1
|
Xie L, Wang L, Liao Y, Yao M, Mai T, Fan R, Han Y, Zhou G. Therapeutic potential of short-chain fatty acids for acute lung injury: a systematic review and meta-analysis of preclinical animal studies. Front Nutr 2025; 11:1528200. [PMID: 39845918 PMCID: PMC11752998 DOI: 10.3389/fnut.2024.1528200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Background Short-chain fatty acids (SCFAs), derived from the fermentation of dietary fiber by intestinal commensal bacteria, have demonstrated protective effects against acute lung injury (ALI) in animal models. However, the findings have shown variability across different studies. It is necessary to conduct a comprehensive evaluation of the efficacy of these treatments and their consistency. Objective This systematic review and meta-analysis aimed to explore the effects of SCFAs on ALI based on preclinical research evidence, in order to provide new treatment strategies for ALI. Methods We included studies that tested the effects of SCFAs on ALI in animal models. This study was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. A comprehensive search for relevant studies was conducted in the PubMed, Embase, Web of Science, Cochrane Library, and China National Knowledge Infrastructure (CNKI) databases up to February 2024. The data were extracted in accordance with the established selection criteria, and the risk of bias was evaluated for each study. Results A total of 16 articles were finally included in the meta-analysis. The results indicated that the SCFAs significantly reduced lung wet-to-dry weight (SMD = -2.75, 95% CI = -3.46 to -2.03, p < 0.00001), lung injury scores (SMD = -5.07, 95% CI = -6.25 to -3.89, p < 0.00001), myeloperoxidase (SMD = -3.37, 95% CI = -4.05 to -2.70, p < 0.00001), tumor necrosis factor-alpha (SMD = -3.31, 95% CI = -4.45 to -2.16, p < 0.00001) and malondialdehyde (SMD = -3.91, 95% CI = -5.37 to -2.44, p < 0.00001) levels in animal models of ALI. The results of the subgroup analysis indicated that the efficacy of SCFAs varies significantly with dosage and duration of treatment. Conclusion SCFAs can reduce inflammation and oxidative stress in animal models of ALI. The clinical efficacy of SCFAs for ALI deserves further in-depth research. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=584008, CRD42024584008.
Collapse
Affiliation(s)
- Liying Xie
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Linyan Wang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongxin Liao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Miaoen Yao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tong Mai
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rongrong Fan
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yun Han
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gengbiao Zhou
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
2
|
Zhao J, Jiang L, He W, Han D, Yang X, Wu L, Zhong H. Clostridium butyricum, a future star in sepsis treatment. Front Cell Infect Microbiol 2024; 14:1484371. [PMID: 39711782 PMCID: PMC11659258 DOI: 10.3389/fcimb.2024.1484371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/19/2024] [Indexed: 12/24/2024] Open
Abstract
Sepsis is a systemic inflammatory response syndrome of multiorgan failure caused by dysregulation of the host response to infection and is a major cause of death in critically ill patients. In recent years, with the continuous development of sequencing technology, the intestinal microecology of this disease has been increasingly studied. The gut microbiota plays a host-protective role mainly through the maintenance of normal immune function and the intestinal barrier. Recent evidence suggests that intestinal flora dysbiosis plays a crucial role in sepsis. Clostridium butyricum (C. butyricum), which has been used as a probiotic in poultry feed since its discovery, has been found to play a potential protective role in intestinal infections, inflammatory bowel disease (IBD), colorectal cancer, and other diseases in recent studies. In this review, we continue to focus on the important role and mechanism of C. butyricum as a probiotic in human diseases, especially intestinal diseases. Additionally, we evaluate the research progress of C. butyricum in treatment of sepsis to identify more therapeutic targets for the clinical treatment of sepsis.
Collapse
Affiliation(s)
- Jinglin Zhao
- Medical Laboratory, Kunming Children’s Hospital, Children’s Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Li Jiang
- Medical Laboratory, Kunming Children’s Hospital, Children’s Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Weizhi He
- Medical Laboratory, Kunming Children’s Hospital, Children’s Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Dingrui Han
- Medical Laboratory, Kunming Children’s Hospital, Children’s Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Xuan Yang
- Medical Laboratory, Kunming Children’s Hospital, Children’s Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Liuli Wu
- The Affiliated Hospital of Kunming University of Science and Technology, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Haiyan Zhong
- Medical Laboratory, Kunming Children’s Hospital, Children’s Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
3
|
Patil RS, Maloney ME, Lucas R, Fulton DJR, Patel V, Bagi Z, Kovacs-Kasa A, Kovacs L, Su Y, Verin AD. Zinc-Dependent Histone Deacetylases in Lung Endothelial Pathobiology. Biomolecules 2024; 14:140. [PMID: 38397377 PMCID: PMC10886568 DOI: 10.3390/biom14020140] [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: 12/31/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/25/2024] Open
Abstract
A monolayer of endothelial cells (ECs) lines the lumen of blood vessels and, as such, provides a semi-selective barrier between the blood and the interstitial space. Compromise of the lung EC barrier due to inflammatory or toxic events may result in pulmonary edema, which is a cardinal feature of acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS). The EC functions are controlled, at least in part, via epigenetic mechanisms mediated by histone deacetylases (HDACs). Zinc-dependent HDACs represent the largest group of HDACs and are activated by Zn2+. Members of this HDAC group are involved in epigenetic regulation primarily by modifying the structure of chromatin upon removal of acetyl groups from histones. In addition, they can deacetylate many non-histone histone proteins, including those located in extranuclear compartments. Recently, the therapeutic potential of inhibiting zinc-dependent HDACs for EC barrier preservation has gained momentum. However, the role of specific HDAC subtypes in EC barrier regulation remains largely unknown. This review aims to provide an update on the role of zinc-dependent HDACs in endothelial dysfunction and its related diseases. We will broadly focus on biological contributions, signaling pathways and transcriptional roles of HDACs in endothelial pathobiology associated mainly with lung diseases, and we will discuss the potential of their inhibitors for lung injury prevention.
Collapse
Affiliation(s)
- Rahul S. Patil
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - McKenzie E. Maloney
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Rudolf Lucas
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - David J. R. Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Vijay Patel
- Department of Cardiothoracic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Anita Kovacs-Kasa
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Laszlo Kovacs
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Alexander D. Verin
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
4
|
Niu K, Yang L, Song W, Liu Z, Yuan J, Zhang H, Zhang W, Wang J, Tao K. A COMPARATIVE ANALYSIS TO DETERMINE THE OPTIMUM HISTONE DEACETYLASE INHIBITORS AND ADMINISTRATION ROUTE FOR IMPROVING SURVIVAL AND ORGAN INJURY IN RATS AFTER HEMORRHAGIC SHOCK. Shock 2023; 60:75-83. [PMID: 37141162 PMCID: PMC10417212 DOI: 10.1097/shk.0000000000002136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/20/2023] [Indexed: 05/05/2023]
Abstract
ABSTRACT Objective: Histone deacetylase inhibitors (HDACIs) have been reported to improve survival in rats with hemorrhagic shock (HS). However, no consensus exists on the most effective HDACIs and their administration routes. We herein aimed to determine the optimal HDACIs and administration route in rats with HS. Methods: Survival analysis: In experiment I, male Sprague-Dawley rats were subjected to HS (mean arterial pressure [MAP] was maintained at 30-40 mm Hg for 20 min), and intravenously injected with the following agents (n = 8 per group): (1) no treatment, (2) vehicle (VEH), (3) entinostat (MS-275), (4) [ N -((6-(Hydroxyamino)-6-oxohexyl)oxy)-3,5-dimethylbenzamide] (LMK-235), (5) tubastatin A, (6) trichostatin A (TSA), and (7) sirtinol. In experiment II, rats were intraperitoneally injected with TSA. Mechanism research: In experiments I and II, rats were observed for 3 h, after which blood samples and liver, heart, and lung tissues were harvested. Results: In experiment I, 75% rats in the VEH group but only 25% rats in the LMK-235 and sirtinol groups died within ≤5 h of treatment, whereas the survival of rats in the MS-275, tubastatin A, and TSA groups was significantly prolonged. MS-275, LMK-235, tubastatin A, and TSA significantly reduced histopathological scores, apoptosis cell numbers, and inflammatory cytokine levels. In experiment II, the survival was longer after i.v. TSA treatment than after i.p. TSA treatment, and the IL-6 levels in the heart were significantly lower in rat who received i.p. TSA treatment than in those who received i.v. TSA treatment. Conclusions: The i.v. effect was superior to the i.p. effect, while nonselective and isoform-specific classes I and IIb HDACIs had similar effects.
Collapse
|
5
|
Liu Y, Yang H, Zhu F, Ouyang Y, Pan P. Inhibition of STAT3 phosphorylation by colchicine regulates NLRP3 activation to alleviate sepsis-induced acute lung injury. Inflammopharmacology 2023:10.1007/s10787-023-01199-9. [PMID: 37115345 DOI: 10.1007/s10787-023-01199-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/17/2023] [Indexed: 04/29/2023]
Abstract
The pharmacotherapeutic mechanism of colchicine, a tricyclic, lipid-soluble alkaloid extracted from the plant of the Lily family Colchicum autumnale, has not been fully understood in diverse disorders, including sepsis-induced acute lung injury (ALI). The study aimed at exploring the impact of colchicine on sepsis-induced ALI and the relevant mechanisms. Colchicine significantly attenuated ALI in mice caused by sepsis by alleviating respiratory dysfunction and pulmonary edema in mice, inhibiting NLRP3 inflammasome formation, and reducing oxidative stress, pyroptosis, and apoptosis of murine alveolar macrophage (J774A.1) cells. The targets of colchicine were predicted in the superPRED database and intersected with the differentially expressed genes in the GSE5883 and GSE129775 datasets. The major targets were subjected to protein-protein interaction network generation and Kyoto Encyclopedia of Genes and Genomes enrichment analysis. It was thus found that colchicine inhibited STAT3 phosphorylation but did not alter STAT3 total protein expression. Phosphorylated STAT3 recruited EP300 to form a complex to promote histone H3 acetylation and histone H4 acetylation of NLRP3 promoter, leading to pyroptosis of J774A.1 cells. In conclusion, inhibition of STAT3 phosphorylation by colchicine represses NLRP3 promoter acetylation via the STAT3/EP300 complex, thereby alleviating ALI caused by sepsis.
Collapse
Affiliation(s)
- Yuanshui Liu
- Department of Respiratory Medicine, Key Cite of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, People's Republic of China
- Department of Emergency Medicine, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, Hainan, People's Republic of China
| | - Hang Yang
- Department of Respiratory Medicine, Key Cite of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, People's Republic of China
| | - Fei Zhu
- Department of Respiratory Medicine, Key Cite of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, People's Republic of China
| | - Yanhong Ouyang
- Department of Emergency Medicine, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, Hainan, People's Republic of China
| | - Pinhua Pan
- Department of Respiratory Medicine, Key Cite of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Kaifu District, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
6
|
Liu MY, Ju YN, Jia BW, Sun XK, Qiu L, Liu HY, Xu GX, Tai QH, Tan J, Gao W. Inhibition of DNA methylation attenuates lung ischemia-reperfusion injury after lung transplantation. J Int Med Res 2023; 51:3000605231153587. [PMID: 36756846 PMCID: PMC9912569 DOI: 10.1177/03000605231153587] [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] [Indexed: 02/10/2023] Open
Abstract
OBJECTIVE DNA methylation plays an important role in inflammation and oxidative stress. This study aimed to investigate the effect of inhibiting DNA methylation on lung ischemia-reperfusion injury (LIRI). METHODS We adopted a completely random design for our study. Thirty-two rats were randomized into the sham, LIRI, azathioprine (AZA), and pluripotin (SC1) groups. The rats in the LIRI, AZA, and SC1 groups received left lung transplantation and intravenous injection of saline, AZA, and SC1, respectively. After 24 hours of reperfusion, histological injury, the arterial oxygen partial pressure to fractional inspired oxygen ratio, the wet/dry weight ratio, protein and cytokine concentrations in lung tissue, and DNA methylation in lung tissue were evaluated. The pulmonary endothelium that underwent hypoxemia and reoxygenation was treated with AZA or SC1. Endothelial apoptosis, chemokines, reactive oxygen species, nuclear factor-κB, and apoptotic proteins in the endothelium were studied. RESULTS Inhibition of DNA methylation by AZA attenuated lung injury, inflammation, and the oxidative stress response, but SC1 aggravated LIRI injury. AZA significantly improved endothelial function, suppressed apoptosis and necrosis, reduced chemokines, and inhibited nuclear factor-κB. CONCLUSIONS Inhibition of DNA methylation ameliorates LIRI and apoptosis and improves pulmonary function via the regulation of inflammation and oxidative stress.
