1
|
Pang X, Liu X. Immune Dysregulation in Chronic Obstructive Pulmonary Disease. Immunol Invest 2024; 53:652-694. [PMID: 38573590 DOI: 10.1080/08820139.2024.2334296] [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] [Indexed: 04/05/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease whose incidence increase with age and is characterised by chronic inflammation and significant immune dysregulation. Inhalation of toxic substances cause oxidative stress in the lung tissue as well as airway inflammation, under the recruitment of chemokines, immune cells gathered and are activated to play a defensive role. However, persistent inflammation damages the immune system and leads to immune dysregulation, which is mainly manifested in the reduction of the body's immune response to antigens, and immune cells function are impaired, further destroy the respiratory defensive system, leading to recurrent lower respiratory infections and progressive exacerbation of the disease, thus immune dysregulation play an important role in the pathogenesis of COPD. This review summarizes the changes of innate and adaptive immune-related cells during the pathogenesis of COPD, aiming to control COPD airway inflammation and improve lung tissue remodelling by regulating immune dysregulation, for further reducing the risk of COPD progression and opening new avenues of therapeutic intervention in COPD.
Collapse
Affiliation(s)
- Xichen Pang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaoju Liu
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
2
|
Zheng S, Bulut GB, Kummarapurugu AB, Ma J, Voynow JA. Neutrophil Elastase Degrades Histone Deacetylases and Sirtuin 1 in Primary Human Monocyte Derived Macrophages. Int J Mol Sci 2024; 25:4265. [PMID: 38673851 PMCID: PMC11050352 DOI: 10.3390/ijms25084265] [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: 03/05/2024] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Neutrophil elastase (NE) is taken up by macrophages, retains intracellular protease activity, and induces a pro-inflammatory phenotype. However, the mechanism of NE-induced pro-inflammatory polarization of macrophages is not well understood. We hypothesized that intracellular NE degrades histone deacetylases (HDAC) and Sirtuins, disrupting the balance of lysine acetylation and deacetylation and resulting in nuclear to cytoplasmic translocation of a major alarmin, High Mobility Group Box 1 (HMGB1), a pro-inflammatory response in macrophages. Human blood monocytes were obtained from healthy donors or from subjects with cystic fibrosis (CF) or chronic obstructive pulmonary disease (COPD). Monocytes were differentiated into blood monocyte derived macrophages (BMDMs) in vitro. Human BMDMs were exposed to NE or control vehicle, and the abundance of HDACs and Sirtuins was determined by Western blotting of total cell lysates or nuclear extracts or determined by ELISA. HDAC, Sirtuin, and Histone acetyltransferase (HAT) activities were measured. NE degraded most HDACs and Sirtuin (Sirt)1, resulting in decreased HDAC and sirtuin activities, with minimal change in HAT activity. We then evaluated whether the NE-induced loss of Sirt activity or loss of HDAC activities would alter the cellular localization of HMGB1. NE treatment or treatment with Trichostatin A (TSA), a global HDAC inhibitor, both increased HMGB1 translocation from the nucleus to the cytoplasm, consistent with HMGB1 activation. NE significantly degraded Class I and II HDAC family members and Sirt 1, which shifted BMDMs to a pro-inflammatory phenotype.
Collapse
Affiliation(s)
| | | | | | | | - Judith A. Voynow
- Department of Pediatric Pulmonary Medicine, Children’s Hospital of Richmond at Virginia Commonwealth University, Richmond, VA 23219, USA; (S.Z.); (G.B.B.); (A.B.K.); (J.M.)
| |
Collapse
|
3
|
Wang L, Yu Q, Xiao J, Chen Q, Fang M, Zhao H. Cigarette Smoke Extract-Treated Mouse Airway Epithelial Cells-Derived Exosomal LncRNA MEG3 Promotes M1 Macrophage Polarization and Pyroptosis in Chronic Obstructive Pulmonary Disease by Upregulating TREM-1 via m 6A Methylation. Immune Netw 2024; 24:e3. [PMID: 38725674 PMCID: PMC11076299 DOI: 10.4110/in.2024.24.e3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/13/2023] [Accepted: 01/02/2024] [Indexed: 05/12/2024] Open
Abstract
Cigarette smoke extract (CSE)-treated mouse airway epithelial cells (MAECs)-derived exosomes accelerate the progression of chronic obstructive pulmonary disease (COPD) by upregulating triggering receptor expressed on myeloid cells 1 (TREM-1); however, the specific mechanism remains unclear. We aimed to explore the potential mechanisms of CSE-treated MAECs-derived exosomes on M1 macrophage polarization and pyroptosis in COPD. In vitro, exosomes were extracted from CSE-treated MAECs, followed by co-culture with macrophages. In vivo, mice exposed to cigarette smoke (CS) to induce COPD, followed by injection or/and intranasal instillation with oe-TREM-1 lentivirus. Lung function and pathological changes were evaluated. CD68+ cell number and the levels of iNOS, TNF-α, IL-1β (M1 macrophage marker), and pyroptosis-related proteins (NOD-like receptor family pyrin domain containing 3, apoptosis-associated speck-like protein containing a caspase-1 recruitment domain, caspase-1, cleaved-caspase-1, gasdermin D [GSDMD], and GSDMD-N) were examined. The expression of maternally expressed gene 3 (MEG3), spleen focus forming virus proviral integration oncogene (SPI1), methyltransferase 3 (METTL3), and TREM-1 was detected and the binding relationships among them were verified. MEG3 increased N6-methyladenosine methylation of TREM-1 by recruiting SPI1 to activate METTL3. Overexpression of TREM-1 or METTL3 negated the alleviative effects of MEG3 inhibition on M1 polarization and pyroptosis. In mice exposed to CS, EXO-CSE further aggravated lung injury, M1 polarization, and pyroptosis, which were reversed by MEG3 inhibition. TREM-1 overexpression negated the palliative effects of MEG3 inhibition on COPD mouse lung injury. Collectively, CSE-treated MAECs-derived exosomal long non-coding RNA MEG3 may expedite M1 macrophage polarization and pyroptosis in COPD via the SPI1/METTL3/TREM-1 axis.
Collapse
Affiliation(s)
- Lijing Wang
- Department of Geriatrics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qiao Yu
- Department of Geriatrics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jian Xiao
- Department of Geriatrics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qiong Chen
- Department of Geriatrics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Min Fang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, the “Double-First Class” Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha 410219, China
| | - Hongjun Zhao
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
4
|
Rojas A, Lindner C, Schneider I, Gonzalez I, Uribarri J. The RAGE Axis: A Relevant Inflammatory Hub in Human Diseases. Biomolecules 2024; 14:412. [PMID: 38672429 PMCID: PMC11048448 DOI: 10.3390/biom14040412] [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: 03/04/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
In 1992, a transcendental report suggested that the receptor of advanced glycation end-products (RAGE) functions as a cell surface receptor for a wide and diverse group of compounds, commonly referred to as advanced glycation end-products (AGEs), resulting from the non-enzymatic glycation of lipids and proteins in response to hyperglycemia. The interaction of these compounds with RAGE represents an essential element in triggering the cellular response to proteins or lipids that become glycated. Although initially demonstrated for diabetes complications, a growing body of evidence clearly supports RAGE's role in human diseases. Moreover, the recognizing capacities of this receptor have been extended to a plethora of structurally diverse ligands. As a result, it has been acknowledged as a pattern recognition receptor (PRR) and functionally categorized as the RAGE axis. The ligation to RAGE leads the initiation of a complex signaling cascade and thus triggering crucial cellular events in the pathophysiology of many human diseases. In the present review, we intend to summarize basic features of the RAGE axis biology as well as its contribution to some relevant human diseases such as metabolic diseases, neurodegenerative, cardiovascular, autoimmune, and chronic airways diseases, and cancer as a result of exposure to AGEs, as well as many other ligands.
Collapse
Affiliation(s)
- Armando Rojas
- Biomedical Research Laboratories, Faculty of Medicine, Catholic University of Maule, Talca 34600000, Chile; (A.R.); (I.G.)
| | - Cristian Lindner
- Department of Radiology, Faculty of Medicine, University of Concepción, Concepción 4030000, Chile;
| | - Ivan Schneider
- Centre of Primary Attention, South Metropolitan Health Service, Santiago 3830000, Chile;
| | - Ileana Gonzalez
- Biomedical Research Laboratories, Faculty of Medicine, Catholic University of Maule, Talca 34600000, Chile; (A.R.); (I.G.)
| | - Jaime Uribarri
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10021, USA
| |
Collapse
|
5
|
Zhang J, Ren G, Huang T, Sang Y, Zhong Y, Yi Y. miRNA-363-3p Hinders Proliferation, Migration, Invasion and Autophagy of Thyroid Cancer Cells by Controlling SYT1 Transcription to affect NF-κB. Endocr Metab Immune Disord Drug Targets 2024; 24:153-162. [PMID: 37150983 DOI: 10.2174/1871530323666230504112553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/28/2023] [Accepted: 03/10/2023] [Indexed: 05/09/2023]
Abstract
BACKGROUND Thyroid cancer (TC) is a frequent endocrine malignant tumor with various pathologic types. miRNA-363-3p plays a pivotal part in the occurrence, development, prognosis, and treatment of cancer. OBJECTIVE To explore the mechanism of miRNA-363-3p in TC and provide a new idea for targeted therapy of TC. METHODS Differential miRNAs and downstream target mRNAs in TC tissues were predicted with bioinformatics analysis. Expression levels of miRNA-363-3p and Synaptotagmin I (SYT1) in TC cells were ascertained by qRT-PCR. Cell migration, invasion, and proliferation were detected by wound healing assay, transwell assay, colony formation assay, CCK-8, and BrdU fluorescence experiment, respectively. Flow cytometry was utilized to detect the levels of apoptosis and necrosis. Immunofluorescence assay was used for detecting autophagosome formation in cells, and the expression levels of autophagy-related proteins, as well as NF-κB related proteins, were measured by western blot. Dual-luciferase reporter gene assay was applied for detecting the interaction between miRNA-363-3p and SYT1. RESULTS miRNA-363-3p was prominently down-regulated in TC cells. miRNA-363-3p overexpression suppressed migration, invasion, and proliferation, promoting apoptosis and necrosis of TC cells. As the downstream target of miRNA-363-3p, SYT1 was up-regulated in TC cells. SYT1 overexpression reversed the inhibition of TC cell proliferation, invasion, migration, and autophagy mediated by miRNA-363-3p overexpression. In addition, miRNA-363-3p overexpression inhibited the activation of the NF-κB pathway in cells, while further overexpression of SYT1 weakened the inhibition of miRNA-363-3p overexpression on the NF-κB pathway. CONCLUSION miRNA-363-3p affected the NF-κB signaling pathway by down-regulating SYT1 expression to inhibit the malignant progression of TC cells, providing theoretical support for the treatment of TC.
