1
|
Fukuda A, Mizuno T, Yoshida T, Sunami K, Kubo T, Koyama T, Yonemori K, Okusaka T, Kato K, Ohe Y, Yatabe Y, Yamamoto N. Upfront liquid biopsy in patients with advanced solid tumors who were not feasible for tissue-based next-generation sequencing. Jpn J Clin Oncol 2025:hyaf065. [PMID: 40251768 DOI: 10.1093/jjco/hyaf065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 02/03/2025] [Accepted: 04/09/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Liquid biopsy has been developed as an alternative to tissue-based sequencing for detecting genomic alterations in solid tumors. However, the clinical utility of liquid biopsy in patients with solid tumors for whom tissue-based next-generation sequencing (NGS) is infeasible has not been well-characterized, particularly in previously untreated individuals. METHODS This prospective study evaluated the clinical impact of liquid biopsy, focusing on six solid tumor types. Overall, 109 patients were enrolled and underwent liquid biopsy using Guardant360 (Guardant Health, Redwood City, CA, USA). Among these, 94 (86.3%) patients were previously untreated. RESULTS The most common cancer type was non-small cell lung cancer (n = 57, 52.3%), followed by pancreatic (n = 35, 32.1%), biliary tract (n = 8, 7.3%), gastric (n = 5, 4.6%), colorectal (n = 3, 2.8%), and triple-negative breast (n = 1, 0.9%) cancers. The success rate of liquid biopsy was 99.1%, and the median turnaround time from blood collection to results was 7 days (range: 5-22 days). Actionable alterations were detected in 31 (28.4%) patients, and 8.3% of them received matched therapy based on alterations identified by liquid biopsy. Among previously untreated patients, actionable mutations were identified in 29.8%, and 8.5% received matched therapy. CONCLUSIONS In patients with advanced solid tumors for which tissue-based NGS is not feasible, performing upfront liquid biopsy could lead to the detection of actionable alterations and help guide targeted therapies. CLINICAL TRIAL REGISTRY UMIN Clinical Trials Registry (UMIN000041722).
Collapse
Affiliation(s)
- Akito Fukuda
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Takaaki Mizuno
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Kuniko Sunami
- Department of Laboratory Medicine, National Cancer Center Hospital, Tokyo, Japan
| | - Takashi Kubo
- Department of Laboratory Medicine, National Cancer Center Hospital, Tokyo, Japan
| | - Takafumi Koyama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Kan Yonemori
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ken Kato
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Noboru Yamamoto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
2
|
Xu M, Ren M, Zhang X, Peng W, Li H, Liao W, Xie J, Zhang X. Mesenchymal stem cell-derived small extracellular vesicles restored nasal barrier function in allergic rhinitis via miR-143-GSK3B in human nasal epithelial cells. J Allergy Clin Immunol 2025; 155:1236-1249.e5. [PMID: 39542141 DOI: 10.1016/j.jaci.2024.10.034] [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: 12/04/2023] [Revised: 08/05/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND The nasal epithelial barrier is the first line of defense against the deep entry of pathogens or aeroallergens and is more critical in allergic rhinitis (AR). Restoring epithelial barrier dysfunction might be a promising strategy for AR. Recent studies reported that mesenchymal stem cell-derived small extracellular vesicles (MSC-sEV) potentially inhibit the inflammation response and promote tissue regeneration. However, their effect on nasal epithelial cells remains unknown. OBJECTIVES This study sought to describe the therapeutic effect of MSC-sEV on AR, particularly focusing their effect on nasal epithelial cells and underlying molecular mechanisms. METHODS We utilized an ovalbumin-induced mouse model to study AR. Both primary and immortalized human nasal epithelial cells (HNEpC) were used to further validate the therapeutic effects of MSC-sEV on epithelial cell function. Then we constructed microRNA (miR)-143 overexpressing and low-expressing HNEpC and MSC-sEV to elucidate molecular mechanisms. Transcriptome analysis was performed to identify the downstream pathways involved. RESULTS MSC-sEV successfully maintained nasal barrier integrity in AR mouse model. The MSC-sEV therapeutic effect on the nasal barrier was substantiated in HNEpC. Mechanistically, miR-143 was a candidate mediator of the above effects. Subsequently, transfecting HNEpC with miR-143 partially mimicked the restoring effect of MSC-sEV. MSC-sEV overexpressing miR-143 exerted more therapeutic effects on tight junctions and barrier integrity. Moreover, miR-143 regulated the glycogen synthase kinase-3β (GSK3B) pathway. CONCLUSIONS Our results indicated that MSC-sEV mitigated AR and restored nasal epithelial barrier dysfunction through the miR-143-GSK3B axis, which suggested that MSC-sEV have the remarkable ability to treat AR.
Collapse
Affiliation(s)
- Meiqian Xu
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China
| | - Mei Ren
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China
| | - Xinyin Zhang
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China
| | - Wenxu Peng
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China
| | - Hao Li
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China
| | - Wenjing Liao
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China.
| | - Jianlei Xie
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China.
| | - Xiaowen Zhang
- The State Key Laboratory of Respiratory Disease, Department of Otolaryngology, Head and Neck Surgery, Laboratory of Otolaryngology-Head and Neck Surgery, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Innovation and Transformation Platform of Upper Airway Disease in Guangdong Province, Guangzhou, China.
| |
Collapse
|
3
|
Dabo AJ, Raghavan S, Ezegbunam W, Thankachen J, Evgrafov O, Majka S, Geraghty P, Foronjy RF. Cigarette smoke alters calcium flux to induce PP2A membrane trafficking and endothelial cell permeability. Sci Rep 2024; 14:28012. [PMID: 39543165 PMCID: PMC11564810 DOI: 10.1038/s41598-024-77776-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/25/2024] [Indexed: 11/17/2024] Open
Abstract
Alveolar capillary barrier disruption induces local edema and inflammation that impairs pulmonary function and promotes alveolar destruction in COPD. This study aimed to determine how cigarette smoke modulated the serine-threonine phosphatase protein phosphatase 2 A (PP2A) to alter the barrier function of human lung microvascular endothelial cells (HLMVECs). Cigarette smoke exposure lowered overall PP2A activity and enhanced endothelial permeability in HLMVECs. However, directly decreasing PP2A activity with Fostriecin significantly reduced endothelial cell permeability. Protein fractionation studies determined that cigarette smoke diminished cytosolic PP2A activity but increased membrane and cytoskeletal activity. These changes coincided with the translocation of PP2A to the membrane, which reduced occludin phosphorylation in the membrane. Cigarette smoke decreased protein tyrosine phosphatase 1B (PTP1B) activity, a PP2A activator which also counters calcium intracellular influx. The decrease in PTP1B activity correlated with reduced calcium efflux in endothelial cells and these changes in calcium flux regulated PP2A activity. Indeed, culturing endothelial cells in low calcium medium prevented the decrease in cytosolic PP2A activity mediated by cigarette smoke. Together, these findings outline a mechanism whereby cigarette smoke acts via calcium to traffic PP2A from the cytosol to the membrane where it dephosphorylates occludin to increase endothelial cell permeability.
Collapse
Affiliation(s)
- Abdoulaye J Dabo
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
| | - Sonya Raghavan
- Division of Pulmonary, and Critical Care Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wendy Ezegbunam
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
| | - Jincy Thankachen
- Division of Pulmonary, and Critical Care Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Oleg Evgrafov
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
| | - Sue Majka
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
| | - Patrick Geraghty
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
| | - Robert F Foronjy
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA.
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA.
- Division of Pulmonary & Critical Care Medicine, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY, 11203, USA.
| |
Collapse
|
4
|
Zhou H, Wu R, Li H. Downregulation of HDAC6 mitigates lung ischemia/reperfusion injury depending on activation of Nrf2/HO-1 signaling pathway and inactivation of ERK/NF-κB signaling pathway. Tissue Cell 2024; 89:102446. [PMID: 38936199 DOI: 10.1016/j.tice.2024.102446] [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: 12/01/2023] [Revised: 05/22/2024] [Accepted: 06/14/2024] [Indexed: 06/29/2024]
Abstract
INTRODUCTION Lung ischemia/reperfusion injury (LIRI) is a pathological process caused by the deficiency and subsequent reperfusion of oxygen and blood to the lung. Literature reports that the catalytic activity and expression of HDAC6 can be induced in response to IRI. HDAC6 inhibition confers protective effects against a series of IRI and also exerts pulmonary protection against various lung damage. The present study was formulated to investigate the functional role of HDAC6 inhibitor in LIRI and to probe into the intrinsic mechanisms underlying the protective role of HDAC6 inhibitor against LIRI. METHODS Lung epithelial cell line MLE-12 cells were subjected to H/R injury to construct in vitro cell culture model of LIRI. For functional experiments, MLE-12 cells were pre-treated with various concentrations of selective HDAC6 inhibitor ACY-1215 (1, 5, 10 μM) to evaluate the biological role of HDAC6 in LIRI. For rescue experiments, MLE-12 cells were pre-treated with Nrf2 inhibitor ML385 (10 μM) or ERK activator LM22B-10 (50 μM) to discuss the molecular mechanisms. RESULTS It was verified that HDAC6 inhibition repressed H/R-induced apoptosis, oxidative stress, inflammation and mitochondrial dysfunction of MLE-12 cells. HDAC6 inhibition activated Nrf2/HO-1 signaling pathway and inactivated ERK/NF-κB signaling pathway in MLE-12 cells. The repressing effects of HDAC6 inhibition on H/R-induced apoptosis, oxidative stress, inflammation and mitochondrial dysfunction of MLE-12 cells were partially abolished upon pre-treatment with Nrf2 inhibitor ML385 or ERK activator LM22B-10. CONCLUSION HDAC6 inhibition may mitigate H/R-induced lung epithelial cell injury depending on activation of Nrf2/HO-1 signaling pathway and inactivation of ERK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Hong Zhou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710065, PR China
| | - Rui Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710065, PR China
| | - Hong Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710065, PR China.
| |
Collapse
|
5
|
Li X, Saiyin H, Chen X, Yu Q, Ma L, Liang W. Ketamine impairs growth cone and synaptogenesis in human GABAergic projection neurons via GSK-3β and HDAC6 signaling. Mol Psychiatry 2024; 29:1647-1659. [PMID: 36414713 PMCID: PMC11371642 DOI: 10.1038/s41380-022-01864-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/23/2022]
Abstract
The growth cone guides the axon or dendrite of striatal GABAergic projection neurons that protrude into the midbrain and cortex and form complex neuronal circuits and synaptic networks in a developing brain, aberrant projections and synaptic connections in the striatum related to multiple brain disorders. Previously, we showed that ketamine, an anesthetic, reduced dendritic growth, dendritic branches, and spine density in human striatal GABAergic neurons. However, whether ketamine affects the growth cone, the synaptic connection of growing striatal GABAergic neurons has not been tested. Using human GABAergic projection neurons derived from human inducible pluripotent stem cells (hiPSCs) and embryonic stem cells (ES) in vitro, we tested ketamine effects on the growth cones and synapses in developing GABAergic neurons by assessing the morphometry and the glycogen synthase kinase-3 (GSK-3) and histone deacetylase 6 (HDAC6) pathway. Ketamine exposure impairs growth cone formation, synaptogenesis, dendritic development, and maturation via ketamine-mediated activation of GSK-3 pathways and inhibiting HDAC6, an essential stabilizing protein for dendritic morphogenesis and synapse maturation. Our findings identified a novel ketamine neurotoxic pathway that depends on GSK-3β and HDAC6 signaling, suggesting that microtubule acetylation is a potential target for reducing ketamine's toxic effect on GABAergic projection neuronal development.
