, pathogenesis of COVID-19 and its genomic variants. Accordingly, there has been development of a variety of therapeutic interventions, ranging from mask wearing to vaccination to medication. This review summarizes the current understanding of molecular pathogenesis of COVID-19, effects of comorbidities on COVID-19, and prospective therapeutic strategies for the prevention and treatment of this contagious disease.
Collapse
Affiliation(s)
- Pulak R. Manna
- Department of Internal Medicine, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX 79430, USA,*To whom correspondence should be addressed: Pulak R. Manna, Department of Internal Medicine, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX 79430, USA; Tel: +1-806-743-3573, Fax: +1-806-743-3143, E-mail:
| | - Zackery C. Gray
- Department of Internal Medicine, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX 79430, USA
| | - Malabika Sikdar
- Department of Zoology, Dr. Hari Singh Gour Vishwavidyalaya, Sagar, MP 470003, India
| | - Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX 79430, USA,Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA,Neurology, Departments of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA,Public Health Department of the Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA,Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA,Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
35
|
Khair S, Brenner LA, Koval M, Samuelson D, Cucinello-Regland JA, Anton P, Piano MR, Simon L, Crotty K, Sharieh F, Travers JB, Singh V, Cannon A, Kim A, McCullough RL, Yeligar SM, Wyatt TA, McMahan RH, Choudhry MA, Kovacs EJ. New insights into the mechanism of alcohol-mediated organ damage via its impact on immunity, metabolism, and repair pathways: A summary of the 2021 Alcohol and Immunology Research Interest Group (AIRIG) meeting. Alcohol 2022; 103:1-7. [PMID: 35659577 PMCID: PMC9994264 DOI: 10.1016/j.alcohol.2022.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 01/14/2023]
Abstract
On November 19th, 2021, the annual Alcohol and Immunology Research Interest Group (AIRIG) meeting was held at Loyola University Chicago Health Sciences Campus in Maywood, Illinois. The 2021 meeting focused on how alcohol misuse is linked to immune system derangements, leading to tissue and organ damage, and how this research can be translated into improving treatment of alcohol-related disease. This meeting was divided into three plenary sessions: the first session focused on how alcohol misuse affects different parts of the immune system, the second session presented research on mechanisms of organ damage from alcohol misuse, and the final session highlighted research on potential therapeutic targets for treating alcohol-mediated tissue damage. Diverse areas of alcohol research were covered during the meeting, from alcohol's effect on pulmonary systems and neuroinflammation to epigenetic changes, senescence markers, and microvesicle particles. These presentations yielded a thoughtful discussion on how the findings can lead to therapeutic treatments for people suffering from alcohol-related diseases.
Collapse
Affiliation(s)
- Shanawaj Khair
- Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States; Alcohol Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States; Molecular Biology Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States; Medical Scientist Training Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States
| | - Lisa A Brenner
- VA Rocky Mountain Mental Illness Research Education and Clinical Center, Rocky Mountain Regional Veterans Affairs (VA) Medical Center, Aurora, CO, United States; Department of Physical Medicine and Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Department of Psychiatry and Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Military and Veteran Microbiome: Consortium for Research and Education, Aurora, CO, United States
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Derrick Samuelson
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jessica A Cucinello-Regland
- Department of Physiology, Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Paige Anton
- Department of Pharmaceutical Sciences, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States
| | - Mariann R Piano
- Center for Research Development and Scholarship, Vanderbilt University School of Nursing, Nashville, TN, United States
| | - Liz Simon
- Department of Physiology, Comprehensive Alcohol HIV/AIDs Research Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Kathryn Crotty
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Farah Sharieh
- Department of Orthopedic Surgery and Rehabilitation, Loyola University Medical Center, Maywood, IL, United States; Alcohol Research Program, Loyola University Chicago Stritch School of Medicine, Maywood, IL, United States
| | | | - Vaibhav Singh
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Abigail Cannon
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL, United States
| | - Adam Kim
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Rebecca L McCullough
- Department of Pharmaceutical Sciences, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States
| | - Samantha M Yeligar
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Todd A Wyatt
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States; Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Rachel H McMahan
- Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States; Alcohol Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States
| | - Mashkoor A Choudhry
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL, United States; Integrative Cell Biology Program, Loyola University Chicago Health Sciences Campus, Maywood, IL, United States
| | - Elizabeth J Kovacs
- Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States; Alcohol Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States; Molecular Biology Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States; Medical Scientist Training Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, United States; VA Rocky Mountain Mental Illness Research Education and Clinical Center, Rocky Mountain Regional Veterans Affairs (VA) Medical Center, Aurora, CO, United States.