Collapse
Affiliation(s)
- Ming-yuan Liu
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Ying-nan Ju
- Department of Intensive Care Unit, The Third Affiliated Hospital
of Harbin Medical University, Harbin, Heilongjiang, China
| | - Bao-wei Jia
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Xi-kun Sun
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Lin Qiu
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Heng-yu Liu
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Guang-xiao Xu
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Qi-hang Tai
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Jing Tan
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China
| | - Wei Gao
- Department of Anesthesiology, The Second Affiliated Hospital of
Harbin Medical University, Harbin, Heilongjiang, China,Wei Gao, Department of Anesthesiology, The
Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin,
Heilongjiang 150081, China.
| |
Collapse
|
7
|
See KC. Personalizing Care for Critically Ill Adults Using Omics: A Concise Review of Potential Clinical Applications. Cells 2023; 12:541. [PMID: 36831207 PMCID: PMC9954497 DOI: 10.3390/cells12040541] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Current guidelines for critically ill patients use broad recommendations to promote uniform protocols for the management of conditions such as acute kidney injury, acute respiratory distress syndrome, and sepsis. Although these guidelines have enabled the substantial improvement of care, mortality for critical illness remains high. Further outcome improvement may require personalizing care for critically ill patients, which involves tailoring management strategies for different patients. However, the current understanding of disease heterogeneity is limited. For critically ill patients, genomics, transcriptomics, proteomics, and metabolomics have illuminated such heterogeneity and unveiled novel biomarkers, giving clinicians new means of diagnosis, prognosis, and monitoring. With further engineering and economic development, omics would then be more accessible and affordable for frontline clinicians. As the knowledge of pathophysiological pathways mature, targeted treatments can then be developed, validated, replicated, and translated into clinical practice.
Collapse
Affiliation(s)
- Kay Choong See
- Division of Respiratory and Critical Care Medicine, Department of Medicine, National University Hospital, Singapore 119228, Singapore
| |
Collapse
|
8
|
Schneider J, Sundaravinayagam D, Blume A, Marg A, Grunwald S, Metzler E, Escobar H, Müthel S, Wang H, Wollersheim T, Weber-Carstens S, Akalin A, Di Virgilio M, Tursun B, Spuler S. Disintegration of the NuRD Complex in Primary Human Muscle Stem Cells in Critical Illness Myopathy. Int J Mol Sci 2023; 24:2772. [PMID: 36769095 PMCID: PMC9916927 DOI: 10.3390/ijms24032772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Critical illness myopathy (CIM) is an acquired, devastating, multifactorial muscle-wasting disease with incomplete recovery. The impact on hospital costs and permanent loss of quality of life is enormous. Incomplete recovery might imply that the function of muscle stem cells (MuSC) is impaired. We tested whether epigenetic alterations could be in part responsible. We characterized human muscle stem cells (MuSC) isolated from early CIM and analyzed epigenetic alterations (CIM n = 15, controls n = 21) by RNA-Seq, immunofluorescence, analysis of DNA repair, and ATAC-Seq. CIM-MuSC were transplanted into immunodeficient NOG mice to assess their regenerative potential. CIM-MuSC exhibited significant growth deficits, reduced ability to differentiate into myotubes, and impaired DNA repair. The chromatin structure was damaged, as characterized by alterations in mRNA of histone 1, depletion or dislocation of core proteins of nucleosome remodeling and deacetylase complex, and loosening of multiple nucleosome-spanning sites. Functionally, CIM-MuSC had a defect in building new muscle fibers. Further, MuSC obtained from the electrically stimulated muscle of CIM patients was very similar to control MuSC, indicating the impact of muscle contraction in the onset of CIM. CIM not only affects working skeletal muscle but has a lasting and severe epigenetic impact on MuSC.
Collapse
Affiliation(s)
- Joanna Schneider
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
- Charité Universitätsmedizin Berlin, Department of Pediatric Neurology, 13353 Berlin, Germany
- Berlin Institute of Health–Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Devakumar Sundaravinayagam
- Laboratory of DNA Repair and Maintenance of Genome Stability, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13092 Berlin, Germany
| | - Alexander Blume
- Berlin Institute of Medical Systems Biology (BIMSB), Max Delbruck Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Andreas Marg
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Stefanie Grunwald
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Eric Metzler
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Helena Escobar
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Stefanie Müthel
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
- Berlin Institute of Medical Systems Biology (BIMSB), Max Delbruck Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Haicui Wang
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Tobias Wollersheim
- Berlin Institute of Health–Universitätsmedizin Berlin, 10117 Berlin, Germany
- Charité Universitätsmedizin Berlin, Department of Anesthesiology and Operative Intensive Care Medicine, 13353 Berlin, Germany
| | - Steffen Weber-Carstens
- Charité Universitätsmedizin Berlin, Department of Anesthesiology and Operative Intensive Care Medicine, 13353 Berlin, Germany
| | - Altuna Akalin
- Berlin Institute of Medical Systems Biology (BIMSB), Max Delbruck Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Michela Di Virgilio
- Laboratory of DNA Repair and Maintenance of Genome Stability, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13092 Berlin, Germany
| | - Baris Tursun
- Berlin Institute of Medical Systems Biology (BIMSB), Max Delbruck Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| |
Collapse
|
9
|
Truong N, Goodis CC, Cottingham AL, Shaw JR, Fletcher S, Pearson RM. Modified Suberoylanilide Hydroxamic Acid Reduced Drug-Associated Immune Cell Death and Organ Damage under Lipopolysaccharide Inflammatory Challenge. ACS Pharmacol Transl Sci 2022; 5:1128-1141. [PMID: 36407956 PMCID: PMC9667537 DOI: 10.1021/acsptsci.2c00119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Indexed: 11/29/2022]
Abstract
Histone deacetylase inhibitors (HDACi) induce potent anti-inflammatory responses when used to treat inflammatory diseases. Suberoylanilide hydroxamic acid (SAHA), a pan-HDACi, decreases pro-inflammatory cytokine levels and attenuates cytokine storm in sepsis; however, its toxicity profile toward immune cells has limited its use as a sepsis therapeutic. Here, we developed a modification to SAHA by para-hydroxymethylating the capping group to generate SAHA-OH. We discovered that SAHA-OH provides a favorable improvement to the toxicity profile compared to SAHA. SAHA-OH significantly reduced primary macrophage apoptosis and splenic B cell death as well as mitigated organ damage using a lipopolysaccharide (LPS)-induced endotoxemia mouse model. Furthermore, SAHA-OH retained anti-inflammatory responses similar to SAHA as measured by reductions in LPS-induced proinflammatory cytokine secretions in vitro and in vivo. These effects were attributed to a decreased selectivity of HDAC1, 2, 3, 8 and an increased selectivity for HDAC6 for SAHA-OH as determined by IC50 values. Our results support the potential for SAHA-OH to modulate acute proinflammatory responses while mitigating SAHA-associated drug toxicity for use in the treatment of inflammation-associated diseases and conditions.
Collapse
Affiliation(s)
- Nhu Truong
- Department
of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland 21201, United States
| | - Christopher C. Goodis
- Department
of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland 21201, United States
| | - Andrea L. Cottingham
- Department
of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland 21201, United States
| | - Jacob R. Shaw
- Department
of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
| | - Steven Fletcher
- Department
of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland 21201, United States
| | - Ryan M. Pearson
- Department
of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland 21201, United States
- Department
of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
- Marlene
and Stewart Greenbaum Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
| |
Collapse
|
10
|
Yin X, Xu X, Li H, Jiang N, Wang J, Lu Z, Xiong N, Gong Y. Evaluation of early antibiotic use in patients with non-severe COVID-19 without bacterial infection. Int J Antimicrob Agents 2022; 59:106462. [PMID: 34695565 PMCID: PMC8536497 DOI: 10.1016/j.ijantimicag.2021.106462] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVES The use of antibiotics was common in some countries during the early phase of the coronavirus disease 2019 (COVID-19) pandemic, but adequate evaluation remains lacking. This study aimed to evaluate the effect of early antibiotic use in patients with non-severe COVID-19 admitted without bacterial infection. METHODS This multi-centre retrospective cohort study included 1,373 inpatients with non-severe COVID-19 admitted without bacterial infection. Patients were divided into two groups according to their exposure to antibiotics within 48 h of admission. The outcomes were progression to severe COVID-19, length of stay >15 days and mortality rate. A mixed-effect Cox model and random effect logistic regression were used to explore the association between early antibiotic use and outcomes. RESULTS During the 30-day follow-up period, the proportion of patients who progressed to severe COVID-19 in the early antibiotic use group was almost 1.4 times that of the comparison group. In the mixed-effect model, the early use of antibiotics was associated with higher probability of developing severe COVID-19 and staying in hospital for >15 days. However, there was no significant association between early use of antibiotics and mortality. Analysis with propensity-score-matched cohorts displayed similar results. In subgroup analysis, patients receiving any class of antibiotic were at increased risk of adverse health outcomes. Azithromycin did not improve disease progression and length of stay in patients with COVID-19. CONCLUSIONS It is suggested that antibiotic use should be avoided unless absolutely necessary in patients with non-severe COVID-19, particularly in the early stages.
Collapse
Affiliation(s)
- Xiaoxv Yin
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xing Xu
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Hui Li
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Nan Jiang
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jing Wang
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zuxun Lu
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| | - Yanhong Gong
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
11
|
Hao Y, Wang Z, Wang X, Zhan W, Wu D. OGDH is involved in sepsis induced acute lung injury through the MAPK pathway. J Thorac Dis 2021; 13:5042-5054. [PMID: 34527342 PMCID: PMC8411135 DOI: 10.21037/jtd-21-948] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/02/2021] [Indexed: 11/06/2022]
Abstract
Background Acute lung injury (ALI) induced by sepsis is a common cause of death in clinical practice, and there remains a lack of clinical effective treatment. Cecal ligation and puncture (CLP) is a classic animal model of sepsis, which can induce ALI. Studies have shown that in the lung injury cell model, OGDH (oxoglutarate dehydrogenase) transcription is up-regulated, which is a potential therapeutic target for acute pneumonia. The purpose of this study was to confirm the effects of OGDH on lung injury and inflammation in animal and cell models, and to explore its mechanism. Methods By analyzing the GSE16650 gene set, the upregulated OGDH gene was detected in the lung injury cell model. In a sepsis animal model established by CLP and a lung injury cell model, RT-PCR, immunohistochemistry, WB, and other techniques were used to verify the upregulation of OGDH expression, which was then was down-regulated with shRNA to confirm its relationship with ALI. Further, ELISA, RT-PCR, and WB were used to detect the effect of OGDH on the expression of pro-inflammatory factors including IL-1β, IL-6, IL-18, and TNF-α. The downstream pathway of OGDH was predicted using KEGG and GSEA tools and verified by WB and immunofluorescence. Results The results showed OGDH was highly expressed in a lung injury cell model and the lung tissue of ALI mice induced by CLP, and downregulation of OGDH alleviated sepsis induced ALI. In animal models and cell models, the expression of OGDH was positively correlated with the expression of pro-inflammatory factors. OGDH may act through the MAPK pathway. Conclusions Under the pathological condition of sepsis, OGDH amplifies the inflammatory response through the MAPK pathway, releases pro-inflammatory factors, and induces ALI.