Collapse
Affiliation(s)
- Xuan Li
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, China
- Department of Anesthesiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Hexige Saiyin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Xinyu Chen
- Department of Anatomy and Histology & Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qiong Yu
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Lixiang Ma
- Department of Anatomy and Histology & Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Weimin Liang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Balczon R, Lin MT, Voth S, Nelson AR, Schupp JC, Wagener BM, Pittet JF, Stevens T. Lung endothelium, tau, and amyloids in health and disease. Physiol Rev 2024; 104:533-587. [PMID: 37561137 PMCID: PMC11281824 DOI: 10.1152/physrev.00006.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/26/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023] Open
Abstract
Lung endothelia in the arteries, capillaries, and veins are heterogeneous in structure and function. Lung capillaries in particular represent a unique vascular niche, with a thin yet highly restrictive alveolar-capillary barrier that optimizes gas exchange. Capillary endothelium surveys the blood while simultaneously interpreting cues initiated within the alveolus and communicated via immediately adjacent type I and type II epithelial cells, fibroblasts, and pericytes. This cell-cell communication is necessary to coordinate the immune response to lower respiratory tract infection. Recent discoveries identify an important role for the microtubule-associated protein tau that is expressed in lung capillary endothelia in the host-pathogen interaction. This endothelial tau stabilizes microtubules necessary for barrier integrity, yet infection drives production of cytotoxic tau variants that are released into the airways and circulation, where they contribute to end-organ dysfunction. Similarly, beta-amyloid is produced during infection. Beta-amyloid has antimicrobial activity, but during infection it can acquire cytotoxic activity that is deleterious to the host. The production and function of these cytotoxic tau and amyloid variants are the subject of this review. Lung-derived cytotoxic tau and amyloid variants are a recently discovered mechanism of end-organ dysfunction, including neurocognitive dysfunction, during and in the aftermath of infection.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Mike T Lin
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Sarah Voth
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States
| | - Amy R Nelson
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Jonas C Schupp
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University, New Haven, Connecticut, United States
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Hannover, Germany
| | - Brant M Wagener
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| |
Collapse
|
7
|
Zhang Q, Yan L, Lu Y, Liu X, Yin Y, Wang Q, Gu X, Zhou X. HDAC6-selective inhibitor CAY10603 ameliorates cigarette smoke-induced small airway remodeling by regulating epithelial barrier dysfunction and reversing. Respir Res 2024; 25:66. [PMID: 38317159 PMCID: PMC10840206 DOI: 10.1186/s12931-024-02688-3] [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: 09/23/2023] [Accepted: 01/12/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Small airway remodelling is a vital characteristic of chronic obstructive pulmonary disease (COPD), which is mainly caused by epithelial barrier dysfunction and epithelial-mesenchymal transition (EMT). Recent studies have indicated that histone deacetylase 6 (HDAC6) plays an important role in the dysregulation of epithelial function. In this study, we investigated the therapeutic effects and underlying mechanisms of an inhibitor with high selectivity for HDAC6 in COPD. METHODS Cigarette smoke (CS) exposure was used to establish a CS-induced COPD mouse model. CAY10603 at doses of 2.5 and 10 mg/kg was injected intraperitoneally on alternate days. The protective effects of CAY10603 against CS-induced emphysema, epithelial barrier function and small airway remodeling were evaluated using hematoxylin and eosin (H&E) staining, Masson's trichrome staining, immunohistochemical staining, and western blot. The human lung bronchial epithelial cell line (HBE) was used to elucidate the underlying molecular mechanism of action of CAY10603. RESULTS HDAC6 levels in the lung homogenates of CS-exposed mice were higher than that those in control mice. Compared to the CS group, the mean linear intercept (MLI) of the CAY10603 treatment group decreased and the mean alveolar number (MAN)increased. Collagen deposition was reduced in groups treated with CAY10603. The expression of α-SMA was markedly upregulated in the CS group, which was reversed by CAY10603 treatment. Conversely, E-cadherin expression in the CS group was further downregulated, which was reversed by CAY10603 treatment. CAY10603 affects the tight junction protein expression of ZO-1 and occludin. ZO-1 and occludin expression were markedly downregulated in the CS group. After CAY10603treatment, the protein expression level of ZO-1 and occludin increased significantly. In HBE cells, Cigarette smoke extract (CSE) increased HDAC6 levels. CAY10603 significantly attenuated the release of TGF-β1 induced by CSE. CAY10603 significantly increased the E-cadherin levels in TGF-β1 treated HBE cells, while concurrently attenuated α-SMA expression. This effect was achieved through the suppression of Smad2 and Smad3 phosphorylation. CAY10603 also inhibited TGF-β1 induced cell migration. CONCLUSIONS These findings suggested that CAY10603 inhibited CS induced small airway remodelling by regulating epithelial barrier dysfunction and reversing EMT via the TGF-β1/Smad2/3 signalling pathway.
Collapse
Affiliation(s)
- Qin Zhang
- National Center for Respiratory Medicine, Shenyang, China
- State Key Laboratory of Respiratory Health and Multimorbidity, Shenyang, China
- National Clinical Research Center for Respiratory Diseases, Shenyang, China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Shenyang, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Liming Yan
- Department of Pulmonary and Critical Care Medicine, Fourth Hospital of China Medical University, Shenyang, China
| | - Ye Lu
- Department of Respiratory and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaodong Liu
- Department of Respiratory and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yan Yin
- Department of Respiratory and Critical Care Medicine, First Hospital of China Medical University, Shenyang, China
| | - Qiuyue Wang
- Department of Respiratory and Critical Care Medicine, First Hospital of China Medical University, Shenyang, China
| | - Xiu Gu
- Department of Pulmonary and Critical Care Medicine, Fourth Hospital of China Medical University, Shenyang, China
| | - Xiaoming Zhou
- Respiratory Department, Center for Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
8
|
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
|
9
|
Xue Y, Gan B, Zhou Y, Wang T, Zhu T, Peng X, Zhang X, Zhou Y. Advances in the Mechanistic Study of the Control of Oxidative Stress Injury by Modulating HDAC6 Activity. Cell Biochem Biophys 2023; 81:127-139. [PMID: 36749475 PMCID: PMC9925596 DOI: 10.1007/s12013-022-01125-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/14/2022] [Indexed: 02/08/2023]
Abstract
Oxidative stress is defined as an injury resulting from a disturbance in the dynamic equilibrium of the redox environment due to the overproduction of active/radical oxygen exceeding the antioxidative ability of the body. This is a key step in the development of various diseases. Oxidative stress is modulated by different factors and events, including the modification of histones, which are the cores of nucleosomes. Histone modification includes acetylation and deacetylation of certain amino acid residues; this process is catalyzed by different enzymes. Histone deacetylase 6 (HDAC6) is a unique deacetylating protease that also catalyzes the deacetylation of different nonhistone substrates to regulate various physiologic processes. The intimate relationship between HDAC6 and oxidative stress has been demonstrated by different studies. The present paper aims to summarize the data obtained from a mechanistic study of HDAC6 and oxidative stress to guide further investigations on mechanistic characterization and drug development.
Collapse
Affiliation(s)
- Yuanye Xue
- Department of Pathophysiology, Guangdong Medical University, Dongguan, 523808, China
| | - Bing Gan
- The Third Affiliated Hospital of Guangdong Medical University, Fo Shan, 528000, Guangdong, China
| | - Yanxing Zhou
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Tingyu Wang
- Department of Pathophysiology, Guangdong Medical University, Dongguan, 523808, China
| | - Tong Zhu
- Department of Pathophysiology, Guangdong Medical University, Dongguan, 523808, China
| | - Xinsheng Peng
- Biomedical Innovation Center, Guangdong Medical University, Dongguan, 523808, China.
- Institute of Marine Medicine, Guangdong Medical University, Zhanjiang, 524023, China.
| | - Xiangning Zhang
- Department of Pathophysiology, Guangdong Medical University, Dongguan, 523808, China.
| | - Yanfang Zhou
- Department of Pathophysiology, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
10
|
Zeng LH, Fatima M, Syed S, Shaukat S, Mahdy A, Hussain N, Al Haddad AH, Said AS, Alqahtani A, Alqahtani T, Majeed A, Tariq M, Hussain M. Anti-inflammatory and anti-oxidant properties of Ipomoea nil (Linn.) Roth significantly alleviates cigarette smoke (CS)-induced acute lung injury via possibly inhibiting the NF-κB pathway. Biomed Pharmacother 2022; 155:113267. [DOI: 10.1016/j.biopha.2022.113267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/13/2022] [Accepted: 06/06/2022] [Indexed: 11/26/2022] Open
|
11
|
White A, Wang Z, Wang X, King M, Guo C, Mantsounga C, Ayala A, Morrison AR, Choudhary G, Sellke F, Chambers E, Ware LB, Rounds S, Lu Q. NLRP3 inflammasome activation in cigarette smoke priming for Pseudomonas aeruginosa-induced acute lung injury. Redox Biol 2022; 57:102467. [PMID: 36175355 PMCID: PMC9618465 DOI: 10.1016/j.redox.2022.102467] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 02/04/2023] Open
Abstract
It is increasingly recognized that cigarette smoke (CS) exposure increases the incidence and severity of acute respiratory distress syndrome (ARDS) in critical ill humans and animals. However, the mechanism(s) is not well understood. This study aims to investigate mechanism underlying the priming effect of CS on Pseudomonas aeruginosa-triggered acute lung injury, by using pre-clinic animal models and genetically modified mice. We demonstrated that CS impaired P. aeruginosa-induced mitophagy flux, promoted p62 accumulation, and exacerbated P. aeruginosa-triggered mitochondrial damage and NLRP3 inflammasome activation in alveolar macrophages; an effect associated with increased acute lung injury and mortality. Pharmacological inhibition of caspase-1, a component of inflammasome, attenuated CS primed P. aeruginosa-triggered acute lung injury and improved animal survival. Global or myeloid-specific knockout of IL-1β, a downstream component of inflammasome activation, also attenuated CS primed P. aeruginosa-triggered acute lung injury. Our results suggest that NLRP3 inflammasome activation is an important mechanism for CS primed P. aeruginosa-triggered acute lung injury. (total words: 155).
Collapse
Affiliation(s)
- Alexis White
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Zhengke Wang
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Xing Wang
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Michelle King
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Cynthia Guo
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Chris Mantsounga
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Alfred Ayala
- Department of Surgery, The Warren Alpert Medical School of Brown University and Lifespan-Rhode Island Hospital, Providence, RI, USA
| | - Alan R Morrison
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA; Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Gaurav Choudhary
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA; Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Frank Sellke
- Cardiothoracic Surgery, The Warren Alpert Medical School of Brown University and Lifespan-Rhode Island Hospital, Providence, RI, USA
| | - Eboni Chambers
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Lorraine B Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Sharon Rounds
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA; Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Qing Lu
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA; Department of Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
12
|
Zemskov EA, Gross CM, Aggarwal S, Zemskova MA, Wu X, Gu C, Wang T, Tang H, Black SM. NF-κB-dependent repression of Sox18 transcription factor requires the epigenetic regulators histone deacetylases 1 and 2 in acute lung injury. Front Physiol 2022; 13:947537. [PMID: 35991176 PMCID: PMC9386230 DOI: 10.3389/fphys.2022.947537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/11/2022] [Indexed: 11/30/2022] Open
Abstract
In acute lung injury (ALI), the NF-κB-mediated downregulation of Sox18 gene expression leads to the disruption of the pulmonary endothelial barrier. Previous studies have suggested that the action of NF-κB as a transcriptional repressor also requires the action of class I histone deacetylases (HDACs). Thus, the purpose of this study was to investigate and further delineate the mechanism of Sox18 repression during lipopolysaccharide (LPS) induced ALI. Using selective inhibitors and specific siRNA-driven depletion of HDACs 1-3 in human lung microvascular endothelial cells (HLMVEC) we were able to demonstrate a critical role for HDACs 1 and 2 in the LPS-mediated repression of Sox18 gene expression and the loss of endothelial monolayer integrity. Moreover, our data demonstrate that HDAC1 associates with a transcription-repressive complex within the NF-κB-binding site of Sox18 promoter. Further, we were able to show that the selective inhibitor of HDAC1, tacedinaline, significantly reduced the endothelial permeability and injury associated with LPS challenge in the mouse lung. Taken together, our data demonstrate, for the first time, that transcription repressors HDACs 1 and 2 are involved in pathological mechanism of ALI and can be considered as therapeutic targets.