| |
Collapse
|
36
|
Sang W, Chen S, Lin L, Wang N, Kong X, Ye J. Antioxidant mitoquinone ameliorates EtOH-LPS induced lung injury by inhibiting mitophagy and NLRP3 inflammasome activation. Front Immunol 2022; 13:973108. [PMID: 36059543 PMCID: PMC9436256 DOI: 10.3389/fimmu.2022.973108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 07/29/2022] [Indexed: 12/02/2022] Open
Abstract
Chronic ethanol abuse is a systemic disorder and a risk factor for acute respiratory distress syndrome (ARDS) and chronic obstructive pulmonary disease (COPD). However, the mechanisms involved are unknown. One explanation is that ethanol produces damaging reactive oxygen species (ROS) and disturbs the balance of mitochondria within the lungs to promote a pro-injury environment. We hypothesized that targeting an antioxidant to the mitochondria would prevent oxidative damage and attenuate EtOH-LPS-induced lung injury. To test this, we investigated the effects of mitochondria-targeted ubiquinone, Mitoquinone (MitoQ) on ethanol-sensitized lung injury induced by LPS. Lung inflammation, ROS, mitochondria function, and mitophagy were assessed. We demonstrated that chronic ethanol feeding sensitized the lung to LPS-induced lung injury with significantly increased reactive oxygen species ROS level and mitochondrial injury as well as lung cellular NLRP3 inflammasome activation. These deleterious effects were attenuated by MitoQ administration in mice. The protective effects of MitoQ are associated with decreased cellular mitophagy and NLRP3 inflammasome activation in vivo and in vitro. Taken together, our results demonstrated that ethanol aggravated LPS-induced lung injury, and antioxidant MitoQ protects from EtOH-LPS-induced lung injury, probably through reducing mitophagy and protecting mitochondria, followed by NLRP3 inflammasome activation. These results will provide the prevention and treatment of ethanol intake effects with new ideas.
Collapse
Affiliation(s)
- Wenhua Sang
- School of Basic Medical Sciences, Institute of Hypoxia Research, Cixi Biomedical Institute, Wenzhou Medical University, Wenzhou, China
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Sha Chen
- School of Basic Medical Sciences, Institute of Hypoxia Research, Cixi Biomedical Institute, Wenzhou Medical University, Wenzhou, China
| | - Lidan Lin
- School of Basic Medical Sciences, Institute of Hypoxia Research, Cixi Biomedical Institute, Wenzhou Medical University, Wenzhou, China
| | - Nan Wang
- School of Basic Medical Sciences, Institute of Hypoxia Research, Cixi Biomedical Institute, Wenzhou Medical University, Wenzhou, China
| | - Xiaoxia Kong
- School of Basic Medical Sciences, Institute of Hypoxia Research, Cixi Biomedical Institute, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Xiaoxia Kong, ; Jinyan Ye,
| | - Jinyan Ye
- Department of Respiratory Medicine and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Xiaoxia Kong, ; Jinyan Ye,
| |
Collapse
|
37
|
Park SH, Lee YS, Sim J, Seo S, Seo W. Alcoholic liver disease: a new insight into the pathogenesis of liver disease. Arch Pharm Res 2022; 45:447-459. [PMID: 35761115 DOI: 10.1007/s12272-022-01392-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 06/10/2022] [Indexed: 11/02/2022]
Abstract
Excessive alcohol consumption contributes to a broad clinical spectrum of liver diseases, from simple steatosis to end-stage hepatocellular carcinoma. The liver is the primary organ that metabolizes ingested alcohol and is exquisitely sensitive to alcohol intake. Alcohol metabolism is classified into two pathways: oxidative and non-oxidative alcohol metabolism. Both oxidative and non-oxidative alcohol metabolisms and their metabolites have toxic consequences for multiple organs, including the liver, adipose tissue, intestine, and pancreas. Although many studies have focused on the effects of oxidative alcohol metabolites on liver damage, the importance of non-oxidative alcohol metabolites in cellular damage has also been discovered. Furthermore, extrahepatic alcohol effects are crucial for providing additional information necessary for the progression of alcoholic liver disease. Therefore, studying the effects of alcohol-producing metabolites and interorgan crosstalk between the liver and peripheral organs that express ethanol-metabolizing enzymes will facilitate a comprehensive understanding of the pathogenesis of alcoholic liver disease. This review focuses on alcohol-metabolite-associated hepatotoxicity due to oxidative and non-oxidative alcohol metabolites and the role of interorgan crosstalk in alcoholic liver disease pathogenesis.
Collapse
Affiliation(s)
- Seol Hee Park
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea
| | - Young-Sun Lee
- Department of Internal Medicine, Korea University Medical Center, Seoul, Republic of Korea
| | - Jaemin Sim
- Lab of Hepatotoxicity, College of Pharmacy, Ewha Womans University, #52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03765, Republic of Korea
| | - Seonkyung Seo
- Lab of Hepatotoxicity, College of Pharmacy, Ewha Womans University, #52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03765, Republic of Korea
| | - Wonhyo Seo
- Lab of Hepatotoxicity, College of Pharmacy, Ewha Womans University, #52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03765, Republic of Korea.