Collapse
Affiliation(s)
- Yuewei Hao
- Department of Emergency, Shandong Second Provincial General Hospital, Jinan, China
| | - Zheng Wang
- Prehospital Emergency, Shandong Second Provincial General Hospital, Jinan, China
| | - Xinfang Wang
- Neonatal Intensive Care Unit, Shandong Second Provincial General Hospital, Jinan, China
| | - Wenming Zhan
- Department of Internal Medicine-Cardiovascular, Xishui People's Hospital, Xishui, China
| | - Dianshui Wu
- Department of Clinical Laboratory, Shandong Second Provincial General Hospital, Jinan, China
| |
Collapse
|
12
|
Falcão-Holanda RB, Brunialti MKC, Jasiulionis MG, Salomão R. Epigenetic Regulation in Sepsis, Role in Pathophysiology and Therapeutic Perspective. Front Med (Lausanne) 2021; 8:685333. [PMID: 34322502 PMCID: PMC8312749 DOI: 10.3389/fmed.2021.685333] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis is characterized by an initial hyperinflammatory response, with intense cell activation and cytokine storm. In parallel, a prolonged compensatory anti-inflammatory response, known as immunological tolerance, can lead to immunosuppression. Clinically, this condition is associated with multiple organ failure, resulting in the patient's death. The mechanisms underlying the pathophysiology of sepsis are not yet fully understood, but evidence is strong showing that epigenetic changes, including DNA methylation and post-translational modifications of histones, modulate the inflammatory response of sepsis. During the onset of infection, host cells undergo epigenetic changes that favor pathogen survival. Besides, epigenetic changes in essential genes also orchestrate the patient's inflammatory response. In this review, we gathered studies on sepsis and epigenetics to show the central role of epigenetic mechanisms in various aspects of the pathogenesis of sepsis and the potential of epigenetic interventions for its treatment.
Collapse
Affiliation(s)
- Renata Brito Falcão-Holanda
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Milena Karina Colo Brunialti
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Miriam Galvonas Jasiulionis
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Reinaldo Salomão
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
13
|
Histone Deacetylase 7 Inhibition in a Murine Model of Gram-Negative Pneumonia-Induced Acute Lung Injury. Shock 2021; 53:344-351. [PMID: 31083049 DOI: 10.1097/shk.0000000000001372] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Pulmonary infections remain the most common cause of Acute Respiratory Distress Syndrome (ARDS), a pulmonary inflammatory disease with high mortality, for which no targeted therapy currently exists. We have previously demonstrated an ameliorated syndrome with early, broad spectrum Histone Deacetylase (HDAC) inhibition in a murine model of gram-negative pneumonia-induced Acute Lung Injury (ALI), the underlying pulmonary pathologic phenotype leading to ARDS. With the current project we aim to determine if selective inhibition of a specific HDAC leads to a similar pro-survival phenotype, potentially pointing to a future therapeutic target. METHODS C57Bl/6 mice underwent endotracheal instillation of 30×10Escherichia coli (strain 19138) versus saline (n = 24). Half the infected mice were administered Trichostatin A (TSA) 30 min later. All animals were sacrificed 6 h later for tissue sampling and HDAC quantification, while another set of animals (n = 24) was followed to determine survival. Experiments were repeated with selective siRNA inhibition of the HDAC demonstrating the greatest inhibition versus scrambled siRNA (n = 24). RESULTS TSA significantly ameliorated the inflammatory phenotype and improved survival in infected-ALI mice, and HDAC7 was the HDAC with the greatest transcription and protein translation suppression. Similar results were obtained with selective HDAC7 siRNA inhibition compared with scrambled siRNA. CONCLUSION HDAC7 appears to play a key role in the inflammatory response that leads to ALI after gram-negative pneumonia in mice.
Collapse
|
14
|
Abstract
OBJECTIVES Recent evidence from the fields of microbiology and immunology, as well as a small number of human sepsis studies, suggest that epigenetic regulation may play a central role in the pathogenesis of sepsis. The term "epigenetics" refers to regulatory mechanisms that control gene expression but are not related to changes in DNA sequence. These include DNA methylation, histone modifications, and regulation of transcription via non-coding RNAs. Epigenetic modifications, occurring in response to external stressors, lead to changes in gene expression, and thus lie at the intersection between genetics and the environment. In this review, we examine data from in vitro studies, animal studies, and the existing human sepsis studies in epigenetics to demonstrate that epigenetic mechanisms are likely central to the pathogenesis of sepsis and that epigenetic therapies may have potential in the treatment of sepsis and its associated organ failures. DATA SOURCES Online search of published scientific literature via Pubmed using the term "epigenetics" in combination with the terms "sepsis", "infection", "bacterial infection", "viral infection", "critical illness", "acute respiratory distress syndrome", and "acute lung injury". STUDY SELECTION Articles were chosen for inclusion based on their relevance to sepsis, acute inflammation, sepsis-related immune suppression, and sepsis-related organ failure. Reference lists were reviewed to identify additional relevant articles. DATA EXTRACTION Relevant data was extracted and synthesized for narrative review. DATA SYNTHESIS Epigenetic regulation is a key determinant of gene expression in sepsis. At the onset of infection, host-pathogen interactions often result in epigenetic alterations to host cells that favor pathogen survival. In parallel, the host inflammatory response is characterized by epigenetic modifications in key regulatory genes, including tumor necrosis factor and interleukin-1β. In human sepsis patients, multiple epigenetic modifying enzymes show differential expression in early sepsis, suggesting a role for epigenetics in coordinating the response to infection. In the later stages of sepsis, epigenetic modifications accompany endotoxin tolerance and the immune-suppressed state. In animal models, treatment with epigenetic modifiers can mitigate the effects of sepsis and improve survival as well as reverse sepsis-associated organ injury. CONCLUSIONS Epigenetic modifications are associated with key phases of sepsis, from the host-pathogen interaction, to acute inflammation, to immune suppression. Epigenetic markers show promise in the diagnosis and prognosis of sepsis and epigenetic modifying agents show promise as therapeutic tools in animal models of sepsis. Human studies in the area of epigenetics are sorely lacking and should be a priority for sepsis researchers.
Collapse
|
15
|
Wei X, Tsai T, Howe S, Zhao J. Weaning Induced Gut Dysfunction and Nutritional Interventions in Nursery Pigs: A Partial Review. Animals (Basel) 2021; 11:1279. [PMID: 33946901 PMCID: PMC8146462 DOI: 10.3390/ani11051279] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 01/08/2023] Open
Abstract
Weaning is one of the most stressful events in the life of a pig. Unsuccessful weaning often leads to intestinal and immune system dysfunctions, resulting in poor growth performance as well as increased morbidity and mortality. The gut microbiota community is a complex ecosystem and is considered an "organ," producing various metabolites with many beneficial functions. In this review, we briefly introduce weaning-associated gut microbiota dysbiosis. Then, we explain the importance of maintaining a balanced gut microbiota. Finally, we discuss dietary supplements and their abilities to restore intestinal balance and improve the growth performance of weaning pigs.
Collapse
Affiliation(s)
| | | | | | - Jiangchao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR 72701, USA; (X.W.); (T.T.); (S.H.)
| |
Collapse
|
16
|
Bossardi Ramos R, Adam AP. Molecular Mechanisms of Vascular Damage During Lung Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:95-107. [PMID: 34019265 DOI: 10.1007/978-3-030-68748-9_6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A variety of pulmonary and systemic insults promote an inflammatory response causing increased vascular permeability, leading to the development of acute lung injury (ALI), a condition necessitating hospitalization and intensive care, or the more severe acute respiratory distress syndrome (ARDS), a disease with a high mortality rate. Further, COVID-19 pandemic-associated ARDS is now a major cause of mortality worldwide. The pathogenesis of ALI is explained by injury to both the vascular endothelium and the alveolar epithelium. The disruption of the lung endothelial and epithelial barriers occurs in response to both systemic and local production of pro-inflammatory cytokines. Studies that evaluate the association of genetic polymorphisms with disease risk did not yield many potential therapeutic targets to treat and revert lung injury. This failure is probably due in part to the phenotypic complexity of ALI/ARDS, and genetic predisposition may be obscured by the multiple environmental and behavioral risk factors. In the last decade, new research has uncovered novel epigenetic mechanisms that control ALI/ARDS pathogenesis, including histone modifications and DNA methylation. Enzyme inhibitors such as DNMTi and HDACi may offer new alternative strategies to prevent or reverse the vascular damage that occurs during lung injury. This review will focus on the latest findings on the molecular mechanisms of vascular damage in ALI/ARDS, the genetic factors that might contribute to the susceptibility for developing this disease, and the epigenetic changes observed in humans, as well as in experimental models of ALI/ADRS.
Collapse
Affiliation(s)
- Ramon Bossardi Ramos
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| | - Alejandro Pablo Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA. .,Department of Ophthalmology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
17
|
Yao Y, Cai X, Fei W, Ye Y, Zhao M, Zheng C. The role of short-chain fatty acids in immunity, inflammation and metabolism. Crit Rev Food Sci Nutr 2020; 62:1-12. [PMID: 33261516 DOI: 10.1080/10408398.2020.1854675] [Citation(s) in RCA: 360] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Short-chain fatty acids (SCFAs) are carboxylic acids with carbon atom numbers less than 6, which are important metabolites of gut microbiome. Existing research shows that SCFAs play a vital role in the health and disease of the host. First, SCFAs are the key energy source for colon and ileum cells, and affect the intestinal epithelial barrier and defense functions by regulating related gene expression. Second, SCFAs regulate the function of innate immune cells to participate in the immune system, such as macrophages, neutrophils and dendritic cells. Third, SCFAs can also regulate the differentiation of T cells and B cells and the antigen-specific adaptive immunity mediated by them. Besides, SCFAs are raw materials for sugar and lipid synthesis, which provides a theoretical basis for studying the potential role of SCFAs in regulating energy homeostasis and metabolism. There are also studies showing that SCFAs inhibit tumor cell proliferation and promote apoptosis. In this article, we summarized in detail the role of SCFAs in immunity, inflammation and metabolism, and briefly introduced the role of SCFAs in tumor cell survival. It provides a systematic theoretical basis for the study of SCFAs as potential drugs to promote human health.
Collapse
Affiliation(s)
- Yao Yao
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Xiaoyu Cai
- Department of Pharmacy, Hangzhou First People's Hospital, Hangzhou, China
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Yiqing Ye
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Mengdan Zhao
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| |
Collapse
|
18
|
Kim SM, DeFazio JR, Hyoju SK, Sangani K, Keskey R, Krezalek MA, Khodarev NN, Sangwan N, Christley S, Harris KG, Malik A, Zaborin A, Bouziat R, Ranoa DR, Wiegerinck M, Ernest JD, Shakhsheer BA, Fleming ID, Weichselbaum RR, Antonopoulos DA, Gilbert JA, Barreiro LB, Zaborina O, Jabri B, Alverdy JC. Fecal microbiota transplant rescues mice from human pathogen mediated sepsis by restoring systemic immunity. Nat Commun 2020; 11:2354. [PMID: 32393794 PMCID: PMC7214422 DOI: 10.1038/s41467-020-15545-w] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 03/11/2020] [Indexed: 12/13/2022] Open
Abstract
Death due to sepsis remains a persistent threat to critically ill patients confined to the intensive care unit and is characterized by colonization with multi-drug-resistant healthcare-associated pathogens. Here we report that sepsis in mice caused by a defined four-member pathogen community isolated from a patient with lethal sepsis is associated with the systemic suppression of key elements of the host transcriptome required for pathogen clearance and decreased butyrate expression. More specifically, these pathogens directly suppress interferon regulatory factor 3. Fecal microbiota transplant (FMT) reverses the course of otherwise lethal sepsis by enhancing pathogen clearance via the restoration of host immunity in an interferon regulatory factor 3-dependent manner. This protective effect is linked to the expansion of butyrate-producing Bacteroidetes. Taken together these results suggest that fecal microbiota transplantation may be a treatment option in sepsis associated with immunosuppression. Sepsis due to multidrug resistant pathogens is the most common cause of death in intensive care units. Here, the authors report that fecal microbiota transplant (FMT) can rescue mice from lethal sepsis of pathogens isolated from stool of a critically ill patient and show that FMT reverses the immunosuppressive effect induced by the pathogen community.