Collapse
Affiliation(s)
- Jizong Zhang
- Department of General Surgery, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing City, Jiangsu Province, 210009, China
| | - Guanghui Ren
- Department of General Surgery, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing City, Jiangsu Province, 210009, China
| | - Tao Huang
- Department of General Surgery, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing City, Jiangsu Province, 210009, China
| | - Yiming Sang
- Department of General Surgery, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing City, Jiangsu Province, 210009, China
| | - Yan Zhong
- Department of General Surgery, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing City, Jiangsu Province, 210009, China
| | - Yongxiang Yi
- Department of General Surgery, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing City, Jiangsu Province, 210009, China
| |
Collapse
|
6
|
Yıldız S, Kılıçaslan AK, Emir BS, Uğur K, Kılıç F. Serum HMGB1 and Beclin 1 Levels in Patients with a Diagnosis of Schizophrenia. TURK PSIKIYATRI DERGISI = TURKISH JOURNAL OF PSYCHIATRY 2024; 35:1-7. [PMID: 38556931 PMCID: PMC11003366 DOI: 10.5080/u27030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 05/06/2022] [Indexed: 04/02/2024]
Abstract
OBJECTIVE It is known that inflammation plays a role in the etiopathogenesis of schizophrenia. In this study, we examined high mobility group box 1 protein (HMGB1) and Beclin 1 levels and their relationship with clinical variables in patients with schizophrenia. METHOD Forty-three patients with schizophrenia and 43 healthy controls were included in this study. The patients were administered sociodemographic data form, the Positive Negative Symptoms Assessment Scale (PANSS) and the Clinical Global Impressions (CGI) scale. After the scales were filled, venous blood samples were taken from both the patient and control groups to measure serum HMGB1 and Beclin 1 levels. Serum samples obtained at the end of centrifugation were measured by Enzyme-Linked ImmunoSorbent Assay (ELISA) method. RESULTS The mean serum HMGB1 levels were significantly increased and the mean serum Beclin 1 levels were significantly decreased in the schizophrenia group compared to the control group. In addition, a negative correlation was found between HMGB1 and Beclin 1 levels. CONCLUSION In conclusion, current research shows that HMGB1 is increased and Beclin 1 is decreased in patients with schizophrenia, and these findings may contribute to the role of autophagy in the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Sevler Yıldız
- Assoc. Prof., Binali Yıldırım University, Faculty of Medicine, Department of Psychiatry, Erzincan
| | | | - Burcu Sırlıer Emir
- Psychiatrist, Elazığ Fethi Sekin City Hospital, Department of Psychiatry, Elazığ
| | - Kerim Uğur
- Assoc. Prof., Turgut Özal University, Faculty of Medicine, Department of Psychiatry, Malatya
| | - Faruk Kılıç
- Assoc. Prof., Süleyman Demirel University, Faculty of Medicine, Department of Psychiatry, Isparta, Turkey
| |
Collapse
|
7
|
Cui X, Yao A, Jia L. Starvation insult induces the translocation of high mobility group box 1 to cytosolic compartments in glioma. Oncol Rep 2023; 50:216. [PMID: 37888772 PMCID: PMC10636726 DOI: 10.3892/or.2023.8653] [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: 05/03/2023] [Accepted: 10/04/2023] [Indexed: 10/28/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a highly conserved and ubiquitous nuclear protein in eukaryotic cells. In response to stress, it transfers from the nucleus to the cytoplasm and finally, to the extracellular matrix, participating in inflammation and carcinogenesis. Increased HMGB1 protein levels are frequently associated with the reduced survival of patients with glioma. HMGB1 plays contextual roles depending on its subcellular localization. However, the mechanisms underlying its subcellular localization and secretion remain unclear. In the present study, the subcellular localization and secretion of HMGB1 in starved glioma cells were investigated using immunofluorescence microscopy, enzyme‑linked immunosorbent assay, subcellular fractionation, western blotting and immunoelectron microscopy. The results demonstrated that starvation induced HMGB1 translocation from the nucleus to the cytoplasm and finally, to the extracellular milieu in glioma cells. HMGB1 was localized in the mitochondria, endoplasmic reticulum (ER), peroxisomes, autophagosomes, lysosomes, endosomes and the cytoskeleton. Immunoelectron microscopy confirmed that HMGB1 was present within or around cytosolic compartments. Subcellular fractionation further demonstrated that HMGB1 transferred to membrane‑bound compartments. In addition, HMGB1 was localized to specific contact areas between the ER and mitochondria, known as mitochondria‑associated membranes. On the whole, the results of the present study suggest that starvation induces HMGB1 secretion, which can be inhibited through the suppression of autophagy. Starvation insult induces HMGB1 translocation to the cytosolic compartments of glioma cells, and autophagy may be involved in the extracellular secretion of HMGB1 in starved glioma cells.
Collapse
Affiliation(s)
- Xiaohang Cui
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Anhui Yao
- Department of Neurosurgery, 988th Hospital of Joint Logistic Support Force of PLA, Zhengzhou, Henan 450053, P.R. China
| | - Liyun Jia
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
8
|
Meng X, Guo S, Zhang X, Jiao B, Yang X, Li M, Li C, He J, Chen S, Peng C, Shao H, Jia Q. HMGB1 inhibition reduces TDI-induced occupational asthma through ROS/AMPK/autophagy pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 266:115575. [PMID: 37839183 DOI: 10.1016/j.ecoenv.2023.115575] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 10/17/2023]
Abstract
Exposure to toluene diisocyanate (TDI) can cause pulmonary diseases such as asthma. Inhibition of high mobility group box 1 protein (HMGB1) has been found to be protective against the toxic effects of TDI on human bronchial epithelial (HBE) cells. Here, we evaluated the in vivo positive roles of HMGB1 in the TDI-caused asthma mice and explored its underlying mechanisms in HBE cells. We found that suppression of HMGB1 obviously alleviated airway inflammation, airway hyperresponsiveness, and airway remodeling in the lung tissue of the asthma mice. The in vitro results showed that inhibition of HMGB1 ameliorated TDI-induced reactive oxygen species (ROS) release, inflammatory response, and activation of autophagy in HBE cells. At the molecular level, inhibition of HMGB1 decreased the expressions of HMGB1, Toll-like receptor 4, Vimentin and matrix metalloproteinase-9 proteins, activated NF-κB and NOD-like receptor protein 3 (NLRP3) inflammasome, and increased E-cadherin expression. Importantly, activation of autophagy could lead to the overactivation of NLRP3 inflammasome in TDI-induced asthma. These results suggest that inhibition of HMGB1 can alleviate TDI-induced asthma through ROS/AMPK/autophagy pathways, which may provide valuable evidence for the pathogenesis and therapeutic targets of TDI-induced asthma.
Collapse
Affiliation(s)
- Xiangjing Meng
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Sumei Guo
- Erqi District Center for Disease Control and Prevention, Zhengzhou, Henan 450052, China
| | - Xiaoxia Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Bo Jiao
- National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention (CDC), Beijing 100050, China
| | - Xiaohan Yang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Ming Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Chao Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Jin He
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Shangya Chen
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Cheng Peng
- Eusyn Institute of Health Science, Brisbane, Queensland 4108, Australia
| | - Hua Shao
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China.
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China.
| |
Collapse
|
9
|
Schröder L, Rupp ABA, Gihr KME, Kobilay M, Domroese CM, Mallmann MR, Holdenrieder S. Immunogenic Biomarkers HMGB1 and sRAGE Are Potential Diagnostic Tools for Ovarian Malignancies. Cancers (Basel) 2023; 15:5081. [PMID: 37894448 PMCID: PMC10605106 DOI: 10.3390/cancers15205081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND High mobility group box 1 (HMGB1), soluble receptor of advanced glycation end products (sRAGE) and programmed cell death markers PD-1 and PD-L1 are immunogenic serum biomarkers that may serve as novel diagnostic tools for cancer diagnosis. METHODS We investigated the four markers in sera of 231 women, among them 76 with ovarian cancer, 87 with benign diseases and 68 healthy controls, using enzyme immunoassays. Discrimination between groups was calculated using receiver operating characteristic (ROC) curves and sensitivities at fixed 90% and 95% specificities. RESULTS HMGB1 levels were significantly elevated and sRAGE levels were decreased in cancer patients as compared to benign and healthy controls. In consequence, the ratio of HMGB1 and sRAGE discriminated best between diagnostic groups. The areas under the curve (AUCs) of the ROC curves for differentiation of cancer vs. healthy were 0.77 for HMGB1, 0.65 for sRAGE and 0.78 for the HMGB1/sRAGE ratio, and slightly lower for the differentiation of cancer vs. benigns with 0.72 for HMGB1, 0.61 for sRAGE and 0.74 for the ratio of both. The highest sensitivities for cancer detection at 90% specificity versus benign diseases were achieved using HMGB1 with 41.3% and the HMGB1/sRAGE ratio with 39.2%, followed by sRAGE with 18.9%. PD-1 showed only minor and PD-L1 no power for discrimination between ovarian cancer and benign diseases. CONCLUSION HMGB1 and sRAGE have differential diagnostic potential for ovarian cancer detection and warrant inclusion in further validation studies.