Collapse
Affiliation(s)
- Evgeny A. Zemskov
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Christine M. Gross
- Department of Medicine at Broward Health Medical Center, Fort Lauderdale, FL, United States
| | - Saurabh Aggarwal
- Department of Anesthesiology, The University of Alabama, Birmingham, AL, United States
| | - Marina A. Zemskova
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
| | - Xiaomin Wu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, AZ, United States
| | - Chenxin Gu
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ting Wang
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, United States
| | - Haiyang Tang
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, United States
| | - Stephen M. Black
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, United States
| |
Collapse
|
13
|
Wu X, Hussain M, Syed SK, Saadullah M, Alqahtani AM, Alqahtani T, Aldahish A, Fatima M, Shaukat S, Hussain L, Jamil Q, Mukhtar I, Khan KUR, Zeng LH. Verapamil attenuates oxidative stress and inflammatory responses in cigarette smoke (CS)-induced murine models of acute lung injury and CSE-stimulated RAW 264.7 macrophages via inhibiting the NF-κB pathway. Biomed Pharmacother 2022; 149:112783. [PMID: 35299124 DOI: 10.1016/j.biopha.2022.112783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/21/2022] [Accepted: 02/28/2022] [Indexed: 01/09/2023] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), severe form of ALI, are characterized by overwhelming of lung inflammation, and no treatment is currently available to treat ALI/ARDS. Cigarette smoke (CS) is one of the prime causes to induce ALI/ARDS via oxidative stress. Despite extensive research, no appropriate therapy is currently available to treat ALI/ARDS. Hence, new potential approaches are needed to treat ALI/ARDS. Consequently, this project was designed to explore the protective effects of verapamil against CS-induced ALI by in vivo and in vitro method. In vivo data obtained from respiratory mechanics, pulmonary morphometric analyses and lung histopathology revealed that verapamil dose-dependently and strikingly decreased the lung weight coefficient, attenuated the albumin exudation into lungs, minimized the infiltration of macrophages and neutrophils into lungs, reduced the pro-inflammatory cytokines (tumour necrosis factor-α (TNF-α), interleukin-6 (IL-6) and keratinocyte chemoattractant (KC)) production, and improved the hypoxemia and lung histopathological changes. Similarly, verapamil also reduced the production of TNF-α, IL-6 and KC from cigarette smoke extract (CSE)-stimulated RAW 264.7 macrophage. Importantly, verapamil dose-dependently and remarkably suppressed the CS-induced oxidative stress via not only reducing the myeloperoxidase (MPO) activity of lungs, total oxidative stress (TOS) and malondialdehyde (MDA) content in the lungs and supernatant of RAW 264.7 macrophage but also improving total antioxidant capacity (TAC) and superoxide dismutase (SOD) production. Finally, verapamil strikingly decreased the NF-κB expression both in in vivo and in vitro models. Hence, verapamil has positive therapeutic effects against CS-induced ALI via suppressing uncontrolled inflammatory response, oxidative stress and NF-κB p65 signaling.
Collapse
Affiliation(s)
- Ximei Wu
- Department of Pharmacology, Zhejiang University City College, 51 Huzhou Street, Hangzhou 310015, China.
| | - Musaddique Hussain
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan.
| | - Shahzada Khurram Syed
- Department of Basic Medical Sciences, School of Health Sciences, University of Management and Technology Lahore, 54000, Pakistan
| | - Malik Saadullah
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Ali M Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Taha Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Afaf Aldahish
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Mobeen Fatima
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Saira Shaukat
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Liaqat Hussain
- Department of Pharmacology, Government College University, Faisalabad 38000, Pakistan
| | - Qurratulain Jamil
- Department of Pharmacy Practice, Faculty of Pharmacy, The Islamia University of Bahawalpur, Pakistan
| | - Imran Mukhtar
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan; Sir Sadiq Muhammad Khan Abassi post Graduate Medical College, The Islamia University of Bahawalpur, Pakistan
| | - Kashif-Ur-Rehman Khan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Ling-Hui Zeng
- Department of Pharmacology, Zhejiang University City College, 51 Huzhou Street, Hangzhou 310015, China
| |
Collapse
|
14
|
Rounds S, Lu Q, Siamwala J. CIGARETTE SMOKING INCREASES THE RISK OF ACUTE RESPIRATORY DISTRESS SYNDROME. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2022; 132:224-235. [PMID: 36196201 PMCID: PMC9480561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Cigarette smoking (CS) remains a cause of considerable morbidity and mortality, despite recent progress in smoking cessation in the United States. Epidemiologic studies in humans have reported associations between CS and development of acute respiratory distress syndrome (ARDS) after a number of inciting risk factors. We have assessed the effects of CS exposure on lung vascular permeability and inflammation in mice and found that both acute and sustained CS exposure increased the severity of acute lung injury caused by subsequent intrapulmonary instillation of lipopolysaccharide. In addition to enhanced inflammation, CS exposure directly impaired lung endothelial cell barrier function. Our results indicate that mouse strains differ in susceptibility to CS exacerbation of acute lung injury and that there are differences in transcriptomic effects of CS. These results demonstrate the biologic basis for the association of CS with development of ARDS. We propose that CS be considered a cause of heterogeneity of ARDS phenotypes and that this be recorded as a risk factor in the design of clinical trials.
Collapse
|
15
|
Karki P, Birukova AA. Microtubules as Major Regulators of Endothelial Function: Implication for Lung Injury. Front Physiol 2021; 12:758313. [PMID: 34777018 PMCID: PMC8582326 DOI: 10.3389/fphys.2021.758313] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/06/2021] [Indexed: 01/04/2023] Open
Abstract
Endothelial dysfunction has been attributed as one of the major complications in COVID-19 patients, a global pandemic that has already caused over 4 million deaths worldwide. The dysfunction of endothelial barrier is characterized by an increase in endothelial permeability and inflammatory responses, and has even broader implications in the pathogenesis of acute respiratory syndromes such as ARDS, sepsis and chronic illnesses represented by pulmonary arterial hypertension and interstitial lung disease. The structural integrity of endothelial barrier is maintained by cytoskeleton elements, cell-substrate focal adhesion and adhesive cell junctions. Agonist-mediated changes in endothelial permeability are directly associated with reorganization of actomyosin cytoskeleton leading to cell contraction and opening of intercellular gaps or enhancement of cortical actin cytoskeleton associated with strengthening of endothelial barrier. The role of actin cytoskeleton remodeling in endothelial barrier regulation has taken the central stage, but the impact of microtubules in this process remains less explored and under-appreciated. This review will summarize the current knowledge on the crosstalk between microtubules dynamics and actin cytoskeleton remodeling, describe the signaling mechanisms mediating this crosstalk, discuss epigenetic regulation of microtubules stability and its nexus with endothelial barrier maintenance, and overview a role of microtubules in targeted delivery of signaling molecules regulating endothelial permeability and inflammation.
Collapse
Affiliation(s)
- Pratap Karki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Anna A Birukova
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
16
|
Wang Y, Kuang X, Yin Y, Han N, Chang L, Wang H, Hou Y, Li H, Li Z, Liu Y, Hao Y, Wei Y, Wang X, Jia Z. Tongxinluo prevents chronic obstructive pulmonary disease complicated with atherosclerosis by inhibiting ferroptosis and protecting against pulmonary microvascular barrier dysfunction. Biomed Pharmacother 2021; 145:112367. [PMID: 34740097 DOI: 10.1016/j.biopha.2021.112367] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/09/2021] [Accepted: 10/20/2021] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular comorbidities are pervasive in chronic obstructive pulmonary disease (COPD) and often result in serious adverse cardiovascular events. Tongxinluo (TXL) has been clinically verified to treat atherosclerosis (AS), improve lung function and alleviate dyspnoea. The present study aimed to explore the effect of lung microvascular barrier dysfunction on AS in COPD and the potential pulmonary protective mechanisms of TXL in COPD complicated with AS. COPD complicated with AS was induced in mice by cigarette smoke (CS) exposure and high-fat diet (HFD) feeding. The mice were treated with atorvastatin (ATO), TXL or combination therapy (ATO+TXL) for 20 weeks. Pulmonary function, lung pathology, serum lipid levels, atherosclerotic plaque area and indicators of barrier function, oxidative stress and ferroptosis in lung tissue were evaluated. In vitro, human pulmonary microvascular endothelial cells (HPMECs) were pretreated with TXL for 4 h and then incubated with cigarette smoke extract (CSE) and homocysteine (Hcy) for 36 h to induce barrier dysfunction. Then the indicators of barrier function, oxidative stress and ferroptosis were measured. The results demonstrate that CS aggravated dyslipidaemia, atherosclerotic plaque formation, pulmonary function decline, pathological injury, barrier dysfunction, oxidative stress and ferroptosis in the HFD-fed mice. However, these abnormalities were partially reversed by ATO and TXL. Similar results were observed in vitro. In conclusion, pulmonary microvascular barrier dysfunction plays an important role by which COPD affects the progression of AS, and ferroptosis may be involved. Moreover, TXL delays the progression of AS and reduces cardiovascular events by protecting the pulmonary microvascular barrier and inhibiting ferroptosis.
Collapse
Affiliation(s)
- Yafen Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050090, Hebei, China
| | - Xiangnan Kuang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050090, Hebei, China
| | - Yujie Yin
- Hebei Yiling Pharmaceutical Research Institute, Shijiazhuang 050035, China
| | - Ningxin Han
- Graduate School, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Liping Chang
- Hebei Yiling Pharmaceutical Research Institute, Shijiazhuang 050035, China
| | - Hongtao Wang
- Hebei Yiling Pharmaceutical Research Institute, Shijiazhuang 050035, China
| | - Yunlong Hou
- Hebei Yiling Pharmaceutical Research Institute, Shijiazhuang 050035, China
| | - Huixin Li
- Hebei Yiling Pharmaceutical Research Institute, Shijiazhuang 050035, China
| | - Zhen Li
- Graduate School, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Yi Liu
- Graduate School, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Yuanjie Hao
- Graduate School, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Yaru Wei
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050090, Hebei, China
| | - Xiaoqi Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050090, Hebei, China
| | - Zhenhua Jia
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050090, Hebei, China; Hebei Yiling Pharmaceutical Research Institute, Shijiazhuang 050035, China; Department of Cardiology, Affiliated Yiling Hospital of Hebei University of Chinese Medicine, Shijiazhuang 050091, Hebei, China
| |
Collapse
|
17
|
Kim JS, Anderson MR, Bernstein EJ, Oelsner EC, Raghu G, Noth I, Tsai MY, Salvatore M, Austin JHM, Hoffman EA, Barr RG, Podolanczuk AJ. Associations of D-Dimer with Computed Tomographic Lung Abnormalities, Serum Biomarkers of Lung Injury, and Forced Vital Capacity: MESA Lung Study. Ann Am Thorac Soc 2021; 18:1839-1848. [PMID: 33861685 PMCID: PMC8641831 DOI: 10.1513/annalsats.202012-1557oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 04/08/2021] [Indexed: 12/30/2022] Open
Abstract
Rationale: The coagulation cascade may play a role in the pathogenesis of interstitial lung disease through increased production of thrombin and fibrin deposition. Whether circulating coagulation cascade factors are linked to lung inflammation and scarring among community-dwelling adults is unknown. Objectives: To test the hypothesis that higher baseline D-dimer concentrations are associated with markers of early lung injury and scarring. Methods: Using the MESA (Multi-Ethnic Study of Atherosclerosis) cohort (n = 6,814), we examined associations of baseline D-dimer concentrations with high attenuation areas from examination 1 (2000-2002; n = 6,184) and interstitial lung abnormalities from examination 5 computed tomographic (CT) scans (2010-2012; n = 2,227), and serum MMP-7 (matrix metalloproteinase-7) and SP-A (surfactant protein-A) from examination 1 (n = 1,098). We examined longitudinal change in forced vital capacity (FVC) from examinations 3-6 (2004-2018, n = 3,562). We used linear logistic regression and linear mixed models to examine associations and adjust for potential confounders. Results: The mean (standard deviation) age of the cohort was 62 (10) years, and the D-dimer concentration was 0.35 (0.69) ug/ml. For every 10% increase in D-dimer concentration, there was an increase in high attenuation area percentage of 0.27 (95% confidence interval (CI), 0.08-0.47) after adjustment for covariates. Associations were stronger among those older than 65 years (P values for interaction < 0.001). A 10% increase in D-dimer concentration was associated with an odds ratio of 1.05 for interstitial lung abnormalities (95% CI, 0.99-1.11). Higher D-dimer concentrations were associated with higher serum MMP-7 and a faster decline in FVC. D-dimer was not associated with SP-A. Conclusions: Higher D-dimer concentrations were associated with a greater burden of lung parenchymal abnormalities detected on CT scan, MMP-7, and FVC decline among community-dwelling adults.