Collapse
Affiliation(s)
- Sangman M Kim
- Committee on Immunology, University of Chicago, Chicago, IL, USA.,Department of Medicine, University of Chicago, Chicago, IL, USA.,Department of Biology, University of San Francisco, San Francisco, CA, USA
| | - Jennifer R DeFazio
- Department of Surgery, University of Chicago, Chicago, IL, USA.,Department of Surgery, Columbia University, New York, NY, USA
| | - Sanjiv K Hyoju
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Kishan Sangani
- Committee on Immunology, University of Chicago, Chicago, IL, USA.,Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Robert Keskey
- Committee on Immunology, University of Chicago, Chicago, IL, USA.,Department of Surgery, University of Chicago, Chicago, IL, USA
| | | | - Nikolai N Khodarev
- Department of Radiation and Cellular Oncology and The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Naseer Sangwan
- Department of Surgery, University of Chicago, Chicago, IL, USA.,Argonne National Laboratory, Argonne, IL, USA
| | - Scott Christley
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | | | - Ankit Malik
- Committee on Immunology, University of Chicago, Chicago, IL, USA.,Department of Medicine, University of Chicago, Chicago, IL, USA
| | | | - Romain Bouziat
- Committee on Immunology, University of Chicago, Chicago, IL, USA.,Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Diana R Ranoa
- Department of Radiation and Cellular Oncology and The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Mara Wiegerinck
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Jordan D Ernest
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | | | - Irma D Fleming
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Dionysios A Antonopoulos
- Department of Medicine, University of Chicago, Chicago, IL, USA.,Argonne National Laboratory, Argonne, IL, USA
| | - Jack A Gilbert
- Department of Surgery, University of Chicago, Chicago, IL, USA.,Argonne National Laboratory, Argonne, IL, USA
| | - Luis B Barreiro
- Department of Medicine, University of Chicago, Chicago, IL, USA.,Department of Genetics, Sainte-Justine Hospital Research Centre, University of Montreal, Montreal, QC, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Olga Zaborina
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Bana Jabri
- Committee on Immunology, University of Chicago, Chicago, IL, USA. .,Department of Medicine, University of Chicago, Chicago, IL, USA. .,Department of Pathology, University of Chicago, Chicago, IL, USA. .,Department of Pediatrics, Section of Gastroenterology, Hepatology and Nutrition, University of Chicago, Chicago, IL, USA.
| | - John C Alverdy
- Department of Surgery, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
19
|
Hwang JS, Kim E, Hur J, Yoon TJ, Seo HG. Ring finger protein 219 regulates inflammatory responses by stabilizing sirtuin 1. Br J Pharmacol 2020; 177:4601-4614. [PMID: 32220064 DOI: 10.1111/bph.15060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 03/11/2020] [Accepted: 03/14/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND PURPOSE Ring finger protein 219 (RNF219), a protein containing the C3 HC4 -type RING-HC motif, has been identified as a binding partner of the histone deacetylase sirtuin 1 (SIRT1). To explore the functions of RNF219, we examined its possible roles in the cellular responses to inflammation. EXPERIMENTAL APPROACH Effects of RNF219 on SIRT1 were studied in vitro using RAW264.7 cells and in male BALB/c mice, treated with LPS or IFN-γ. Western blots, RT-PCR, co-immunoprecipitation and ubiquitination assays were used, along with LC-MS/MS analysis. In vivo, survival and serum cytokines and tissue levels of RNF219 and SIRT1 were measured. KEY RESULTS Binding of RNF219 to SIRT1 inhibited degradation of SIRT1 by preventing its ubiquitination, thereby prolonging SIRT1-mediated anti-inflammatory signalling. LPS caused RNF219 deacetylation, leading to instability of RNF219 and preventing its association with SIRT1. Accordingly, the acetylation status of RNF219 is a critical determinant in its interaction with SIRT1, affecting the response to inflammatory stimuli. The deacetylase inhibitor trichostatin A, increased acetylation and stability of RNF219 and survival of mice injected with LPS, through the interaction of RNF219 with SIRT1. CONCLUSION AND IMPLICATIONS RNF219 is involved in a novel mechanism to stabilize SIRT1 protein by protein-protein interaction, leading to the resolution of cellular inflammation. These observations provide new insights into the function of RNF219 in modulation of cellular inflammation, and may aid and encourage the development of new anti-inflammatory drugs.
Collapse
Affiliation(s)
- Jung Seok Hwang
- College of Sang-Huh Life Sciences, Konkuk University, Seoul, Republic of Korea
| | - Eunsu Kim
- College of Sang-Huh Life Sciences, Konkuk University, Seoul, Republic of Korea
| | - Jinwoo Hur
- College of Sang-Huh Life Sciences, Konkuk University, Seoul, Republic of Korea
| | - Taek Joon Yoon
- Department of Food Science and Nutrition, Yuhan University, Bucheon-si, Republic of Korea
| | - Han Geuk Seo
- College of Sang-Huh Life Sciences, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
20
|
Fu J, Li G, Wu X, Zang B. Sodium Butyrate Ameliorates Intestinal Injury and Improves Survival in a Rat Model of Cecal Ligation and Puncture-Induced Sepsis. Inflammation 2020; 42:1276-1286. [PMID: 30903547 DOI: 10.1007/s10753-019-00987-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sepsis is a life-threatening condition with a high rate of mortality. Unfortunately, very few therapies can improve outcomes in patients with sepsis. Butyrate, which is the most potent histone deacetylase (HDAC) inhibitor among short-chain fatty acids, exerts anti-inflammatory effects in a variety of inflammatory diseases. Butyrate might thus be valuable in the treatment of sepsis, in which inhibition of overwhelming cytokine release is vitally important. Sepsis was induced in 7- to 8-week-old Sprague-Dawley rats by cecal ligation and puncture (CLP) with a 21-g double-puncture technique. Rats received an intravenous injection of normal saline (vehicle) or sodium butyrate (200 mg/kg) after CLP and were sacrificed 12 h later. Hematoxylin and eosin staining was performed to observe the intestinal mucosal morphology. RT-PCR and ELISA were used to determine the intestinal inflammatory response in vivo. Intestinal permeability was evaluated by measuring fluorescein isothiocyanate dextran (FD-4) absorption in vivo, and tight junction protein expression was examined by western blot. NF-κB p65 activities were assessed by western blot and immunohistochemistry. Sodium butyrate treatment improved the survival rate of CLP rats and alleviated sepsis-induced intestinal mucosal injury. Proinflammatory cytokine expression was lower in butyrate-treated rats than in the vehicle group. FD-4 leakage from the intestinal tract was reduced, and the expression levels of the tight junction proteins claudin-1 and ZO-1 were also restored in rats that received sodium butyrate treatment. These effects were associated with less NF-κB p65 nuclear translocation, whereas the expression of Iκ-Bα was not affected or even increased. Sodium butyrate mitigates the inflammatory response and maintains intestinal barrier function in polymicrobial sepsis partly through inhibition of NF-κB activation and may serve as a novel therapy for sepsis.
Collapse
Affiliation(s)
- Jiahong Fu
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China
| | - Guofu Li
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China
| | - Xingmao Wu
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China
| | - Bin Zang
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
21
|
Hamam HJ, Palaniyar N. Post-Translational Modifications in NETosis and NETs-Mediated Diseases. Biomolecules 2019; 9:E369. [PMID: 31416265 PMCID: PMC6723044 DOI: 10.3390/biom9080369] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 12/19/2022] Open
Abstract
: Neutrophils undergo a unique form of cell death that generates neutrophil extracellular traps (NETs) that may help to neutralize invading pathogens and restore homeostasis. However, uncontrolled NET formation (NETosis) can result in numerous diseases that adversely affect health. Recent studies further elucidate the mechanistic details of the different forms of NETosis and their common end structure, as NETs were constantly found to contain DNA, modified histones and cytotoxic enzymes. In fact, emerging evidence reveal that the post translational modifications (PTMs) of histones in neutrophils have a critical role in regulating neutrophil death. Histone citrullination is shown to promote a rapid form of NET formation independent of NADPH oxidase (NOX), which relies on calcium influx. Interestingly, few studies suggest an association between histone citrullination and other types of PTMs to control cell survival and death, such as histone methylation. Even more exciting is the finding that histone acetylation has a biphasic effect upon NETosis, where histone deacetylase (HDAC) inhibitors promote baseline, NOX-dependent and -independent NETosis. However, increasing levels of histone acetylation suppresses NETosis, and to switch neutrophil death to apoptosis. Interestingly, in the presence of NETosis-promoting stimuli, high levels of HDACis limit both NETosis and apoptosis, and promote neutrophil survival. Recent studies also reveal the importance of the PTMs of neutrophils in influencing numerous pathologies. Histone modifications in NETs can act as a double-edged sword, as they are capable of altering multiple types of neutrophil death, and influencing numerous NET-mediated diseases, such as acute lung injury (ALI), thrombosis, sepsis, systemic lupus erythematosus, and cancer progression. A clear understanding of the role of different PTMs in neutrophils would be important for an understanding of the molecular mechanisms of NETosis, and to appropriately treat NETs-mediated diseases.
Collapse
Affiliation(s)
- Hussein J Hamam
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Nades Palaniyar
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
22
|
Menden H, Xia S, Mabry SM, Noel-MacDonnell J, Rajasingh J, Ye SQ, Sampath V. Histone deacetylase 6 regulates endothelial MyD88-dependent canonical TLR signaling, lung inflammation, and alveolar remodeling in the developing lung. Am J Physiol Lung Cell Mol Physiol 2019; 317:L332-L346. [PMID: 31268348 DOI: 10.1152/ajplung.00247.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Lung endothelial cell (EC) immune activation during bacterial sepsis contributes to acute lung injury and bronchopulmonary dysplasia in premature infants. The epigenetic regulators of sepsis-induced endothelial immune activation, lung inflammation, and alveolar remodeling remain unclear. Herein, we examined the role of the cytoplasmic histone deacetylase, HDAC6, in regulating EC Toll-like receptor 4 (TLR4) signaling and modulating sepsis-induced lung injury in a neonatal model of sterile sepsis. In human primary microvascular endothelial cells (HPMEC), lipopolysaccharide (LPS)-induced MAPK, IKK-β, and p65 phosphorylation as well as inflammatory cytokine expression were exaggerated with the HDAC6 inhibitor tubastatin A, and by dominant-negative HDAC6 with a mutated catalytic domain 2. Expression of HDAC6 wild-type protein suppressed LPS-induced myeloid differentiation primary response 88 (MyD88) acetylation, p65 (Lys310) acetylation, MyD88/TNF receptor-associated factor 6 (TRAF6) coimmunoprecipitation, and proinflammatory TLR4 signaling in HPMEC. In a neonatal mouse model of sepsis, the HDAC6 inhibitor tubastatin A amplified lung EC TLR4 signaling and vascular permeability. HDAC6 inhibition augmented LPS-induced MyD88 acetylation, MyD88/TRAF6 binding, p65 acetylation, canonical TLR4 signaling, and inflammation in the developing lung. Sepsis-induced decreases in the fibroblast growth factors FGF2 and FGF7 and increase in matrix metalloproteinase-9 were worsened with HDAC6 inhibition, while elastin expression was equally suppressed. Exaggerated sepsis-induced acute lung inflammation observed with HDAC6 inhibition worsened alveolar simplification evidenced by increases in mean linear intercepts and decreased radial alveolar counts. Our studies reveal that HDAC6 is a constitutive negative regulator of cytoplasmic TLR4 signaling in EC and the developing lung. The therapeutic efficacy of augmenting HDAC6 activity in neonatal sepsis to prevent lung injury needs to be evaluated.
Collapse
Affiliation(s)
- Heather Menden
- Division of Neonatology, Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Sheng Xia
- Division of Neonatology, Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Sherry M Mabry
- Division of Neonatology, Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Janelle Noel-MacDonnell
- Division of Health Services and Outcomes Research, Children's Mercy Hospital, Kansas City, Missouri
| | - Johnson Rajasingh
- Department of Cardiovascular Medicine, Kansas University Medical Center, Kansas City, Missouri
| | - Shui Qing Ye
- Department of Biomedical and Health Informatics, University of Missouri at Kansas City, Kansas City, Missouri
| | - Venkatesh Sampath
- Division of Neonatology, Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| |
Collapse
|
23
|
Abstract
Sepsis, the 10th leading cause of death, is the most expensive condition in the United States. The immune response in sepsis transitions from hyperinflammatory to a hypoinflammatory and immunosuppressive phase; individual variations regarding timing and overlap between hyper- and hypoinflammation exist in a number of patients. While one third of the sepsis-related deaths occur during hyperinflammation, majority of the sepsis-mortality occurs during the hypoinflammatory phase. Currently, no phase-specific molecular-based therapies exist to treat sepsis. Coordinated epigenetic and metabolic perturbations orchestrate this shift from hyper- to hypoinflammation in innate immune cells during sepsis. These epigenetic and metabolic changes during sepsis progression and therapeutic opportunities they pose are described in this review.
