1
|
Yu X, Wu W, Hao J, Zhou Y, Yu D, Ding W, Zhang X, Liu G, Wang J. Ginger protects against vein graft remodeling by precisely modulating ferroptotic stress in vascular smooth muscle cell dedifferentiation. J Pharm Anal 2025; 15:101053. [PMID: 39974619 PMCID: PMC11835576 DOI: 10.1016/j.jpha.2024.101053] [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: 03/20/2024] [Revised: 06/28/2024] [Accepted: 07/22/2024] [Indexed: 02/21/2025] Open
Abstract
Vein graft (VG) failure (VGF) is associated with VG intimal hyperplasia, which is characterized by abnormal accumulation of vascular smooth muscle cells (VSMCs). Most neointimal VSMCs are derived from pre-existing VSMCs via a process of VSMC phenotypic transition, also known as dedifferentiation. There is increasing evidence to suggest that ginger or its bioactive ingredients may block VSMC dedifferentiation, exerting vasoprotective functions; however, the precise mechanisms have not been fully characterized. Therefore, we investigated the effect of ginger on VSMC phenotypic transition in VG remodeling after transplantation. Ginger significantly inhibited neointimal hyperplasia and promoted lumen (L) opening in a 3-month VG, which was primarily achieved by reducing ferroptotic stress. Ferroptotic stress is a pro-ferroptotic state. Contractile VSMCs did not die but instead gained a proliferative capacity and switched to the secretory type, forming neointima (NI) after vein transplantation. Ginger and its two main vasoprotective ingredients (6-gingerol and 6-shogaol) inhibit VSMC dedifferentiation by reducing ferroptotic stress. Network pharmacology analysis revealed that 6-gingerol inhibits ferroptotic stress by targeting P53, while 6-shogaol inhibits ferroptotic stress by targeting 5-lipoxygenase (Alox5), both promoting ferroptosis. Furthermore, both ingredients co-target peroxisome proliferator-activated receptor gamma (PPARγ), decreasing PPARγ-mediated nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1 (Nox1) expression. Nox1 promotes intracellular reactive oxygen species (ROS) production and directly induces VSMC dedifferentiation. In addition, Nox1 is a ferroptosis-promoting gene that encourages ferroptotic stress production, indirectly leading to VSMC dedifferentiation. Ginger, a natural multi-targeted ferroptotic stress inhibitor, finely and effectively prevents VSMC phenotypic transition and protects against venous injury remodeling.
Collapse
Affiliation(s)
- Xiaoyu Yu
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Weiwei Wu
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Jingjun Hao
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Yuxin Zhou
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Deyang Yu
- Department of Emergency Surgery, Qingdao Central Hospital, Qingdao, Shandong, 266071, China
| | - Wei Ding
- Department of General Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266071, China
| | - Xuejuan Zhang
- Department of General Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266071, China
| | - Gaoli Liu
- Department of Cardiac Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266071, China
| | - Jianxun Wang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| |
Collapse
|
2
|
Parker D, Muhkopadyay S, Sivaraman V. Alcohol activates cannabinoid receptor 1 and 2 in a model of pathogen induced pulmonary inflammation. Toxicol Lett 2024; 401:24-34. [PMID: 39251147 PMCID: PMC11527581 DOI: 10.1016/j.toxlet.2024.08.012] [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: 02/14/2024] [Revised: 07/31/2024] [Accepted: 08/21/2024] [Indexed: 09/11/2024]
Abstract
Alcohol use disorder (AUD) is defined as patterns of alcohol misuse and affects over 30 million people in the US. AUD is a systemic disease with the epidemiology of acute lung injury and excessive alcohol use established in the literature. However, the distinct mechanisms by which alcohol induces the risk of pulmonary inflammation are less clear. A compelling body of evidence shows that cannabinoid receptors (CB1R and CB2R) play a relevant role in AUD. For this study, we investigated the role of CBR signaling in pulmonary immune activation. Using a human macrophage cell line, we evaluated the expression of CBR1 and CBR2 after cells were exposed to EtOH, +/- cannabinoid agonists and antagonists by flow cytometry. We also evaluated the expression of cannabinoid receptors from the lungs of adolescent mice exposed to acute binge EtOH +/- cannabinoid agonists and antagonists at both resting state and after microbial challenge via western blot, rt-PCR, cytokine analysis, and histology. Our results suggest that EtOH exposure modulates the expression of CBR1 and CBR2. Second, EtOH may contribute to the release of DAMPs and other proinflammatory cytokines, Finally, microbial challenge induces pulmonary inflammation in acute binge EtOH-exposed mice, and this observed immune activation may be CBR-dependent. We have shown that adolescent binge drinking primes the lung to subsequent microbial infection in adulthood and this response can be mitigated with cannabinoid antagonists. These novel findings may provide a framework for developing potential novel therapeutics in AUD research.
Collapse
MESH Headings
- Animals
- Receptor, Cannabinoid, CB2/metabolism
- Receptor, Cannabinoid, CB2/agonists
- Receptor, Cannabinoid, CB2/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB1/genetics
- Humans
- Ethanol/toxicity
- Lung/drug effects
- Lung/metabolism
- Lung/immunology
- Lung/pathology
- Mice, Inbred C57BL
- Pneumonia/chemically induced
- Pneumonia/metabolism
- Male
- Mice
- Cytokines/metabolism
- Macrophages/drug effects
- Macrophages/metabolism
- Macrophages/immunology
- Disease Models, Animal
- Cannabinoid Receptor Agonists/pharmacology
- Binge Drinking/complications
- Binge Drinking/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- De'Jana Parker
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Somnath Muhkopadyay
- The Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Vijay Sivaraman
- Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA.
| |
Collapse
|
3
|
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
|
4
|
Crotty KM, Kabir SA, Chang SS, Mehta AJ, Yeligar SM. Pioglitazone reverses alcohol-induced alterations in alveolar macrophage mitochondrial phenotype. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:810-826. [PMID: 38499395 DOI: 10.1111/acer.15300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND People with alcohol use disorder (AUD) have an increased risk of developing pneumonia and pulmonary diseases. Alveolar macrophages (AMs) are immune cells of the lower respiratory tract that are necessary for clearance of pathogens. However, alcohol causes AM oxidative stress, mitochondrial damage and dysfunction, and diminished phagocytic capacity, leading to lung injury and immune suppression. METHODS AMs were isolated by bronchoalveolar lavage from people with AUD and male and female C57BL/6J mice given chronic ethanol (20% w/v, 12 weeks) in drinking water. The peroxisome proliferator-activated receptor γ ligand, pioglitazone, was used to treat human AMs ex vivo (10 μM, 24 h) and mice in vivo by oral gavage (10 mg/kg/day). Levels of AM mitochondrial superoxide and hypoxia-inducible factor-1 alpha (HIF-1α) mRNA, a marker of oxidative stress, were measured by fluorescence microscopy and RT-qPCR, respectively. Mouse AM phagocytic ability was determined by internalized Staphylococcus aureus, and mitochondrial capacity, dependency, and flexibility for glucose, long-chain fatty acid, and glutamine oxidation were measured using an extracellular flux analyzer. In vitro studies used a murine AM cell line, MH-S (±0.08% ethanol, 72 h) to investigate mitochondrial fuel oxidation and ATP-linked respiration. RESULTS Pioglitazone treatment decreased mitochondrial superoxide in AMs from people with AUD and ethanol-fed mice and HIF-1α mRNA in ethanol-fed mouse lungs. Pioglitazone also reversed mouse AM glutamine oxidation and glucose or long-chain fatty acid flexibility to meet basal oxidation needs. In vitro, ethanol decreased the rate of AM mitochondrial and total ATP production, and pioglitazone improved changes in glucose and glutamine oxidation. CONCLUSIONS Pioglitazone reversed chronic alcohol-induced oxidative stress in human AM and mitochondrial substrate oxidation flexibility and superoxide levels in mouse AM. Decreased ethanol-induced AM HIF-1α mRNA with pioglitazone suggests that this pathway may be a focus for metabolic-targeted therapeutics to improve morbidity and mortality in people with AUD.
Collapse
Affiliation(s)
- Kathryn M Crotty
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
| | - Shayaan A Kabir
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
| | - Sarah S Chang
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
| | - Ashish J Mehta
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
| | - Samantha M Yeligar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
| |
Collapse
|
5
|
Grander C, Meyer M, Steinacher D, Claudel T, Hausmann B, Pjevac P, Grabherr F, Oberhuber G, Grander M, Brigo N, Jukic A, Schwärzler J, Weiss G, Adolph TE, Trauner M, Tilg H. 24-Norursodeoxycholic acid ameliorates experimental alcohol-related liver disease and activates hepatic PPARγ. JHEP Rep 2023; 5:100872. [PMID: 37818230 PMCID: PMC10561126 DOI: 10.1016/j.jhepr.2023.100872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/23/2023] [Accepted: 07/12/2023] [Indexed: 10/12/2023] Open
Abstract
Background & Aims Alcohol-related liver disease (ALD) is a global healthcare challenge with limited treatment options. 24-Norursodeoxycholic acid (NorUDCA) is a synthetic bile acid with anti-inflammatory properties in experimental and human cholestatic liver diseases. In the present study, we explored the efficacy of norUDCA in experimental ALD. Methods NorUDCA was tested in a preventive and therapeutic setting in an experimental ALD model (Lieber-DeCarli diet enriched with ethanol). Liver disease was phenotypically evaluated using histology and biochemical methods, and anti-inflammatory properties and peroxisome proliferator-activated receptor gamma activation by norUDCA were evaluated in cellular model systems. Results NorUDCA administration ameliorated ethanol-induced liver injury, reduced hepatocyte death, and reduced the expression of hepatic pro-inflammatory cytokines including tumour necrosis factor (Tnf), Il-1β, Il-6, and Il-10. NorUDCA shifted hepatic macrophages towards an anti-inflammatory M2 phenotype. Further, norUDCA administration altered the composition of the intestinal microbiota, specifically increasing the abundance of Roseburia, Enterobacteriaceae, and Clostridum spp. In a therapeutic model, norUDCA also ameliorated ethanol-induced liver injury. Moreover, norUDCA suppressed lipopolysaccharide-induced IL-6 expression in human peripheral blood mononuclear cells and evoked peroxisome proliferator-activated receptor gamma activation. Conclusions NorUDCA ameliorated experimental ALD, protected against hepatic inflammation, and affected gut microbial commensalism. NorUDCA could serve as a novel therapeutic agent in the future management of patients with ALD. Impact and implications Alcohol-related liver disease is a global healthcare concern with limited treatment options. 24-Norursodeoxycholic acid (NorUDCA) is a modified bile acid, which was proven to be effective in human cholestatic liver diseases. In the present study, we found a protective effect of norUDCA in experimental alcoholic liver disease. For patients with ALD, norUDCA could be a potential new treatment option.