Collapse
Affiliation(s)
- John S. Kim
- Department of Medicine, University of Virginia, Charlottesville, Virginia
- Department of Medicine
| | | | | | | | - Ganesh Raghu
- Department of Medicine, University of Washington, Seattle, Washington
| | - Imre Noth
- Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | | | | | - Eric A. Hoffman
- Departments of Radiology, Medicine, and Biomedical Engineering, University of Iowa Carver College of Medicine, Iowa City, Iowa; and
| | - R. Graham Barr
- Department of Medicine
- Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, New York
| | - Anna J. Podolanczuk
- Department of Medicine
- Department of Medicine, Weill Cornell Medical Center, New York, New York
| |
Collapse
|
18
|
Xie X, Zhong Z, Zhao W, Wu S, Liu J. The Differences and Changes of Semi-Quantitative and Quantitative CT Features of Coronavirus Disease 2019 Pneumonia in Patients With or Without Smoking History. Front Med (Lausanne) 2021; 8:663514. [PMID: 34568353 PMCID: PMC8455871 DOI: 10.3389/fmed.2021.663514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/30/2021] [Indexed: 01/08/2023] Open
Abstract
Objective: To assess CT features of COVID-19 patients with different smoking status using quantitative and semi-quantitative technologies and to investigate changes of CT features in different disease states between the two groups. Methods: 30 COVID-19 patients with current smoking status (29 men, 1 woman) admitted in our database were enrolled as smoking group and 56 COVID-19 patients without smoking history (24 men, 32 women) admitted during the same period were enrolled as a control group. Twenty-seven smoking cases and 55 control cases reached recovery standard and were discharged. Initial and follow-up CT during hospitalization and follow-up CT after discharge were acquired. Thirty quantitative features, including the ratio of infection volume and visual-assessed interstitial changes score including total score, score of ground glass opacity, consolidation, septal thickening, reticulation and honeycombing sign, were analyzed. Results: Initial CT images of the smoking group showed higher scores of septal thickening [4.5 (0–5) vs. 0 (0–4), p = 0.001] and reticulation [0 (0–5.25) vs 0 (0–0), p = 0.001] as well as higher total score [7 (5–12.25) vs. 6 (5–7), p = 0.008] with statistical significance than in the control group. The score of reticulation was higher in the smoking group than in the control group when discharged [0.89 (0–0) vs. 0.09 (0–0), p = 0.02]. The score of septal thickening tended to be higher in the smoking group than the control group [4 (0–4) vs. 0 (0–4), p = 0.007] after being discharged. Quantitative CT features including infection ratio of whole lung and left lung as well as infection ratio within HU (−750, −300) and within HU (−300, 49) were higher in the control group of initial CT with statistical differences. The infection ratio of whole lung and left lung, infection ratio within HU (−750), and within HU (−750, −300) were higher in the control group with statistical differences when discharged. This trend turned adverse after discharge and the values of quantitative features were generally higher in the smoking group than in the control group without statistical differences. Conclusions: Patients with a history of smoking presented more severe interstitial manifestations and more residual lesion after being discharged. More support should be given for COVID-19 patients with a smoking history during hospitalization and after discharge.
Collapse
Affiliation(s)
- Xingzhi Xie
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Zheng Zhong
- Department of Radiology, First Hospital of Changsha, Changsha, China.,Changsha Public Health Treatment Center, Changsha, China
| | - Wei Zhao
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Shangjie Wu
- Department of Respiratory Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Jun Liu
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha, China.,Department of Radiology Quality Control Center, Changsha, China
| |
Collapse
|
19
|
Li J, Fang Y, Wu D. Mechanical forces and metabolic changes cooperate to drive cellular memory and endothelial phenotypes. CURRENT TOPICS IN MEMBRANES 2021; 87:199-253. [PMID: 34696886 PMCID: PMC8639155 DOI: 10.1016/bs.ctm.2021.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Endothelial cells line the innermost layer of arterial, venous, and lymphatic vascular tree and accordingly are subject to hemodynamic, stretch, and stiffness mechanical forces. Normally quiescent, endothelial cells have a hemodynamic set point and become "activated" in response to disturbed hemodynamics, which may signal impending nutrient or gas depletion. Endothelial cells in the majority of tissue beds are normally inactivated and maintain vessel barrier functions, are anti-inflammatory, anti-coagulant, and anti-thrombotic. However, under aberrant mechanical forces, endothelial signaling transforms in response, resulting cellular changes that herald pathological diseases. Endothelial cell metabolism is now recognized as the primary intermediate pathway that undergirds cellular transformation. In this review, we discuss the various mechanical forces endothelial cells sense in the large vessels, microvasculature, and lymphatics, and how changes in environmental mechanical forces result in changes in metabolism, which ultimately influence cell physiology, cellular memory, and ultimately disease initiation and progression.
Collapse
Affiliation(s)
- Jin Li
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - Yun Fang
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - David Wu
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States.
| |
Collapse
|
20
|
Affiliation(s)
- Kazuhiro Ito
- Imperial College, National Heart & Lung Institute,, London, United Kingdom of Great Britain and Northern Ireland;
| |
Collapse
|
21
|
Su Y, Han W, Kovacs-Kasa A, Verin AD, Kovacs L. HDAC6 Activates ERK in Airway and Pulmonary Vascular Remodeling of COPD. Am J Respir Cell Mol Biol 2021; 65:603-614. [PMID: 34280336 DOI: 10.1165/rcmb.2020-0520oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a multisystemic respiratory disease which is associated with progressive airway and pulmonary vascular remodeling due to the increased proliferation of bronchial and pulmonary arterial smooth muscle cells (BSMCs and PASMCs) and overproduction of extracellular matrix (ECM), e.g., collagen. Cigarette smoke (CS) and several mediators such as PDGF and IL-6 play critical role in the COPD pathogenesis. Histone deacetylase 6 (HDAC6) has been shown to be implicated in vascular remodeling. However, the HDAC6 signaling in airway and pulmonary vascular remodeling of COPD and the underlying mechanisms remain undetermined. Here we show that HDAC6 expression is upregulated in lungs of COPD patients and animal model. We also found that cigarette smoke extract (CSE), PDGF and IL-6 increase the protein levels and activation of HDAC6 in BSMCs and PASMCs. Furthermore, CSE and these stimulants induced deacetylation and phosphorylation of ERK1/2 and increased collagen synthesis and proliferation of BSMCs and PASMCs which were prevented by HDAC6 inhibition. Inhibition of ERK1/2 also diminished the CSE, PDGF and IL-6-caused elevation in collagen levels and cell proliferation. Pharmacological HDAC6 inhibition by tubastatin A prevented the CS-stimulated increases in the thickness of the bronchial and pulmonary arterial wall, airway resistance, emphysema as well as right ventricular (RV) systolic pressure (RVSP) and RV hypertrophy in rat model of COPD. These data demonstrate that the upregulated HDAC6 governs the collagen synthesis and proliferation of BSMCs and PASMCs leading to airway and vascular remodeling in COPD.
Collapse
Affiliation(s)
- Yunchao Su
- Augusta University Medical College of Georgia, 160343, Department of Pharmacology, Augusta, Georgia, United States
| | - Weihong Han
- Augusta University, 1421, Augusta, Georgia, United States
| | | | | | - Laszlo Kovacs
- Augusta University, 1421, Augusta, Georgia, United States;
| |
Collapse
|
22
|
Rounds S, Lu Q. Where There's Smoke, There's Fire. Chest 2021; 158:1301-1302. [PMID: 33036072 DOI: 10.1016/j.chest.2020.07.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 11/30/2022] Open
Affiliation(s)
- Sharon Rounds
- Department of Medicine and of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Vascular Research Laboratory, Providence VA Medical Center, Providence, RI.
| | - Qing Lu
- Department of Medicine and of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Vascular Research Laboratory, Providence VA Medical Center, Providence, RI
| |
Collapse
|
23
|
Kovacs-Kasa A, Kovacs L, Cherian-Shaw M, Patel V, Meadows ML, Fulton DJ, Su Y, Verin AD. Inhibition of Class IIa HDACs improves endothelial barrier function in endotoxin-induced acute lung injury. J Cell Physiol 2021; 236:2893-2905. [PMID: 32959895 PMCID: PMC9946131 DOI: 10.1002/jcp.30053] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/21/2022]
Abstract
Acute lung injury (ALI) is an acute inflammatory process arises from a wide range of lung insults. A major cause of ALI is dysfunction of the pulmonary vascular endothelial barrier but the mechanisms involved are incompletely understood. The therapeutic potential of histone deacetylase (HDAC) inhibitors for the treatment of cardiovascular and inflammatory diseases is increasingly apparent, but the mechanisms by which HDACs regulate pulmonary vascular barrier function remain to be resolved. We found that specific Class IIa HDACs inhibitor, TMP269, significantly attenuated the lipopolysaccharide (LPS)-induced human lung microvascular endothelial cells (HLMVEC) barrier compromise in vitro and improved vascular barrier integrity and lung function in murine model of ALI in vivo. TMP269 decreased LPS-induced myosin light chain phosphorylation suggesting the role for Class IIa HDACs in LPS-induced cytoskeleton reorganization. TMP269 did not affect microtubule structure and tubulin acetylation in contrast to the HDAC6-specific inhibitor, Tubastatin A suggesting that Class IIa HDACs and HDAC6 (Class IIb) regulate endothelial cytoskeleton and permeability via different mechanisms. Furthermore, LPS increased the expression of ArgBP2 which has recently been attributed to HDAC-mediated activation of Rho. Depletion of ArgBP2 abolished the ability of LPS to disrupt barrier function in HLMVEC and both TMP269 and Tubastatin A decreased the level of ArgBP2 expression after LPS stimulation suggesting that both Class IIa and IIb HDACs regulate endothelial permeability via ArgBP2-dependent mechanism. Collectively, our data strongly suggest that Class IIa HDACs are involved in LPS-induced ALI in vitro and in vivo via specific mechanism which involved contractile responses, but not microtubule reorganization.
Collapse
Affiliation(s)
- Anita Kovacs-Kasa
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Laszlo Kovacs
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Mary Cherian-Shaw
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Vijay Patel
- Department of Cardiothoracic Surgery, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Mary L. Meadows
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - David J. Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Yunchao Su
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Alexander D. Verin
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
24
|
Drummer C, Saaoud F, Shao Y, Sun Y, Xu K, Lu Y, Ni D, Atar D, Jiang X, Wang H, Yang X. Trained Immunity and Reactivity of Macrophages and Endothelial Cells. Arterioscler Thromb Vasc Biol 2021; 41:1032-1046. [PMID: 33380171 PMCID: PMC7904591 DOI: 10.1161/atvbaha.120.315452] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/14/2020] [Indexed: 12/15/2022]
Abstract
Innate immune cells can develop exacerbated immunologic response and long-term inflammatory phenotype following brief exposure to endogenous or exogenous insults, which leads to an altered response towards a second challenge after the return to a nonactivated state. This phenomenon is known as trained immunity (TI). TI is not only important for host defense and vaccine response but also for chronic inflammations such as cardiovascular and metabolic diseases such as atherosclerosis. TI can occur in innate immune cells such as monocytes/macrophages, natural killer cells, endothelial cells (ECs), and nonimmune cells, such as fibroblast. In this brief review, we analyze the significance of TI in ECs, which are also considered as innate immune cells in addition to macrophages. TI can be induced by a variety of stimuli, including lipopolysaccharides, BCG (bacillus Calmette-Guerin), and oxLDL (oxidized low-density lipoprotein), which are defined as risk factors for cardiovascular and metabolic diseases. Furthermore, TI in ECs is functional for inflammation effectiveness and transition to chronic inflammation. Rewiring of cellular metabolism of the trained cells takes place during induction of TI, including increased glycolysis, glutaminolysis, increased accumulation of tricarboxylic acid cycle metabolites and acetyl-coenzyme A production, as well as increased mevalonate synthesis. Subsequently, this leads to epigenetic remodeling, resulting in important changes in chromatin architecture that enables increased gene transcription and enhanced proinflammatory immune response. However, TI pathways and inflammatory pathways are separated to ensure memory stays when inflammation undergoes resolution. Additionally, reactive oxygen species play context-dependent roles in TI. Therefore, TI plays significant roles in EC and macrophage pathology and chronic inflammation. However, further characterization of TI in ECs and macrophages would provide novel insights into cardiovascular disease pathogenesis and new therapeutic targets. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Charles Drummer
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Fatma Saaoud
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Ying Shao
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Yu Sun
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Keman Xu
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Yifan Lu
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Dong Ni
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Diana Atar
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaohua Jiang
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Hong Wang
- Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Cardiovascular Research Center, Centers for Inflammation, Translational and Clinical Lung Research and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
25
|
Zhang Q, Wang Y, Qu D, Yu J, Yang J. Role of HDAC6 inhibition in sepsis-induced acute respiratory distress syndrome (Review). Exp Ther Med 2021; 21:422. [PMID: 33747162 DOI: 10.3892/etm.2021.9866] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) induced by sepsis contributes remarkably to the high mortality rate observed in intensive care units, largely due to a lack of effective drug therapies. Histone deacetylase 6 (HDAC6) is a class-IIb deacetylase that modulates non-nuclear protein functions via deacetylation and ubiquitination. Importantly, HDAC6 has been shown to exert anti-cancer, anti-neurodegeneration, and immunological effects, and several HDAC6 inhibitors have now entered clinical trials. It has also been recently shown to modulate inflammation, and HDAC6 inhibition has been demonstrated to markedly suppress experimental sepsis. The present review summarizes the role of HDAC6 in sepsis-induced inflammation and endothelial barrier dysfunction in recent years. It is proposed that HDAC6 inhibition predominantly ameliorates sepsis-induced ARDS by directly attenuating inflammation, which modulates the innate and adaptive immunity, transcription of pro-inflammatory genes, and protects endothelial barrier function. HDAC6 inhibition protects against sepsis-induced ARDS, thereby making HDAC6 a promising therapeutic target. However, HDAC inhibition may be associated with adverse effects on the embryo sac and oocyte, necessitating further studies.