Collapse
Affiliation(s)
- Vidula Vachharajani
- Department of Anesthesiology, Wake Forest School of Medicine,
Winston-Salem, NC, USA,Department of Internal Medicine, Wake Forest School of Medicine,
Winston-Salem, NC, USA,Vidula Vachharajani, Critical Care
Medicine/Respiratory Institute, Inflammation and Immunity/Lerner Research
Institute, Cleveland Lerner College of Medicine, 9500 Euclid Avenue, Cleveland,
OH, USA.
| | - Charles E McCall
- Department of Internal Medicine, Wake Forest School of Medicine,
Winston-Salem, NC, USA
| |
Collapse
|
24
|
Valproic acid attenuates sepsis-induced myocardial dysfunction in rats by accelerating autophagy through the PTEN/AKT/mTOR pathway. Life Sci 2019; 232:116613. [PMID: 31265853 DOI: 10.1016/j.lfs.2019.116613] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/27/2019] [Accepted: 06/27/2019] [Indexed: 01/15/2023]
Abstract
AIMS Sepsis is a leading cause of death and disability worldwide. Autophagy may play a protective role in sepsis-induced myocardial dysfunction (SIMD). The present study investigated whether valproic acid (VPA), a class I histone deacetylase (HDAC) inhibitor, can attenuate SIMD by accelerating autophagy. MAIN METHODS A sepsis model was established via the cecum ligation and puncture of male Sprague-Dawley rats. Cardiac injuries were measured using serum markers, echocardiographic cardiac parameters, and hematoxylin and eosin staining. Cardiac mitochondria injuries were detected with transmission electron microscopy, adenosine triphosphate (ATP) and cardiac mitochondrial DNA (mtDNA) contents. Cardiac oxidative levels were measured using redox markers in the cardiac homogenate. Real-time polymerase chain reaction (RT-PCR) and Western blot were performed to detect the expression levels of relative genes and proteins. HDAC binding to the phosphatase and tensin homolog deleted on chromosome ten (PTEN) promoters and histone acetylation levels of the PTEN promoters were analyzed via chromatin immunoprecipitation and quantitative RT-PCR. KEY FINDINGS VPA can ameliorate SIMD by enhancing the autophagy level of the myocardium to reduce mitochondrial damage, oxidative stress, and myocardial inflammation in septic rats. Moreover, this study demonstrated that VPA induces autophagy by inhibiting HDAC1- and HDAC3-mediated PTEN expression in the myocardial tissues of septic rats. SIGNIFICANCE This study found that VPA attenuates SIMD through myocardial autophagy acceleration by increasing PTEN expression and inhibiting the AKT/mTOR pathway. These findings preliminarily suggest that VPA may be a potential approach for the intervention and treatment of SIMD.
Collapse
|
25
|
Cross D, Drury R, Hill J, Pollard AJ. Epigenetics in Sepsis: Understanding Its Role in Endothelial Dysfunction, Immunosuppression, and Potential Therapeutics. Front Immunol 2019; 10:1363. [PMID: 31275313 PMCID: PMC6591469 DOI: 10.3389/fimmu.2019.01363] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 05/29/2019] [Indexed: 12/11/2022] Open
Abstract
Sepsis has a complex pathophysiology in which both excessive and refractory inflammatory responses are hallmark features. Pro-inflammatory cytokine responses during the early stages are responsible for significant endothelial dysfunction, loss of endothelial integrity, and organ failure. In addition, it is now well-established that a substantial number of sepsis survivors experience ongoing immunological derangement and immunosuppression following a septic episode. The underpinning mechanisms of these phenomena are incompletely understood yet they contribute to a significant proportion of sepsis-associated mortality. Epigenetic mechanisms including DNA methylation, histone modifications, and non-coding RNAs, have an increasingly clear role in modulating inflammatory and other immunological processes. Recent evidence suggests epigenetic mechanisms are extensively perturbed as sepsis progresses, and particularly play a role in endothelial dysfunction and immunosuppression. Whilst therapeutic modulation of the epigenome is still in its infancy, there is substantial evidence from animal models that this approach could reap benefits. In this review, we summarize research elucidating the role of these mechanisms in several aspects of sepsis pathophysiology including tissue injury and immunosuppression. We also evaluate pre-clinical evidence for the use of "epi-therapies" in the treatment of poly-microbial sepsis.
Collapse
Affiliation(s)
- Deborah Cross
- Oxford Vaccine Group, Department of Paediatrics, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | | | | | | |
Collapse
|
26
|
Xu M, Wang C, Li N, Wang J, Zhang Y, Deng X. Intraperitoneal Injection of Acetate Protects Mice Against Lipopolysaccharide (LPS)‑Induced Acute Lung Injury Through Its Anti-Inflammatory and Anti-Oxidative Ability. Med Sci Monit 2019; 25:2278-2288. [PMID: 30921298 PMCID: PMC6450300 DOI: 10.12659/msm.911444] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND As a member of short-chain fatty acids, acetate exhibits anti-inflammatory capacity. The present study aimed to investigate the effect of acetate on lipopolysaccharide (LPS)-induced acute lung injury (ALI) and explored its underlying mechanism. MATERIAL AND METHODS Acetate (250 mM, 400 µL) was given intraperitoneally 30 minutes after LPS (5 mg/kg) intratracheal injection. Lung tissues and bronchoalveolar lavage fluid (BALF) were collected 6 hours after the challenge of LPS. The histopathology scores, wet-to-dry weight ratios, protein content, and cytokine levels in BALF were assessed. RESULTS The acetate treatment resulted in improved lung pathological score, alleviated LPS-induced microvascular permeability, and suppressed the production of reactive oxygen species. Furthermore, acetate decreased the level of pro-inflammatory cytokines and chemokines in the lungs and BALF, consistent with the declined immune cell counting found in BALF. In addition, phosphorylation levels of mitogen-activated protein kinase (MAPK) pathway in lung tissues were downregulated by acetate. CONCLUSIONS These results suggested that acetate exerts its protective effects via anti-inflammatory and anti-oxidant activities on LPS-induced ALI.
Collapse
Affiliation(s)
- Mengda Xu
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Changli Wang
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Na Li
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Jun Wang
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Yan Zhang
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| | - Xiaoming Deng
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China (mainland)
| |
Collapse
|
27
|
Cross talk between neutrophils and the microbiota. Blood 2019; 133:2168-2177. [PMID: 30898860 DOI: 10.1182/blood-2018-11-844555] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 12/24/2018] [Indexed: 12/18/2022] Open
Abstract
The microbiota has emerged as an important regulator of the host immunity by the induction, functional modulation, or suppression of local and systemic immune responses. In return, the host immune system restricts translocation and fine tunes the composition and distribution of the microbiota to maintain a beneficial symbiosis. This paradigm applies to neutrophils, a critical component of the innate immunity, allowing their production and function to be influenced by microbial components and metabolites derived from the microbiota, and engaging them in the process of microbiota containment and regulation. The cross talk between neutrophils and the microbiota adjusts the magnitude of neutrophil-mediated inflammation on challenge while preventing neutrophil responses against commensals under steady state. Here, we review the major molecular and cellular mediators of the interactions between neutrophils and the microbiota and discuss their interplay and contribution in chronic inflammatory diseases and cancer.
Collapse
|
28
|
von Knethen A, Brüne B. Histone Deacetylation Inhibitors as Therapy Concept in Sepsis. Int J Mol Sci 2019; 20:ijms20020346. [PMID: 30654448 PMCID: PMC6359123 DOI: 10.3390/ijms20020346] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/11/2019] [Accepted: 01/12/2019] [Indexed: 12/15/2022] Open
Abstract
Sepsis is characterized by dysregulated gene expression, provoking a hyper-inflammatory response occurring in parallel to a hypo-inflammatory reaction. This is often associated with multi-organ failure, leading to the patient’s death. Therefore, reprogramming of these pro- and anti-inflammatory, as well as immune-response genes which are involved in acute systemic inflammation, is a therapy approach to prevent organ failure and to improve sepsis outcomes. Considering epigenetic, i.e., reversible, modifications of chromatin, not altering the DNA sequence as one tool to adapt the expression profile, inhibition of factors mediating these changes is important. Acetylation of histones by histone acetyltransferases (HATs) and initiating an open-chromatin structure leading to its active transcription is counteracted by histone deacetylases (HDACs). Histone deacetylation triggers a compact nucleosome structure preventing active transcription. Hence, inhibiting the activity of HDACs by specific inhibitors can be used to restore the expression profile of the cells. It can be assumed that HDAC inhibitors will reduce the expression of pro-, as well as anti-inflammatory mediators, which blocks sepsis progression. However, decreased cytokine expression might also be unfavorable, because it can be associated with decreased bacterial clearance.
Collapse
Affiliation(s)
- Andreas von Knethen
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt/Main, 60590 Frankfurt, Germany.
- Fraunhofer⁻IME, Project Group Translational Medicine and Pharmacology (TMP), 60596 Frankfurt, Germany.
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt/Main, 60590 Frankfurt, Germany.
- Fraunhofer⁻IME, Project Group Translational Medicine and Pharmacology (TMP), 60596 Frankfurt, Germany.
| |
Collapse
|
29
|
Karki P, Ke Y, Tian Y, Ohmura T, Sitikov A, Sarich N, Montgomery CP, Birukova AA. Staphylococcus aureus-induced endothelial permeability and inflammation are mediated by microtubule destabilization. J Biol Chem 2019; 294:3369-3384. [PMID: 30622143 DOI: 10.1074/jbc.ra118.004030] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 12/13/2018] [Indexed: 12/18/2022] Open
Abstract
Staphylococcus aureus is a major etiological agent of sepsis and induces endothelial cell (EC) barrier dysfunction and inflammation, two major hallmarks of acute lung injury. However, the molecular mechanisms of bacterial pathogen-induced EC barrier disruption are incompletely understood. Here, we investigated the role of microtubules (MT) in the mechanisms of EC barrier compromise caused by heat-killed S. aureus (HKSA). Using a customized monolayer permeability assay in human pulmonary EC and MT fractionation, we observed that HKSA-induced barrier disruption is accompanied by MT destabilization and increased histone deacetylase-6 (HDAC6) activity resulting from elevated reactive oxygen species (ROS) production. Molecular or pharmacological HDAC6 inhibition rescued barrier function in HKSA-challenged vascular endothelium. The HKSA-induced EC permeability was associated with impaired MT-mediated delivery of cytoplasmic linker-associated protein 2 (CLASP2) to the cell periphery, limiting its interaction with adherens junction proteins. HKSA-induced EC barrier dysfunction was also associated with increased Rho GTPase activity via activation of MT-bound Rho-specific guanine nucleotide exchange factor-H1 (GEF-H1) and was abolished by HDAC6 down-regulation. HKSA activated the NF-κB proinflammatory pathway and increased the expression of intercellular and vascular cell adhesion molecules in EC, an effect that was also HDAC6-dependent and mediated, at least in part, by a GEF-H1/Rho-dependent mechanism. Of note, HDAC6 knockout mice or HDAC6 inhibitor-treated WT mice were partially protected from vascular leakage and inflammation caused by both HKSA or methicillin-resistant S. aureus (MRSA). Our results indicate that S. aureus-induced, ROS-dependent up-regulation of HDAC6 activity destabilizes MT and thereby activates the GEF-H1/Rho pathway, increasing both EC permeability and inflammation.