| |
Collapse
|
38
|
Crotty KM, Yeligar SM. Hyaladherins May be Implicated in Alcohol-Induced Susceptibility to Bacterial Pneumonia. Front Immunol 2022; 13:865522. [PMID: 35634317 PMCID: PMC9133445 DOI: 10.3389/fimmu.2022.865522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/15/2022] [Indexed: 11/13/2022] Open
Abstract
Although the epidemiology of bacterial pneumonia and excessive alcohol use is well established, the mechanisms by which alcohol induces risk of pneumonia are less clear. Patterns of alcohol misuse, termed alcohol use disorders (AUD), affect about 15 million people in the United States. Compared to otherwise healthy individuals, AUD increase the risk of respiratory infections and acute respiratory distress syndrome (ARDS) by 2-4-fold. Levels and fragmentation of hyaluronic acid (HA), an extracellular glycosaminoglycan of variable molecular weight, are increased in chronic respiratory diseases, including ARDS. HA is largely involved in immune-assisted wound repair and cell migration. Levels of fragmented, low molecular weight HA are increased during inflammation and decrease concomitant with leukocyte levels following injury. In chronic respiratory diseases, levels of fragmented HA and leukocytes remain elevated, inflammation persists, and respiratory infections are not cleared efficiently, suggesting a possible pathological mechanism for prolonged bacterial pneumonia. However, the role of HA in alcohol-induced immune dysfunction is largely unknown. This mini literature review provides insights into understanding the role of HA signaling in host immune defense following excessive alcohol use. Potential therapeutic strategies to mitigate alcohol-induced immune suppression in bacterial pneumonia and HA dysregulation are also discussed.
Collapse
Affiliation(s)
- Kathryn M Crotty
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, United States.,Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Samantha M Yeligar
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, United States.,Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| |
Collapse
|
39
|
Nissen CG, Mosley DD, Kharbanda KK, Katafiasz DM, Bailey KL, Wyatt TA. Malondialdehyde Acetaldehyde-Adduction Changes Surfactant Protein D Structure and Function. Front Immunol 2022; 13:866795. [PMID: 35669781 PMCID: PMC9164268 DOI: 10.3389/fimmu.2022.866795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/13/2022] [Indexed: 11/23/2022] Open
Abstract
Alcohol consumption with concurrent cigarette smoking produces malondialdehyde acetaldehyde (MAA)-adducted lung proteins. Lung surfactant protein D (SPD) supports innate immunity via bacterial aggregation and lysis, as well as by enhancing macrophage-binding and phagocytosis. MAA-adducted SPD (SPD-MAA) has negative effects on lung cilia beating, macrophage function, and epithelial cell injury repair. Because changes in SPD multimer structure are known to impact SPD function, we hypothesized that MAA-adduction changes both SPD structure and function. Purified human SPD and SPD-MAA (1 mg/mL) were resolved by gel filtration using Sephadex G-200 and protein concentration of each fraction determined by Bradford assay. Fractions were immobilized onto nitrocellulose by slot blot and assayed by Western blot using antibodies to SPD and to MAA. Binding of SPD and SPD-MAA was determined fluorometrically using GFP-labeled Streptococcus pneumoniae (GFP-SP). Anti-bacterial aggregation of GFP-SP and macrophage bacterial phagocytosis were assayed by microscopy and permeability determined by bacterial phosphatase release. Viral injury was measured as LDH release in RSV-treated airway epithelial cells. Three sizes of SPD were resolved by gel chromatography as monomeric, trimeric, and multimeric forms. SPD multimer was the most prevalent, while the majority of SPD-MAA eluted as trimer and monomer. SPD dose-dependently bound to GFP-SP, but SPD-MAA binding to bacteria was significantly reduced. SPD enhanced, but MAA adduction of SPD prevented, both aggregation and macrophage phagocytosis of GFP-SP. Likewise, SPD increased bacterial permeability while SPD-MAA did not. In the presence of RSV, BEAS-2B cell viability was enhanced by SPD, but not protected by SPD-MAA. Our results demonstrate that MAA adduction changes the quaternary structure of SPD from multimer to trimer and monomer leading to a decrease in the native anti-microbial function of SPD. These findings suggest one mechanism for increased pneumonia observed in alcohol use disorders.