Collapse
Affiliation(s)
- Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Moritz Meyer
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Daniel Steinacher
- Hans Popper Laboratory of Molecular Hepatology, Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Thierry Claudel
- Hans Popper Laboratory of Molecular Hepatology, Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Bela Hausmann
- Joint Microbiome Facility of the Medical University of Vienna, The University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Petra Pjevac
- Joint Microbiome Facility of the Medical University of Vienna, The University of Vienna, Vienna, Austria
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Georg Oberhuber
- INNPATH, Tirol-Kliniken University Hospital Innsbruck, Innsbruck, Austria
| | - Manuel Grander
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University Innsbruck, Innsbruck, Austria
| | - Natascha Brigo
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University Innsbruck, Innsbruck, Austria
| | - Almina Jukic
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Julian Schwärzler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University Innsbruck, Innsbruck, Austria
| | - Timon E. Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Department of Internal Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Moser SE, Brown AM, Clark BC, Arnold WD, Baumann CW. Neuromuscular mechanisms of weakness in a mouse model of chronic alcoholic myopathy. Alcohol Clin Exp Res 2022; 46:1636-1647. [PMID: 35869821 PMCID: PMC9804636 DOI: 10.1111/acer.14907] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Weakness is a common clinical symptom reported in individuals with chronic alcohol use disorder. However, it remains unclear whether low strength in these individuals is directly related to excessive ethanol intake, other deleterious factors (lifestyle, environment, genetics, etc.), or a combination of both. Therefore, we examined whether (and how) ethanol reduces the muscle's force-producing capacity using a controlled in vivo preclinical mouse model of excessive ethanol intake. METHODS To establish whether chronic ethanol consumption causes weakness, C57BL/6 female mice consumed 20% ethanol for 40 weeks (following a 2-week ethanol ramping period), and various measures of muscular force were quantified. Functional measures included all-limb grip strength and in vivo contractility of the left ankle dorsiflexors and plantarflexors. Once confirmed that mice consuming ethanol were weaker than age-matched controls, we sought to determine the potential neuromuscular mechanisms of muscle dysfunction by assessing neuromuscular excitation, muscle quantity, and muscle quality. RESULTS Mice consuming chronic ethanol were 13 to 16% weaker (p ≤ 0.016) than controls (i.e., mice consuming 100% water) with the negative impact of ethanol on voluntary grip strength (ƞ2 = 0.603) being slightly larger than that of electrically stimulated muscle contractility (ƞ2 = 0.482). Relative to controls, lean mass and muscle wet masses were 9 to 16% lower in ethanol-consuming mice (p ≤ 0.048, ƞ2 ≥ 0.268). No significant changes were observed between groups for indices of neuromuscular excitation at the level of the motor unit, neuromuscular junction, or plasmalemma (p ≥ 0.259, ƞ2 ≤ 0.097), nor was muscle quality altered after 40 weeks of 20% ethanol consumption (p ≥ 0.695, ƞ2 ≤ 0.012). CONCLUSIONS Together, these findings establish that chronic ethanol consumption in mice induces a substantial weakness in vivo that we interpret to be primarily due to muscle atrophy (i.e., reduced muscle quantity) and possibly, to a lesser degree, loss of central neural drive.
Collapse
Affiliation(s)
- Samantha E. Moser
- Honors Tutorial CollegeOhio UniversityAthensOhioUSA,Ohio Musculoskeletal and Neurological Institute (OMNI)Ohio UniversityAthensOhioUSA
| | - Austin M. Brown
- Honors Tutorial CollegeOhio UniversityAthensOhioUSA,Ohio Musculoskeletal and Neurological Institute (OMNI)Ohio UniversityAthensOhioUSA
| | - Brian C. Clark
- Ohio Musculoskeletal and Neurological Institute (OMNI)Ohio UniversityAthensOhioUSA,Department of Biomedical SciencesOhio UniversityAthensOhioUSA
| | - W. David Arnold
- Department of NeurologyThe Ohio State University Wexner Medical CenterColumbusOhioUSA,Department of Physical Medicine and RehabilitationThe Ohio State University Wexner Medical CenterColumbusOhioUSA,Department of NeuroscienceThe Ohio State University Wexner Medical CenterColumbusOhioUSA,Department of Physiology and Cell BiologyThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Cory W. Baumann
- Ohio Musculoskeletal and Neurological Institute (OMNI)Ohio UniversityAthensOhioUSA,Department of Biomedical SciencesOhio UniversityAthensOhioUSA
| |
Collapse
|
9
|
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
|
10
|
Morris NL, Michael DN, Crotty KM, Chang SS, Yeligar SM. Alcohol-Induced Glycolytic Shift in Alveolar Macrophages Is Mediated by Hypoxia-Inducible Factor-1 Alpha. Front Immunol 2022; 13:865492. [PMID: 35634337 PMCID: PMC9130492 DOI: 10.3389/fimmu.2022.865492] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/15/2022] [Indexed: 12/20/2022] Open
Abstract
Excessive alcohol use increases the risk of developing respiratory infections partially due to impaired alveolar macrophage (AM) phagocytic capacity. Previously, we showed that chronic ethanol (EtOH) exposure led to mitochondrial derangements and diminished oxidative phosphorylation in AM. Since oxidative phosphorylation is needed to meet the energy demands of phagocytosis, EtOH mediated decreases in oxidative phosphorylation likely contribute to impaired AM phagocytosis. Treatment with the peroxisome proliferator-activated receptor gamma (PPARγ) ligand, pioglitazone (PIO), improved EtOH-mediated decreases in oxidative phosphorylation. In other models, hypoxia-inducible factor-1 alpha (HIF-1α) has been shown to mediate the switch from oxidative phosphorylation to glycolysis; however, the role of HIF-1α in chronic EtOH mediated derangements in AM has not been explored. We hypothesize that AM undergo a metabolic shift from oxidative phosphorylation to a glycolytic phenotype in response to chronic EtOH exposure. Further, we speculate that HIF-1α is a critical mediator of this metabolic switch. To test these hypotheses, primary mouse AM (mAM) were isolated from a mouse model of chronic EtOH consumption and a mouse AM cell line (MH-S) were exposed to EtOH in vitro. Expression of HIF-1α, glucose transporters (Glut1 and 4), and components of the glycolytic pathway (Pfkfb3 and PKM2), were measured by qRT-PCR and western blot. Lactate levels (lactate assay), cell energy phenotype (extracellular flux analyzer), glycolysis stress tests (extracellular flux analyzer), and phagocytic function (fluorescent microscopy) were conducted. EtOH exposure increased expression of HIF-1α, Glut1, Glut4, Pfkfb3, and PKM2 and shifted AM to a glycolytic phenotype. Pharmacological stabilization of HIF-1α via cobalt chloride treatment in vitro mimicked EtOH-induced AM derangements (increased glycolysis and diminished phagocytic capacity). Further, PIO treatment diminished HIF-1α levels and reversed glycolytic shift following EtOH exposure. These studies support a critical role for HIF-1α in mediating the glycolytic shift in energy metabolism of AM during excessive alcohol use.
Collapse
Affiliation(s)
- Niya L Morris
- 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
| | - David N Michael
- 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
| | - 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
| | - Sarah S Chang
- 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
|
11
|
Wigger GW, Bouton TC, Jacobson KR, Auld SC, Yeligar SM, Staitieh BS. The Impact of Alcohol Use Disorder on Tuberculosis: A Review of the Epidemiology and Potential Immunologic Mechanisms. Front Immunol 2022; 13:864817. [PMID: 35432348 PMCID: PMC9009367 DOI: 10.3389/fimmu.2022.864817] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
Globally, an estimated 107 million people have an alcohol use disorder (AUD) leading to 2.8 million premature deaths each year. Tuberculosis (TB) is one of the leading causes of death globally and over 8% of global TB cases are estimated to be attributable to AUD. Social determinants of health such as poverty and undernutrition are often shared among those with AUD and TB and could explain the epidemiologic association between them. However, recent studies suggest that these shared risk factors do not fully account for the increased risk of TB in people with AUD. In fact, AUD has been shown to be an independent risk factor for TB, with a linear increase in the risk for TB with increasing alcohol consumption. While few studies have focused on potential biological mechanisms underlying the link between AUD and TB, substantial overlap exists between the effects of alcohol on lung immunity and the mechanisms exploited by Mycobacterium tuberculosis (Mtb) to establish infection. Alcohol misuse impairs the immune functions of the alveolar macrophage, the resident innate immune effector in the lung and the first line of defense against Mtb in the lower respiratory tract. Chronic alcohol ingestion also increases oxidative stress in the alveolar space, which could in turn facilitate Mtb growth. In this manuscript, we review the epidemiologic data that links AUD to TB. We discuss the existing literature on the potential mechanisms by which alcohol increases the risk of TB and review the known effects of alcohol ingestion on lung immunity to elucidate other mechanisms that Mtb may exploit. A more in-depth understanding of the link between AUD and TB will facilitate the development of dual-disease interventions and host-directed therapies to improve lung health and long-term outcomes of TB.