Collapse
Affiliation(s)
- Pratap Karki
- From the Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Yunbo Ke
- the Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Yufeng Tian
- the Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, and
| | - Tomomi Ohmura
- the Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, and
| | - Albert Sitikov
- the Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, and
| | - Nicolene Sarich
- the Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, and
| | - Christopher P Montgomery
- the Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, and.,the Department of Critical Care Medicine, Nationwide Children's Hospital, Columbus, Ohio 43205
| | - Anna A Birukova
- From the Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201,
| |
Collapse
|
30
|
Wienhold SM, Macrì M, Nouailles G, Dietert K, Gurtner C, Gruber AD, Heimesaat MM, Lienau J, Schumacher F, Kleuser B, Opitz B, Suttorp N, Witzenrath M, Müller-Redetzky HC. Ventilator-induced lung injury is aggravated by antibiotic mediated microbiota depletion in mice. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018; 22:282. [PMID: 30373626 PMCID: PMC6206919 DOI: 10.1186/s13054-018-2213-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Antibiotic exposure alters the microbiota, which can impact the inflammatory immune responses. Critically ill patients frequently receive antibiotic treatment and are often subjected to mechanical ventilation, which may induce local and systemic inflammatory responses and development of ventilator-induced lung injury (VILI). The aim of this study was to investigate whether disruption of the microbiota by antibiotic therapy prior to mechanical ventilation affects pulmonary inflammatory responses and thereby the development of VILI. METHODS Mice underwent 6-8 weeks of enteral antibiotic combination treatment until absence of cultivable bacteria in fecal samples was confirmed. Control mice were housed equally throughout this period. VILI was induced 3 days after completing the antibiotic treatment protocol, by high tidal volume (HTV) ventilation (34 ml/kg; positive end-expiratory pressure = 2 cmH2O) for 4 h. Differences in lung function, oxygenation index, pulmonary vascular leakage, macroscopic assessment of lung injury, and leukocyte and lymphocyte differentiation were assessed. Control groups of mice ventilated with low tidal volume and non-ventilated mice were analyzed accordingly. RESULTS Antibiotic-induced microbiota depletion prior to HTV ventilation led to aggravation of VILI, as shown by increased pulmonary permeability, increased oxygenation index, decreased pulmonary compliance, enhanced macroscopic lung injury, and increased cytokine/chemokine levels in lung homogenates. CONCLUSIONS Depletion of the microbiota by broad-spectrum antibiotics prior to HTV ventilation renders mice more susceptible to developing VILI, which could be clinically relevant for critically ill patients frequently receiving broad-spectrum antibiotics.
Collapse
Affiliation(s)
- Sandra-Maria Wienhold
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Mario Macrì
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Present address: Scuola di specializzazione in Anestesia, Rianimazione e Terapia Intensiva, Università degli Studi di Milano, Milan, Italy
| | - Geraldine Nouailles
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kristina Dietert
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Corinne Gurtner
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany.,Present address: Institute of Animal Pathology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Markus M Heimesaat
- Institute for Microbiology and Infection Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jasmin Lienau
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Fabian Schumacher
- Department of Nutritional Toxicology, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.,Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Burkhard Kleuser
- Department of Nutritional Toxicology, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Bastian Opitz
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Martin Witzenrath
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany. .,Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
| | - Holger C Müller-Redetzky
- Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
31
|
Van Wyngene L, Vandewalle J, Libert C. Reprogramming of basic metabolic pathways in microbial sepsis: therapeutic targets at last? EMBO Mol Med 2018; 10:e8712. [PMID: 29976786 PMCID: PMC6079534 DOI: 10.15252/emmm.201708712] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 04/27/2018] [Accepted: 05/25/2018] [Indexed: 12/15/2022] Open
Abstract
Sepsis is a highly lethal and urgent unmet medical need. It is the result of a complex interplay of several pathways, including inflammation, immune activation, hypoxia, and metabolic reprogramming. Specifically, the regulation and the impact of the latter have become better understood in which the highly catabolic status during sepsis and its similarity with starvation responses appear to be essential in the poor prognosis in sepsis. It seems logical that new interventions based on the recognition of new therapeutic targets in the key metabolic pathways should be developed and may have a good chance to penetrate to the bedside. In this review, we concentrate on the pathological changes in metabolism, observed during sepsis, and the presumed underlying mechanisms, with a focus on the level of the organism and the interplay between different organ systems.
Collapse
Affiliation(s)
- Lise Van Wyngene
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jolien Vandewalle
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Claude Libert
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
32
|
Kovacs L, Kovacs-Kasa A, Verin AD, Fulton D, Lucas R, Su Y. Histone deacetylases in vascular permeability and remodeling associated with acute lung injury. ACTA ACUST UNITED AC 2018; 2. [PMID: 32099966 DOI: 10.20517/2574-1209.2018.06] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Acute lung injury (ALI) is a severe progressive disorder that arises from a wide range of causes such as toxins or inflammation, resulting in significant morbidity and mortality. There are no effective therapeutic options apart from mechanical ventilation strategies. While the mechanisms that govern the clinically relevant process of increased EC permeability and remodeling associated with ALI are under intense investigation, our knowledge of the processes that determine barrier enhancement or preservation are far from completion. Recently, epigenetic mechanisms have emerged as a major regulator of enduring changes in cell behavior and the therapeutic potential of inhibiting histone deacetylases (HDACs) for the treatment of cardiovascular and inflammatory diseases has gained remarkable attention. Although HDACs have been shown to play an important role in regulating EC barrier function, the involved HDAC subtypes and mechanisms remain undefined. Further investigation of the HDAC signaling may provide therapeutic approaches for the prevention and treatment of ALI.
Collapse
Affiliation(s)
- Laszlo Kovacs
- Department of Pharmacology & Toxicology, Augusta University, Augusta, GA 30912
| | | | - Alexander D Verin
- Vascular Biology Center, Augusta University, Augusta, GA 30912.,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - David Fulton
- Department of Pharmacology & Toxicology, Augusta University, Augusta, GA 30912.,Vascular Biology Center, Augusta University, Augusta, GA 30912
| | - Rudolf Lucas
- Department of Pharmacology & Toxicology, Augusta University, Augusta, GA 30912.,Vascular Biology Center, Augusta University, Augusta, GA 30912.,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Yunchao Su
- Department of Pharmacology & Toxicology, Augusta University, Augusta, GA 30912.,Vascular Biology Center, Augusta University, Augusta, GA 30912.,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912.,Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia 30912
| |
Collapse
|
33
|
|
34
|
Gonçalves P, Araújo JR, Di Santo JP. A Cross-Talk Between Microbiota-Derived Short-Chain Fatty Acids and the Host Mucosal Immune System Regulates Intestinal Homeostasis and Inflammatory Bowel Disease. Inflamm Bowel Dis 2018; 24:558-572. [PMID: 29462379 DOI: 10.1093/ibd/izx029] [Citation(s) in RCA: 277] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Indexed: 12/22/2022]
Abstract
Gut microbiota has a fundamental role in the energy homeostasis of the host and is essential for proper "education" of the immune system. Intestinal microbial communities are able to ferment dietary fiber releasing short-chain fatty acids (SCFAs). The SCFAs, particularly butyrate (BT), regulate innate and adaptive immune cell generation, trafficing, and function. For example, BT has an anti-inflammatory effect by inhibiting the recruitment and proinflammatory activity of neutrophils, macrophages, dendritic cells, and effector T cells and by increasing the number and activity of regulatory T cells. Gut microbial dysbiosis, ie, a microbial community imbalance, has been suggested to play a role in the development of inflammatory bowel disease (IBD). The relationship between dysbiosis and IBD has been difficult to prove, especially in humans, and is probably complex and dynamic, rather than one of a simple cause and effect relationship. However, IBD patients have dysbiosis with reduced numbers of SCFAs-producing bacteria and reduced BT concentration that is linked to a marked increase in the number of proinflammatory immune cells in the gut mucosa of these patients. Thus, microbial dysbiosis and reduced BT concentration may be a factor in the emergence and severity of IBD. Understanding the relationship between microbial dysbiosis and reduced BT concentration to IBD may lead to novel therapeutic interventions.
Collapse
Affiliation(s)
- Pedro Gonçalves
- Innate Immunity Unit, Institut Pasteur, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U1223, Paris, France
| | - João Ricardo Araújo
- Molecular Microbial Pathogenesis Unit, Institut Pasteur, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM) U1202, Paris, France
| | - James P Di Santo
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1223, Paris, France
| |
Collapse
|
35
|
Lu Q, Gottlieb E, Rounds S. Effects of cigarette smoke on pulmonary endothelial cells. Am J Physiol Lung Cell Mol Physiol 2018; 314:L743-L756. [PMID: 29351435 DOI: 10.1152/ajplung.00373.2017] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cigarette smoking is the leading cause of preventable disease and death in the United States. Cardiovascular comorbidities associated with both active and secondhand cigarette smoking indicate the vascular toxicity of smoke exposure. Growing evidence supports the injurious effect of cigarette smoke on pulmonary endothelial cells and the roles of endothelial cell injury in development of acute respiratory distress syndrome (ARDS), emphysema, and pulmonary hypertension. This review summarizes results from studies of humans, preclinical animal models, and cultured endothelial cells that document toxicities of cigarette smoke exposure on pulmonary endothelial cell functions, including barrier dysfunction, endothelial activation and inflammation, apoptosis, and vasoactive mediator production. The discussion is focused on effects of cigarette smoke-induced endothelial injury in the development of ARDS, emphysema, and vascular remodeling in chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Qing Lu
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center , Providence, Rhode Island.,Department of Medicine, Alpert Medical School of Brown University , Providence, Rhode Island
| | - Eric Gottlieb
- Department of Medicine, Alpert Medical School of Brown University , Providence, Rhode Island
| | - Sharon Rounds
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center , Providence, Rhode Island.,Department of Medicine, Alpert Medical School of Brown University , Providence, Rhode Island
| |
Collapse
|
36
|
Kasotakis G, Galvan MD, Osathanugrah P, Dharia N, Bufe L, Breed Z, Mizgerd JP, Remick DG. Timing of valproic acid in acute lung injury: prevention is the best therapy? J Surg Res 2017; 220:206-212. [PMID: 29180183 DOI: 10.1016/j.jss.2017.06.088] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/09/2017] [Accepted: 06/29/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND Acute lung injury and respiratory distress syndrome is characterized by uncontrolled inflammation of the lungs after a severe inflammatory stimulus. We have previously demonstrated an ameliorated syndrome and improved survival in mice with early administration of valproic acid (VPA), a broad-spectrum histone deacetylase inhibitor, while studies in humans have shown no benefit when anti-inflammatories are administered late. The current study tested the hypothesis that early treatment would improve outcomes in our gram-negative pneumonia-induced acute lung injury. MATERIALS AND METHODS Mice (C57BL/6) had 50 × 106 Escherichia coli (strain 19,138) instilled endotracheally and VPA (250 mg/kg) administered intraperitoneally 3, 4, 6, and 9 h (n = 12/group) later. Six hours after VPA administration, the animals were sacrificed, and bronchoalveolar lavage (BAL) fluid interleukin-6 (IL-6), tumor necrosis factor, neutrophils and macrophages as well as the E coli colony-forming units were quantified. Plasma IL-6 was also measured. A separate group of mice (n = 12/group) were followed prospectively for 7 days to assess survival. RESULTS BAL IL-6 and tumor necrosis factor as well as plasma IL-6 were significantly lower in the animals administered VPA within 3 h (P < 0.05) but not when administered later (4, 6, 9 h). There was no difference in the BAL E coli colony-forming units, macrophage, or neutrophil numbers at any time point. Survival improved only when VPA was administered within 3 h. CONCLUSIONS A narrow therapeutic window exists in this murine model of gram-negative pneumonia-induced acute lung injury and likely explains the lack of response in studies with late administration of anti-inflammatory therapies in clinical studies.