Collapse
Affiliation(s)
- Qinghua Zhang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Danhua Qu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Jinyan Yu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Junling Yang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
26
|
Haddad C, Bou Malhab S, Sacre H, Salameh P. Smoking and COVID-19: A Scoping Review. Tob Use Insights 2021; 14:1179173X21994612. [PMID: 33642886 PMCID: PMC7890709 DOI: 10.1177/1179173x21994612] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/25/2021] [Indexed: 01/08/2023] Open
Abstract
Smoking affects a person's overall health and damages nearly every organ of the body. Since smoking tobacco affects and damages the lungs, it increases the risk of respiratory infections and makes it easier for the coronavirus disease (COVID-19) to invade the lung tissue, causing more severe symptoms and increasing the risk of death. However, debates are still ongoing as to the effect of cigarette smoking on vulnerability to COVID-19. Some studies, where active smokers were underrepresented among patients with COVID-19, claimed that a "smoker's paradox" may exist in COVID-19 and that smokers are protected from infection and severe complications of COVID-19. However, other studies reported the opposite trend. The objective of this study is to review the findings of epidemiological and in vitro studies about the association between smoking and the risk of contracting COVID-19, taking into account disease severity. Several epidemiological studies have found a higher smoking prevalence among COVID-19 infected patients. Also, studies had shown that people with respiratory diseases caused by tobacco use are at higher risk of developing severe COVID-19 symptoms. Studies have shown that in vitro, the acute exposure allows for more severe proximal airway epithelial disease from SARS-CoV-2 by reducing the mucosal innate immune response and the proliferation of airway basal stem cells and has implications for disease spread and severity in people exposed to cigarette smoke, with a more severe viral infection and cell death. Smoker patients with different comorbidities are at higher risk of contracting the COVID-19 virus and have a worse prognosis for the virus as well as for their comorbidities. Further investigations of the interaction between smoking and COVID-19 are warranted to accurately assess the risk of contracting COVID-19 among smokers, and the progression to mechanical ventilation or death in patients who suffer from it.
Collapse
Affiliation(s)
- Chadia Haddad
- Research Department, Psychiatric Hospital of the Cross, Jal Eddib, Lebanon
- INSERM, Univ. Limoges, CH Esquirol, IRD, U1094 Tropical Neuroepidemiology, Institute of Epidemiology and Tropical Neurology, GEIST, Limoges, France
| | - Sandrella Bou Malhab
- Ecole Doctorale des Sciences et Technologie, Lebanese University, Hadath, Lebanon
| | - Hala Sacre
- INSPECT-LB: Institut National de Santé Publique, Épidémiologie Clinique et Toxicologie-Liban, Beirut, Lebanon
| | - Pascale Salameh
- INSPECT-LB: Institut National de Santé Publique, Épidémiologie Clinique et Toxicologie-Liban, Beirut, Lebanon
- Faculty of Pharmacy, Lebanese University, Hadat, Lebanon
- School of Medicine, University of Nicosia, Cyprus
| |
Collapse
|
27
|
Wang Y, Liu YJ, Xu DF, Zhang H, Xu CF, Mao YF, Lv Z, Zhu XY, Jiang L. DRD1 downregulation contributes to mechanical stretch-induced lung endothelial barrier dysfunction. Am J Cancer Res 2021; 11:2505-2521. [PMID: 33456556 PMCID: PMC7806475 DOI: 10.7150/thno.46192] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 12/05/2020] [Indexed: 01/11/2023] Open
Abstract
Rationale: The lung-protective effects of dopamine and its role in the pathology of ventilator-induced lung injury (VILI) are emerging. However, the underlying mechanisms are still largely unknown. Objective: To investigate the contribution of dopamine receptor dysregulation in the pathogenesis of VILI and therapeutic potential of dopamine D1 receptor (DRD1) agonist in VILI. Methods: The role of dopamine receptors in mechanical stretch-induced endothelial barrier dysfunction and lung injury was studied in DRD1 knockout mice, in isolated mouse lung vascular endothelial cells (MLVECs), and in lung samples from patients who underwent pulmonary lobectomy with mechanical ventilation for different time periods. Measurements and Main Results: DRD1 was downregulated in both surgical patients and mice exposed to mechanical ventilation. Prophylactic administration of dopamine or DRD1 agonist attenuated mechanical stretch-induced lung endothelial barrier dysfunction and lung injury. By contrast, pulmonary knockdown or global knockout of DRD1 exacerbated these effects. Prophylactic administration of dopamine attenuated mechanical stretch-induced α-tubulin deacetylation and subsequent endothelial hyperpermeability through DRD1 signaling. We identified that cyclic stretch-induced glycogen-synthase-kinase-3β activation led to phosphorylation and activation of histone deacetylase 6 (HDAC6), which resulted in deacetylation of α-tubulin. Upon activation, DRD1 signaling attenuated mechanical stretch-induced α-tubulin deacetylation and subsequent lung endothelial barrier dysfunction through cAMP/exchange protein activated by cAMP (EPAC)-mediated inactivation of HDAC6. Conclusions: This work identifies a novel protective role for DRD1 against mechanical stretch-induced lung endothelial barrier dysfunction and lung injury. Further study of the mechanisms involving DRD1 in the regulation of microtubule stability and interference with DRD1/cAMP/EPAC/HDAC6 signaling may provide insight into therapeutic approaches for VILI.
Collapse
|
28
|
Karki P, Ke Y, Zhang CO, Li Y, Tian Y, Son S, Yoshimura A, Kaibuchi K, Birukov KG, Birukova AA. SOCS3-microtubule interaction via CLIP-170 and CLASP2 is critical for modulation of endothelial inflammation and lung injury. J Biol Chem 2021; 296:100239. [PMID: 33372035 PMCID: PMC7949054 DOI: 10.1074/jbc.ra120.014232] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 11/23/2020] [Accepted: 12/28/2020] [Indexed: 12/12/2022] Open
Abstract
Proinflammatory cytokines such as IL-6 induce endothelial cell (EC) barrier disruption and trigger an inflammatory response in part by activating the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway. The protein suppressor of cytokine signaling-3 (SOCS3) is a negative regulator of JAK-STAT, but its role in modulation of lung EC barrier dysfunction caused by bacterial pathogens has not been investigated. Using human lung ECs and EC-specific SOCS3 knockout mice, we tested the hypothesis that SOCS3 confers microtubule (MT)-mediated protection against endothelial dysfunction. SOCS3 knockdown in cultured ECs or EC-specific SOCS3 knockout in mice resulted in exacerbated lung injury characterized by increased permeability and inflammation in response to IL-6 or heat-killed Staphylococcus aureus (HKSA). Ectopic expression of SOCS3 attenuated HKSA-induced EC dysfunction, and this effect required assembled MTs. SOCS3 was enriched in the MT fractions, and treatment with HKSA disrupted SOCS3-MT association. We discovered that-in addition to its known partners gp130 and JAK2-SOCS3 interacts with MT plus-end binding proteins CLIP-170 and CLASP2 via its N-terminal domain. The resulting SOCS3-CLIP-170/CLASP2 complex was essential for maximal SOCS3 anti-inflammatory effects. Both IL-6 and HKSA promoted MT disassembly and disrupted SOCS3 interaction with CLIP-170 and CLASP2. Moreover, knockdown of CLIP-170 or CLASP2 impaired SOCS3-JAK2 interaction and abolished the anti-inflammatory effects of SOCS3. Together, these findings demonstrate for the first time an interaction between SOCS3 and CLIP-170/CLASP2 and reveal that this interaction is essential to the protective effects of SOCS3 in lung endothelium.
Collapse
Affiliation(s)
- Pratap Karki
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yunbo Ke
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Chen-Ou Zhang
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yue Li
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yufeng Tian
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Sophia Son
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University, Tokyo, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University, Nagoya, Japan
| | - Konstantin G Birukov
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Anna A Birukova
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
29
|
Wang Z, White A, Wang X, Ko J, Choudhary G, Lange T, Rounds S, Lu Q. Mitochondrial Fission Mediated Cigarette Smoke-induced Pulmonary Endothelial Injury. Am J Respir Cell Mol Biol 2020; 63:637-651. [PMID: 32672471 DOI: 10.1165/rcmb.2020-0008oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cigarette smoke (CS) exposure increases the risk for acute respiratory distress syndrome in humans and promotes alveolar-capillary barrier permeability and acute lung injury in animal models. However, the underlying mechanisms are not well understood. Mitochondrial fusion and fission are essential for mitochondrial homeostasis in health and disease. In this study, we hypothesized that CS caused endothelial injury via an imbalance of mitochondrial fusion and fission and resultant mitochondrial oxidative stress and dysfunction. We noted that CS altered mitochondrial morphology by shortening mitochondrial networks and causing perinuclear accumulation of damaged mitochondria in primary rat lung microvascular endothelial cells. We also found that CS increased mitochondrial fission likely by decreasing Drp1-S637 and increasing FIS1, Drp1-S616 phosphorylation, mitochondrial translocation, and tetramerization and reduced mitochondrial fusion likely by decreasing Mfn2 in lung microvascular endothelial cells and mouse lungs. CS also caused aberrant mitophagy, increased mitochondrial oxidative stress, and reduced mitochondrial respiration. An inhibitor of mitochondrial fission and a mitochondria-specific antioxidant prevented CS-induced increased endothelial barrier dysfunction and apoptosis. Our data suggest that excessive mitochondrial fission and resultant oxidative stress are essential mediators of CS-induced endothelial injury and that inhibition of mitochondrial fission and mitochondria-specific antioxidants may be useful therapeutic strategies for CS-induced endothelial injury and associated pulmonary diseases.
Collapse
Affiliation(s)
- Zhengke Wang
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
| | - Alexis White
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
| | - Xing Wang
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island.,Haihe Hospital, Tianjin University, Tianjin, China; and
| | - Junsuk Ko
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island
| | - Gaurav Choudhary
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island.,Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Thilo Lange
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, 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
| | - 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
| |
Collapse
|
30
|
Cigarette smoke extract induces airway epithelial cell death via repressing PRMT6/AKT signaling. Aging (Albany NY) 2020; 12:24301-24317. [PMID: 33260152 PMCID: PMC7762507 DOI: 10.18632/aging.202210] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a severe public health threat world-wide. Cigarette smoke (CS)-induced airway epithelial cell death is a major pathway of pathogenesis in emphysema, a subtype of COPD. Protein arginine methyltransferase 6 (PRMT6) is a type I PRMT that catalyzes mono- and di-methylation on arginine residues within histone and non-histone proteins to modulate a variety of life processes, such as apoptosis. However, its role in CS-induced lung epithelial death has not been fully elucidated. Here we report that PRMT6 was decreased in mouse lung tissues from a cigarette smoke extract (CSE)-mediated experimental emphysematous model and in CSE treated or cigarette smoke exposed lung epithelial cells. Depletion of PRMT6 increased the protein levels of phosphatase PTEN and PI3K regulatory subunit p85 but decreased a downstream kinase PDK1, resulting in AKT dephosphorylation and thereafter, lung epithelial cell death. Knockout of PRMT6 inhibited epithelial survival and promoted CSE-mediated epithelial cell death, while ectopic expression of PRMT6 protein partially reversed epithelial cell death via PI3K/AKT-mediated cell survival signaling in CSE cellular models. These findings demonstrate that PRMT6 plays a crucial role in CS-induced bronchial epithelial cell death that may be a potential therapeutic target against the airway cell death in CS-induced COPD.