Collapse
Affiliation(s)
- Gregory W Wigger
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Tara C Bouton
- Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Karen R Jacobson
- Section of Infectious Diseases, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Sara C Auld
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States.,Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Samantha M Yeligar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States.,Atlanta VA Medical Center, Atlanta, GA, United States
| | - Bashar S Staitieh
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
12
|
Development of mode of action networks related to the potential role of PPARγ in respiratory diseases. Pharmacol Res 2021; 172:105821. [PMID: 34403731 DOI: 10.1016/j.phrs.2021.105821] [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: 02/04/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 11/30/2022]
Abstract
The peroxisome proliferator-activated receptor γ (PPARγ) is a key transcription factor, operating at the intercept of metabolic control and immunomodulation. It is ubiquitously expressed in multiple tissues and organs, including lungs. There is a growing body of information supporting the role of PPARγ signalling in respiratory diseases. The aim of the present study was to develop mode of action (MoA) networks reflecting the relationships between PPARγ signalling and the progression/alleviation of a spectrum of lung pathologies. Data mining was performed using the resources of the NIH PubMed and PubChem information systems. By linking available data on pathological/therapeutic effects of PPARγ modulation, knowledge-based MoA networking at different levels of biological organization (molecular, cellular, tissue, organ, and system) was performed. Multiple MoA networks were developed to relate PPARγ modulation to the progress or the alleviation of pulmonary disorders, triggered by diverse pathogenic, genetic, chemical, or mechanical factors. Pharmacological targeting of PPARγ signalling was discussed with regard to ligand- and cell type-specific effects in the context of distinct disease inductor- and disease stage-dependent patterns. The proposed MoA networking analysis allows for a better understanding of the potential role of PPARγ modulation in lung pathologies. It presents a mechanistically justified basis for further computational, experimental, and clinical monitoring studies on the dynamic control of PPARγ signalling in respiratory diseases.
Collapse
|
13
|
Yeligar SM, Mehta AJ, Harris FL, Brown LAS, Hart CM. Pioglitazone Reverses Alcohol-Induced Alveolar Macrophage Phagocytic Dysfunction. THE JOURNAL OF IMMUNOLOGY 2021; 207:483-492. [PMID: 34193599 DOI: 10.4049/jimmunol.2000565] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/09/2021] [Indexed: 01/02/2023]
Abstract
Alcohol use disorders (AUD) increase susceptibility to respiratory infections by 2- to 4-fold in part because of impaired alveolar macrophage (AM) immune function. Alcohol causes AM oxidative stress, diminishing AM phagocytic capacity and clearance of microbes from the alveolar space. Alcohol increases AM NADPH oxidases (Noxes), primary sources of AM oxidative stress, and reduces peroxisome proliferator-activated receptor γ (PPARγ) expression, a critical regulator of AM immune function. To investigate the underlying mechanisms of these alcohol-induced AM derangements, we hypothesized that alcohol stimulates CCAAT/enhancer-binding protein β (C/EBPβ) to suppress Nox-related microRNAs (miRs), thereby enhancing AM Nox expression, oxidative stress, and phagocytic dysfunction. Furthermore, we postulated that pharmacologic PPARγ activation with pioglitazone would inhibit C/EBPβ and attenuate alcohol-induced AM dysfunction. AM isolated from human AUD subjects or otherwise healthy control subjects were examined. Compared with control AM, alcohol activated AM C/EBPβ, decreased Nox1-related miR-1264 and Nox2-related miR-107, and increased Nox1, Nox2, and Nox4 expression and activity. These alcohol-induced AM derangements were abrogated by inhibition of C/EBPβ, overexpression of miR-1264 or miR-107, or pioglitazone treatment. These findings define novel molecular mechanisms of alcohol-induced AM dysfunction mediated by C/EBPβ and Nox-related miRs that are amenable to therapeutic targeting with PPARγ ligands. These results demonstrate that PPARγ ligands provide a novel and rapidly translatable strategy to mitigate susceptibility to respiratory infections and related morbidity in individuals with AUD.
Collapse
Affiliation(s)
- Samantha M Yeligar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA; .,Atlanta Veterans Affairs Health Care System, Decatur, GA; and
| | - Ashish J Mehta
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA.,Atlanta Veterans Affairs Health Care System, Decatur, GA; and
| | - Frank L Harris
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University and Children's Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, GA
| | - Lou Ann S Brown
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University and Children's Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, GA
| | - C Michael Hart
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA.,Atlanta Veterans Affairs Health Care System, Decatur, GA; and
| |
Collapse
|
14
|
Morris NL, Harris FL, Brown LAS, Yeligar SM. Alcohol induces mitochondrial derangements in alveolar macrophages by upregulating NADPH oxidase 4. Alcohol 2021; 90:27-38. [PMID: 33278514 DOI: 10.1016/j.alcohol.2020.11.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/11/2020] [Accepted: 11/25/2020] [Indexed: 12/15/2022]
Abstract
Excessive alcohol users have increased risk of developing respiratory infections in part due to oxidative stress-induced alveolar macrophage (AM) phagocytic dysfunction. Chronic ethanol exposure increases cellular oxidative stress in AMs via upregulation of NADPH oxidase (Nox) 4, and treatment with the peroxisome proliferator-activated receptor gamma (PPARγ) ligand, rosiglitazone, decreases ethanol-induced Nox4. However, the mechanism by which ethanol induces Nox4 expression and the PPARγ ligand reverses this defect has not been elucidated. Since microRNA (miR)-92a has been predicted to target Nox4 for destabilization, we hypothesized that ethanol exposure decreases miR-92a expression and leads to Nox4 upregulation. Previous studies have implicated mitochondrial-derived oxidative stress in AM dysfunction. We further hypothesized that ethanol increases mitochondrial-derived AM oxidative stress and dysfunction via miR-92a, and that treatment with the PPARγ ligand, pioglitazone, could reverse these derangements. To test these hypotheses, a mouse AM cell line, MH-S cells, was exposed to ethanol in vitro, and primary AMs were isolated from a mouse model of chronic ethanol consumption to measure Nox4, mitochondrial target mRNA (qRT-PCR) and protein levels (confocal microscopy), mitochondria-derived reactive oxygen species (confocal immunofluorescence), mitochondrial fission (electron microscopy), and mitochondrial bioenergetics (extracellular flux analyzer). Ethanol exposure increased Nox4, enhanced mitochondria-derived oxidative stress, augmented mitochondrial fission, and impaired mitochondrial bioenergetics. Transfection with a miR-92a mimic in vitro or pioglitazone treatment in vivo diminished Nox4 levels, resulting in improvements in these ethanol-mediated derangements. These findings demonstrate that pioglitazone may provide a novel therapeutic approach to mitigate ethanol-induced AM mitochondrial derangements.
Collapse
|
15
|
Potential Effects of Nutraceuticals in Retinopathy of Prematurity. Life (Basel) 2021; 11:life11020079. [PMID: 33499180 PMCID: PMC7912639 DOI: 10.3390/life11020079] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 02/07/2023] Open
Abstract
Retinopathy of prematurity (ROP), the most common cause of childhood blindness, is a hypoxia-induced eye disease characterized by retinal neovascularization. In the normal retina, a well-organized vascular network provides oxygen and nutrients as energy sources to maintain a normal visual function; however, it is disrupted when pathological angiogenesis is induced in ROP patients. Under hypoxia, inadequate oxygen and energy supply lead to oxidative stress and stimulate neovasculature formation as well as affecting the function of photoreceptors. In order to meet the metabolic needs in the developing retina, protection against abnormal vascular formation is one way to manage ROP. Although current treatments provide beneficial effects in reducing the severity of ROP, these invasive therapies may also induce life-long consequences such as systemic structural and functional complications as well as neurodevelopment disruption in the developing infants. Nutritional supplements for the newborns are a novel concept for restoring energy supply by protecting the retinal vasculature and may lead to better ROP management. Nutraceuticals are provided in a non-invasive manner without the developmental side effects associated with current treatments. These nutraceuticals have been investigated through various in vitro and in vivo methods and are indicated to protect retinal vasculature. Here, we reviewed and discussed how the use of these nutraceuticals may be beneficial in ROP prevention and management.
Collapse
|
16
|
McMahan RH, Afshar M, Amedee AM, Bishehsari F, Carr RM, Coleman LG, Herrnreiter CJ, Lewis SL, Mandrekar P, McCullough RL, Morris NL, Vasiliou V, Wang HJ, Yeligar SM, Choudhry MA, Kovacs EJ. Summary of the 2019 alcohol and immunology research interest group (AIRIG) meeting: Alcohol-mediated mechanisms of multiple organ injury. Alcohol 2020; 87:89-95. [PMID: 32353591 PMCID: PMC7483664 DOI: 10.1016/j.alcohol.2020.04.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 12/30/2022]
Abstract
On November 15, 2019, the 24th annual Alcohol and Immunology Research Interest Group (AIRIG) meeting was held as a satellite conference during the annual Society for Leukocyte Biology meeting in Boston, Massachusetts. The 2019 meeting focused on alcohol, immunity, and organ damage, and included two plenary sessions. The first session highlighted new research exploring the mechanisms of alcohol-induced inflammation and liver disease, including effects on lipidomics and lipophagy, regulatory T cells, epigenetics, epithelial cells, and age-related changes in the gut. The second session covered alcohol-induced injury of other organs, encompassing diverse areas of research ranging from neurodegeneration, to lung barrier function, to colon carcinogenesis, to effects on viral infection. The discussions also highlighted current laboratory and clinical research used to identify biomarkers of alcohol use and disease.