Collapse
Affiliation(s)
- George Kasotakis
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts.
| | - Manuel D Galvan
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Paw Osathanugrah
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Neerav Dharia
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Lauren Bufe
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Zachary Breed
- Department of Surgery, Boston University School of Medicine, Boston, Massachusetts
| | - Joseph P Mizgerd
- Departments of Medicine, Microbiology and Biochemistry, Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Daniel G Remick
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
37
|
Valproic acid mitigates the inflammatory response and prevents acute respiratory distress syndrome in a murine model of Escherichia coli pneumonia at the expense of bacterial clearance. J Trauma Acute Care Surg 2017; 82:758-765. [PMID: 28099388 DOI: 10.1097/ta.0000000000001389] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Histone deacetylase inhibitors (HDACI) are members of a family of epigenetic modifying agents with broad anti-inflammatory properties. These anti-inflammatory properties may have important therapeutic implications in acute respiratory distress syndrome (ARDS). However, administration of HDACI may create an immunosuppressive environment conducive to bacterial growth. Accordingly, the aim of the current study is to investigate the effect of HDACI valproic acid (VPA) on host inflammatory response and bacterial burden in a murine model of Escherichia coli pneumonia-induced ARDS. METHODS ARDS was induced in male C57BL6 mice (n = 24) by endotracheal instillation of 3 × 10 E. coli. VPA (250 mg/kg) was administered 30 minutes after E. coli instillation in the intervention group. Blood samples were collected at 3 and 6 hours, and animals were sacrificed at 6 hours. Bronchoalveolar lavage (BAL) was performed, and tissue specimens were harvested. Cytokine levels were measured in blood and BAL, and so was transalveolar protein transit. Cell counts and colony forming units were quantified in BAL fluid. RESULTS VPA reduced neutrophil influx into the lungs and local tissue destruction through decreased myeloperoxidase activity. It also ameliorated the pulmonary and systemic inflammatory response. This led to greater bacterial proliferation in the pulmonary parenchyma. CONCLUSION Administration of VPA in a clinically relevant bacterial model of murine ARDS mitigates the host inflammatory response, essentially preventing ARDS, but creates an immunosuppressive environment that favors bacterial overgrowth.
Collapse
|
38
|
Borgas D, Chambers E, Newton J, Ko J, Rivera S, Rounds S, Lu Q. Cigarette Smoke Disrupted Lung Endothelial Barrier Integrity and Increased Susceptibility to Acute Lung Injury via Histone Deacetylase 6. Am J Respir Cell Mol Biol 2017; 54:683-96. [PMID: 26452072 DOI: 10.1165/rcmb.2015-0149oc] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Epidemiologic evidence indicates that cigarette smoke (CS) is associated with the development of acute lung injury (ALI). We have previously shown that brief CS exposure exacerbates lipopolysaccharide (LPS)-induced ALI in vivo and endothelial barrier dysfunction in vitro. In this study, we found that CS also exacerbated Pseudomonas-induced ALI in mice. We demonstrated that lung microvascular endothelial cells (ECs) isolated from mice exposed to CS had a greater permeability or incomplete recovery after challenges by LPS and thrombin. Histone deacetylase (HDAC) 6 deacetylates proteins essential for maintenance of endothelial barrier function. We found that HDAC6 phosphorylation at serine-22 was increased in lung tissues of mice exposed to CS and in lung ECs exposed to cigarette smoke extract (CSE). Inhibition of HDAC6 attenuated CSE-induced increases in EC permeability and CS priming of ALI. Similar barrier protection was provided by the microtubule stabilizer taxol, which preserved α-tubulin acetylation. CSE decreased α-tubulin acetylation and caused microtubule depolymerization. In coordination with increased HDAC6 phosphorylation, CSE inhibited Akt and activated glycogen synthase kinase (GSK)-3β; these effects were ameliorated by the antioxidant N-acetyl cysteine. Our results suggest that CS increases lung EC permeability, thereby enhancing susceptibility to ALI, likely through oxidative stress-induced Akt inactivation and subsequent GSK-3β activation. Activated GSK-3β may activate HDAC6 via phosphorylation of serine-22, leading to α-tubulin deacetylation and microtubule disassembly. Inhibition of HDAC6 may be a novel therapeutic option for ALI in cigarette smokers.
Collapse
Affiliation(s)
- Diana Borgas
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Eboni Chambers
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Julie Newton
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Junsuk Ko
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Stephanie Rivera
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Sharon Rounds
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Qing Lu
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
39
|
Xie M, Chen HH, Nie SP, Yin JY, Xie MY. Gamma-Aminobutyric Acid Increases the Production of Short-Chain Fatty Acids and Decreases pH Values in Mouse Colon. Molecules 2017; 22:molecules22040653. [PMID: 28425937 PMCID: PMC6154700 DOI: 10.3390/molecules22040653] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 04/12/2017] [Accepted: 04/14/2017] [Indexed: 02/06/2023] Open
Abstract
Gamma-Aminobutyric acid (GABA) could regulate physiological functions in the gastrointestinal tract. The present study aimed to investigate the effect of GABA on colon health in mice. The female Kunming mice were given GABA at doses of 5, 10, 20 and 40 mg/kg/d for 14 days. Afterwards, the short-chain fatty acids (SCFAs) concentrations, pH values, colon index, colon length and weight of colonic and cecal contents were determined to evaluate the effects of GABA on colon health. The results showed that intake of GABA could increase the concentrations of acetate, propionate, butyrate and total SCFAs in colonic and cecal contents, as well as the weight of colonic and cecal contents. The colon index and length of the 40 mg/kg/d GABA-treated group were significantly higher than those of the control group (p < 0.05). In addition, decrease of pH values in colonic and cecal contents was also observed. These results suggest that GABA may improve colon health.
Collapse
Affiliation(s)
- Min Xie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China.
| | - Hai-Hong Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China.
| | - Shao-Ping Nie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China.
| | - Jun-Yi Yin
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China.
| | - Ming-Yong Xie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
40
|
Impact of the microbial derived short chain fatty acid propionate on host susceptibility to bacterial and fungal infections in vivo. Sci Rep 2016; 6:37944. [PMID: 27897220 PMCID: PMC5126587 DOI: 10.1038/srep37944] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 11/02/2016] [Indexed: 12/21/2022] Open
Abstract
Short chain fatty acids (SCFAs) produced by intestinal microbes mediate anti-inflammatory effects, but whether they impact on antimicrobial host defenses remains largely unknown. This is of particular concern in light of the attractiveness of developing SCFA-mediated therapies and considering that SCFAs work as inhibitors of histone deacetylases which are known to interfere with host defenses. Here we show that propionate, one of the main SCFAs, dampens the response of innate immune cells to microbial stimulation, inhibiting cytokine and NO production by mouse or human monocytes/macrophages, splenocytes, whole blood and, less efficiently, dendritic cells. In proof of concept studies, propionate neither improved nor worsened morbidity and mortality parameters in models of endotoxemia and infections induced by gram-negative bacteria (Escherichia coli, Klebsiella pneumoniae), gram-positive bacteria (Staphylococcus aureus, Streptococcus pneumoniae) and Candida albicans. Moreover, propionate did not impair the efficacy of passive immunization and natural immunization. Therefore, propionate has no significant impact on host susceptibility to infections and the establishment of protective anti-bacterial responses. These data support the safety of propionate-based therapies, either via direct supplementation or via the diet/microbiota, to treat non-infectious inflammation-related disorders, without increasing the risk of infection.
Collapse
|
41
|
Obi AT, Andraska E, Kanthi Y, Luke CE, Elfline M, Madathilparambil S, Siahaan TJ, Jaffer FA, Wakefield TW, Raghavendran K, Henke PK. Gram-Negative Pneumonia Alters Large-Vein Cell-Adhesion Molecule Profile and Potentiates Experimental Stasis Venous Thrombosis. J Vasc Res 2016; 53:186-195. [PMID: 27771726 DOI: 10.1159/000447299] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 05/28/2016] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND/AIMS Pneumonia is a significant risk factor for the development of venous thrombosis (VT). Cell-adhesion molecules (CAMs) are linked to the pathogenesis of both pneumonia and VT. We hypothesized that remote infection would confer a prothrombogenic milieu via systemic elevation of CAMs. METHODS Lung injury was induced in wild-type (C57BL/6) mice by lung contusion or intratracheal inoculation with Klebsiella pneumoniae or saline controls. K. pneumoniae-treated mice and controls additionally underwent inferior vena cava (IVC) ligation to generate VT. RESULTS Lung-contusion mice demonstrated no increase in E-selectin or P-selectin whereas mice infected with K. pneumoniae demonstrated increased circulating P-selectin, ICAM-1, VCAM-1 and thrombin-antithrombin (TAT) complexes. Mice with pneumonia formed VT 3 times larger than controls, demonstrated significantly more upregulation of vein-wall and systemic CAMs, and formed erythrocyte-rich thrombi. CONCLUSION Elevated CAM expression was identified in mice with pneumonia, but not lung contusion, indicating that the type of inflammatory stimulus and the presence of infection drive the vein-wall response. Elevation of CAMs was associated with amplified VT and may represent an alternate mechanism by which to target the prevention of VT.
Collapse
Affiliation(s)
- Andrea T Obi
- Conrad Jobst Vascular Research Laboratory, University of Michigan Medical School, Ann Arbor, Mich., USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
Patel U, Rajasingh S, Samanta S, Cao T, Dawn B, Rajasingh J. Macrophage polarization in response to epigenetic modifiers during infection and inflammation. Drug Discov Today 2016; 22:186-193. [PMID: 27554801 DOI: 10.1016/j.drudis.2016.08.006] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/14/2016] [Accepted: 08/12/2016] [Indexed: 12/17/2022]
Abstract
Macrophages are a heterogeneous population of phagocytic cells present in all tissues. Recently, several drugs that target the epigenetic machinery have emerged as attractive molecules for treating infection and inflammation by modulating macrophages. Treatment of lipopolysaccharide (LPS)-challenged macrophages with epigenetic modifiers leads to phenotype switching. This could provide stimulatory/destructive (M1) or suppressive/protective (M2) therapeutic strategies, which are crucial in the cytokine milieu in which the macrophages reside. In this review, we provide an overview of macrophage functional diversity during various diseases, including infection, as well as the current status in the development and clinical utility of epigenetic modifiers.
Collapse
Affiliation(s)
- Urmi Patel
- Department of Internal Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sheeja Rajasingh
- Department of Internal Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Saheli Samanta
- Department of Internal Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Thuy Cao
- Department of Internal Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Buddhadeb Dawn
- Department of Internal Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Johnson Rajasingh
- Department of Internal Medicine, Cardiovascular Research Institute, University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
44
|
Spence S, Greene MK, Fay F, Hams E, Saunders SP, Hamid U, Fitzgerald M, Beck J, Bains BK, Smyth P, Themistou E, Small DM, Schmid D, O'Kane CM, Fitzgerald DC, Abdelghany SM, Johnston JA, Fallon PG, Burrows JF, McAuley DF, Kissenpfennig A, Scott CJ. Targeting Siglecs with a sialic acid-decorated nanoparticle abrogates inflammation. Sci Transl Med 2016; 7:303ra140. [PMID: 26333936 DOI: 10.1126/scitranslmed.aab3459] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sepsis is the most frequent cause of death in hospitalized patients, and severe sepsis is a leading contributory factor to acute respiratory distress syndrome (ARDS). At present, there is no effective treatment for these conditions, and care is primarily supportive. Murine sialic acid-binding immunoglobulin-like lectin-E (Siglec-E) and its human orthologs Siglec-7 and Siglec-9 are immunomodulatory receptors found predominantly on hematopoietic cells. These receptors are important negative regulators of acute inflammatory responses and are potential targets for the treatment of sepsis and ARDS. We describe a Siglec-targeting platform consisting of poly(lactic-co-glycolic acid) nanoparticles decorated with a natural Siglec ligand, di(α2→8) N-acetylneuraminic acid (α2,8 NANA-NP). This nanoparticle induced enhanced oligomerization of the murine Siglec-E receptor on the surface of macrophages, unlike the free α2,8 NANA ligand. Furthermore, treatment of murine macrophages with these nanoparticles blocked the production of lipopolysaccharide-induced inflammatory cytokines in a Siglec-E-dependent manner. The nanoparticles were also therapeutically beneficial in vivo in both systemic and pulmonary murine models replicating inflammatory features of sepsis and ARDS. Moreover, we confirmed the anti-inflammatory effect of these nanoparticles on human monocytes and macrophages in vitro and in a human ex vivo lung perfusion (EVLP) model of lung injury. We also established that interleukin-10 (IL-10) induced Siglec-E expression and α2,8 NANA-NP further augmented the expression of IL-10. Indeed, the effectiveness of the nanoparticle depended on IL-10. Collectively, these results demonstrated a therapeutic effect of targeting Siglec receptors with a nanoparticle-based platform under inflammatory conditions.