Collapse
Affiliation(s)
- Claire G. Nissen
- Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE, United States
| | - Deanna D. Mosley
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kusum K. Kharbanda
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Dawn M. Katafiasz
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kristina L. Bailey
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Todd A. Wyatt
- Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| |
Collapse
|
40
|
Substance use disorders and COVID-19: An analysis of nation-wide Veterans Health Administration electronic health records. Drug Alcohol Depend 2022; 234:109383. [PMID: 35279457 PMCID: PMC8891118 DOI: 10.1016/j.drugalcdep.2022.109383] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Substance use disorders (SUD) elevate the risk for COVID-19 hospitalization, but studies are inconsistent on the relationship of SUD to COVID-19 mortality. METHODS Veterans Health Administration (VHA) patients treated in 2019 and evaluated in 2020 for COVID-19 (n=5,556,315), of whom 62,303 (1.1%) tested positive for COVID-19 (COVID-19+). Outcomes were COVID-19+ by 11/01/20, hospitalization, ICU admission, or death within 60 days of a positive test. Main predictors were any ICD-10-CM SUDs, with substance-specific SUDs (cannabis, cocaine, opioid, stimulant, sedative) explored individually. Logistic regression produced unadjusted and covariate-adjusted odds ratios (OR; aOR). RESULTS Among COVID-19+ patients, 19.25% were hospitalized, 7.71% admitted to ICU, and 5.84% died. In unadjusted models, any SUD and all substance-specific SUDs except cannabis use disorder were associated with COVID-19+(ORs=1.06-1.85); adjusted models produced similar results. Any SUD and all substance-specific SUDs were associated with hospitalization (aORs: 1.24-1.91). Any SUD, cocaine and opioid disorder were associated with ICU admission in unadjusted but not adjusted models. Any SUD, cannabis, cocaine, and stimulant disorders were inversely associated with mortality in unadjusted models (OR=0.27-0.46). After adjustment, associations with mortality were no longer significant. In ad hoc analyses, adjusted odds of mortality were lower among the 49.9% of COVID-19+ patients with SUD who had SUD treatment in 2019, but not among those without such treatment. CONCLUSIONS In VHA patients, SUDs are associated with COVID-19 hospitalization but not COVID-19 mortality. SUD treatment may provide closer monitoring of care, ensuring that these patients received needed medical attention, enabling them to ultimately survive serious illness.
Collapse
|
41
|
Danışman M, İspir GZ, Oğur B. Delirium Tremens and Central Pontine Myelinolysis in a Patient with Alcohol Use Disorder and Pneumonia: a Case Report and a Narrative Review. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2022; 90:288-290. [PMID: 35483887 DOI: 10.1055/a-1778-3727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Delirium tremens (DT) is a serious condition occurring in alcohol withdrawal syndrome. Alcohol consumption may also cause additional health problems, such as respiratory infections or neuropsychiatric conditions such as central pontine myelinolysis. In this clinical scenario, managing DT can be expected to be more compelling and complex. Alcohol decreases coughing and mucociliary clearance and disrupts the immunity of the respiratory system. CASE Here we report on a middle-aged man with alcohol use disorder who had developed DT due to alcohol withdrawal and comorbid pneumonia. DISCUSSION AND CONCLUSION In this paper, DT, the relation between respiratory infections and alcohol intake, and the correlation of alcohol consumption and central pontine myelinolysis (CPM) are discussed. Also, the literature on alcohol consumption and the additional respiratory and neurologic problems resulting from it are presented.
Collapse
Affiliation(s)
- Mustafa Danışman
- AMATEM (Alcohol and Drug Addiction Research, Treatment and Education Center), Ankara Training and Research Hospital, Ankara, Turkey
| | - Gamze Zengin İspir
- Department of Psychiatry, University of Health Science-Diskapi Yildirim Beyazit Teaching and Research Hospital, Ankara, Turkey
| | - Begüm Oğur
- Department of Psychiatry, University of Health Science-Gulhane Teaching and Research Hospital, Ankara, Turkey
| |
Collapse
|
42
|
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
|
43
|
Aitkens L, Waller JL, Baer SL, Mohammed A, Tran S, Siddiqui B, Padala S, Young L, Kheda M, Bollag WB. Psoriasis and Pneumonia in the End-stage Renal Disease Population. Am J Med Sci 2022; 364:29-35. [DOI: 10.1016/j.amjms.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/21/2021] [Accepted: 03/08/2022] [Indexed: 12/01/2022]
|
44
|
Hu J, Gu L, Shao Y, Zhang R, Qi T, Sun J, Wang Z, Song W, Tang Y, Wang J, Xu S, Yang J, Shen Y, Liu L, Chen J, Lu H. Long-term case-fatality rate of nontuberculous mycobacterial disease in people living with HIV. Infect Dis Poverty 2022; 11:16. [PMID: 35130974 PMCID: PMC8822711 DOI: 10.1186/s40249-022-00942-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Background Few data are available regarding the long-term case-fatality rate (CFR) among people living with HIV (PLWH) with nontuberculous mycobacteria (NTM) disease. The aim of this study is to analyze the long-term CFR in patients with NTM disease and to identify risk factors for their death. Methods A retrospective cohort study of 379 cases of microbiologically confirmed NTM disease in PLWH was conducted from January 1, 2012, to December 31, 2020, in Shanghai, China. We used Kaplan–Meier survival analysis and the log-rank test to compare the long-term CFR in patients with disseminated NTM (DNTM) and localized NTM disease. Univariate Cox proportional hazards regression analysis and a stepwise Cox proportional hazards regression model were used to estimate the predictors of long-term CFR. Results The cohort was followed up for a median of 26 months. The total CFR was 15.7% by one year and increased to 22.6% at 5 years after the diagnosis of NTM disease. The 5-year CFR of PLWH with DNTM was significantly higher than that of PLWH with localized NTM (26.7% vs 19.6% for DNTM and localized NTM disease, respectively). Older age [hazard ratio (HR) = 1.04, 95% confidence interval (CI): 1.02–1.06, P < 0.001], comorbidity (HR = 2.05, 95% CI: 1.21–3.49, P < 0.01), DNTM (HR = 2.08, 95% CI: 1.17–3.68, P < 0.05), and HIV viral load (HR = 1.32, 95% CI: 1.12–1.55, P < 0.001) were all independent risk factors for long-term CFR. In the subgroup analysis, time to culture positivity was negatively correlated with CFR in patients with DNTM (HR = 0.90, 95% CI: 0.82–0.98, P < 0.05). Conclusions NTM was associated with a high long-term CFR in PLWH. Further approaches to prevent NTM disease in PLWH are urgently needed. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Jingjing Hu
- School of Public Health, Bengbu Medical College, Bengbu, 233000, Anhui, China.,Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Ling Gu
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Yueming Shao
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Renfang Zhang
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Tangkai Qi
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Jianjun Sun
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Zhenyan Wang
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Wei Song
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Yang Tang
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Jiangrong Wang
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Shuibao Xu
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Junyang Yang
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Yinzhong Shen
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China
| | - Li Liu
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China.