Collapse
Affiliation(s)
- Rachel H McMahan
- Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Alcohol Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.
| | - Majid Afshar
- Division of Pulmonary and Critical Care Medicine, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA; Alcohol Research Program, Burn and Shock Trauma Research Institute, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA
| | - Angela M Amedee
- Department of Microbiology, Immunology, and Parasitology and the Comprehensive Alcohol Research Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Faraz Bishehsari
- Department of Internal Medicine, Division of Gastroenterology, Rush University Medical Center, Chicago, IL, USA
| | - Rotonya M Carr
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leon G Coleman
- Department of Pharmacology, Bowles Center for Alcohol Studies, UNC-Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Caroline J Herrnreiter
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA
| | - Sloan L Lewis
- Department of Molecular Biology and Biochemistry, University of California-Irvine, Irvine, CA, USA
| | - Pranoti Mandrekar
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Rebecca L McCullough
- Alcohol Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Niya L Morris
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep, Emory University and Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - H Joe Wang
- Division of Metabolism and Health Effects, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, USA
| | - Samantha M Yeligar
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep, Emory University and Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| | - Mashkoor A Choudhry
- Alcohol Research Program, Burn and Shock Trauma Research Institute, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA; Department of Surgery, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA
| | - Elizabeth J Kovacs
- Department of Surgery, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Alcohol Research Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Immunology Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
17
|
Sun X, Lu Q, Yegambaram M, Kumar S, Qu N, Srivastava A, Wang T, Fineman JR, Black SM. TGF-β1 attenuates mitochondrial bioenergetics in pulmonary arterial endothelial cells via the disruption of carnitine homeostasis. Redox Biol 2020; 36:101593. [PMID: 32554303 PMCID: PMC7303661 DOI: 10.1016/j.redox.2020.101593] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 12/23/2022] Open
Abstract
Transforming growth factor beta-1 (TGF-β1) signaling is increased and mitochondrial function is decreased in multiple models of pulmonary hypertension (PH) including lambs with increased pulmonary blood flow (PBF) and pressure (Shunt). However, the potential link between TGF-β1 and the loss of mitochondrial function has not been investigated and was the focus of our investigations. Our data indicate that exposure of pulmonary arterial endothelial cells (PAEC) to TGF-β1 disrupted mitochondrial function as determined by enhanced mitochondrial ROS generation, decreased mitochondrial membrane potential, and disrupted mitochondrial bioenergetics. These events resulted in a decrease in cellular ATP levels, decreased hsp90/eNOS interactions and attenuated shear-mediated NO release. TGF-β1 induced mitochondrial dysfunction was linked to a nitration-mediated activation of Akt1 and the subsequent mitochondrial translocation of endothelial NO synthase (eNOS) resulting in the nitration of carnitine acetyl transferase (CrAT) and the disruption of carnitine homeostasis. The increase in Akt1 nitration correlated with increased NADPH oxidase activity associated with increased levels of p47phox, p67phox, and Rac1. The increase in NADPH oxidase was associated with a decrease in peroxisome proliferator-activated receptor type gamma (PPARγ) and the PPARγ antagonist, GW9662, was able to mimic the disruptive effect of TGF-β1 on mitochondrial bioenergetics. Together, our studies reveal for the first time, that TGF-β1 can disrupt mitochondrial function through the disruption of cellular carnitine homeostasis and suggest that stimulating carinitine homeostasis may be an avenue to treat pulmonary vascular disease.
Collapse
Affiliation(s)
- Xutong Sun
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA
| | - Qing Lu
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA
| | - Manivannan Yegambaram
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA
| | - Sanjiv Kumar
- Center for Blood Disorders, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Ning Qu
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA
| | - Anup Srivastava
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA
| | - Ting Wang
- Department of Internal Medicine University of Arizona, Phoenix, AZ, 85004, The Department of Pediatrics and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jeffrey R Fineman
- Department of Internal Medicine University of Arizona, Phoenix, AZ, 85004, The Department of Pediatrics and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Stephen M Black
- Department of Medicine, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
18
|
Zhang W, Chihade DB, Xie J, Chen CW, Ramonell KM, Liang Z, Coopersmith CM, Ford ML. Preexisting malignancy abrogates the beneficial effects of CXCR4 blockade during sepsis. J Leukoc Biol 2020; 107:485-495. [PMID: 31985098 DOI: 10.1002/jlb.3a1019-502r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 10/21/2018] [Accepted: 12/30/2019] [Indexed: 11/06/2022] Open
Abstract
Patients with cancer are at an increased risk of developing and dying from sepsis. We previously reported that blockade of the chemokine receptor CXCR4 resulted in decreased CD4+ T cell exhaustion and improved survival in a model of polymicrobial sepsis in previously healthy mice. Here, we sought to determine whether CXCR4 blockade could improve mortality and immune dysregulation during sepsis complicated with malignancy. Results in animals inoculated with a lung cancer cell line and subjected to CLP 3 weeks later indicated that CXCR4 was up-regulated on naïve and central memory T cells following sepsis. Of note, and in contrast to results in previously healthy mice, CXCR4 blockade failed to improve survival in cancer septic animals; instead, it actually significantly worsened survival. In the setting of cancer, CXCR4 blockade failed to result in T cell egress from the bone marrow, reverse lymphopenia in the spleen, or reverse T cell exhaustion. Mechanistically, elevated expression of CD69 on naïve T cells in the bone marrow of cancer septic animals was associated with their inability to egress from the bone marrow in the setting of CXCR4 blockade. In conclusion, these results illuminate the differential impact of CXCR4 blockade on sepsis pathophysiology in the setting of cancer and highlight the need for personalized therapy during sepsis.
Collapse
Affiliation(s)
- Wenxiao Zhang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA.,Department of Critical Care Medicine, People's Hospital of Zhengzhou University (Henan Provincial People's Hospital), Zhengzhou, China
| | - Deena B Chihade
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jianfeng Xie
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ching-Wen Chen
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kimberly M Ramonell
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Zhe Liang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Craig M Coopersmith
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA.,Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mandy L Ford
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA.,Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
19
|
El-Saka MH, Madi NM, Ibrahim RR, Alghazaly GM, Elshwaikh S, El-Bermawy M. The ameliorative effect of angiotensin 1-7 on experimentally induced-preeclampsia in rats: Targeting the role of peroxisome proliferator-activated receptors gamma expression & asymmetric dimethylarginine. Arch Biochem Biophys 2019; 671:123-129. [PMID: 31295432 DOI: 10.1016/j.abb.2019.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/02/2019] [Accepted: 07/07/2019] [Indexed: 01/10/2023]
Abstract
This study was designed to explore the effect of angiotensin 1-7 (Ang 1-7) on experimentally induced-preeclampsia in Wistar rats targeting the role of peroxisome proliferator-activated receptors gamma expression (PPARs-γ) & asymmetric dimethylarginine (ADMA). 30 female Wistar rats were divided into three groups: Normal pregnant (NP), preeclampsia (PE), and preeclampsia treated with Ang 1-7 (PE + Ang 1-7) groups. Reduced uterine perfusion pressure (RUPP) model was induced on GD14. On GD18, protein in urine, urine volume and urinary sodium excretion were determined. On GD19, the systolic blood pressure (SBP) was measured, and the gene expression of PPARs-γ were determined. The serum samples were separated for determination of Ang 1-7, ADMA, soluble fms-like tyrosine kinase (sFlt-1), vascular endothelial growth factor (VEGF), nitric oxide (NO) products, endothelial nitric oxide synthase (eNOS) activity, interleukin-6 (IL-6), interleukin-10 (IL-10), malondialdehyde (MDA), and total anti-oxidant capacity (T-AOC). Compared to NP group, SBP, urine protein, serum levels of ADMA, sFlt-1, IL-6 and MDA significantly increased, while expression of PPARs-γ, serum levels of Ang 1-7, VEGF, NO products, eNOS, IL-10 and T-AOC significantly decreased in PE group, while treatment of Ang 1-7 significantly ameliorated all these studied parameters as compared to PE group. We concluded that Ang 1-7 attenuated the symptoms of preeclampsia, which might be via increasing the expression of PPARs-γ and reduction of ADMA levels which could explain its anti-hypertensive, anti-angiogenic, anti-inflammatory and antioxidant effects.
Collapse
Affiliation(s)
- Mervat H El-Saka
- The Department of Physiology, Faculty of Medicine, Tanta University, Egypt.
| | - Nermin M Madi
- The Department of Physiology, Faculty of Medicine, Tanta University, Egypt
| | - Rowida Raafat Ibrahim
- The Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Tanta University, Egypt
| | | | - Shereef Elshwaikh
- The Department of Gynecology and Obstetric, Faculty of Medicine, Tanta University, Egypt
| | - Manal El-Bermawy
- The Department of Anatomy, Faculty of Medicine, Tanta University, Egypt
| |
Collapse
|
20
|
Akiyama TE, Skelhorne-Gross GE, Lightbody ED, Rubino RE, Shi JY, McNamara LA, Sharma N, Zycband EI, Gonzalez FJ, Liu H, Woods JW, Chang CH, Berger JP, Nicol CJB. Endothelial Cell-Targeted Deletion of PPAR γ Blocks Rosiglitazone-Induced Plasma Volume Expansion and Vascular Remodeling in Adipose Tissue. J Pharmacol Exp Ther 2019; 368:514-523. [PMID: 30606762 PMCID: PMC11047031 DOI: 10.1124/jpet.118.250985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 12/06/2018] [Indexed: 12/13/2022] Open
Abstract
Thiazolidinediones (TZDs) are peroxisome proliferator-activated receptor γ (PPARγ) agonists that represent an effective class of insulin-sensitizing agents; however, clinical use is associated with weight gain and peripheral edema. To elucidate the role of PPARγ expression in endothelial cells (ECs) in these side effects, EC-targeted PPARγ knockout (Pparg ΔEC) mice were placed on a high-fat diet to promote PPARγ agonist-induced plasma volume expansion, and then treated with the TZD rosiglitazone. Compared with Pparg-floxed wild-type control (Pparg f/f) mice, Pparg ΔEC treated with rosiglitazone are resistant to an increase in extracellular fluid, water content in epididymal and inguinal white adipose tissue, and plasma volume expansion. Interestingly, histologic assessment confirmed significant rosiglitazone-mediated capillary dilation within white adipose tissue of Pparg f/f mice, but not Pparg ΔEC mice. Analysis of ECs isolated from untreated mice in both strains suggested the involvement of changes in endothelial junction formation. Specifically, compared with cells from Pparg f/f mice, Pparg ΔEC cells had a 15-fold increase in focal adhesion kinase, critically important in EC focal adhesions, and >3-fold significant increase in vascular endothelial cadherin, the main component of focal adhesions. Together, these results indicate that rosiglitazone has direct effects on the endothelium via PPARγ activation and point toward a critical role for PPARγ in ECs during rosiglitazone-mediated plasma volume expansion.