Collapse
Affiliation(s)
- Lars Schröder
- Department of Obstetrics and Gynecology, University Hospital Cologne, 50931 Cologne, Germany
- Department of Obstetrics and Gynecology, Ketteler Hospital, 63071 Offenbach, Germany
| | - Alexander B. A. Rupp
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
- Munich Biomarker Research Center, Institute of Laboratory Medicine, German Heart Centre, Technical University Munich, 80636 Munich, Germany
| | - Kathrin M. E. Gihr
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Makbule Kobilay
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Christian M. Domroese
- Department of Obstetrics and Gynecology, University Hospital Cologne, 50931 Cologne, Germany
| | - Michael R. Mallmann
- Department of Obstetrics and Gynecology, University Hospital Cologne, 50931 Cologne, Germany
| | - Stefan Holdenrieder
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
- Munich Biomarker Research Center, Institute of Laboratory Medicine, German Heart Centre, Technical University Munich, 80636 Munich, Germany
- CEBIO GmbH—Center for Evaluation of Biomarkers, 81679 Munich, Germany
| |
Collapse
|
10
|
Lei Y, Zhu Y, Mallah MA, Lu P, Yang L, He X, Shang P, Chen Y, Zhou X, Feng F, Zhang Q. The activation of SIRT1 ameliorates BPDE-induced inflammatory damage in BEAS-2B cells via HMGB1/TLR4/NF-κB pathway. ENVIRONMENTAL TOXICOLOGY 2023; 38:2429-2439. [PMID: 37436145 DOI: 10.1002/tox.23878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/08/2023] [Accepted: 06/05/2023] [Indexed: 07/13/2023]
Abstract
Benzo(a)pyrene-7,8-dihydrodiol-9,10-epoxide (BPDE), the metabolite of environmental pollutant benzo(a)pyrene (B(a)P) could induce pulmonary toxicity and inflammation. SIRT1, an NAD+ -dependent histone deacetylase, is known to regulate inflammation in the occurrence and development of various diseases, but its effects on BPDE-induced acute lung injury are still unknown. The present study aimed to explore the role of SIRT1 in BPDE-induced acute lung injury. Here, human bronchial epithelial (HBE) cells (BEAS-2B) cells were stimulated with BPDE at different concentrations (0.50, 0.75, and 1.00 μmol/L) for 24 h, we found that the levels of cytokines in the supernatant were increased and the expression of SIRT1 in cells was down-regulated, at the same time, BPDE stimulation up-regulated the protein expression of HMGB1, TLR4, and p-NF-κBp65 in BEAS-2B cells. Then the activator and inhibitor of SIRT1 were used before BPDE exposure, it was shown that the activation of SIRT1 significantly attenuated the levels of inflammatory cytokines and HMGB1, and reduced the expression of HMGB1, AC-HMGB1, TLR4, and p-NF-κBp65 protein; while these results were reversed by the inhibition of SIRT1. This study revealed that the SIRT1 activation may protect against BPDE-induced inflammatory damage in BEAS-2B cells by regulating the HMGB1/TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Yanting Lei
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Yonghang Zhu
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Manthar Ali Mallah
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Ping Lu
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Liu Yang
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Xi He
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Pingping Shang
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute, CNC, Zhengzhou, China
| | - Yusong Chen
- Quality Supervision & Test Center, China National Tobacco Corporation Shandong Branch, Jinan, China
| | - Xiaolei Zhou
- Department of Pulmonary Medicine, Henan Provincial Chest Hospital, Zhengzhou University, Zhengzhou, China
| | - Feifei Feng
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, China
| | - Qiao Zhang
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, China
| |
Collapse
|
11
|
Suarez JS, Novelli F, Goto K, Ehara M, Steele M, Kim JH, Zolondick AA, Xue J, Xu R, Saito M, Pastorino S, Minaai M, Takanishi Y, Emi M, Pagano I, Wakeham A, Berger T, Pass HI, Gaudino G, Mak TW, Carbone M, Yang H. HMGB1 released by mesothelial cells drives the development of asbestos-induced mesothelioma. Proc Natl Acad Sci U S A 2023; 120:e2307999120. [PMID: 37729199 PMCID: PMC10523480 DOI: 10.1073/pnas.2307999120] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/09/2023] [Indexed: 09/22/2023] Open
Abstract
Asbestos is the main cause of malignant mesothelioma. Previous studies have linked asbestos-induced mesothelioma to the release of HMGB1 from the nucleus to the cytoplasm, and from the cytoplasm to the extracellular space. In the cytoplasm, HMGB1 induces autophagy impairing asbestos-induced cell death. Extracellularly, HMGB1 stimulates the secretion of TNFα. Jointly, these two cytokines kick-start a chronic inflammatory process that over time promotes mesothelioma development. Whether the main source of extracellular HMGB1 were the mesothelial cells, the inflammatory cells, or both was unsolved. This information is critical to identify the targets and design preventive/therapeutic strategies to interfere with asbestos-induced mesothelioma. To address this issue, we developed the conditional mesothelial HMGB1-knockout (Hmgb1ΔpMeso) and the conditional myelomonocytic-lineage HMGB1-knockout (Hmgb1ΔMylc) mouse models. We establish here that HMGB1 is mainly produced and released by the mesothelial cells during the early phases of inflammation following asbestos exposure. The release of HMGB1 from mesothelial cells leads to atypical mesothelial hyperplasia, and in some animals, this evolves over the years into mesothelioma. We found that Hmgb1ΔpMeso, whose mesothelial cells cannot produce HMGB1, show a greatly reduced inflammatory response to asbestos, and their mesothelial cells express and secrete significantly reduced levels of TNFα. Moreover, the tissue microenvironment in areas of asbestos deposits displays an increased fraction of M1-polarized macrophages compared to M2 macrophages. Supporting the biological significance of these findings, Hmgb1ΔpMeso mice showed a delayed and reduced incidence of mesothelioma and an increased mesothelioma-specific survival. Altogether, our study provides a biological explanation for HMGB1 as a driver of asbestos-induced mesothelioma.
Collapse
Affiliation(s)
- Joelle S. Suarez
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
| | - Flavia Novelli
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
| | - Keisuke Goto
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima734-8551, Japan
| | - Michiko Ehara
- Department of Oral Pathology, Division of Oral Pathogenesis and Disease Control, School of Dentistry, Asahi University, Mizuho Gifu501-0296, Japan
| | - Mika Steele
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
| | - Jin-Hee Kim
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
| | - Alicia A. Zolondick
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI96822
| | - Jiaming Xue
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
- John A. Burns, School of Medicine, University of Hawai’i, Honolulu, HI96813
| | - Ronghui Xu
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
| | - Mai Saito
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
| | - Sandra Pastorino
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
| | - Michael Minaai
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
| | - Yasutaka Takanishi
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
| | - Mitsuru Emi
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
| | - Ian Pagano
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
| | - Andrew Wakeham
- Princess Margaret Cancer Center, University Health Network, Toronto, ONM5G 2M9, Canada
| | - Thorsten Berger
- Princess Margaret Cancer Center, University Health Network, Toronto, ONM5G 2M9, Canada
| | - Harvey I. Pass
- Department of Cardiothoracic Surgery, New York University, New York, NY10016
| | - Giovanni Gaudino
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
| | - Tak W. Mak
- Princess Margaret Cancer Center, University Health Network, Toronto, ONM5G 2M9, Canada
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR999077, China
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR999077, China
| | - Michele Carbone
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
| | - Haining Yang
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI96813
| |
Collapse
|
12
|
Wang X, Hu B, Hu H, Zhou S, Yin M, Cheng X, Zhang Z, Liu H. Tannic Acid Suppresses HBV Replication via the Regulation of NF-κB, MAPKs, and Autophagy in HepG2.2.15 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37450882 DOI: 10.1021/acs.jafc.3c00863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Hepatitis B virus (HBV) infection is a serious global health problem that threatens the health of human. Tannic acid (TA), a natural polyphenol in foods, fruits, and plants, exhibits a variety of bioactive functions. In our research, we decide to explore the pharmacological mechanism of TA against HBV replication. Our results showed that TA effectively reduced the content of HBV DNA and viral antigens (HBsAg and HBeAg) in HepG2.2.15 cells. Meanwhile, TA significantly decreased the mRNA expression of HBV RNA, which include total HBV RNA, HBV pregenomic RNA, and HBV precore mRNA. Besides, TA evidently downregulated the activity of HBV promoters in HepG2.2.15 cells. Furthermore, we found that TA upregulated the expression of IL-8, TNF-α, IFN-α, and IFN-α-mediated antiviral effectors in HepG2.2.15 cells. On the contrary, TA downregulated the expression of IL-10 and hepatic nuclear factor 4 (HNF4α). In addition, TA activated the NF-κB and MAPK pathways that contributed to the inhibition of HBV replication. Finally, TA treatment led to the occurrence of autophagy, which accelerated the elimination of HBV components in HepG2.2.15 cells. Taken together, our results elucidated the suppressive effect of TA on HBV replication and provided inspiration for its clinical application in HBV treatment.