| | - Jun Chen
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai, 201508, China.
| | - Hongzhou Lu
- School of Public Health, Bengbu Medical College, Bengbu, 233000, Anhui, China. .,Shenzhen Third People's Hospital, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518000, China.
| |
Collapse
|
45
|
Chen L, Li L, Song Y, Lv T. Blocking SphK1/S1P/S1PR1 Signaling Pathway Alleviates Lung Injury Caused by Sepsis in Acute Ethanol Intoxication Mice. Inflammation 2021; 44:2170-2179. [PMID: 34109517 PMCID: PMC8189277 DOI: 10.1007/s10753-021-01490-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/12/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022]
Abstract
Acute ethanol intoxication increases the risk of sepsis and aggravates the symptoms of sepsis and lung injury. Therefore, this study aimed to explore whether sphingosine kinase 1 (SphK1)/sphingosine-1-phosphate (S1P)/S1P receptor 1 (S1PR1) signaling pathway functions in lung injury caused by acute ethanol intoxication-enhanced sepsis, as well as its underlying mechanism. The acute ethanol intoxication model was simulated by intraperitoneally administering mice with 32% ethanol solution, and cecal ligation and puncture (CLP) was used to construct the sepsis model. The lung tissue damage was observed by hematoxylin-eosin (H&E) staining, and the wet-to-dry (W/D) ratio was used to evaluate the degree of pulmonary edema. Inflammatory cell counting and protein concentration in bronchoalveolar lavage fluid (BALF) were, respectively, detected by hemocytometer and bicinchoninic acid (BCA) method. The levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1β, and IL-18 in BALF were detected by their commercial enzyme-linked immunosorbent assay (ELISA) kits. The myeloperoxidase (MPO) activity and expression of apoptosis-related proteins and SphK1/S1P/S1PR1 pathway-related proteins were, respectively, analyzed by MPO ELISA kit and Western blot analysis. The cell apoptosis in lung tissues was observed by TUNEL assay. Acute ethanol intoxication (EtOH) decreased the survival rate of mice and exacerbated the lung injury caused by sepsis through increasing pulmonary vascular permeability, neutrophil infiltration, release of inflammatory factors, and cell apoptosis. In addition, EtOH could activate the SphK1/S1P/S1PR1 pathway in CLP mice. However, PF-543, as a specific inhibitor of SphK1, could partially reverse the deleterious effects on lung injury of CLP mice. PF-543 alleviated lung injury caused by sepsis in acute ethanol intoxication rats by suppressing the SphK1/S1P/S1PR1 signaling pathway.
Collapse
Affiliation(s)
- Liang Chen
- Department of Respiratory and Critical Care Medicine, The Affiliated No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jiangsu, China
| | - Lingling Li
- Department of Respiratory and Critical Care Medicine, The Affiliated No.1 People's Hospital of Nanjing Medical University, Huaian, 223300, Jiangsu, China
| | - Yong Song
- Jinling Clinical Medical College, Nanjing Medical University, 305 Zhongshan East Road, Xuanwu District, Nanjing City, 210002, Jiangsu Province, China.
| | - Tangfeng Lv
- Jinling Clinical Medical College, Nanjing Medical University, 305 Zhongshan East Road, Xuanwu District, Nanjing City, 210002, Jiangsu Province, China.