Collapse
Affiliation(s)
- Taro E Akiyama
- Cardiometabolic Disorders Department, Merck Research Laboratories, Kenilworth, New Jersey (T.E.A., L.A.M., N.S., E.I.Z., H.L., J.W.W., C.H.C., J.P.B.); Department of Pathology and Molecular Medicine (G.E.S.-G., E.D.L., C.J.B.N.), Cancer Biology and Genetics Division, Cancer Research Institute (R.E.R., C.J.B.N.), and Department of Biomedical and Molecular Sciences (J.Y.S., C.J.B.N.), Queen's University, Kingston, Ontario, Canada; National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts (J.P.B.)
| | - Graham E Skelhorne-Gross
- Cardiometabolic Disorders Department, Merck Research Laboratories, Kenilworth, New Jersey (T.E.A., L.A.M., N.S., E.I.Z., H.L., J.W.W., C.H.C., J.P.B.); Department of Pathology and Molecular Medicine (G.E.S.-G., E.D.L., C.J.B.N.), Cancer Biology and Genetics Division, Cancer Research Institute (R.E.R., C.J.B.N.), and Department of Biomedical and Molecular Sciences (J.Y.S., C.J.B.N.), Queen's University, Kingston, Ontario, Canada; National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts (J.P.B.)
| | - Elizabeth D Lightbody
- Cardiometabolic Disorders Department, Merck Research Laboratories, Kenilworth, New Jersey (T.E.A., L.A.M., N.S., E.I.Z., H.L., J.W.W., C.H.C., J.P.B.); Department of Pathology and Molecular Medicine (G.E.S.-G., E.D.L., C.J.B.N.), Cancer Biology and Genetics Division, Cancer Research Institute (R.E.R., C.J.B.N.), and Department of Biomedical and Molecular Sciences (J.Y.S., C.J.B.N.), Queen's University, Kingston, Ontario, Canada; National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts (J.P.B.)
| | - Rachel E Rubino
- Cardiometabolic Disorders Department, Merck Research Laboratories, Kenilworth, New Jersey (T.E.A., L.A.M., N.S., E.I.Z., H.L., J.W.W., C.H.C., J.P.B.); Department of Pathology and Molecular Medicine (G.E.S.-G., E.D.L., C.J.B.N.), Cancer Biology and Genetics Division, Cancer Research Institute (R.E.R., C.J.B.N.), and Department of Biomedical and Molecular Sciences (J.Y.S., C.J.B.N.), Queen's University, Kingston, Ontario, Canada; National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts (J.P.B.)
| | - Jia Yue Shi
- Cardiometabolic Disorders Department, Merck Research Laboratories, Kenilworth, New Jersey (T.E.A., L.A.M., N.S., E.I.Z., H.L., J.W.W., C.H.C., J.P.B.); Department of Pathology and Molecular Medicine (G.E.S.-G., E.D.L., C.J.B.N.), Cancer Biology and Genetics Division, Cancer Research Institute (R.E.R., C.J.B.N.), and Department of Biomedical and Molecular Sciences (J.Y.S., C.J.B.N.), Queen's University, Kingston, Ontario, Canada; National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts (J.P.B.)
| | - Lesley A McNamara
- Cardiometabolic Disorders Department, Merck Research Laboratories, Kenilworth, New Jersey (T.E.A., L.A.M., N.S., E.I.Z., H.L., J.W.W., C.H.C., J.P.B.); Department of Pathology and Molecular Medicine (G.E.S.-G., E.D.L., C.J.B.N.), Cancer Biology and Genetics Division, Cancer Research Institute (R.E.R., C.J.B.N.), and Department of Biomedical and Molecular Sciences (J.Y.S., C.J.B.N.), Queen's University, Kingston, Ontario, Canada; National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts (J.P.B.)
| | - Neelam Sharma
- Cardiometabolic Disorders Department, Merck Research Laboratories, Kenilworth, New Jersey (T.E.A., L.A.M., N.S., E.I.Z., H.L., J.W.W., C.H.C., J.P.B.); Department of Pathology and Molecular Medicine (G.E.S.-G., E.D.L., C.J.B.N.), Cancer Biology and Genetics Division, Cancer Research Institute (R.E.R., C.J.B.N.), and Department of Biomedical and Molecular Sciences (J.Y.S., C.J.B.N.), Queen's University, Kingston, Ontario, Canada; National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts (J.P.B.)
| | - Emanuel I Zycband
- Cardiometabolic Disorders Department, Merck Research Laboratories, Kenilworth, New Jersey (T.E.A., L.A.M., N.S., E.I.Z., H.L., J.W.W., C.H.C., J.P.B.); Department of Pathology and Molecular Medicine (G.E.S.-G., E.D.L., C.J.B.N.), Cancer Biology and Genetics Division, Cancer Research Institute (R.E.R., C.J.B.N.), and Department of Biomedical and Molecular Sciences (J.Y.S., C.J.B.N.), Queen's University, Kingston, Ontario, Canada; National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts (J.P.B.)
| | - Frank J Gonzalez
- Cardiometabolic Disorders Department, Merck Research Laboratories, Kenilworth, New Jersey (T.E.A., L.A.M., N.S., E.I.Z., H.L., J.W.W., C.H.C., J.P.B.); Department of Pathology and Molecular Medicine (G.E.S.-G., E.D.L., C.J.B.N.), Cancer Biology and Genetics Division, Cancer Research Institute (R.E.R., C.J.B.N.), and Department of Biomedical and Molecular Sciences (J.Y.S., C.J.B.N.), Queen's University, Kingston, Ontario, Canada; National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts (J.P.B.)
| | - Haiying Liu
- Cardiometabolic Disorders Department, Merck Research Laboratories, Kenilworth, New Jersey (T.E.A., L.A.M., N.S., E.I.Z., H.L., J.W.W., C.H.C., J.P.B.); Department of Pathology and Molecular Medicine (G.E.S.-G., E.D.L., C.J.B.N.), Cancer Biology and Genetics Division, Cancer Research Institute (R.E.R., C.J.B.N.), and Department of Biomedical and Molecular Sciences (J.Y.S., C.J.B.N.), Queen's University, Kingston, Ontario, Canada; National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts (J.P.B.)
| | - John W Woods
- Cardiometabolic Disorders Department, Merck Research Laboratories, Kenilworth, New Jersey (T.E.A., L.A.M., N.S., E.I.Z., H.L., J.W.W., C.H.C., J.P.B.); Department of Pathology and Molecular Medicine (G.E.S.-G., E.D.L., C.J.B.N.), Cancer Biology and Genetics Division, Cancer Research Institute (R.E.R., C.J.B.N.), and Department of Biomedical and Molecular Sciences (J.Y.S., C.J.B.N.), Queen's University, Kingston, Ontario, Canada; National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts (J.P.B.)
| | - C H Chang
- Cardiometabolic Disorders Department, Merck Research Laboratories, Kenilworth, New Jersey (T.E.A., L.A.M., N.S., E.I.Z., H.L., J.W.W., C.H.C., J.P.B.); Department of Pathology and Molecular Medicine (G.E.S.-G., E.D.L., C.J.B.N.), Cancer Biology and Genetics Division, Cancer Research Institute (R.E.R., C.J.B.N.), and Department of Biomedical and Molecular Sciences (J.Y.S., C.J.B.N.), Queen's University, Kingston, Ontario, Canada; National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts (J.P.B.)
| | - Joel P Berger
- Cardiometabolic Disorders Department, Merck Research Laboratories, Kenilworth, New Jersey (T.E.A., L.A.M., N.S., E.I.Z., H.L., J.W.W., C.H.C., J.P.B.); Department of Pathology and Molecular Medicine (G.E.S.-G., E.D.L., C.J.B.N.), Cancer Biology and Genetics Division, Cancer Research Institute (R.E.R., C.J.B.N.), and Department of Biomedical and Molecular Sciences (J.Y.S., C.J.B.N.), Queen's University, Kingston, Ontario, Canada; National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts (J.P.B.)
| | - Christopher J B Nicol
- Cardiometabolic Disorders Department, Merck Research Laboratories, Kenilworth, New Jersey (T.E.A., L.A.M., N.S., E.I.Z., H.L., J.W.W., C.H.C., J.P.B.); Department of Pathology and Molecular Medicine (G.E.S.-G., E.D.L., C.J.B.N.), Cancer Biology and Genetics Division, Cancer Research Institute (R.E.R., C.J.B.N.), and Department of Biomedical and Molecular Sciences (J.Y.S., C.J.B.N.), Queen's University, Kingston, Ontario, Canada; National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Takeda Pharmaceuticals International, Inc., Cambridge, Massachusetts (J.P.B.)