Collapse
Affiliation(s)
- Xuefeng Wang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Baifei Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Haiming Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Shuhan Zhou
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Mingzhu Yin
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Xue Cheng
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Zhigang Zhang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Hongtao Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| |
Collapse
|
13
|
Geng C, Wang X, Chen J, Sun N, Wang Y, Li Z, Han L, Hou S, Fan H, Li N, Gong Y. Repetitive Low-Level Blast Exposure via Akt/NF-κB Signaling Pathway Mediates the M1 Polarization of Mouse Alveolar Macrophage MH-S Cells. Int J Mol Sci 2023; 24:10596. [PMID: 37445774 DOI: 10.3390/ijms241310596] [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: 04/19/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 07/15/2023] Open
Abstract
Repetitive low-level blast (rLLB) exposure is a potential risk factor for the health of soldiers or workers who are exposed to it as an occupational characteristic. Alveolar macrophages (AMs) are susceptible to external blast waves and produce pro-inflammatory or anti-inflammatory effects. However, the effect of rLLB exposure on AMs is still unclear. Here, we generated rLLB waves through a miniature manual Reddy-tube and explored their effects on MH-S cell morphology, phenotype transformation, oxidative stress status, and apoptosis by immunofluorescence, real-time quantitative PCR (qPCR), western blotting (WB) and flow cytometry. Ipatasertib (GDC-0068) or PDTC was used to verify the role of the Akt/NF-κB signaling pathway in these processes. Results showed that rLLB treatment could cause morphological irregularities and cytoskeletal disorders in MH-S cells and promote their polarization to the M1 phenotype by increasing iNOS, CD86 and IL-6 expression. The molecular mechanism is through the Akt/NF-κB signaling pathway. Moreover, we found reactive oxygen species (ROS) burst, Ca2+ accumulation, mitochondrial membrane potential reduction, and early apoptosis of MH-S cells. Taken together, our findings suggest rLLB exposure may cause M1 polarization and early apoptosis of AMs. Fortunately, it is blocked by specific inhibitors GDC-0068 or PDTC. This study provides a new treatment strategy for preventing and alleviating health damage in the occupational population caused by rLLB exposure.
Collapse
Affiliation(s)
- Chenhao Geng
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Xinyue Wang
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Jiale Chen
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Na Sun
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yuru Wang
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Zizheng Li
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Lu Han
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Shike Hou
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Ning Li
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yanhua Gong
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| |
Collapse
|
14
|
Golub A, Ordak M, Nasierowski T, Bujalska-Zadrozny M. Advanced Biomarkers of Hepatotoxicity in Psychiatry: A Narrative Review and Recommendations for New Psychoactive Substances. Int J Mol Sci 2023; 24:ijms24119413. [PMID: 37298365 DOI: 10.3390/ijms24119413] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
One of the factors that increase the effectiveness of the pharmacotherapy used in patients abusing various types of new psychoactive substances (NPSs) is the proper functioning of the liver. However, the articles published to date on NPS hepatotoxicity only address non-specific hepatic parameters. The aim of this manuscript was to review three advanced markers of hepatotoxicity in psychiatry, namely, osteopontin (OPN), high-mobility group box 1 protein (HMGB1) and glutathione dehydrogenase (GDH, GLDH), and, on this basis, to identify recommendations that should be included in future studies in patients abusing NPSs. This will make it possible to determine whether NPSs do indeed have a hepatotoxic effect or whether other factors, such as additional substances taken or hepatitis C virus (HCV) infection, are responsible. NPS abusers are at particular risk of HCV infection, and for this reason, it is all the more important to determine what factors actually show a hepatotoxic effect in them.
Collapse
Affiliation(s)
- Aniela Golub
- Department of Pharmacotherapy and Pharmaceutical Care, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland
| | - Michal Ordak
- Department of Pharmacotherapy and Pharmaceutical Care, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland
| | - Tadeusz Nasierowski
- Department of Psychiatry, Faculty of Pharmacy, Medical University of Warsaw, Nowowiejska 27 Str., 00-665 Warsaw, Poland
| | - Magdalena Bujalska-Zadrozny
- Department of Pharmacotherapy and Pharmaceutical Care, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland
| |
Collapse
|
15
|
Akkermansia muciniphila Ameliorates Lung Injury in Smoke-Induced COPD Mice by IL-17 and Autophagy. Cell Microbiol 2023. [DOI: 10.1155/2023/4091825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Objective. Smoking is a primary hazard factor for chronic obstructive pulmonary disease (COPD), which induced a decrease in intestinal Akkermansia muciniphila abundance and Th17 imbalance in COPD. This study analyzed the changes of gut microbiota metabolism and Akkermansia abundance in patients with smoking-related COPD and explored the potential function of Akkermansia muciniphila in smoke-induced COPD mice. Methods. Gut microbiota diversity and metabolic profile were analyzed by 16S rRNA sequence and metabolomics in COPD patients. The IL-1β, IL-17, TNF-α, and IL-6 levels were tested by ELISA. Lung tissue damage was observed by HE staining. The expression of cleave-caspase 3, trophoblast antigen 2 (TROP2), and LC3 in lung tissues were analyzed by IHC or IF. The p-mTOR, mTOR, p62, and LC3 expression in lung tissues were tested by western blot. Results. The levels of IL-17, IL-1β, TNF-α, and IL-6 in the peripheral blood of COPD patients increased significantly. The number and alpha diversity of gut microbiota were decreased in COPD patients. The abundance of Akkermansia muciniphila in gut of COPD patients was decreased, and the metabolic phenotype and retinol metabolism were changed. In the retinol metabolism, the retinol and retinal were significantly changed. Akkermansia muciniphila could improve the alveolar structure and inflammatory cell infiltration in lung tissue, reduce the IL-17, TNF-α, and IL-6 levels in peripheral blood, promote the p-mTOR expression, and inhibit the expression of autophagy-related proteins in smoke-induced COPD mice. Conclusion. The number and alpha diversity of gut microbiota were decreased in patients with smoking-related COPD, accompanied by decreased abundance of Akkermansia muciniphila, and altered retinol metabolism function. Gut Akkermansia muciniphila ameliorated lung injury in smoke-induced COPD mice by inflammation and autophagy.
Collapse
|
16
|
Zhao Y, Li R. HMGB1 is a promising therapeutic target for asthma. Cytokine 2023; 165:156171. [PMID: 36924610 DOI: 10.1016/j.cyto.2023.156171] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023]
Abstract
High-mobility group box protein 1 (HMGB1) is a non-histone deoxyribonucleic acid-binding nuclear protein. In physiological state it is involved in gene transctioripn regulation and cell replication, differentiation and maturation. HMGB1 is actively secreted into the extracellular space in the form of intracellular vesicles, upon stimulation of inflammation and infection, by monocytes, macrophages, dendritic cells (DCs), and other immune cells, and can also be passively released by necrotic or injured cells. After binding with the corresponding receptors, HMGB1 can activate the downstream substrate and trigger a series of biological effects. HMGB1 was mainly dependent on toll-like re ceptors (TLR) 2 and 4, and receptors for advanced glycation end products (RAGE) to trigger intracellular signal transduction, and mediate innate and adoptive immune responses. Besides these, studies have reported the participation of TLR3, TLR9, T-cell immunoglobulin mucin (TIM) 3, CD24, anti-N-methyl-D-aspartate receptor (NMDAR) in Th2 inflammatory response, eosinophilic airway inflammation, and airway hyperresponsiveness, mediated by HMGB1 in asthma. Both clinical and experimental studies suggested that HMGB1 was involved in the pathogenesis of asthma probably by regulating the downstream signaling pathways via corresponding receptors. This article reviews the role of HMGB1 in pathogenesis of asthma, and provides a new theoretical basis for the diagnosis and treatment of asthma.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Critical Care Medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology; Hubei Clinical Research Center for Infectious Diseases; Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences; Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, 430023, PR China; The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, PR China
| | - Ruiting Li
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China.
| |
Collapse
|
17
|
Zeng X, Xin J, Liu K, Deng W, Liu F. The protective role of HMGB1 in affecting the balance between autophagy and pyroptosis to maintain neutrophils homeostasis during β-glucan-induced mice lung inflammation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 252:114637. [PMID: 36774802 DOI: 10.1016/j.ecoenv.2023.114637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
Fungal contamination is omnipresent, and inhalation of fungi-contaminated organic dust leads to hypersensitivity pneumonitis (HP), in which neutrophils played a pivotal role. Existing studies have suggested that cell homeostasis is crucial for the pathogenesis of the inflammatory disease. Although HMGB1 has been shown to contribute to suppressing HP, there is a lack of studies on its mechanisms, especially the regulation of neutrophil homeostasis. This study aims to investigate how HMGB1 regulates neutrophil function by affecting neutrophil homeostasis, and then affects lung inflammation induced by β-glucan, the exposure marker of fungi. Our results showed that deficient HMGB1 led to neutrophil death by disrupting the balance between autophagy and pyroptosis after β-glucan treatment. And HMGB1 deficiency exacerbated the β-glucan-induced lung inflammation and neutrophil dysfunction both in vivo and in vitro. Furthermore, HMGB1 contributed to remodeling neutrophil function by restricting autophagy and aggravating pyroptosis β-glucan exposure. Our funding suggested that HMGB1 deficiency could break the balance between autophagy and pyroptosis towards pyroptosis to cause neutrophil dysfunction during the exacerbated inflammatory response, which provides insights into the pathogenesis of HP and the potential biological targets for its treatment. DATA AVAILABILITY: The datasets used during the current study are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Xinning Zeng
- School of Public Health, Jinzhou Medical University, Jinzhou, PR China; Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China
| | - Jiaxuan Xin
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China
| | - Kaiyue Liu
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China
| | - Wei Deng
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China
| | - Fangwei Liu
- Division of Pneumoconiosis, School of Public Health, China Medical University, Shenyang, PR China.
| |
Collapse
|
18
|
Dihydromyricetin inhibits Hepatitis B virus replication by activating NF-κB, MAPKs, and autophagy in HepG2.2.15 cells. Mol Biol Rep 2023; 50:1403-1414. [PMID: 36474061 DOI: 10.1007/s11033-022-07971-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 09/21/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hepatitis B virus (HBV) infection is a severe global health problem, and there has been no effective method to eliminate HBV. This study was designed to explore the pharmacological mechanism of Dihydromyricetin (DHM) treatment on HBV replication in vitro. METHODS AND RESULTS DHM is a flavonoid compound from Ampelopsis grossedentata. Using HepG2.2.15 cells, which can stably express HBV in vitro, we demonstrated that DHM treatment dramatically reduced HBV replication and secretions of HBsAg and HBeAg. Meanwhile, DHM inhibited mRNA expression of HBV RNAs in HepG2.2.15 cells, including Total HBV RNA, HBV pregenomic RNA (pgRNA), and HBV precore mRNA (pcRNA). Also, DHM elevated the mRNA expressions of inflammatory cytokines and antiviral effectors. In contrast, DHM decreased the mRNA level of HNF4α, which positively correlated with HBV replication. Further studies show that the activation of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathway played a critical role in DHM-initiated inhibition of HBV replication in HepG2.2.15 cells. Besides, activated autophagy was another contributor that may accelerate the clearance of HBV components. CONCLUSION In summary, DHM could suppress HBV replication by activating NF-κB, MAPKs, and autophagy in HepG2.2.15 cells. Our studies shed light on the future application of DHM for the clinical treatment of HBV infection.