Collapse
|
31
|
Chen X, He Y, Fu W, Sahebkar A, Tan Y, Xu S, Li H. Histone Deacetylases (HDACs) and Atherosclerosis: A Mechanistic and Pharmacological Review. Front Cell Dev Biol 2020; 8:581015. [PMID: 33282862 PMCID: PMC7688915 DOI: 10.3389/fcell.2020.581015] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis (AS), the most common underlying pathology for coronary artery disease, is a chronic inflammatory, proliferative disease in large- and medium-sized arteries. The vascular endothelium is important for maintaining vascular health. Endothelial dysfunction is a critical early event leading to AS, which is a major risk factor for stroke and myocardial infarction. Accumulating evidence has suggested the critical roles of histone deacetylases (HDACs) in regulating vascular cell homeostasis and AS. The purpose of this review is to present an updated view on the roles of HDACs (Class I, Class II, Class IV) and HDAC inhibitors in vascular dysfunction and AS. We also elaborate on the novel therapeutic targets and agents in atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaona Chen
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanhong He
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenjun Fu
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Polish Mother's Memorial Hospital Research Institute, Łódź, Poland
| | - Yuhui Tan
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Suowen Xu
- Department of Endocrinology, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hong Li
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
32
|
Abouhish H, Thounaojam MC, Jadeja RN, Gutsaeva DR, Powell FL, Khriza M, Martin PM, Bartoli M. Inhibition of HDAC6 Attenuates Diabetes-Induced Retinal Redox Imbalance and Microangiopathy. Antioxidants (Basel) 2020; 9:antiox9070599. [PMID: 32660051 PMCID: PMC7402090 DOI: 10.3390/antiox9070599] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/20/2022] Open
Abstract
We investigated the contributing role of the histone deacetylase 6 (HDAC6) to the early stages of diabetic retinopathy (DR). Furthermore, we examined the mechanism of action of HDAC6 in human retinal endothelial cells (HuREC) exposed to glucidic stress. Streptozotocin-induced diabetic rats (STZ-rats), a rat model of type 1 diabetes, were used as model of DR. HDAC6 expression and activity were increased in human diabetic postmortem donors and STZ-rat retinas and were augmented in HuREC exposed to glucidic stress (25 mM glucose). Administration of the HDAC6 specific inhibitor Tubastatin A (TS) (10 mg/kg) prevented retinal microvascular hyperpermeability and up-regulation of inflammatory markers. Furthermore, in STZ-rats, TS decreased the levels of senescence markers and rescued the expression and activity of the histone deacetylase sirtuin 1 (SIRT1), while downregulating the levels of free radicals and of the redox stress markers 4-hydroxynonenal (4-HNE) and nitrotyrosine (NT). The antioxidant effects of TS, consequent to HDAC6 inhibition, were associated with preservation of Nrf2-dependent gene expression and up-regulation of thioredoxin-1 activity. In vitro data, obtained from HuREC, exposed to glucidic stress, largely replicated the in vivo results further confirming the antioxidant effects of HDAC6 inhibition by TS in the diabetic rat retina. In summary, our data implicate HDAC6 activation in mediating hyperglycemia-induced retinal oxidative/nitrative stress leading to retinal microangiopathy and, potentially, DR.
Collapse
Affiliation(s)
- Hossameldin Abouhish
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (H.A.); (M.C.T.); (D.R.G.)
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Menaka C. Thounaojam
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (H.A.); (M.C.T.); (D.R.G.)
| | - Ravirajsinh N. Jadeja
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.N.J.); (F.L.P.); (P.M.M.)
| | - Diana R. Gutsaeva
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (H.A.); (M.C.T.); (D.R.G.)
| | - Folami L. Powell
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.N.J.); (F.L.P.); (P.M.M.)
| | - Mohamed Khriza
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Pamela M. Martin
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (R.N.J.); (F.L.P.); (P.M.M.)
| | - Manuela Bartoli
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (H.A.); (M.C.T.); (D.R.G.)
- Correspondence: ; Tel.: +706-721-9797 or +706-721-7910; Fax: +706-721-9799
| |
Collapse
|
33
|
Traboulsi H, Cherian M, Abou Rjeili M, Preteroti M, Bourbeau J, Smith BM, Eidelman DH, Baglole CJ. Inhalation Toxicology of Vaping Products and Implications for Pulmonary Health. Int J Mol Sci 2020; 21:E3495. [PMID: 32429092 PMCID: PMC7278963 DOI: 10.3390/ijms21103495] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/28/2020] [Accepted: 05/12/2020] [Indexed: 12/17/2022] Open
Abstract
E-cigarettes have a liquid that may contain flavors, solvents, and nicotine. Heating this liquid generates an aerosol that is inhaled into the lungs in a process commonly referred to as vaping. E-cigarette devices can also contain cannabis-based products including tetrahydrocannabinol (THC), the psychoactive component of cannabis (marijuana). E-cigarette use has rapidly increased among current and former smokers as well as youth who have never smoked. The long-term health effects are unknown, and emerging preclinical and clinical studies suggest that e-cigarettes may not be harmless and can cause cellular alterations analogous to traditional tobacco smoke. Here, we review the historical context and the components of e-cigarettes and discuss toxicological similarities and differences between cigarette smoke and e-cigarette aerosol, with specific reference to adverse respiratory outcomes. Finally, we outline possible clinical disorders associated with vaping on pulmonary health and the recent escalation of acute lung injuries, which led to the declaration of the vaping product use-associated lung injury (EVALI) outbreak. It is clear there is much about vaping that is not understood. Consequently, until more is known about the health effects of vaping, individual factors that need to be taken into consideration include age, current and prior use of combustible tobacco products, and whether the user has preexisting lung conditions such as asthma and chronic obstructive pulmonary disease (COPD).
Collapse
Affiliation(s)
- Hussein Traboulsi
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (H.T.); (M.A.R.); (M.P.); (J.B.); (B.M.S.)
| | - Mathew Cherian
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (M.C.); (D.H.E.)
| | - Mira Abou Rjeili
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (H.T.); (M.A.R.); (M.P.); (J.B.); (B.M.S.)
- Respiratory Epidemiology and Clinical Research Unit, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Matthew Preteroti
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (H.T.); (M.A.R.); (M.P.); (J.B.); (B.M.S.)
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Jean Bourbeau
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (H.T.); (M.A.R.); (M.P.); (J.B.); (B.M.S.)
- Respiratory Epidemiology and Clinical Research Unit, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Benjamin M. Smith
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (H.T.); (M.A.R.); (M.P.); (J.B.); (B.M.S.)
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (M.C.); (D.H.E.)
- Respiratory Epidemiology and Clinical Research Unit, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - David H. Eidelman
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (M.C.); (D.H.E.)
| | - Carolyn J. Baglole
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (H.T.); (M.A.R.); (M.P.); (J.B.); (B.M.S.)
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (M.C.); (D.H.E.)
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada
| |
Collapse
|
34
|
Ran J, Liu M, Feng J, Li H, Ma H, Song T, Cao Y, Zhou P, Wu Y, Yang Y, Yang Y, Yu F, Guo H, Zhang L, Xie S, Li D, Gao J, Zhang X, Zhu X, Zhou J. ASK1-Mediated Phosphorylation Blocks HDAC6 Ubiquitination and Degradation to Drive the Disassembly of Photoreceptor Connecting Cilia. Dev Cell 2020; 53:287-299.e5. [PMID: 32275885 DOI: 10.1016/j.devcel.2020.03.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 01/20/2020] [Accepted: 03/12/2020] [Indexed: 01/08/2023]
Abstract
Retinopathy of prematurity (ROP) is a leading cause of childhood blindness. However, the pathogenesis and molecular mechanisms underlying ROP remain elusive. Herein, using the oxygen-induced retinopathy (OIR) mouse model of ROP, we demonstrate that disassembly of photoreceptor connecting cilia is an early event in response to oxygen changes. Histone deacetylase 6 (HDAC6) is upregulated in the retina of OIR mice and accumulates in the transition zone of connecting cilia. We also show that in response to oxygen changes, apoptosis signal-regulating kinase 1 (ASK1) is activated and phosphorylates HDAC6, blocking its ubiquitination by von Hippel-Lindau and subsequent degradation by the proteasome. Moreover, depletion of HDAC6 or inhibition of the ASK1/HDAC6 axis protects mice from oxygen-change-induced pathological changes of photoreceptors. These findings reveal a critical role for ASK1/HDAC6-mediated connecting cilium disassembly in the OIR mouse model of ROP and suggest a potential value of ASK1/HDAC6-targeted agents for prevention of this disease.
Collapse
Affiliation(s)
- Jie Ran
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Min Liu
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China.
| | - Jie Feng
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Haixia Li
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Huixian Ma
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Ting Song
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Yu Cao
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Peng Zhou
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Yuhan Wu
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Yunfan Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yang Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Fan Yu
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Heng Guo
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Liang Zhang
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Songbo Xie
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Dengwen Li
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jinmin Gao
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Xiaomin Zhang
- Eye Institute, School of Optometry and Ophthalmology, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Xueliang Zhu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jun Zhou
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China; State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
35
|
Chambers ED, White A, Vang A, Wang Z, Ayala A, Weng T, Blackburn M, Choudhary G, Rounds S, Lu Q. Blockade of equilibrative nucleoside transporter 1/2 protects against Pseudomonas aeruginosa-induced acute lung injury and NLRP3 inflammasome activation. FASEB J 2020; 34:1516-1531. [PMID: 31914698 PMCID: PMC7045807 DOI: 10.1096/fj.201902286r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 11/11/2022]
Abstract
Pseudomonas aeruginosa infections are increasingly multidrug resistant and cause healthcare-associated pneumonia, a major risk factor for acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). Adenosine is a signaling nucleoside with potential opposing effects; adenosine can either protect against acute lung injury via adenosine receptors or cause lung injury via adenosine receptors or equilibrative nucleoside transporter (ENT)-dependent intracellular adenosine uptake. We hypothesized that blockade of intracellular adenosine uptake by inhibition of ENT1/2 would increase adenosine receptor signaling and protect against P. aeruginosa-induced acute lung injury. We observed that P. aeruginosa (strain: PA103) infection induced acute lung injury in C57BL/6 mice in a dose- and time-dependent manner. Using ENT1/2 pharmacological inhibitor, nitrobenzylthioinosine (NBTI), and ENT1-null mice, we demonstrated that ENT blockade elevated lung adenosine levels and significantly attenuated P. aeruginosa-induced acute lung injury, as assessed by lung wet-to-dry weight ratio, BAL protein levels, BAL inflammatory cell counts, pro-inflammatory cytokines, and pulmonary function (total lung volume, static lung compliance, tissue damping, and tissue elastance). Using both agonists and antagonists directed against adenosine receptors A2AR and A2BR, we further demonstrated that ENT1/2 blockade protected against P. aeruginosa -induced acute lung injury via activation of A2AR and A2BR. Additionally, ENT1/2 chemical inhibition and ENT1 knockout prevented P. aeruginosa-induced lung NLRP3 inflammasome activation. Finally, inhibition of inflammasome prevented P. aeruginosa-induced acute lung injury. Our results suggest that targeting ENT1/2 and NLRP3 inflammasome may be novel strategies for prevention and treatment of P. aeruginosa-induced pneumonia and subsequent ARDS.