| |
Collapse
|
21
|
Yeligar SM, Ward JM, Harris FL, Brown LAS, Guidot DM, Cribbs SK. Dysregulation of Alveolar Macrophage PPARγ, NADPH Oxidases, and TGFβ 1 in Otherwise Healthy HIV-Infected Individuals. AIDS Res Hum Retroviruses 2017; 33:1018-1026. [PMID: 28314381 DOI: 10.1089/aid.2016.0030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Despite antiretroviral therapy (ART), respiratory infections increase mortality in individuals living with chronic human immunodeficiency virus (HIV) infection. In experimental and clinical studies of chronic HIV infection, alveolar macrophages (AMs) exhibit impaired phagocytosis and bacterial clearance. Peroxisome proliferator-activated receptor (PPAR)γ, NADPH oxidase (Nox) isoforms Nox1, Nox2, Nox4, and transforming growth factor-beta 1 (TGFβ1) are critical mediators of AM oxidative stress and phagocytic dysfunction. Therefore, we hypothesized that HIV alters AM expression of these targets, resulting in chronic lung oxidative stress and subsequent immune dysfunction. A cross-sectional study of HIV-infected (n = 22) and HIV-uninfected (n = 6) subjects was conducted. Bronchoalveolar lavage (BAL) was performed, and AMs were isolated. Lung H2O2 generation was determined by measuring H2O2 in the BAL fluid. In AMs, PPARγ, Nox1, Nox2, Nox4, and TGFβ1 mRNA (quantitative real-time polymerase chain reaction) and protein (fluorescent immunomicroscopy) levels were assessed. Compared with HIV-uninfected (control) subjects, HIV-infected subjects were relatively older and the majority were African American; ∼86% were on ART, and their median CD4 count was 445, with a median viral load of 0 log copies/ml. HIV infection was associated with increased H2O2 in the BAL, decreased AM mRNA and protein levels of PPARγ, and increased AM mRNA and protein levels of Nox1, Nox2, Nox4, and TGFβ1. PPARγ attenuation and increases in Nox1, Nox2, Nox4, and TGFβ1 contribute to AM oxidative stress and immune dysfunction in the AMs of otherwise healthy HIV-infected subjects. These findings provide novel insights into the molecular mechanisms by which HIV increases susceptibility to pulmonary infections.
Collapse
Affiliation(s)
- Samantha M. Yeligar
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory + Children's Healthcare of Atlanta Center for Developmental Lung Biology, Emory University, Atlanta, Georgia
| | - Janine M. Ward
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory + Children's Healthcare of Atlanta Center for Developmental Lung Biology, Emory University, Atlanta, Georgia
| | - Frank L. Harris
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory + Children's Healthcare of Atlanta Center for Developmental Lung Biology, Emory University, Atlanta, Georgia
| | - Lou Ann S. Brown
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory + Children's Healthcare of Atlanta Center for Developmental Lung Biology, Emory University, Atlanta, Georgia
| | - David M. Guidot
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
- Atlanta VA Medical Center, Decatur, Georgia
| | - Sushma K. Cribbs
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, Georgia
- Atlanta VA Medical Center, Decatur, Georgia
| |
Collapse
|
22
|
Nebivolol prevents ethanol-induced reactive oxygen species generation and lipoperoxidation in the rat kidney by regulating NADPH oxidase activation and expression. Eur J Pharmacol 2017; 799:33-40. [DOI: 10.1016/j.ejphar.2017.01.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 12/20/2022]
|
23
|
Margoles LM, Mittal R, Klingensmith NJ, Lyons JD, Liang Z, Serbanescu MA, Wagener ME, Coopersmith CM, Ford ML. Chronic Alcohol Ingestion Delays T Cell Activation and Effector Function in Sepsis. PLoS One 2016; 11:e0165886. [PMID: 27861506 PMCID: PMC5115670 DOI: 10.1371/journal.pone.0165886] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 10/19/2016] [Indexed: 12/21/2022] Open
Abstract
Sepsis is the leading cause of death in intensive care units in the US, and it is known that chronic alcohol use is associated with higher incidence of sepsis, longer ICU stays, and higher mortality from sepsis. Both sepsis and chronic alcohol use are associated with immune deficits such as decreased lymphocyte numbers, impaired innate immunity, delayed-type hypersensitivity reactions, and susceptibility to infections; however, understanding of specific pathways of interaction or synergy between these two states of immune dysregulation is lacking. This study therefore sought to elucidate mechanisms underlying the immune dysregulation observed during sepsis in the setting of chronic alcohol exposure. Using a murine model of chronic ethanol ingestion followed by sepsis induction via cecal ligation and puncture, we determined that while CD4+ and CD8+ T cells isolated from alcohol fed mice eventually expressed the same cellular activation markers (CD44, CD69, and CD43) and effector molecules (IFN-γ, TNF) as their water fed counterparts, there was an overall delay in the acquisition of these phenotypes. This early lag in T cell activation was associated with significantly reduced IL-2 production at a later timepoint in both the CD4+ and CD8+ T cell compartments in alcohol sepsis, as well as with a reduced accumulation of CD8dim activated effectors. Taken together, these data suggest that delayed T cell activation may result in qualitative differences in the immune response to sepsis in the setting of chronic alcohol ingestion.
Collapse
Affiliation(s)
- Lindsay M. Margoles
- Division of Infectious Diseases, Emory University, Atlanta, GA, United States of America
| | - Rohit Mittal
- Department of Surgery, Emory University, Atlanta, GA, United States of America
| | | | - John D. Lyons
- Department of Surgery, Emory University, Atlanta, GA, United States of America
| | - Zhe Liang
- Department of Surgery, Emory University, Atlanta, GA, United States of America
| | - Mara A. Serbanescu
- Department of Surgery, Emory University, Atlanta, GA, United States of America
| | - Maylene E. Wagener
- Department of Surgery, Emory University, Atlanta, GA, United States of America
- Emory Transplant Center, Emory University, Atlanta, GA, United States of America
| | - Craig M. Coopersmith
- Department of Surgery, Emory University, Atlanta, GA, United States of America
- Emory Critical Care Center, Emory University, Atlanta, GA, United States of America
| | - Mandy L. Ford
- Department of Surgery, Emory University, Atlanta, GA, United States of America
- Emory Transplant Center, Emory University, Atlanta, GA, United States of America
| |
Collapse
|
24
|
Yeligar SM, Chen MM, Kovacs EJ, Sisson JH, Burnham EL, Brown LAS. Alcohol and lung injury and immunity. Alcohol 2016; 55:51-59. [PMID: 27788778 DOI: 10.1016/j.alcohol.2016.08.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 07/07/2016] [Accepted: 08/24/2016] [Indexed: 02/06/2023]
Abstract
Annually, excessive alcohol use accounts for more than $220 billion in economic costs and 80,000 deaths, making excessive alcohol use the third leading lifestyle-related cause of death in the US. Patients with an alcohol-use disorder (AUD) also have an increased susceptibility to respiratory pathogens and lung injury, including a 2-4-fold increased risk of acute respiratory distress syndrome (ARDS). This review investigates some of the potential mechanisms by which alcohol causes lung injury and impairs lung immunity. In intoxicated individuals with burn injuries, activation of the gut-liver axis drives pulmonary inflammation, thereby negatively impacting morbidity and mortality. In the lung, the upper airway is the first checkpoint to fail in microbe clearance during alcohol-induced lung immune dysfunction. Brief and prolonged alcohol exposure drive different post-translational modifications of novel proteins that control cilia function. Proteomic approaches are needed to identify novel alcohol targets and post-translational modifications in airway cilia that are involved in alcohol-dependent signal transduction pathways. When the upper airway fails to clear inhaled pathogens, they enter the alveolar space where they are primarily cleared by alveolar macrophages (AM). With chronic alcohol ingestion, oxidative stress pathways in the AMs are stimulated, thereby impairing AM immune capacity and pathogen clearance. The epidemiology of pneumococcal pneumonia and AUDs is well established, as both increased predisposition and illness severity have been reported. AUD subjects have increased susceptibility to pneumococcal pneumonia infections, which may be due to the pro-inflammatory response of AMs, leading to increased oxidative stress.
Collapse
Affiliation(s)
- Samantha M Yeligar
- Department of Medicine, Emory University and Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
| | - Michael M Chen
- Burn and Shock Trauma Research Institute, Alcohol Research Program, Integrative Cell Biology Program, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, USA
| | - Elizabeth J Kovacs
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Joseph H Sisson
- Pulmonary, Critical Care, Sleep and Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ellen L Burnham
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Lou Ann S Brown
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
25
|
Yeligar SM, Mehta AJ, Harris FL, Brown LAS, Hart CM. Peroxisome Proliferator-Activated Receptor γ Regulates Chronic Alcohol-Induced Alveolar Macrophage Dysfunction. Am J Respir Cell Mol Biol 2016; 55:35-46. [PMID: 26677910 PMCID: PMC4942203 DOI: 10.1165/rcmb.2015-0077oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 12/15/2015] [Indexed: 12/13/2022] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR) γ is critical for alveolar macrophage (AM) function. Chronic alcohol abuse causes AM phagocytic dysfunction and susceptibility to respiratory infections by stimulating nicotinamide adenine dinucleotide oxidases (Nox), transforming growth factor-β1, and oxidative stress in the AM. Because PPARγ inhibits Nox expression, we hypothesized that alcohol reduces PPARγ, stimulating AM dysfunction. AMs were examined from: (1) patients with alcoholism or control patients; (2) a mouse model of chronic ethanol consumption; (3) PPARγ knockout mice; or (4) MH-S cells exposed to ethanol in vitro. Alcohol reduced AM PPARγ levels and increased Nox1, -2, and -4, transforming growth factor-β1, oxidative stress, and phagocytic dysfunction. Genetic loss of PPARγ recapitulated, whereas stimulating PPARγ activity attenuated alcohol-mediated alterations in gene expression and phagocytic function, supporting the importance of PPARγ in alcohol-induced AM derangements. Similarly, PPARγ activation in vivo reduced alcohol-mediated impairments in lung bacterial clearance. Alcohol increased levels of microRNA-130a/-301a, which bind to the PPARγ 3' untranslated region to reduce PPARγ expression. MicroRNA-130a/-301a inhibition attenuated alcohol-mediated PPARγ reductions and derangements in AM gene expression and function. Alcohol-induced Toll-like receptor 4 endocytosis was reversed by PPARγ activation. These findings demonstrate that targeting PPARγ provides a novel therapeutic approach for mitigating alcohol-induced AM derangements and susceptibility to lung infection.