Collapse
|
19
|
Cong Z, Yang C, Zeng Z, Wu C, Zhao F, Shen Z, Xiao H, Zhu X. α 1-adrenoceptor stimulation ameliorates lipopolysaccharide-induced lung injury by inhibiting alveolar macrophage inflammatory responses through NF-κB and ERK1/2 pathway in ARDS. Front Immunol 2023; 13:1090773. [PMID: 36685596 PMCID: PMC9853445 DOI: 10.3389/fimmu.2022.1090773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction Catecholamines such as norepinephrine or epinephrine have been reported to participate in the development of acute respiratory distress syndrome (ARDS) by activating adrenergic receptors (ARs). But the role of α1-AR in this process has yet to be elucidated. Methods In this study, ARDS mouse model was induced by intratracheal instillation of lipopolysaccharide. After treatment with α1-AR agonist phenylephrine or antagonist prazosin, lung pathological injury, alveolar barrier disruption and inflammation, and haemodynamic changes were evaluated. Cytokine levels and cell viability of alveolar macrophages were measured in vitro. Nuclear factor κB (NF-κB), mitogen-activated protein kinase, and Akt signalling pathways were analysed by western blot. Results It showed that α1-AR activation alleviated lung injuries, including reduced histopathological damage, cytokine expression, and inflammatory cell infiltration, and improved alveolar capillary barrier integrity of ARDS mice without influencing cardiovascular haemodynamics. In vitro experiments suggested that α1-AR stimulation inhibited secretion of TNF-α, IL-6, CXCL2/MIP-2, and promoted IL-10 secretion, but did not affect cell viability. Moreover, α1-AR stimulation inhibited NF-κB and enhanced ERK1/2 activation without significantly influencing p38, JNK, or Akt activation. Discussion Our studies reveal that α1-AR stimulation could ameliorate lipopolysaccharide-induced lung injury by inhibiting NF-κB and promoting ERK1/2 to suppress excessive inflammatory responses of alveolar macrophages.
Collapse
Affiliation(s)
- Zhukai Cong
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China,Department of Anaesthesiology, Peking University Third Hospital, Beijing, China
| | - Cui Yang
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Zhaojin Zeng
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Changyi Wu
- Department of Anaesthesiology, Peking University Third Hospital, Beijing, China
| | - Feng Zhao
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Ziyuan Shen
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China,National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China,Key Laboratory of Cardiovascular Receptors Research, Beijing, China,*Correspondence: Xi Zhu, ; Han Xiao,
| | - Xi Zhu
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China,*Correspondence: Xi Zhu, ; Han Xiao,
| |
Collapse
|
20
|
Harutyunyan KR, Abrahamyan HT, Adamyan SH, Mkrtchian S, Ter-Markosyan AS. Calcium-Regulating Hormonal System and HMGB1 in Cardiomyopathies. Endocr Metab Immune Disord Drug Targets 2023; 23:115-121. [PMID: 35980074 DOI: 10.2174/1871530322666220817110538] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Calcium ions play a key role in the heart's functional activity. The steadystate levels of calcium are contingent on the calcium regulating hormonal system, impairment of which might result in the development of cardiac pathology. An important role in these processes is also attributed to the specific inflammatory mediator, HMGB1, one of the damage-associated molecular patterns (DAMPs) released by immune cells or cell damage. OBJECTIVE This study investigated the cardioprotective potential of the calcium-regulating hormonal system in cardiomyopathies with an emphasis on the possible role of HMGB1. METHODS Ca2+ and inorganic phosphate levels were determined in the serum using an electrolyte analyzer and spectrophotometric analyzer correspondingly. The 1-34 fragment of parathyroid hormone (PTH), calcitonin, vitamin D, and HMGB1 were detected using ELISA kits. RESULTS The levels of PTH, calcitonin, phosphate, and HMGB1 were found elevated in females suffering from cardiomyopathy. The same tendency was observed in men; however, statistically significant changes were registered only for PTH and phosphate. CONCLUSION It can be suggested that among other reasons, the decrease of the left ventricular function in cardiomyopathy patients can be linked to the high HMGB1, whereas the activation of the calciumregulating system as manifested by the elevated PTH aims at restoration of calcium homeostasis and thus have positive, i.e. cardioprotective consequences.
Collapse
|
21
|
Wu J, Zhao X, Xiao C, Xiong G, Ye X, Li L, Fang Y, Chen H, Yang W, Du X. The role of lung macrophages in chronic obstructive pulmonary disease. Respir Med 2022; 205:107035. [PMID: 36343504 DOI: 10.1016/j.rmed.2022.107035] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/17/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) as a common, preventable and treatable chronic respiratory disease in clinic, gets continuous deterioration and we can't take effective intervention at present. Lung macrophages (LMs) are closely related to the occurrence and development of COPD, but the specific mechanism is not completely clear. In this review we will focus on the role of LMs and potential avenues for therapeutic targeting for LMs in COPD.
Collapse
Affiliation(s)
- Jianli Wu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xia Zhao
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Chuang Xiao
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China
| | - Guosheng Xiong
- Thoracic Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xiulin Ye
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Lin Li
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Yan Fang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Hong Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Weimin Yang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China.
| | - Xiaohua Du
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| |
Collapse
|
22
|
Qian Y, Wang W, Chen D, Zhu Y, Wang Y, Wang X. Cigarette smoking induces the activation of RIP2/caspase-12/NF- κB axis in oral squamous cell carcinoma. PeerJ 2022; 10:e14330. [PMID: 36353608 PMCID: PMC9639427 DOI: 10.7717/peerj.14330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Cigarette smoking is one of the major risk factors for the occurrence and progression of oral squamous cell carcinoma (OSCC). Receptor-interacting protein 2 (RIP2) has been involved in mucosal immunity and homeostasis via a positive regulation of nuclear factor κB (NF-κB) transcription factor activity. Caspase-12 can bind to RIP2 and dampen mucosal immunity. However, the roles of RIP2/NF-κB and caspase-12 in OSCC induced by cigarette smoking remain unknown. Herein, we investigated the effects of cigarette smoking on the RIP2/NF-κB and caspase-12 in human OSCC tissues and OSCC cell lines (HSC-3). We first observed that RIP2 mediated NF-κB activation and caspase-12 upregulation in OSCC patients with cigarette smoking and cigarette smoke extract (CSE)-treated HSC-3 cells, respectively. Moreover, we confirmed that the downregulation of RIP2 by siRNA resulted in the reduction of caspase-12 expression and NF-κB activity in the presence of CSE treatment in vitro. In summary, our results indicated that cigarette smoking induced the activation of the RIP2/caspase-12/NF-κB axis and it played an important role in the development of OSCC. The RIP2/caspase-12/NF-κB axis could be a target for OSCC prevention and treatment in the future.
Collapse
Affiliation(s)
- Yajie Qian
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenmei Wang
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Deyan Chen
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Yanan Zhu
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yong Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Xiang Wang
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
23
|
Lin L, Li J, Song Q, Cheng W, Chen P. The role of HMGB1/RAGE/TLR4 signaling pathways in cigarette smoke-induced inflammation in chronic obstructive pulmonary disease. Immun Inflamm Dis 2022; 10:e711. [PMID: 36301039 PMCID: PMC9552978 DOI: 10.1002/iid3.711] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/10/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common chronic respiratory disease with irreversible and continuous progression. It has become the fifth most burdensome disease and the third most deadly disease globally. Therefore, the prevention and treatment of COPD are urgent, and it is also important to clarify the pathogenesis of it. Smoking is the main and most common risk factor for COPD. Cigarette smoke (CS) can cause lung inflammation and other pathological mechanisms in the airways and lung tissue. Airway inflammation is one of the important mechanisms leading to the pathogenesis of COPD. Recent studies have shown that high mobility group box 1 (HMGB1) is involved in the occurrence and development of respiratory diseases, including COPD. HMGB1 is a typical damage-associated molecular pattern (DAMP) protein, which mainly exerts its activity by binding to the receptor for advanced glycation end products (RAGE) and toll-like receptor 4 (TLR4) and further participate in the process of airway inflammation. Studies have shown that the abnormal expression of HMGB1, RAGE, and TLR4 are related to inflammation in COPD. Herein, we discuss the roles of HMGB1, RAGE, and TLR4 in CS/cigarette smoke extract-induced inflammation in COPD, providing a new target for the diagnosis, treatment and prevention of COPD.