Collapse
Affiliation(s)
- Eboni D. Chambers
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Alpert Medical School of Brown University, Providence, RI 02908
| | - Alexis White
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Alpert Medical School of Brown University, Providence, RI 02908
| | - Alexander Vang
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Alpert Medical School of Brown University, Providence, RI 02908
| | - Zhengke Wang
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Alpert Medical School of Brown University, Providence, RI 02908
| | - Alfred Ayala
- Division of Surgical Research, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02908
| | - Tingting Weng
- Departments of Biochemistry and Molecular Biology, University of Texas-Houston Medical School, Houston, TX 77030
| | - Michael Blackburn
- Departments of Biochemistry and Molecular Biology, University of Texas-Houston Medical School, Houston, TX 77030
| | - Gaurav Choudhary
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Alpert Medical School of Brown University, Providence, RI 02908
| | - Sharon Rounds
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Alpert Medical School of Brown University, Providence, RI 02908
| | - Qing Lu
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Alpert Medical School of Brown University, Providence, RI 02908
| |
Collapse
|
36
|
Pulmonary Endothelial Cell Apoptosis in Emphysema and Acute Lung Injury. ADVANCES IN ANATOMY EMBRYOLOGY AND CELL BIOLOGY 2019; 228:63-86. [PMID: 29288386 DOI: 10.1007/978-3-319-68483-3_4] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Apoptosis plays an essential role in homeostasis and pathogenesis of a variety of human diseases. Endothelial cells are exposed to various environmental and internal stress and endothelial apoptosis is a pathophysiological consequence of these stimuli. Pulmonary endothelial cell apoptosis initiates or contributes to progression of a number of lung diseases. This chapter will focus on the current understanding of the role of pulmonary endothelial cell apoptosis in the development of emphysema and acute lung injury (ALI) and the factors controlling pulmonary endothelial life and death.
Collapse
|
37
|
Zong D, Liu X, Li J, Ouyang R, Chen P. The role of cigarette smoke-induced epigenetic alterations in inflammation. Epigenetics Chromatin 2019; 12:65. [PMID: 31711545 PMCID: PMC6844059 DOI: 10.1186/s13072-019-0311-8] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022] Open
Abstract
Background Exposure to cigarette smoke (CS) is a major threat to human health worldwide. It is well established that smoking increases the risk of respiratory diseases, cardiovascular diseases and different forms of cancer, including lung, liver, and colon. CS-triggered inflammation is considered to play a central role in various pathologies by a mechanism that stimulates the release of pro-inflammatory cytokines. During this process, epigenetic alterations are known to play important roles in the specificity and duration of gene transcription. Main text Epigenetic alterations include three major modifications: DNA modifications via methylation; various posttranslational modifications of histones, namely, methylation, acetylation, phosphorylation, and ubiquitination; and non-coding RNA sequences. These modifications work in concert to regulate gene transcription in a heritable fashion. The enzymes that regulate these epigenetic modifications can be activated by smoking, which further mediates the expression of multiple inflammatory genes. In this review, we summarize the current knowledge on the epigenetic alterations triggered by CS and assess how such alterations may affect smoking-mediated inflammatory responses. Conclusion The recognition of the molecular mechanisms of the epigenetic changes in abnormal inflammation is expected to contribute to the understanding of the pathophysiology of CS-related diseases such that novel epigenetic therapies may be identified in the near future.
Collapse
Affiliation(s)
- Dandan Zong
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Xiangming Liu
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Jinhua Li
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Ruoyun Ouyang
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China
| | - Ping Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China. .,Research Unit of Respiratory Disease, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
38
|
Ko J, Mills T, Huang J, Chen NY, Mertens TCJ, Collum SD, Lee G, Xiang Y, Han L, Zhou Y, Lee CG, Elias JA, Jyothula SSK, Rajagopal K, Karmouty-Quintana H, Blackburn MR. Transforming growth factor β1 alters the 3'-UTR of mRNA to promote lung fibrosis. J Biol Chem 2019; 294:15781-15794. [PMID: 31488543 DOI: 10.1074/jbc.ra119.009148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/27/2019] [Indexed: 12/18/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic disease characterized by the pathological remodeling of air sacs as a result of excessive accumulation of extracellular matrix (ECM) proteins, but the mechanism governing the robust protein expression is poorly understood. Our recent findings demonstrate that alternative polyadenylation (APA) caused by NUDT21 reduction is important for the increased expression of fibrotic mediators and ECM proteins in lung fibroblasts by shortening the 3'-untranslated regions (3'-UTRs) of mRNAs and stabilizing their transcripts, therefore activating pathological signaling pathways. Despite the importance of NUDT21 reduction in the regulation of fibrosis, the underlying mechanisms for the depletion are unknown. We demonstrate here that NUDT21 is depleted by TGFβ1. We found that miR203, which is increased in IPF, was induced by TGFβ1 to target the NUDT21 3'-UTR, thus depleting NUDT21 in human and mouse lung fibroblasts. TGFβ1-mediated NUDT21 reduction was attenuated by the miR203 inhibitor antagomiR203 in fibroblasts. TGFβ1 transgenic mice revealed that TGFβ1 down-regulates NUDT21 in fibroblasts in vivo Furthermore, TGFβ1 promoted differential APA of fibrotic genes, including FGF14, RICTOR, TMOD2, and UCP5, in association with increased protein expression. This unique differential APA signature was also observed in IPF fibroblasts. Altogether, our results identified TGFβ1 as an APA regulator through NUDT21 depletion amplifying pulmonary fibrosis.
Collapse
Affiliation(s)
- Junsuk Ko
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030.,MD Anderson UTHealth Graduate School, the University of Texas Health Science Center, Houston, Texas 77030
| | - Tingting Mills
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030
| | - Jingjing Huang
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210003 Jiangsu, China
| | - Ning-Yuan Chen
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030
| | - Tinne C J Mertens
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030
| | - Scott D Collum
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030.,MD Anderson UTHealth Graduate School, the University of Texas Health Science Center, Houston, Texas 77030
| | - Garam Lee
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030
| | - Yu Xiang
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030
| | - Leng Han
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030.,MD Anderson UTHealth Graduate School, the University of Texas Health Science Center, Houston, Texas 77030
| | - Yang Zhou
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912
| | - Soma S K Jyothula
- Department of Internal Medicine, McGovern Medical School, the University of Texas Health Science Center, Houston, Texas 77030
| | - Keshava Rajagopal
- Department of Internal Medicine, McGovern Medical School, the University of Texas Health Science Center, Houston, Texas 77030
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030.,MD Anderson UTHealth Graduate School, the University of Texas Health Science Center, Houston, Texas 77030
| | - Michael R Blackburn
- Department of Biochemistry and Molecular Biology, the University of Texas Health Science Center, Houston, Texas 77030 .,MD Anderson UTHealth Graduate School, the University of Texas Health Science Center, Houston, Texas 77030
| |
Collapse
|
39
|
Szucs B, Szucs C, Petrekanits M, Varga JT. Molecular Characteristics and Treatment of Endothelial Dysfunction in Patients with COPD: A Review Article. Int J Mol Sci 2019; 20:E4329. [PMID: 31487864 PMCID: PMC6770145 DOI: 10.3390/ijms20184329] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/23/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD) show systemic consequences, such as chronic systemic inflammation leading to changes in the airway, airway penetrability, and endothelial function. Endothelial dysfunction is characterized by a list of alterations of endothelium towards reduced vasodilation, proinflammatory state, detachment and apoptosis of endothelial cells, and development of atherosclerosis. COPD-induced endothelial dysfunction is associated with elevated cardiovascular risk. The increment of physical activities such as pulmonary rehabilitation (PR) training have a significant effect on COPD, thus, PR can be an integrative part of COPD treatment. In this narrative review the focus is on the function of endothelial inflammatory mediators [cytokines, chemokines, and cellular proteases] and pulmonary endothelial cells and endothelial dysfunction in COPD as well as the effects of dysfunction of the endothelium may play in COPD-related pulmonary hypertension. The relationship between smoking and endothelial dysfunction is also discussed. The connection between different pulmonary rehabilitation programs, arterial stiffness and pulse wave velocity (PWV) is presented. Endothelial dysfunction is a significant prognostic factor of COPD, which can be characterized by PWV. We discuss future considerations, like training programs, as an important part of the treatment that has a favorable impact on the endothelial function.
Collapse
Affiliation(s)
- Botond Szucs
- PharmaFlight Research and Training Center, H-4030 Debrecen, Hungary
| | - Csilla Szucs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen H-4032, Hungary
| | - Mate Petrekanits
- Institute of Exercise Physiology and Sport Medicine, University of Physical Education, H-1123 Budapest, Hungary
| | - Janos T Varga
- Department of Pulmonary Rehabilitation, National Koranyi Institute for Pulmonology, H-1121 Budapest, Hungary.
| |
Collapse
|
40
|
McDowell SAC, Quail DF. Immunological Regulation of Vascular Inflammation During Cancer Metastasis. Front Immunol 2019; 10:1984. [PMID: 31497019 PMCID: PMC6712555 DOI: 10.3389/fimmu.2019.01984] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 08/06/2019] [Indexed: 12/30/2022] Open
Abstract
Metastasis is the predominant cause of cancer-related mortality, despite being a highly inefficient process overall. The vasculature is the gatekeeper for tumor cell seeding within the secondary tissue microenvironment—the rate limiting step of the metastatic cascade. Therefore, factors that regulate vascular physiology dramatically influence cancer outcomes. There are a myriad of physiologic circumstances that not only influence the intrinsic capacity of tumor cells to cross the endothelial barrier, but also that regulate vascular inflammation and barrier integrity to enable extravasation into the metastatic niche. These processes are highly dependent on inflammatory cues largely initiated by the innate immune compartment, that are meant to help re-establish tissue homeostasis, but instead become hijacked by cancer cells. Here, we discuss the scientific advances in understanding the interactions between innate immune cells and the endothelium, describe their influence on cancer metastasis, and evaluate potential therapeutic interventions for the alleviation of metastatic disease. By triangulating the relationship between immune cells, endothelial cells, and tumor cells, we will gain greater insight into how to impede the metastatic process by focusing on its most vulnerable phases, thereby reducing metastatic spread and cancer-related mortality.
Collapse
Affiliation(s)
- Sheri A C McDowell
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada.,Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Daniela F Quail
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada.,Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada.,Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
41
|
Wu D, Birukov K. Endothelial Cell Mechano-Metabolomic Coupling to Disease States in the Lung Microvasculature. Front Bioeng Biotechnol 2019; 7:172. [PMID: 31380363 PMCID: PMC6658821 DOI: 10.3389/fbioe.2019.00172] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/03/2019] [Indexed: 12/15/2022] Open
Abstract
Lungs are the most vascular part of humans, accepting the totality of cardiac output in a volume much smaller than the body itself. Due to this cardiac output, the lung microvasculature is subject to mechanical forces including shear stress and cyclic stretch that vary with the cardiac and breathing cycle. Vessels are surrounded by extracellular matrix which dictates the stiffness which endothelial cells also sense and respond to. Shear stress, stiffness, and cyclic stretch are known to influence endothelial cell state. At high shear stress, endothelial cells exhibit cell quiescence marked by low inflammatory markers and high nitric oxide synthesis, whereas at low shear stress, endothelial cells are thought to "activate" into a pro-inflammatory state and have low nitric oxide. Shear stress' profound effect on vascular phenotype is most apparent in the arterial vasculature and in the pathophysiology of vascular inflammation. To conduct the flow of blood from the right heart, the lung microvasculature must be rigid yet compliant. It turns out that excessive substrate rigidity or stiffness is important in the development of pulmonary hypertension and chronic fibrosing lung diseases via excessive cell proliferation or the endothelial-mesenchymal transition. Recently, a new body of literature has evolved that couples mechanical sensing to endothelial phenotypic changes through metabolic signaling in clinically relevant contexts such as pulmonary hypertension, lung injury syndromes, as well as fibrosis, which is the focus of this review. Stretch, like flow, has profound effect on endothelial phenotype; metabolism studies due to stretch are in their infancy.
Collapse
Affiliation(s)
- David Wu
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Konstantin Birukov
- Department of Anesthesia, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
42
|
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
|
43
|
Cigarette Smoke Induced Lung Barrier Dysfunction, EMT, and Tissue Remodeling: A Possible Link between COPD and Lung Cancer. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2025636. [PMID: 31341890 PMCID: PMC6613007 DOI: 10.1155/2019/2025636] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/02/2019] [Indexed: 12/13/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer, closely related to smoking, are major lung diseases affecting millions of individuals worldwide. The generated gas mixture of smoking is proved to contain about 4,500 components such as carbon monoxide, nicotine, oxidants, fine particulate matter, and aldehydes. These components were considered to be the principle factor driving the pathogenesis and progression of pulmonary disease. A large proportion of lung cancer patients showed a history of COPD, which demonstrated that there might be a close relationship between COPD and lung cancer. In the early stages of smoking, lung barrier provoked protective response and DNA repair are likely to suppress these changes to a certain extent. In the presence of long-term smoking exposure, these mechanisms seem to be malfunctioned and lead to disease progression. The infiltration of inflammatory cells to mucosa, submucosa, and glandular tissue caused by inhaled cigarette smoke is responsible for the destruction of matrix, blood supply shortage, and epithelial cell death. Conversely, cancer cells have the capacity to modulate the proliferation of epithelial cells and produce of new vascular networks. Comprehension understanding of mechanisms responsible for both pathologies is necessary for the prevention and treatment of COPD and lung cancer. In this review, we will summarize related articles and give a glance of possible mechanism between cigarette smoking induced COPD and lung cancer.