| |
Collapse
|
46
|
Outcomes following traumatic inhalational airway injury - Predictors of mortality and effect of procedural intervention. Injury 2021; 52:3320-3326. [PMID: 34565616 DOI: 10.1016/j.injury.2021.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/19/2021] [Accepted: 09/12/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Study outcomes, predictors of mortality, and effects of procedural interventions on patients following traumatic inhalational airway injury. STUDY Design: Retrospective study. SETTING National Trauma Data Bank METHODS: Patients over the age of eighteen admitted between 2008 and 2016 to NTDB-participating sites were included. In-hospital mortality and length of stay were the primary outcomes. RESULTS The final study cohort included 13,351 patients. History of active smoking was negatively associated with in-house mortality with an OR of 0.33 (0.25-0.44). History of alcohol use, and presence of significant medical co-morbidities were positively associated with in-house mortality with OR of 5.28 (4.32-6.46) 2.74 (19.4-3.86) respectively. There was little to no association between procedural interventions and in-house mortality. Intubation, laryngobronchoscopy, and tracheostomy had OR of 0.90 (0.67-1.20), 1.02 (0.79-1.30), and 0.94 (0.58-1.51), respectively. However, procedural intervention did affect both the median hospital and ICU lengths of stay of patients. Median hospital and ICU length of stay were shorter for patients receiving endotracheal intubation. Median hospital length of stay was longer for patients undergoing bronchoscopy and laryngoscopy, but median ICU length of stay was shorter for patients undergoing bronchoscopy and laryngoscopy. Patients receiving a tracheostomy have both significantly increased median hospital and ICU lengths of stay. CONCLUSIONS Active smoking was associated with decreased odds of in-hospital mortality, while presence of pre-existing medical comorbidities and history of alcohol use disorder was associated with increased odds of in-hospital mortality. Procedural intervention had little to no association with in-hospital mortality but did affect overall hospital and ICU LOS.
Collapse
|
47
|
Salamanca-González P, Valls-Zamora RM, Pedret-Figuerola A, Sorlí-Aguilar M, Santigosa-Ayala A, Catalin RE, Pallejà-Millán M, Solà-Alberich R, Martin-Lujan F. Effectiveness of a Motivational Nutritional Intervention through Social Networks 2.0 to Increase Adherence to the Mediterranean Diet and Improve Lung Function in Active Smokers: The DIET Study, a Randomized, Controlled and Parallel Clinical Trial in Primary Care. Nutrients 2021; 13:3597. [PMID: 34684600 PMCID: PMC8538243 DOI: 10.3390/nu13103597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/30/2021] [Accepted: 10/08/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Diet can help preserve lung function in smokers, as well as aid individuals who avoid smoking. This study aimed to evaluate the effectiveness of a nutritional intervention, using the Social Networks 2.0 tool, to increase adherence to the Mediterranean diet (MD) and improve lung function in smokers without prior respiratory disease. METHODS A randomized controlled parallel design was used. The participants were assigned to either the intervention or control group. Data from representative smokers without respiratory disease (n = 77) aged 18-70 years were analyzed. The participants completed a validated semi-quantitative food-frequency questionnaire, and their adherence to the diet was evaluated by using the questionnaire called the Mediterranean Diet Adherence Score (MEDAS, with 14 items), which considers ≥9 points to indicate high adherence. The lung function was assessed by spirometry. Associations among variables were determined by logistic regression. RESULTS A comparison of the variables at the end of the study between the control and intervention groups showed that the intervention significantly increased adherence to the MD based on the MEDAS questionnaire (0.69 (2.1) vs. 2.05 (2.03); p = 0.009). Specifically, the consumption of fruits was increased after two years in both groups; however, a more significant increase was detected in the intervention group (121 (178) vs. 12.7 (167) in the control group; p-value = 0.008). In the unadjusted analysis, the intervention only showed a statistical significant increase in the score of adherence to the MD (β: 1.36; 95% CI 0.35; 2.3; p = 0.009), and this increase was maintained after adjusting for age and sex (β: 1.15; 95% CI 0.05; 2.2; p = 0.040) and after adjusting for various sociodemographic, lifestyle and anthropometric variables (β: 1.17; 95% CI 0.02; 2.31; p = 0.046). The pulmonary function parameters improved more in the intervention group; however, no significant differences were observed between the two groups. CONCLUSIONS A nutritional intervention based on a dietetic-nutritional education program resulted in a significant increase in adherence to the MD. However, some evidence suggests that an MD dietary intervention can improve lung function, but in our study, we were not able to demonstrate this. Further research is needed to obtain more robust data and confirm a possible benefit of the program before it can be extended to general practice.
Collapse
Affiliation(s)
- Patricia Salamanca-González
- Functional Nutrition, Oxidation and Cardiovascular Disease Group (NFOC-SALUT), Facultat de Medicina i Ciències de La Salut, Universitat Rovira i Virgili, Sant Llorenç, 21, 43201 Reus, Spain; (P.S.-G.); (R.M.V.-Z.); (A.P.-F.); (R.S.-A.)
- CENIT Research Group, Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP JGol), Gran Via de les Corts Catalanes, 587, 08007 Barcelona, Spain; (M.S.-A.); (A.S.-A.); (R.-E.C.)
| | - Rosa Maria Valls-Zamora
- Functional Nutrition, Oxidation and Cardiovascular Disease Group (NFOC-SALUT), Facultat de Medicina i Ciències de La Salut, Universitat Rovira i Virgili, Sant Llorenç, 21, 43201 Reus, Spain; (P.S.-G.); (R.M.V.-Z.); (A.P.-F.); (R.S.-A.)
| | - Anna Pedret-Figuerola
- Functional Nutrition, Oxidation and Cardiovascular Disease Group (NFOC-SALUT), Facultat de Medicina i Ciències de La Salut, Universitat Rovira i Virgili, Sant Llorenç, 21, 43201 Reus, Spain; (P.S.-G.); (R.M.V.-Z.); (A.P.-F.); (R.S.-A.)
| | - Mar Sorlí-Aguilar
- CENIT Research Group, Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP JGol), Gran Via de les Corts Catalanes, 587, 08007 Barcelona, Spain; (M.S.-A.); (A.S.-A.); (R.-E.C.)
| | - Antoni Santigosa-Ayala
- CENIT Research Group, Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP JGol), Gran Via de les Corts Catalanes, 587, 08007 Barcelona, Spain; (M.S.-A.); (A.S.-A.); (R.-E.C.)