Collapse
Affiliation(s)
- Samantha M. Yeligar
- Emory University, Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory and Children’s Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, Georgia
- Emory University, Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Decatur, Georgia
| | - Ashish J. Mehta
- Emory University, Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Decatur, Georgia
| | - Frank L. Harris
- Emory University, Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory and Children’s Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, Georgia
| | - Lou Ann S. Brown
- Emory University, Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory and Children’s Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, Georgia
| | - C. Michael Hart
- Emory University, Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Atlanta Veterans Affairs and Emory University Medical Centers, Decatur, Georgia
| |
Collapse
|
26
|
Yang XH, Li P, Yin YL, Tu JH, Dai W, Liu LY, Wang SX. Rosiglitazone via PPARγ-dependent suppression of oxidative stress attenuates endothelial dysfunction in rats fed homocysteine thiolactone. J Cell Mol Med 2015; 19:826-35. [PMID: 25656735 PMCID: PMC4395197 DOI: 10.1111/jcmm.12510] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 11/17/2014] [Indexed: 02/03/2023] Open
Abstract
To explore whether rosiglitazone (RSG), a selective peroxisome proliferator-activated receptor γ (PPARγ) agonist, exerts beneficial effects on endothelial dysfunction induced by homocysteine thiolactone (HTL) and to investigate the potential mechanisms. Incubation of cultured human umbilical vein endothelial cells with HTL (1 mM) for 24 hrs significantly reduced cell viabilities assayed by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide, as well as enhanced productions of reactive oxygen species, activation of nuclear factor kappa B, and increased intercellular cell adhesion molecule-1 secretion. Pre-treatment of cells with RSG (0.001–0.1 mM), pyrollidine dithiocarbamate (PDTC, 0.1 mM) or apocynin (0.1 mM) for 1 hr reversed these effects induced by HTL. Furthermore, co-incubation with GW9662 (0.01 mM) abolished the protective effects of RSG on HTL-treated cells. In ex vivo experiments, exposure of isolated aortic rings from. rats to HTL (1 mM) for 1 hr dramatically impaired acetylcholine-induced endothelium-dependent relaxation, reduced release of nitric oxide and activity of superoxide dismutase, and increased malondialdehyde content in aortic tissues. Preincubation of aortic rings with RSG (0.1, 0.3, 1 mM), PDTC or apocynin normalized the disorders induced by HTL. In vivo analysis indicated that administration of RSG (20 mg/kg/d) remarkably suppressed oxidative stress and prevented endothelial dysfunction in rats fed HTL (50 mg/kg/d) for 8 weeks. RSG improves endothelial functions in rats fed HTL, which is related to PPARγ-dependent suppression of oxidative stress.
Collapse
Affiliation(s)
- Xu-Hong Yang
- Department of Pharmacology, Pharmaceutical College, Central South University, Changsha, China
| | | | | | | | | | | | | |
Collapse
|
27
|
Kurtz M, Capobianco E, Careaga V, Martinez N, Mazzucco MB, Maier M, Jawerbaum A. Peroxisome proliferator-activated receptor ligands regulate lipid content, metabolism, and composition in fetal lungs of diabetic rats. J Endocrinol 2014; 220:345-59. [PMID: 24389592 DOI: 10.1530/joe-13-0362] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Maternal diabetes impairs fetal lung development. Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors relevant in lipid homeostasis and lung development. This study aims to evaluate the effect of in vivo activation of PPARs on lipid homeostasis in fetal lungs of diabetic rats. To this end, we studied lipid concentrations, expression of lipid metabolizing enzymes and fatty acid composition in fetal lungs of control and diabetic rats i) after injections of the fetuses with Leukotriene B4 (LTB4, PPARα ligand) or 15deoxyΔ(12,14)prostaglandin J2 (15dPGJ2, PPARγ ligand) and ii) fed during pregnancy with 6% olive oil- or 6% safflower oil-supplemented diets, enriched with PPAR ligands were studied. Maternal diabetes increased triglyceride concentrations and decreased expression of lipid-oxidizing enzymes in fetal lungs of diabetic rats, an expression further decreased by LTB4 and partially restored by 15dPGJ2 in lungs of male fetuses in the diabetic group. In lungs of female fetuses in the diabetic group, maternal diets enriched with olive oil increased triglyceride concentrations and fatty acid synthase expression, while those enriched with safflower oil increased triglyceride concentrations and fatty acid transporter expression. Both olive oil- and safflower oil-supplemented diets decreased cholesterol and cholesteryl ester concentrations and increased the expression of the reverse cholesterol transporter ATP-binding cassette A1 in fetal lungs of female fetuses of diabetic rats. In fetal lungs of control and diabetic rats, the proportion of polyunsaturated fatty acids increased with the maternal diets enriched with olive and safflower oils. Our results revealed important changes in lipid metabolism in fetal lungs of diabetic rats, and in the ability of PPAR ligands to modulate the composition of lipid species relevant in the lung during the perinatal period.
Collapse
Affiliation(s)
- M Kurtz
- Laboratory of Reproduction and Metabolism, CEFyBO-CONICET, School of Medicine and UMYMFOR (CONICET-UBA), Department of Organic Chemistry, School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
28
|
Alcohol induced mitochondrial oxidative stress and alveolar macrophage dysfunction. BIOMED RESEARCH INTERNATIONAL 2014; 2014:371593. [PMID: 24701574 PMCID: PMC3950485 DOI: 10.1155/2014/371593] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 12/27/2013] [Accepted: 12/28/2013] [Indexed: 01/08/2023]
Abstract
An alcohol use disorder increases the risk of invasive and antimicrobial resistant community-acquired pneumonia and tuberculosis. Since the alveolar macrophage (AM) orchestrates the immune response in the alveolar space, understanding the underlying mechanisms by which alcohol suppresses AM phagocytosis is critical to improving clinical outcomes. In the alveolar space, chronic alcohol ingestion causes severe oxidative stress and depletes antioxidants which are critical for AM function. The mitochondrion is important in maintaining cellular redox balance and providing the ATP critical for phagocytosis. The focus of this study was to understand how alcohol triggers mitochondrial reactive oxygen species (ROS), stimulates cellular oxidative stress, and induces AM dysfunction. The current study also investigated the capacity of the mitochondrial targeted antioxidant, mitoTEMPOL (mitoT), in modulating mitochondrial oxidative stress, and AM dysfunction. Using in vitro ethanol exposure and AMs from ethanol-fed mice, ethanol promoted mitochondrial dysfunction including increased mitochondrial ROS, decreased mitochondrial membrane potential, and decreased ATP. Treatment with mitoT reversed these effects. Ethanol-induced decreases in phagocytosis and cell viability were also attenuated with mitoT. Therefore, antioxidants targeted to the mitochondria have the potential to ameliorate ethanol-induced mitochondrial oxidative stress and subsequent decreases in AM phagocytosis and cell viability.
Collapse
|
29
|
Yeligar SM, Harris FL, Hart CM, Brown LAS. Glutathione attenuates ethanol-induced alveolar macrophage oxidative stress and dysfunction by downregulating NADPH oxidases. Am J Physiol Lung Cell Mol Physiol 2014; 306:L429-41. [PMID: 24441868 DOI: 10.1152/ajplung.00159.2013] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Chronic alcohol abuse increases lung oxidative stress and susceptibility to respiratory infections by impairing alveolar macrophage (AM) function. NADPH oxidases (Nox) are major sources of reactive oxygen species in AMs. We hypothesized that treatment with the critical antioxidant glutathione (GSH) attenuates chronic alcohol-induced oxidative stress by downregulating Noxes and restores AM phagocytic function. Bronchoalveolar lavage (BAL) fluid and AMs were isolated from male C57BL/6J mice (8-10 wk) treated ± ethanol in drinking water (20% wt/vol, 12 wk) ± orally gavaged GSH in methylcellulose vehicle (300 mg x kg(-1) x day(-1), during week 12). MH-S cells, a mouse AM cell line, were treated ± ethanol (0.08%, 3 days) ± GSH (500 μM, 3 days or last 1 day of ethanol). BAL and AMs were also isolated from ethanol-fed and control mice ± inoculated airway Klebsiella pneumoniae (200 colony-forming units, 28 h) ± orally gavaged GSH (300 mg/kg, 24 h). GSH levels (HPLC), Nox mRNA (quantitative RT-PCR) and protein levels (Western blot and immunostaining), oxidative stress (2',7'-dichlorofluorescein-diacetate and Amplex Red), and phagocytosis (Staphylococcus aureus internalization) were measured. Chronic alcohol decreased GSH levels, increased Nox expression and activity, enhanced oxidative stress, impaired phagocytic function in AMs in vivo and in vitro, and exacerbated K. pneumonia-induced oxidative stress. Although how oral GSH restored GSH pools in ethanol-fed mice is unknown, oral GSH treatments abrogated the detrimental effects of chronic alcohol exposure and improved AM function. These studies provide GSH as a novel therapeutic approach for attenuating alcohol-induced derangements in AM Nox expression, oxidative stress, dysfunction, and risk for pneumonia.