Collapse
|
44
|
Chen Y, Wu Z, Zhu X, Zhang M, Zang X, Li X, Xu Y. OCT4B-190 protects against ischemic stroke by modulating GSK-3β/HDAC6. Exp Neurol 2019; 316:52-62. [DOI: 10.1016/j.expneurol.2019.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/25/2019] [Accepted: 04/10/2019] [Indexed: 01/09/2023]
|
45
|
Kaufman MJ, Kanayama G, Hudson JI, Pope HG. Supraphysiologic-dose anabolic-androgenic steroid use: A risk factor for dementia? Neurosci Biobehav Rev 2019; 100:180-207. [PMID: 30817935 PMCID: PMC6451684 DOI: 10.1016/j.neubiorev.2019.02.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/13/2019] [Accepted: 02/17/2019] [Indexed: 02/06/2023]
Abstract
Supraphysiologic-dose anabolic-androgenic steroid (AAS) use is associated with physiologic, cognitive, and brain abnormalities similar to those found in people at risk for developing Alzheimer's Disease and its related dementias (AD/ADRD), which are associated with high brain β-amyloid (Aβ) and hyperphosphorylated tau (tau-P) protein levels. Supraphysiologic-dose AAS induces androgen abnormalities and excess oxidative stress, which have been linked to increased and decreased expression or activity of proteins that synthesize and eliminate, respectively, Aβ and tau-P. Aβ and tau-P accumulation may begin soon after initiating supraphysiologic-dose AAS use, which typically occurs in the early 20s, and their accumulation may be accelerated by other psychoactive substance use, which is common among non-medical AAS users. Accordingly, the widespread use of supraphysiologic-dose AAS may increase the numbers of people who develop dementia. Early diagnosis and correction of sex-steroid level abnormalities and excess oxidative stress could attenuate risk for developing AD/ADRD in supraphysiologic-dose AAS users, in people with other substance use disorders, and in people with low sex-steroid levels or excess oxidative stress associated with aging.
Collapse
Affiliation(s)
- Marc J Kaufman
- McLean Imaging Center, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA.
| | - Gen Kanayama
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - James I Hudson
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Harrison G Pope
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
46
|
Amara CS, Ambati CR, Vantaku V, Badrajee Piyarathna DW, Donepudi SR, Ravi SS, Arnold JM, Putluri V, Chatta G, Guru KA, Badr H, Terris MK, Bollag RJ, Sreekumar A, Apolo AB, Putluri N. Serum Metabolic Profiling Identified a Distinct Metabolic Signature in Bladder Cancer Smokers: A Key Metabolic Enzyme Associated with Patient Survival. Cancer Epidemiol Biomarkers Prev 2019; 28:770-781. [PMID: 30642841 DOI: 10.1158/1055-9965.epi-18-0936] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/29/2018] [Accepted: 12/28/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The current system to predict the outcome of smokers with bladder cancer is insufficient due to complex genomic and transcriptomic heterogeneities. This study aims to identify serum metabolite-associated genes related to survival in this population. METHODS We performed LC/MS-based targeted metabolomic analysis for >300 metabolites in serum obtained from two independent cohorts of bladder cancer never smokers, smokers, healthy smokers, and healthy never smokers. A subset of differential metabolites was validated using Biocrates absoluteIDQ p180 Kit. Genes associated with differential metabolites were integrated with a publicly available cohort of The Cancer Genome Atlas (TCGA) to obtain an intersecting signature specific for bladder cancer smokers. RESULTS Forty metabolites (FDR < 0.25) were identified to be differential between bladder cancer never smokers and smokers. Increased abundance of amino acids (tyrosine, phenylalanine, proline, serine, valine, isoleucine, glycine, and asparagine) and taurine were observed in bladder cancer smokers. Integration of differential metabolomic gene signature and transcriptomics data from TCGA cohort revealed an intersection of 17 genes that showed significant correlation with patient survival in bladder cancer smokers. Importantly, catechol-O-methyltransferase, iodotyrosine deiodinase, and tubulin tyrosine ligase showed a significant association with patient survival in publicly available bladder cancer smoker datasets and did not have any clinical association in never smokers. CONCLUSIONS Serum metabolic profiling of bladder cancer smokers revealed dysregulated amino acid metabolism. It provides a distinct gene signature that shows a prognostic value in predicting bladder cancer smoker survival. IMPACT Serum metabolic signature-derived genes act as a predictive tool for studying the bladder cancer progression in smokers.
Collapse
Affiliation(s)
- Chandra Sekhar Amara
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas
| | - Chandrashekar R Ambati
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, Texas
| | - Venkatrao Vantaku
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas
| | | | - Sri Ramya Donepudi
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, Texas
| | - Shiva Shankar Ravi
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas
| | - James M Arnold
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas
| | - Vasanta Putluri
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, Texas
| | - Gurkamal Chatta
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Khurshid A Guru
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Hoda Badr
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | | | | | - Arun Sreekumar
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas
| | - Andrea B Apolo
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| | - Nagireddy Putluri
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
47
|
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
|
48
|
Hussain M, Xu C, Yao M, Zhang Q, Wu J, Wu X, Lu M, Tang L, Wu F, Wu X. CRTH2 antagonist, CT‑133, effectively alleviates cigarette smoke-induced acute lung injury. Life Sci 2019; 216:156-167. [PMID: 30468833 DOI: 10.1016/j.lfs.2018.11.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/16/2018] [Accepted: 11/18/2018] [Indexed: 01/01/2023]
Abstract
AIMS Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), characterized by overwhelming lung inflammation, are associated with high mortality. Cigarette smoke (CS) is one of the major causes of ALI/ARDS. Since high expression of prostaglandin (PG) D2 has been observed in CS-induced lung injury. Currently, no effective pharmacological therapies are available to treat ALI, and supportive therapies remain the mainstay of treatment. Therefore, we investigated the protective effect of CT‑133, a newly discovered selective CRTH2 antagonist, on CS-induced ALI in vivo and in vitro. MAIN METHODS CT‑133 (10 and 30 mg/kg), dexamethasone (1 mg/kg) and normal saline were intratracheally administrated 1 hr prior to whole-body CS-exposure for seven consecutive days to study the key characteristics of ALI. Subsequently, CSE (4%)- and PGD2-stimulated RAW 264.7 macrophages were used to evaluate the protective effect of CT‑133. KEY FINDINGS CT‑133 remarkably attenuated infiltration of inflammatory cells, neutrophils, and macrophages in the BALF, albumin contents, expression of IL‑1β, IL‑6, TNF‑α and KC, lung myeloperoxidase (MPO) activity and lung histopathological alterations caused by CS exposure in mice. Moreover, CT‑133 not only reversed the uncontrolled secretion of IL‑1β, IL-6, TNF‑α and KC from CSE- and PGD2-stimulated RAW 264.7 macrophages but also augmented IL-10 production in both in vivo and in vitro studies. Additionally, CT‑133 alleviated in vitro neutrophil migration chemoattracted by PGD2. SIGNIFICANCE Our results provide the first evidence that targeting CRTH2 could be a new potential therapeutic option to treat CS-induced ALI.
Collapse
Affiliation(s)
- Musaddique Hussain
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China
| | - Chengyun Xu
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China
| | - Minli Yao
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China
| | - Qin Zhang
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China
| | - Junsong Wu
- Department of Critical Care Medicine and Orthopedics, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou City 310009, China
| | - Xiling Wu
- Department of Respiratory Medicine, the Affiliated Children Hospital, Zhejiang University School of Medicine, Hangzhou City 310052, China
| | - Meiping Lu
- Department of Respiratory Medicine, the Affiliated Children Hospital, Zhejiang University School of Medicine, Hangzhou City 310052, China
| | - Lanfang Tang
- Department of Respiratory Medicine, the Affiliated Children Hospital, Zhejiang University School of Medicine, Hangzhou City 310052, China
| | - Fugen Wu
- Department of Pediatrics, The First People's Hospital of Wenling City, Wenling City 317500, China.
| | - Ximei Wu
- Department of Pharmacology, Zhejiang University, School of Medicine, Hangzhou City 310058, China.
| |
Collapse
|
49
|
Karki P, Meliton A, Sitikov A, Tian Y, Ohmura T, Birukova AA. Microtubule destabilization caused by particulate matter contributes to lung endothelial barrier dysfunction and inflammation. Cell Signal 2018; 53:246-255. [PMID: 30339829 DOI: 10.1016/j.cellsig.2018.10.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 10/15/2018] [Accepted: 10/15/2018] [Indexed: 11/25/2022]
Abstract
Exposure to particulate matter (PM) associated with air pollution remains a major public health concern, as it has been linked to significant increase in cardiopulmonary morbidity and mortality. Lung endothelial cell (EC) dysfunction is one of the hallmarks of cardiovascular events of lung exposure to PM. However, the role of PM in acute lung injury (ALI) exacerbation and delayed recovery remains incompletely understood. This study tested a hypothesis that PM augments lung injury and EC barrier dysfunction via microtubule-dependent mechanisms. Our data demonstrate that in pulmonary EC PM caused time- and dose-dependent remodeling of actin cytoskeleton and considerable destabilization of the microtubule (MT) network. These events led to the weakening of cell junctions and formation of actin stress fibers, resulting in disruption of lung EC monolayer and increased permeability. PM also caused ROS-dependent activation of MT-specific deacetylase, HDAC6. Suppression of HDAC6 activity by pharmacological inhibitors or siRNA-based depletion of HDAC6 abolished PM-induced EC permeability increase, which was accompanied by reduced activation of stress kinase signaling, inhibition of Rho cascade, decreased IL-6 production and suppressed activation of its downstream target STAT3. Pretreatment of pulmonary EC with IL-6 inhibitor led to inhibition of STAT3 activity and decreased PM-induced hyper-permeability. Because one of the major activators of Rho-GTPase, GEFH1, is localized on the MT, we examined its involvement in PM-caused EC barrier compromise. Inhibition of GEF-H1 activation significantly attenuated PM-induced permeability increase. Moreover, combined inhibition of IL-6 and GEF-H1 signaling exhibited additive protective effect. Taken together, these results demonstrate a critical involvement of MT-associated signaling in the PM-induced exacerbation of lung EC barrier compromise and inflammatory response.
Collapse
Affiliation(s)
- Pratap Karki
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Angelo Meliton
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, United States
| | - Albert Sitikov
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, United States
| | - Yufeng Tian
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, United States
| | - Tomomi Ohmura
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, United States
| | - Anna A Birukova
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, United States.
| |
Collapse
|
50
|
Rounds S, Lu Q. Cigarette smoke alters lung vascular permeability and endothelial barrier function (2017 Grover Conference Series). Pulm Circ 2018; 8:2045894018794000. [PMID: 30036149 PMCID: PMC6153538 DOI: 10.1177/2045894018794000] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Smoking of tobacco products continues to be widespread, despite recent progress
in decreasing use. Both in the United States and worldwide, cigarette smoking is
a major cause of morbidity and mortality. Growing evidence indicates that acute
respiratory distress syndrome (ARDS) is among the consequences of cigarette
smoking. Based on the topic from the 2017 Grover Conference, we review evidence
that cigarette smoking increases lung vascular permeability using both acute and
longer exposures of mice to cigarette smoke (CS). We also review studies
indicating that CS extract disrupts cultured lung endothelial cell barrier
function through effects on focal adhesion contacts, adherens junctions, actin
cytoskeleton, and microtubules. Among the potentially injurious components of
CS, the reactive aldehyde, acrolein, similarly increases lung vascular
permeability and disrupts barrier function. We speculate that inhibition of
aldehyde-induced lung vascular permeability may prevent CS-induced lung
injury.
Collapse
Affiliation(s)
- Sharon Rounds
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Pulmonary, Critical Care & Sleep Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Qing Lu
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Pulmonary, Critical Care & Sleep Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|