Collapse
Affiliation(s)
- Ling Lin
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Jing Li
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Qing Song
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Wei Cheng
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Ping Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| |
Collapse
|
24
|
Buscetta M, Cristaldi M, Cimino M, La Mensa A, Dino P, Bucchieri F, Rappa F, Amato S, Aronica TS, Pace E, Bertani A, Cipollina C. Cigarette smoke promotes inflammasome-independent activation of caspase-1 and -4 leading to gasdermin D cleavage in human macrophages. FASEB J 2022; 36:e22525. [PMID: 36004615 DOI: 10.1096/fj.202200837r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/02/2022] [Accepted: 08/17/2022] [Indexed: 11/11/2022]
Abstract
Mechanisms and consequences of gasdermin D (GSDMD) activation in cigarette smoke (CS)-associated inflammation and lung disease are unknown. GSDMD is a downstream effector of caspase-1, -8, and -4. Upon cleavage, GSDMD generates pores into cell membranes. Different degrees of GSDMD activation are associated with a range of physiological outputs ranging from cell hyperactivation to pyroptosis. We have previously reported that in human monocyte-derived macrophages CS extract (CSE) inhibits the NLRP3 inflammasome and shifts the response to lipopolysaccharide (LPS) towards the TLR4-TRIF axis leading to activation of caspase-8, which, in turn, activates caspase-1. In the present work, we investigated whether other ASC-dependent inflammasomes could be involved in caspase activation by CSE and whether caspase activation led to GSDMD cleavage and other downstream effects. Presented results demonstrate that CSE promoted ASC-independent activation of caspase-1 leading to GSDMD cleavage and increased cell permeability, in the absence of cell death. GSDMD cleavage was strongly enhanced upon stimulation with LPS+CSE, suggesting a synergistic effect between the two stimuli. Noteworthy, CSE promoted LPS internalization leading to caspase-4 activation, thus contributing to increased GSDMD cleavage. Caspase-dependent GSDMD cleavage was associated with mitochondrial superoxide generation. Increased cleaved GSDMD was found in lung macrophages of smokers compared to ex-smokers and non-smoking controls. Our findings revealed that ASC-independent activation of caspase-1, -4, and -8 and GSDMD cleavage upon exposure to CS may contribute to macrophage dysfunction and feed the chronic inflammation observed in the smokers' lung.
Collapse
Affiliation(s)
| | | | | | - Agnese La Mensa
- Fondazione RiMED, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Paola Dino
- Fondazione RiMED, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Fabio Bucchieri
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Francesca Rappa
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Santina Amato
- Azienda di Rilievo Nazionale ed Alta Specializzazione Ospedali (A.R.N.A.S) "Civico Di Cristina Benfratelli", Palermo, Italy
| | - Tommaso Silvano Aronica
- Azienda di Rilievo Nazionale ed Alta Specializzazione Ospedali (A.R.N.A.S) "Civico Di Cristina Benfratelli", Palermo, Italy
| | - Elisabetta Pace
- Istituto di Farmacologia Traslazionale (IFT)-CNR, Palermo, Italy
| | | | - Chiara Cipollina
- Fondazione RiMED, Palermo, Italy.,Istituto di Farmacologia Traslazionale (IFT)-CNR, Palermo, Italy
| |
Collapse
|
25
|
An integral perspective of canonical cigarette and e-cigarette-related cardiovascular toxicity based on the adverse outcome pathway framework. J Adv Res 2022:S2090-1232(22)00193-X. [PMID: 35998874 DOI: 10.1016/j.jare.2022.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/29/2022] [Accepted: 08/15/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Nowadays, cigarette smoking remains the leading cause of chronic disease and premature death, especially cardiovascular disease. As an emerging tobacco product, e-cigarettes have been advocated as alternatives to canonical cigarettes, and thus may be an aid to promote smoking cessation. However, recent studies indicated that e-cigarettes should not be completely harmless to the cardiovascular system. AIM OF REVIEW This review aimed to build up an integral perspective of cigarettes and e-cigarettes-related cardiovascular toxicity. KEY SCIENTIFIC CONCEPTS OF REVIEW This review adopted the adverse outcome pathway (AOP) framework as a pivotal tool and aimed to elucidate the association between the molecular initiating events (MIEs) induced by cigarette and e-cigarette exposure to the cardiovascular adverse outcome. Since the excessive generation of reactive oxygen species (ROS) has been widely approved to play a critical role in cigarette smoke-related CVD and may also be involved in e-cigarette-induced toxic effects, the ROS overproduction and subsequent oxidative stress are regarded as essential parts of this framework. As far as we know, this should be the first AOP framework focusing on cigarette and e-cigarette-related cardiovascular toxicity, and we hope our work to be a guide in exploring the biomarkers and novel therapies for cardiovascular injury.
Collapse
|
26
|
Chen Z, Wu H, Fan W, Zhang J, Yao Y, Su W, Wang Y, Li P. Naringenin suppresses BEAS-2B-derived extracellular vesicular cargoes disorder caused by cigarette smoke extract thereby inhibiting M1 macrophage polarization. Front Immunol 2022; 13:930476. [PMID: 35924248 PMCID: PMC9342665 DOI: 10.3389/fimmu.2022.930476] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Extracellular vesicles (EVs)-mediated epithelium-macrophage crosstalk has been proved to maintain lung homeostasis in cigarette smoke-induced lung diseases such as chronic obstructive pulmonary disease (COPD). In our previous study, we found that EVs derived from cigarette smoke extract (CSE) treated BEAS-2B promoted M1 macrophage polarization, which probably accelerated the development of inflammatory responses. Naringenin has been proved to suppress M1 macrophage polarization, but whether naringenin regulates macrophage polarization mediated by EVs has not been reported. In this study, we firstly found that EVs derived from naringenin and CSE co-treated BEAS-2B significantly inhibited the expression of CD86 and CD80 and the secretion of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1β, inducible nitric oxide synthase (iNOS), and IL-12 in macrophage induced by EVs derived from CSE-treated BEAS-2B. Further research revealed that naringenin downregulated BEAS-2B-derived EVs miR-21-3p which targeted phosphatase and tensin homolog deleted on chromosome ten/protein kinase B (PTEN/AKT) cascade in macrophages and then suppressed M1 macrophage polarization. Subsequent proteomics suggested that naringenin decreased BEAS-2B-derived EVs poly ADP-ribose polymerase (PARP)1 expression thereby suppressing M1 macrophage polarization probably. Our study provides novel pharmacological references for the mechanism of naringenin in the treatment of cigarette smoke-induced lung inflammatory diseases.
Collapse
|
27
|
GLP-1 mediates the neuroprotective action of crocin against cigarette smoking-induced cognitive disorders via suppressing HMGB1-RAGE/TLR4-NF-κB pathway. Int Immunopharmacol 2022; 110:108995. [PMID: 35785730 DOI: 10.1016/j.intimp.2022.108995] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/05/2022] [Accepted: 06/20/2022] [Indexed: 11/24/2022]
Abstract
Cigarette smoking (CS) has been associated with an increased risk of cognitive disorders. Although HMGB1 has been connected to various neurological ailments, its role in the pathogenesis of CS-induced cognitive impairments is undefined. With the ability of GLP-1 to lower HMGB1 expression and improve learning and memory performance, we sought to assess the potential neuroprotective efficacy of Crocin (Cro) as a GLP-1 stimulator against CS-induced cognitive impairments, with a focus on the HMGB1-RAGE/TLR4-NF-κB pathway. Fifty adult rats were specified into: Control; Cro (30 mg/kg); CS; Cro then CS and CS concurrently with Cro. Cognitive functions were assessed by MWM, EMP, and passive avoidance tests. Hippocampal levels of GLP-1, HMGB1, pro-inflammatory cytokines, and apoptotic markers were detected using ELISA, western blotting, and immunohistochemistry. Hippocampal oxidant/antioxidant status was evaluated via colorimetric determination of MDA and TAC. The results revealed that Cro either before or along with CS produced a significant improvement in learning and memory. Cro markedly hindered HMGB1-RAGE/TLR4-NF-κB pathway through enhancing GLP-1 level and expression, which in turn suppressed TNF-α and IL-1β levels and alleviated CS-induced neuroinflammation. Cro significantly counteracted CS-triggered oxidative stress as evidenced by reducing MDA level and raising TAC. Histopathologically, Cro lessened neuronal apoptosis by lowering Bax/Bcl-2 ratio at hippocampal CA2 region. These findings confirmed a GLP-1-dependent neuroprotective action of Cro against CS-induced cognitive disorders via suppressing HMGB1-RAGE/TLR4-NF-κB axis.
Collapse
|
28
|
Jin B, Li G, Zhou L, Fan Z. Mechanism Involved in Acute Liver Injury Induced by Intestinal Ischemia-Reperfusion. Front Pharmacol 2022; 13:924695. [PMID: 35694264 PMCID: PMC9185410 DOI: 10.3389/fphar.2022.924695] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/05/2022] [Indexed: 12/28/2022] Open
Abstract
Intestinal ischemia-reperfusion (I/R) is a common pathophysiological process, which can occur in many conditions such as acute enteric ischemia, severe burns, small intestinal transplantation, etc,. Ischemia-reperfusion of the intestine is often accompanied by distal organ injury, especially liver injury. This paper outlined the signal pathways and cytokines involved in acute liver injury induced by intestinal I/R: the NF-κB Signaling Pathway, the P66shc Signaling Pathway, the HMGB1 Signaling Pathway, the Nrf2-ARE Signaling Pathway, the AMPK-SIRT-1 Signaling Pathway and other cytokines, providing new ideas for the prevention and treatment of liver injury caused by reperfusion after intestinal I/R.