- Department of Primary Care Camp de Tarragona, Institut Català de la Salut, Doctor Mallafré Guach, 4, 43007 Tarragona, Spain
- Departament de Medicina i Ciències de La Salut, Universitat Rovira i Virgili, Sant Llorenç, 21, 43201 Reus, Spain;
| | - Roxana-Elena Catalin
- CENIT Research Group, Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP JGol), Gran Via de les Corts Catalanes, 587, 08007 Barcelona, Spain; (M.S.-A.); (A.S.-A.); (R.-E.C.)
- Department of Primary Care Camp de Tarragona, Institut Català de la Salut, Doctor Mallafré Guach, 4, 43007 Tarragona, Spain
| | - Meritxell Pallejà-Millán
- Departament de Medicina i Ciències de La Salut, Universitat Rovira i Virgili, Sant Llorenç, 21, 43201 Reus, Spain;
- Research Support Unit Camp of Tarragona, Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP JGol), Camí de Riudoms, 53-55, 43202 Reus, Spain
| | - Rosa Solà-Alberich
- Functional Nutrition, Oxidation and Cardiovascular Disease Group (NFOC-SALUT), Facultat de Medicina i Ciències de La Salut, Universitat Rovira i Virgili, Sant Llorenç, 21, 43201 Reus, Spain; (P.S.-G.); (R.M.V.-Z.); (A.P.-F.); (R.S.-A.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari Sant Joan XXIII, Doctor Mallafré Guasch, 4, 43007 Tarragona, Spain
| | - Francisco Martin-Lujan
- CENIT Research Group, Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP JGol), Gran Via de les Corts Catalanes, 587, 08007 Barcelona, Spain; (M.S.-A.); (A.S.-A.); (R.-E.C.)
- Department of Primary Care Camp de Tarragona, Institut Català de la Salut, Doctor Mallafré Guach, 4, 43007 Tarragona, Spain
- Departament de Medicina i Ciències de La Salut, Universitat Rovira i Virgili, Sant Llorenç, 21, 43201 Reus, Spain;
- Research Support Unit Camp of Tarragona, Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP JGol), Camí de Riudoms, 53-55, 43202 Reus, Spain
| | | |
Collapse
|
48
|
Wyatt TA, Warren KJ, Wetzel TJ, Suwondo T, Rensch GP, DeVasure JM, Mosley DD, Kharbanda KK, Thiele GM, Burnham EL, Bailey KL, Yeligar SM. Malondialdehyde-Acetaldehyde Adduct Formation Decreases Immunoglobulin A Transport across Airway Epithelium in Smokers Who Abuse Alcohol. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1732-1742. [PMID: 34186073 PMCID: PMC8485061 DOI: 10.1016/j.ajpath.2021.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/09/2021] [Accepted: 06/16/2021] [Indexed: 12/16/2022]
Abstract
Alcohol misuse and smoking are risk factors for pneumonia, yet the impact of combined cigarette smoke and alcohol on pneumonia remains understudied. Smokers who misuse alcohol form lung malondialdehyde-acetaldehyde (MAA) protein adducts and have decreased levels of anti-MAA secretory IgA (sIgA). Transforming growth factor-β (TGF-β) down-regulates polymeric Ig receptor (pIgR) on mucosal epithelium, resulting in decreased sIgA transcytosis to the mucosa. It is hypothesized that MAA-adducted lung protein increases TGF-β, preventing expression of epithelial cell pIgR and decreasing sIgA. Cigarette smoke and alcohol co-exposure on sIgA and TGF-β in human bronchoalveolar lavage fluid and in mice instilled with MAA-adducted surfactant protein D (SPD-MAA) were studied herein. Human bronchial epithelial cells (HBECs) and mouse tracheal epithelial cells were treated with SPD-MAA and sIgA and TGF-β was measured. Decreased sIgA and increased TGF-β were observed in bronchoalveolar lavage from combined alcohol and smoking groups in humans and mice. CD204 (MAA receptor) knockout mice showed no changes in sIgA. SPD-MAA decreased pIgR in HBECs. Conversely, SPD-MAA stimulated TGF-β release in both HBECs and mouse tracheal epithelial cells, but not in CD204 knockout mice. SPD-MAA stimulated TGF-β in alveolar macrophage cells. These data show that MAA-adducted surfactant protein stimulates lung epithelial cell TGF-β, down-regulates pIgR, and decreases sIgA transcytosis. These data provide a mechanism for the decreased levels of sIgA observed in smokers who misuse alcohol.