Collapse
Affiliation(s)
- Samantha M Yeligar
- Dept. of Pediatrics, Division of Neonatal-Perinatal Medicine, Emory Univ., 2015 Uppergate Dr., Atlanta, GA 30322.
| | | | | | | |
Collapse
|
30
|
Liang Y, Harris FL, Jones DP, Brown LAS. Alcohol induces mitochondrial redox imbalance in alveolar macrophages. Free Radic Biol Med 2013; 65:1427-1434. [PMID: 24140864 PMCID: PMC3870467 DOI: 10.1016/j.freeradbiomed.2013.10.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 10/07/2013] [Accepted: 10/08/2013] [Indexed: 12/13/2022]
Abstract
Alcohol abuse suppresses the immune responses of alveolar macrophages (AMs) and increases the risk of a respiratory infection via chronic oxidative stress and depletion of critical antioxidants within alveolar cells and the alveolar lining fluid. Although alcohol-induced mitochondrial oxidative stress has been demonstrated, the oxidation of the mitochondrial thioredoxin redox circuit in response to alcohol has not been examined. In vitro ethanol exposure of a mouse AM cell line and AMs from ethanol-fed mice demonstrated NADPH depletion concomitant with oxidation of mitochondrial glutathione and oxidation of the thioredoxin redox circuit system including thioredoxin 2 (Trx2) and thioredoxin 2 reductase (Trx2R). Mitochondrial peroxiredoxins (Prdx's), which are critical for the reduction of the thioredoxin circuit, were irreversibly hyperoxidized to an inactive form. Ethanol also decreased the mRNAs for Trx2, Trx2R, Prdx3, and Prdx5 plus the mitochondrial thiol-disulfide proteins glutaredoxin 2, glutathione reductase, and glutathione peroxidase 2. Thus, the mitochondrial thioredoxin circuit was highly oxidized by ethanol, thereby compromising the mitochondrial antioxidant capacity and ability to detoxify mitochondrial reactive oxygen species. Oxidation of the mitochondrial thioredoxin redox circuit would further compromise the transient oxidation of thiol groups within specific proteins, the basis of redox signaling, and the processes by which cells respond to oxidants. Impaired mitochondria can then jeopardize cellular function of AMs, such as phagocytosis, which may explain the increased risk of respiratory infection in subjects with an alcohol use disorder.
Collapse
Affiliation(s)
- Yan Liang
- Department of Pediatrics, Allergy & Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA; Center for Developmental Lung Biology, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Frank L Harris
- Department of Pediatrics, Allergy & Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA; Center for Developmental Lung Biology, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy & Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Lou Ann S Brown
- Department of Pediatrics, Allergy & Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA; Center for Developmental Lung Biology, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.
| |
Collapse
|
31
|
Dekeyser GJ, Clary CR, Otis JS. Chronic alcohol ingestion delays skeletal muscle regeneration following injury. Regen Med Res 2013; 1:2. [PMID: 25984321 PMCID: PMC4376340 DOI: 10.1186/2050-490x-1-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 12/21/2012] [Indexed: 12/17/2022] Open
Abstract
Background Chronic alcohol ingestion may cause severe biochemical and pathophysiological derangements to skeletal muscle. Unfortunately, these alcohol-induced events may also prime skeletal muscle for worsened, delayed, or possibly incomplete repair following acute injury. As alcoholics may be at increased risk for skeletal muscle injury, our goals were to identify the effects of chronic alcohol ingestion on components of skeletal muscle regeneration. To accomplish this, age- and gender-matched C57Bl/6 mice were provided normal drinking water or water that contained 20% alcohol (v/v) for 18–20 wk. Subgroups of mice were injected with a 1.2% barium chloride (BaCl2) solution into the tibialis anterior (TA) muscle to initiate degeneration and regeneration processes. Body weights and voluntary wheel running distances were recorded during the course of recovery. Muscles were harvested at 2, 7 or 14 days post-injection and assessed for markers of inflammation and oxidant stress, fiber cross-sectional areas, levels of growth and fibrotic factors, and fibrosis. Results Body weights of injured, alcohol-fed mice were reduced during the first week of recovery. These mice also ran significantly shorter distances over the two weeks following injury compared to uninjured, alcoholics. Injured TA muscles from alcohol-fed mice had increased TNFα and IL6 gene levels compared to controls 2 days after injury. Total protein oxidant stress and alterations to glutathione homeostasis were also evident at 7 and 14 days after injury. Ciliary neurotrophic factor (CNTF) induction was delayed in injured muscles from alcohol-fed mice which may explain, in part, why fiber cross-sectional area failed to normalize 14 days following injury. Gene levels of TGFβ1 were induced early following injury before normalizing in muscle from alcohol-fed mice compared to controls. However, TGFβ1 protein content was consistently elevated in injured muscle regardless of diet. Fibrosis was increased in injured, muscle from alcohol-fed mice at 7 and 14 days of recovery compared to injured controls. Conclusions Chronic alcohol ingestion appears to delay the normal regenerative response following significant skeletal muscle injury. This is evidenced by reduced cross-sectional areas of regenerated fibers, increased fibrosis, and altered temporal expression of well-described growth and fibrotic factors.
Collapse
Affiliation(s)
- Graham J Dekeyser
- Department of Kinesiology and Health, Georgia State University, Atlanta, GA 30322 USA
| | | | - Jeffrey S Otis
- Department of Kinesiology and Health, Georgia State University, Atlanta, GA 30322 USA
| |
Collapse
|
32
|
Chronic alcohol ingestion increases mortality and organ injury in a murine model of septic peritonitis. PLoS One 2013; 8:e62792. [PMID: 23717394 PMCID: PMC3661585 DOI: 10.1371/journal.pone.0062792] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 03/25/2013] [Indexed: 12/13/2022] Open
Abstract
Background Patients admitted to the intensive care unit with alcohol use disorders have increased morbidity and mortality. The purpose of this study was to determine how chronic alcohol ingestion alters the host response to sepsis in mice. Methods Mice were randomized to receive either alcohol or water for 12 weeks and then subjected to cecal ligation and puncture. Mice were sacrificed 24 hours post-operatively or followed seven days for survival. Results Septic alcohol-fed mice had a significantly higher mortality than septic water-fed mice (74% vs. 41%, p = 0.01). This was associated with worsened gut integrity in alcohol-fed mice with elevated intestinal epithelial apoptosis, decreased crypt proliferation and shortened villus length. Further, alcohol-fed mice had higher intestinal permeability with decreased ZO-1 and occludin protein expression in the intestinal tight junction. The frequency of splenic and bone marrow CD4+ T cells was similar between groups; however, splenic CD4+ T cells in septic alcohol-fed mice had a marked increase in both TNF and IFN-γ production following ex vivo stimulation. Neither the frequency nor function of CD8+ T cells differed between alcohol-fed and water-fed septic mice. NK cells were decreased in both the spleen and bone marrow of alcohol-fed septic mice. Pulmonary myeloperoxidase levels and BAL levels of G-CSF and TFG-β were higher in alcohol-fed mice. Pancreatic metabolomics demonstrated increased acetate, adenosine, xanthine, acetoacetate, 3-hydroxybutyrate and betaine in alcohol-fed mice and decreased cytidine, uracil, fumarate, creatine phosphate, creatine, and choline. Serum and peritoneal cytokines were generally similar between alcohol-fed and water-fed mice, and there were no differences in bacteremia, lung wet to dry weight, or pulmonary, liver or splenic histology. Conclusions When subjected to the same septic insult, mice with chronic alcohol ingestion have increased mortality. Alterations in intestinal integrity, the host immune response, and pancreatic metabolomics may help explain this differential response.
Collapse
|
33
|
Liang Y, Yeligar SM, Brown LAS. Chronic-alcohol-abuse-induced oxidative stress in the development of acute respiratory distress syndrome. ScientificWorldJournal 2012; 2012:740308. [PMID: 23346021 PMCID: PMC3543796 DOI: 10.1100/2012/740308] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 11/21/2012] [Indexed: 12/13/2022] Open
Abstract
Chronic alcohol ingestion increases the risk of developing acute respiratory distress syndrome (ARDS), a severe form of acute lung injury, characterized by alveolar epithelial and endothelial barrier disruption and intense inflammation. Alcohol abuse is also associated with a higher incidence of sepsis or pneumonia resulting in a higher rate of admittance to intensive care, longer inpatient stays, higher healthcare costs, and a 2-4 times greater mortality rate. Chronic alcohol ingestion induced severe oxidative stress associated with increased ROS generation, depletion of the critical antioxidant glutathione (GSH), and oxidation of the thiol/disulfide redox potential in the alveolar epithelial lining fluid and exhaled breath condensate. Across intracellular and extracellular GSH pools in alveolar type II cells and alveolar macrophages, chronic alcohol ingestion consistently induced a 40-60 mV oxidation of GSH/GSSG suggesting that the redox potentials of different alveolar GSH pools are in equilibrium. Alcohol-induced GSH depletion or oxidation was associated with impaired functions of alveolar type II cells and alveolar macrophages but could be reversed by restoring GSH pools in the alveolar lining fluid. The aims of this paper are to address the mechanisms for alcohol-induced GSH depletion and oxidation and the subsequent effects in alveolar barrier integrity, modulation of the immune response, and apoptosis.
Collapse
Affiliation(s)
- Yan Liang
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University and Children's Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, GA 30322, USA
| | - Samantha M. Yeligar
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University and Children's Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, GA 30322, USA
- Department of Medicine, Atlanta Veterans' Affairs and Emory University Medical Centers, Decatur, GA 30033, USA
| | - Lou Ann S. Brown
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Emory University and Children's Healthcare of Atlanta Center for Developmental Lung Biology, Atlanta, GA 30322, USA
| |
Collapse
|