Collapse
Affiliation(s)
- Binghui Jin
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China.,Department of Central Laboratory, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Guangyao Li
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China.,Department of Central Laboratory, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Lin Zhou
- Department of Outpatient, the NO. 967 Hospital of PLA Joint Logistics Support Force, Dalian Medical University, Dalian, China
| | - Zhe Fan
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China.,Department of Central Laboratory, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| |
Collapse
|
29
|
Huang D, Su L, He C, Chen L, Huang D, Peng J, Yang F, Cao Y, Luo X. Pristimerin alleviates cigarette smoke-induced inflammation in chronic obstructive pulmonary disease via inhibiting NF-κB pathway. Biochem Cell Biol 2022; 100:223-235. [PMID: 35833632 DOI: 10.1139/bcb-2021-0251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cigarette smoke (CS) is a risk factor for chronic obstructive pulmonary disease (COPD), which can exacerbate inflammation and oxidative stress. Pristimerin (Pris) is a natural compound with antioxidant and anti-inflammatory effects. We managed to evaluate the protective effects of Pris on CS-induced COPD. The CS-induced COPD mice model and cell model were constructed. The effects of Pris treatment on lung function, inflammatory cell infiltration, myeloperoxidase (MPO), and pathological changes of lung tissues in mice model were evaluated. The impacts of Pris treatment on inflammatory factors, chemokines, and oxidative stress parameters in mice lung tissues and cells were determined by kits. The viability of human bronchial epithelial cells after Pris treatment was tested by CCK-8. The activation of NF-κB pathway was confirmed by Western blot and immunofluorescence. CS treatment impaired lung function, reduced weight of mice, and enhanced inflammatory cell infiltration, MPO, and lung tissue damage, but these effects of CS were reversed by Pris treatment. Furthermore, Pris treatment downregulated the levels of malondialdehyde, IL-6, IL-1β, TNF-α, CXCL1, and CXLC2, but upregulated superoxide dismutase and catalase levels. Pris treatment could overturn CS-induced activation of the NF-κB pathway. Pris alleviates CS-induced COPD by inactivating NF-κB pathway.
Collapse
Affiliation(s)
- Dongsheng Huang
- Department of Respiratory and Critical Care Medicine, Shenzhen Longhua District Central Hospital, Shenzhen City 518110, Guangdong Province, China
| | - Lianhui Su
- Department of Respiratory and Critical Care Medicine, Shenzhen Longhua District Central Hospital, Shenzhen City 518110, Guangdong Province, China
| | - Chaowen He
- Department of Respiratory and Critical Care Medicine, Shenzhen Longhua District Central Hospital, Shenzhen City 518110, Guangdong Province, China
| | - Licheng Chen
- Department of Respiratory and Critical Care Medicine, Shenzhen Longhua District Central Hospital, Shenzhen City 518110, Guangdong Province, China
| | - Dongxuan Huang
- Department of Respiratory and Critical Care Medicine, Shenzhen Longhua District Central Hospital, Shenzhen City 518110, Guangdong Province, China
| | - Jianfeng Peng
- Department of Respiratory and Critical Care Medicine, Shenzhen Longhua District Central Hospital, Shenzhen City 518110, Guangdong Province, China
| | - Fan Yang
- Department of Respiratory and Critical Care Medicine, Shenzhen Longhua District Central Hospital, Shenzhen City 518110, Guangdong Province, China
| | - Yahui Cao
- Department of Respiratory and Critical Care Medicine, Shenzhen Longhua District Central Hospital, Shenzhen City 518110, Guangdong Province, China
| | - Xiaohua Luo
- Department of Respiratory and Critical Care Medicine, Shenzhen Longhua District Central Hospital, Shenzhen City 518110, Guangdong Province, China
| |
Collapse
|
30
|
Role of Long Noncoding RNAs in Smoking-Induced Lung Cancer: An In Silico Study. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7169353. [PMID: 35529255 PMCID: PMC9070410 DOI: 10.1155/2022/7169353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/13/2022] [Accepted: 04/05/2022] [Indexed: 11/17/2022]
Abstract
The prevalence of lung cancer induced by cigarette smoking has increased over time. Long noncoding (lnc) RNAs, regulatory factors that play a role in human diseases, are commonly dysregulated in lung cancer. Cigarette smoking is closely related to changes in lncRNA expression, which can affect lung cancer. Herein, we assess the mechanism of lung cancer initiation induced by smoking. To calculate the impact of smoking on the survival of patients with lung cancer, we extracted data from The Cancer Genome Atlas and Gene Expression Omnibus databases and identified the differentially expressed genes in the lung cancer tissue compared to the normal lung tissue. Genes positively and negatively associated with smoking were identified. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and Cytoscape analyses were performed to determine the function of the genes and the effects of smoking on the immune microenvironment. lncRNAs corresponding to smoking-associated genes were identified, and a smoking-related lncRNA model was constructed using univariate and multivariate Cox analyses. This model was used to assess the survival of and potential risk in patients who smoked. During screening, 562 differentially expressed genes were identified, and we elucidated that smoking affected the survival of patients 4.5 years after the diagnosis of lung cancer. Furthermore, genes negatively associated with smoking were closely associated with immunity. Twelve immune cell types were also found to infiltrate differentially in smokers and nonsmokers. Thus, the smoking-associated lncRNA model is a good predictor of survival and risk in smokers and may be used as an independent prognostic factor for lung cancer.
Collapse
|
31
|
Moshensky A, Brand CS, Alhaddad H, Shin J, Masso-Silva JA, Advani I, Gunge D, Sharma A, Mehta S, Jahan A, Nilaad S, Olay J, Gu W, Simonson T, Almarghalani D, Pham J, Perera S, Park K, Al-Kolla R, Moon H, Das S, Byun MK, Shah Z, Sari Y, Heller Brown J, Crotty Alexander LE. Effects of mango and mint pod-based e-cigarette aerosol inhalation on inflammatory states of the brain, lung, heart, and colon in mice. eLife 2022; 11:e67621. [PMID: 35411847 PMCID: PMC9005188 DOI: 10.7554/elife.67621] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/04/2022] [Indexed: 12/13/2022] Open
Abstract
While health effects of conventional tobacco are well defined, data on vaping devices, including one of the most popular e-cigarettes which have high nicotine levels, are less established. Prior acute e-cigarette studies have demonstrated inflammatory and cardiopulmonary physiology changes while chronic studies have demonstrated extra-pulmonary effects, including neurotransmitter alterations in reward pathways. In this study we investigated the impact of inhalation of aerosols produced from pod-based, flavored e-cigarettes (JUUL) aerosols three times daily for 3 months on inflammatory markers in the brain, lung, heart, and colon. JUUL aerosol exposure induced upregulation of cytokine and chemokine gene expression and increased HMGB1 and RAGE in the nucleus accumbens in the central nervous system. Inflammatory gene expression increased in the colon, while gene expression was more broadly altered by e-cigarette aerosol inhalation in the lung. Cardiopulmonary inflammatory responses to acute lung injury with lipopolysaccharide were exacerbated in the heart. Flavor-specific findings were detected across these studies. Our findings suggest that daily e-cigarette use may cause neuroinflammation, which may contribute to behavioral changes and mood disorders. In addition, e-cigarette use may cause gut inflammation, which has been tied to poor systemic health, and cardiac inflammation, which leads to cardiovascular disease.
Collapse
Affiliation(s)
- Alex Moshensky
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Cameron S Brand
- Department of Pharmacology, University of California San Diego (UCSD)San DiegoUnited States
| | - Hasan Alhaddad
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of ToledoToledoUnited States
| | - John Shin
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Jorge A Masso-Silva
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Ira Advani
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Deepti Gunge
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Aditi Sharma
- Department of Pathology, University of California San Diego (UCSD)San DiegoUnited States
| | - Sagar Mehta
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Arya Jahan
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Sedtavut Nilaad
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Jarod Olay
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Wanjun Gu
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Tatum Simonson
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Daniyah Almarghalani
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of ToledoToledoUnited States
| | - Josephine Pham
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Samantha Perera
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Kenneth Park
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Rita Al-Kolla
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| | - Hoyoung Moon
- Department of Pharmacology, University of California San Diego (UCSD)San DiegoUnited States
| | - Soumita Das
- Department of Pathology, University of California San Diego (UCSD)San DiegoUnited States
| | - Min Kwang Byun
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
- Division of Pulmonology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of MedicineSeoulRepublic of Korea
| | - Zahoor Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of ToledoToledoUnited States
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of ToledoToledoUnited States
| | - Joan Heller Brown
- Department of Pharmacology, University of California San Diego (UCSD)San DiegoUnited States
| | - Laura E Crotty Alexander
- Pulmonary and Critical Care Section, VA San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary, Critical Care and Sleep Medicine and Section of Physiology, Department of Medicine, University of California San Diego (UCSD)San DiegoUnited States
| |
Collapse
|
32
|
High Mobility Group Box 1: Biological Functions and Relevance in Oxidative Stress Related Chronic Diseases. Cells 2022; 11:cells11050849. [PMID: 35269471 PMCID: PMC8909428 DOI: 10.3390/cells11050849] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/03/2022] [Accepted: 02/26/2022] [Indexed: 01/27/2023] Open
Abstract
In the early 1970s, a group of non-histone nuclear proteins with high electrophoretic mobility was discovered and named high-mobility group (HMG) proteins. High-mobility group box 1 (HMGB1) is the most studied HMG protein that detects and coordinates cellular stress response. The biological function of HMGB1 depends on its subcellular localization and expression. It plays a critical role in the nucleus and cytoplasm as DNA chaperone, chromosome gatekeeper, autophagy maintainer, and protector from apoptotic cell death. HMGB1 also functions as an extracellular alarmin acting as a damage-associated molecular pattern molecule (DAMP). Recent findings describe HMGB1 as a sophisticated signal of danger, with a pleiotropic function, which is useful as a clinical biomarker for several disorders. HMGB1 has emerged as a mediator in acute and chronic inflammation. Furthermore, HMGB1 targeting can induce beneficial effects on oxidative stress related diseases. This review focus on HMGB1 redox status, localization, mechanisms of release, binding with receptors, and its activities in different oxidative stress-related chronic diseases. Since a growing number of reports show the key role of HMGB1 in socially relevant pathological conditions, to our knowledge, for the first time, here we analyze the scientific literature, evaluating the number of publications focusing on HMGB1 in humans and animal models, per year, from 2006 to 2021 and the number of records published, yearly, per disease and category (studies on humans and animal models).