Collapse
Affiliation(s)
- Todd A Wyatt
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska; Department of Environmental, Agricultural and Occupational Health, University of Nebraska Medical Center, Omaha, Nebraska; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska.
| | - Kristi J Warren
- Department of Medicine-Pulmonary Division, University of Utah/VA Salt Lake Health Care System, Salt Lake City, Utah
| | - Tanner J Wetzel
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Troy Suwondo
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Gage P Rensch
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jane M DeVasure
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Deanna D Mosley
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kusum K Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Geoffrey M Thiele
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Ellen L Burnham
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado
| | - Kristina L Bailey
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Samantha M Yeligar
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia; Research Service, Atlanta VA Health Care System, Decatur, Georgia
| |
Collapse
|
49
|
Morojele NK, Shenoi SV, Shuper PA, Braithwaite RS, Rehm J. Alcohol Use and the Risk of Communicable Diseases. Nutrients 2021; 13:3317. [PMID: 34684318 PMCID: PMC8540096 DOI: 10.3390/nu13103317] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/12/2023] Open
Abstract
The body of knowledge on alcohol use and communicable diseases has been growing in recent years. Using a narrative review approach, this paper discusses alcohol's role in the acquisition of and treatment outcomes from four different communicable diseases: these include three conditions included in comparative risk assessments to date-Human Immunodeficiency Virus (HIV)/AIDS, tuberculosis (TB), and lower respiratory infections/pneumonia-as well as Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) because of its recent and rapid ascension as a global health concern. Alcohol-attributable TB, HIV, and pneumonia combined were responsible for approximately 360,000 deaths and 13 million disability-adjusted life years lost (DALYs) in 2016, with alcohol-attributable TB deaths and DALYs predominating. There is strong evidence that alcohol is associated with increased incidence of and poorer treatment outcomes from HIV, TB, and pneumonia, via both behavioral and biological mechanisms. Preliminary studies suggest that heavy drinkers and those with alcohol use disorders are at increased risk of COVID-19 infection and severe illness. Aside from HIV research, limited research exists that can guide interventions for addressing alcohol-attributable TB and pneumonia or COVID-19. Implementation of effective individual-level interventions and alcohol control policies as a means of reducing the burden of communicable diseases is recommended.
Collapse
Affiliation(s)
- Neo K. Morojele
- Department of Psychology, University of Johannesburg, Johannesburg 2006, South Africa
| | - Sheela V. Shenoi
- Section of Infectious Diseases, Department of Medicine, Yale University School of Medicine, New Haven, CT 06510, USA;
- Yale Institute for Global Health, Yale University, New Haven, CT 06520, USA
| | - Paul A. Shuper
- Centre for Addiction and Mental Health, Institute for Mental Health Policy Research and Campbell Family Mental Health Research Institute, Toronto, ON M5S 2S1, Canada; (P.A.S.); (J.R.)
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
- Institute for Collaboration on Health, Intervention, and Policy, University of Connecticut, Storrs, CT 06269, USA
- Alcohol, Tobacco and Other Drug Research Unit, South African Medical Research Council, Pretoria 0001, South Africa
| | - Ronald Scott Braithwaite
- Division of Comparative Effectiveness and Decision Science, Department of Population Health, NYU Grossman School of Medicine, New York University, New York, NY 10013, USA;
| | - Jürgen Rehm
- Centre for Addiction and Mental Health, Institute for Mental Health Policy Research and Campbell Family Mental Health Research Institute, Toronto, ON M5S 2S1, Canada; (P.A.S.); (J.R.)
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
- Center for Interdisciplinary Addiction Research (ZIS), Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany
- Institute of Clinical Psychology and Psychotherapy, Technische Universität Dresden, 01187 Dresden, Germany
- Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
- Program on Substance Abuse, Public Health Agency of Catalonia, 08005 Barcelona, Spain
- Department of International Health Projects, Institute for Leadership and Health Management, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| |
Collapse
|
50
|
Birková A, Hubková B, Čižmárová B, Bolerázska B. Current View on the Mechanisms of Alcohol-Mediated Toxicity. Int J Mol Sci 2021; 22:9686. [PMID: 34575850 PMCID: PMC8472195 DOI: 10.3390/ijms22189686] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
Alcohol is a psychoactive substance that is widely used and, unfortunately, often abused. In addition to acute effects such as intoxication, it may cause many chronic pathological conditions. Some of the effects are very well described and explained, but there are still gaps in the explanation of empirically co-founded dysfunction in many alcohol-related conditions. This work focuses on reviewing actual knowledge about the toxic effects of ethanol and its degradation products.
Collapse
Affiliation(s)
- Anna Birková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, 04011 Kosice, Slovakia
| | - Beáta Hubková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, 04011 Kosice, Slovakia
| | - Beáta Čižmárová
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, 04011 Kosice, Slovakia
| | - Beáta Bolerázska
- 1st Department of Stomatology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, 04011 Kosice, Slovakia
| |
Collapse
|