Collapse
Affiliation(s)
- Shaun Spence
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | | | - François Fay
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK. Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Emily Hams
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Sean P Saunders
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Umar Hamid
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Marianne Fitzgerald
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Jonathan Beck
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | | | - Peter Smyth
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Efrosyni Themistou
- School of Chemistry and Chemical Engineering, Queen's University Belfast, Belfast BT9 5AG, UK
| | - Donna M Small
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Daniela Schmid
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Cecilia M O'Kane
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Denise C Fitzgerald
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Sharif M Abdelghany
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK. Faculty of Pharmacy, University of Jordan, Amman 11942, Jordan
| | - James A Johnston
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK. Inflammation Research, Amgen Inc., Thousand Oaks, CA 91320, USA
| | - Padraic G Fallon
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin 2, Ireland. National Children's Research Centre, Our Lady's Children's Hospital, Dublin 12, Ireland
| | - James F Burrows
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Daniel F McAuley
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Adrien Kissenpfennig
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7AE, UK
| | | |
Collapse
|
45
|
Kim SJ, Park JS, Lee DW, Lee SM. Trichostatin A Protects Liver against Septic Injury through Inhibiting Toll-Like Receptor Signaling. Biomol Ther (Seoul) 2016; 24:387-94. [PMID: 27068262 PMCID: PMC4930282 DOI: 10.4062/biomolther.2015.176] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 11/26/2015] [Accepted: 12/02/2015] [Indexed: 12/04/2022] Open
Abstract
Sepsis, a serious clinical problem, is characterized by a systemic inflammatory response to infection and leads to organ failure. Toll-like receptor (TLR) signaling is intimately implicated in hyper-inflammatory responses and tissue injury during sepsis. Histone deacetylase (HDAC) inhibitors have been reported to exhibit anti-inflammatory properties. The aim of this study was to investigate the hepatoprotective mechanisms of trichostatin A (TSA), a HDAC inhibitor, associated with TLR signaling pathway during sepsis. The anti-inflammatory properties of TSA were assayed in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. Polymicrobial sepsis was induced in mice by cecal ligation and puncture (CLP), a clinically relevant model of sepsis. The mice were intraperitoneally received TSA (1, 2 or 5 mg/kg) 30 min before CLP. The serum and liver samples were collected 6 and 24-h after CLP. TSA inhibited the increased production of tumor necrosis factor (TNF)-α and interleukin (IL)-6 in LPS-stimulated RAW264.7 cells. TSA improved sepsis-induced mortality, attenuated liver injury and decreased serum TNF-α and IL-6 levels. CLP increased the levels of TLR4, TLR2 and myeloid differentiation primary response protein 88 (MyD88) protein expression and association of MyD88 with TLR4 and TLR2, which were attenuated by TSA. CLP increased nuclear translocation of nuclear factor kappa B and decreased cytosolic inhibitor of kappa B (IκB) protein expression, which were attenuated by TSA. Moreover, CLP decreased acetylation of IκB kinase (IKK) and increased association of IKK with IκB and TSA attenuated these alterations. Our findings suggest that TSA attenuates liver injury by inhibiting TLR-mediated inflammatory response during sepsis.
Collapse
Affiliation(s)
- So-Jin Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jin-Sook Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Do-Won Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sun-Mee Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
46
|
Kim SJ, Baek KS, Park HJ, Jung YH, Lee SM. Compound 9a, a novel synthetic histone deacetylase inhibitor, protects against septic injury in mice by suppressing MAPK signalling. Br J Pharmacol 2016; 173:1045-57. [PMID: 26689981 DOI: 10.1111/bph.13414] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 12/07/2015] [Accepted: 12/10/2015] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND AND PURPOSE Sepsis is a life-threatening clinical condition characterized by uncontrolled inflammatory responses and is a major cause of death in intensive care units. Histone deacetylase (HDAC) inhibitors have recently exhibited anti-inflammatory properties. MAPK phosphatase (MKP) suppresses MAPK signalling, which plays an important role in inflammatory responses. The purpose of this study was to investigate the protective mechanisms of Compound 9a, a newly synthetized HDAC inhibitor, against septic injury. EXPERIMENTAL APPROACH The anti-inflammatory properties of Compound 9a were assayed in LPS-stimulated RAW264.7 cells. In vivo, polymicrobial sepsis was induced in C57BL/6 mice by caecal ligation and puncture (CLP). The mice were treated with Compound 9a (i.p., 10 mg∙kg(-1) ) 2 h before and immediately after CLP. KEY RESULTS Compound 9a inhibited the increased production of TNF-α, IL-6 and NO in LPS-stimulated RAW264.7 cells. In mice with CLP, Compound 9a improved survival rate, attenuated organ injuries and decreased serum TNF-α and IL-6 levels. CLP increased expression of toll-like receptor 4, phosphorylated (p)-p38, p-JNK and p-ERK proteins, which was attenuated by Compound 9a. Compound 9a decreased MKP-1 association with HDAC1 and enhanced MKP-1 acetylation and enhanced MKP-1 association with p-p38 and p-ERK. Moreover, the inhibitory effects of Compound 9a on serum cytokine levels and phosphorylation of MAPK were abolished by MKP-1 siRNA. CONCLUSIONS AND IMPLICATIONS Our findings suggest that Compound 9a protected against septic injury by suppressing MAPK-mediated inflammatory signalling.
Collapse
Affiliation(s)
- So-Jin Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, Korea
| | - Ki Seon Baek
- School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, Korea
| | - Hyun-Ju Park
- School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, Korea
| | - Young Hoon Jung
- School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, Korea
| | - Sun-Mee Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, Korea
| |
Collapse
|
47
|
Qiao Y, Qian J, Lu Q, Tian Y, Chen Q, Zhang Y. Protective effects of butyrate on intestinal ischemia–reperfusion injury in rats. J Surg Res 2015; 197:324-30. [DOI: 10.1016/j.jss.2015.04.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/22/2015] [Accepted: 04/09/2015] [Indexed: 12/24/2022]
|
48
|
Thangavel J, Samanta S, Rajasingh S, Barani B, Xuan YT, Dawn B, Rajasingh J. Epigenetic modifiers reduce inflammation and modulate macrophage phenotype during endotoxemia-induced acute lung injury. J Cell Sci 2015; 128:3094-105. [PMID: 26116574 DOI: 10.1242/jcs.170258] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 06/16/2015] [Indexed: 12/27/2022] Open
Abstract
Acute lung injury (ALI) during sepsis is characterized by bilateral alveolar infiltrates, lung edema and respiratory failure. Here, we examined the efficacy the DNA methyl transferase (DNMT) inhibitor 5-Aza 2-deoxycytidine (Aza), the histone deacetylase (HDAC) inhibitor Trichostatin A (TSA), as well as the combination therapy of Aza and TSA (Aza+TSA) provides in the protection of ALI. In LPS-induced mouse ALI, post-treatment with a single dose of Aza+TSA showed substantial attenuation of adverse lung histopathological changes and inflammation. Importantly, these protective effects were due to substantial macrophage phenotypic changes observed in LPS-stimulated macrophages treated with Aza+TSA as compared with untreated LPS-induced macrophages or LPS-stimulated macrophages treated with either drug alone. Further, we observed significantly lower levels of pro-inflammatory molecules and higher levels of anti-inflammatory molecules in LPS-induced macrophages treated with Aza+TSA than in LPS-induced macrophages treated with either drug alone. The protection was ascribed to dual effects by an inhibition of MAPK-HuR-TNF and activation of STAT3-Bcl2 pathways. Combinatorial treatment with Aza+TSA reduces inflammation and promotes an anti-inflammatory M2 macrophage phenotype in ALI, and has a therapeutic potential for patients with sepsis.
Collapse
Affiliation(s)
- Jayakumar Thangavel
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Saheli Samanta
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sheeja Rajasingh
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Bahar Barani
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yu-Ting Xuan
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Buddhadeb Dawn
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Johnson Rajasingh
- Cardiovascular Research Institute, Division of Cardiovascular Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
49
|
Hartman H, Wetterholm E, Thorlacius H, Regnér S. Histone deacetylase regulates trypsin activation, inflammation, and tissue damage in acute pancreatitis in mice. Dig Dis Sci 2015; 60:1284-9. [PMID: 25492506 DOI: 10.1007/s10620-014-3474-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 11/28/2014] [Indexed: 01/24/2023]
Abstract
BACKGROUND The onset of acute pancreatitis (AP) is characterized by early protease activation followed by inflammation and organ damage, but the mechanisms are poorly understood. AIMS We hypothesized that histone deacetylase (HDAC) inhibition might exert protective effects on AP and investigated the role of HDAC in trypsin activation, inflammation, and tissue damage in severe AP. METHODS Male C57Bl/6 mice were treated i.p. with the HDAC inhibitor trichostatin A (2 mg/kg) prior to retrograde infusion of taurocholic acid (5 %) into the pancreatic duct. Serum levels of amylase and interleukin (IL)-6, pancreatic levels of macrophage inflammatory protein-2 (MIP-2) as well as tissue morphology and myeloperoxidase activity in the pancreas and lung were determined 24 h after taurocholate challenge. Trypsin activation was analyzed in isolated acinar cells. Quantitative RT-PCR was used to examine the expression of pro-inflammatory mediators in the pancreas. RESULTS Pretreatment with trichostatin A decreased amylase levels by 70 % and protected against tissue injury in the pancreas. Moreover, HDAC inhibition reduced systemic IL-6 by more than 95 % and pulmonary myeloperoxidase activity by 75 %. Notably, inhibition of HDAC abolished taurocholate-induced gene expression of cyclooxygenase-2, MIP-2, monocyte chemotactic protein-1, IL-6, and IL-1β in the pancreas. In addition, HDAC inhibition reduced cerulein-induced trypsinogen activation in isolated acinar cells. CONCLUSION Our findings show that HDAC regulates trypsin activation, inflammation, and tissue damage in AP. Thus, targeting HDAC could serve as novel therapeutic approach in the management of severe AP.
Collapse
Affiliation(s)
- Hannes Hartman
- Department of Clinical Sciences Malmö, Section of Surgery, Skåne University Hospital, Lund University, Inga-Marie Nilssons Gata 46, 205 02, Malmö, Sweden,
| | | | | | | |
Collapse
|
50
|
Protection against reperfusion lung injury via aborgating multiple signaling cascades by trichostatin A. Int Immunopharmacol 2015; 25:267-75. [PMID: 25698558 DOI: 10.1016/j.intimp.2015.02.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/08/2015] [Accepted: 02/09/2015] [Indexed: 12/20/2022]
Abstract
Trichostatin A (TSA) is a histone deacetylase inhibitor with anti-inflammatory effects. Nonetheless, little information is available about the effect of TSA in ischemia-reperfusion (IR)-induced lung injury. In a perfused rat lung model, IR was induced by 40min of ischemia followed by 60min of reperfusion. The rat lungs were randomly divided into several groups including control, control+TSA (0.1mg/kg), IR, and IR+various dosages of TSA (0.05, 0.075, 0.1mg/kg). Bronchoalveolar lavage fluids and lung tissues were obtained and examined at the end of the experiment. TSA dose-dependently diminished IR-induced increased vascular permeability and edema, pulmonary artery pressure, and histological changes in the lungs. Additionally, TSA suppressed lavage tumor necrosis factor-α and cytokine-induced neutrophil chemoattractant concentrations, cell infiltration, and myeloperoxidase-positive cells in the lung tissue. Furthermore, TSA attenuated the phosphorylation of extracellular signal-regulated kinase, p38, and c-Jun N-terminal kinase, degradation of the inhibitor of nuclear factor (NF)-κB, and nuclear NF-κB levels. TSA also decreased poly (ADP-ribose) polymerase but enhanced acetylated histone H3 acetylation, Bcl-2, and mitogen-activated protein kinase phosphatase-1 (MKP-1) expression in IR lung tissue. Therefore, TSA exerted a protective effect on IR-induced lung injury via increasing histone acetylation and MKP-1 protein expression, repressing NF-κB, mitogen-activated protein kinase, and apoptosis signaling pathways.
Collapse
|