Collapse
|
33
|
Ma Y, He X, Liu X, Long Y, Chen Y. Endothelial Microparticles Derived from Primary Pulmonary Microvascular Endothelial Cells Mediate Lung Inflammation in Chronic Obstructive Pulmonary Disease by Transferring microRNA-126. J Inflamm Res 2022; 15:1399-1411. [PMID: 35250291 PMCID: PMC8896043 DOI: 10.2147/jir.s349818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/12/2022] [Indexed: 12/14/2022] Open
Abstract
Background Extracellular vesicles (EVs) are considered to new types of intercellular communication media, and microRNA is one of the most common transferring components of EVs. This study aimed to explore the potential role of endothelial microparticles (EMPs) derived from primary pulmonary microvascular endothelial cells in regulating lung inflammation of chronic obstructive pulmonary disease (COPD) through transferring microRNA-126 (miR-126). Methods EMPs generated from primary pulmonary microvascular endothelial cells were isolated by gradient centrifugation and characterized by transmission electron microscopy, flow cytometry and Western blotting. EMPs were treated to in vitro and in vivo COPD models induced by cigarette smoke extract (CSE). miR-126 mimics or inhibitors were transfected into EMPs by calcium chloride. Pathological changes of lung tissue, mRNA and protein levels of inflammation-related factors were measured to explore the effect of EMPs transferring miR-126 on CSE-induced inflammation. Results Both in vitro and in vivo studies demonstrated that mRNA and protein levels of inflammation-related factors were significantly increased in COPD group, while EMPs could dramatically reverse these increases. In vitro, overexpression of miR-126 in EMPs decreased HMGB1 expression and magnified the decreasing effect of EMPs on inflammation-related factors. Conclusion The present study reveals that EMPs are capable of alleviating lung inflammation and transferring miR-126 can magnify the anti-inflammatory effect of EMPs, which may provide a novel therapeutic alternative for COPD.
Collapse
Affiliation(s)
- Yiming Ma
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Xue He
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Xiangming Liu
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Yingjiao Long
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, People’s Republic of China
| | - Yan Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, People’s Republic of China
- Correspondence: Yan Chen; Yingjiao Long, Email ;
| |
Collapse
|
34
|
Zhang M, Lu Y, Liu L, Zhang X, Ning J. Role and mechanism of miR-181a-5p in mice with chronic obstructive pulmonary disease by regulating HMGB1 and the NF-κB pathway. Cells Tissues Organs 2022:000522155. [DOI: 10.1159/000522155] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/16/2022] [Indexed: 11/19/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common respiratory disease. This study explored the mechanism of miR-181a-5p in the inflammatory response in COPD mice. COPD mouse models were established by cigarette smoke (CS) exposure following pretreatment with recombinant adeno-associated virus (rAAv)-miR-181a-5p, si-HMGB1 (high mobility group box 1), and NF-κB pathway inhibitor PDTC, respectively. Pathological changes of lung tissues were determined by HE staining. BALF was collected to count total cells, neutrophils and lymphocytes using a Countess II automatic cell counter. Expressions of NE and inflammatory factors (TNF-α, IL-6, IL-8 and IFN-γ) were detected by ELISA. Binding relationship between miR-181a-5p and HMGB1 was predicted on Starbase (http://starbase.sysu.edu.cn/index.php) and validated by dual-luciferase assay. miR-181a-5p expression was detected by RT-qPCR, and expressions of HMGB1, IκBα, p-IκBα were detected by Western blot. The expression level of miR-181a-5p was lower in lung tissues. miR-181a-5p overexpression alleviated inflammatory response and pathological changes of lung tissues in COPD mice, with decreased pulmonary inflammation scores, total cells, neutrophils, and lymphocytes and expressions of NE and inflammatory factors. HMGB1 expression level was increased in COPD mice. miR-181a-5p targeted HMGB1. si-HMGB1 relieved inflammatory responses in COPD mice. NF-κB was activated in COPD mice, evidenced by degraded IκBα and increased p-IκBα level. si-HMGB1 significantly restrained the activation of NF-κB pathway. Briefly, miR-181a-5p targets HMGB1 to inhibit the NF-κB pathway, thus alleviating the inflammatory response in COPD mice.
Collapse
|
35
|
To Predict Anti-Inflammatory and Immunomodulatory Targets of Guizhi Decoction in Treating Asthma Based on Network Pharmacology, Molecular Docking, and Experimental Validation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9033842. [PMID: 34966437 PMCID: PMC8712140 DOI: 10.1155/2021/9033842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/09/2021] [Accepted: 11/30/2021] [Indexed: 12/23/2022]
Abstract
Asthma, characterized by the continuous inflammatory response caused by a variety of immune cells, is one of the most common chronic respiratory diseases worldwide. Relevant clinical trials proved that the traditional Chinese medicine formula Guizhi Decoction (GZD) had multitarget and multichannel functions, which might be an effective drug for asthma. However, the effective ingredients and mechanisms of GZD against asthma are still unclear. Therefore, network pharmacology, molecular docking, and cell experiments were performed to explore the antiasthma effects and potential mechanisms of GZD. First, we applied the TCMSP database and literature to obtain the bioactivated ingredients in GZD. SwissTargetPrediction, TCMSP, GeneCards, OMIM, PharmGkb, TTD, DrugBank, and STRING database were used to get core genes. In addition, the key pathways were analyzed by the DAVID database. Molecular docking was used to predict whether the important components could act on the core target proteins directly. Finally, qPCR was carried out to verify the network pharmacology results and the possible mechanisms of GZD in the treatment of asthma. We collected 134 active ingredients in GZD, 959 drug targets, and 3223 disease targets. 431 intersection genes were screened for subsequent analysis. Through GO and KEGG analyses, enriched pathways related to inflammation and immune regulation were presented. Through the qPCR method to verify the role of essential genes, we found that GZD had an excellent anti-inflammatory effect. Direct or indirect inhibition of MAPK and NF-κB pathways might be one of the crucial mechanisms of GZD against asthma. GZD might be a promising potential drug for the treatment of asthma. This article provided a reference for the clinical application of GZD.
Collapse
|
36
|
Tong LQ, Sui YF, Jiang SN, Yin YH. The Association Between Lung Fluorodeoxyglucose Metabolism and Smoking History in 347 Healthy Adults. J Asthma Allergy 2021; 14:301-308. [PMID: 33840997 PMCID: PMC8032449 DOI: 10.2147/jaa.s302602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/04/2021] [Indexed: 01/28/2023] Open
Abstract
Objective This study aimed to evaluate the relationship between fluorodeoxyglucose metabolism and smoking history in healthy adults by analyzing lung standardized uptake value (SUV). Methods The 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) studies of 347 patients who did not show signs of having malignant diseases or lung inflammation were retrospectively evaluated. Four circular regions of interest (ROI) were manually drawn on the upper and lower lung regions. The averages of maximum SUV (SUVmax-avr) and mean SUV (SUVmean-avr) were calculated, and the mean values of each parameter for non-smokers, ex-smokers, and current smokers were compared. The correlation between SUVmax-avr and smoking history (tobacco burden and the duration of smoking cessation) was assessed based on present smoking status. The ex-smokers and current smokers were divided into three groups according to their tobacco burden, and the SUVmax-avrs of the two groups were compared. Results Both the mean values of SUVmax-avr and SUVmean-avr increased based on smoking history, with non-smokers having the lowest values and current smokers the highest. Tobacco burden had a positive correlation with SUVmax-avr in current smokers (r = 0.474, P< 0.001). However, neither tobacco burden (r = 0171, P = 0.162) nor duration of smoking cessation (r = 0.212, P = 0.082) had a significant correlation with SUVmax-avr in ex-smokers. The mean SUVmax-avr of current smokers was significantly higher than that of ex-smokers in patients with a medium or large tobacco burden (P = 0.012, P< 0.001, respectively). Although there was no significant difference between the mean SUVmax-avrs of ex-smokers and current smokers in patients with a small tobacco burden (P = 0.888), the mean SUVmax-avrs of both ex-smokers and current smokers with a small tobacco burden were significantly higher than that of non-smokers (P< 0. 001, P< 0.001, respectively). Conclusion The findings indicate that lung SUV increases in current heavy smokers and partially decreases after the cessation of smoking, which is in line with previous reports studied by analyzingfluorodeoxyglucose (FDG) metabolism of lung specimens.
Collapse
Affiliation(s)
- Liang-Qian Tong
- Department of Nuclear Medicine, Central South University Xiangya School Affiliated Haikou Hospital, Haikou, Hainan, 570208, People's Republic of China
| | - Yan-Fang Sui
- Department of Rehabilitation Medicine, Central South University Xiangya School Affiliated Haikou Hospital, Haikou, Hainan, 570208, People's Republic of China
| | - Sheng-Nan Jiang
- Department of Nuclear Medicine, Central South University Xiangya School Affiliated Haikou Hospital, Haikou, Hainan, 570208, People's Republic of China
| | - Yan-Hai Yin
- Department of Nuclear Medicine, Hainan Medicine College Affiliated Hainan Hospital, Haikou, Hainan, 570311, People's Republic of China
| |
Collapse
|
37
|
Li H, Song S, Kong Z, Zhu Z, Liu Y, Zuo S, Yin S. Regulatory Effects of Andrographolide on Lung Tissue Inflammation and Th17/Treg in Rats with Chronic Obstructive Pulmonary Disease Induced by Smoking and Lipopolysaccharide. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The pathogenesis of Chronic obstructive pulmonary disease (COPD) is complex, and lung tissue inflammation and Th17/Treg imbalance are the key factors causing lung dysfunction. We constructed a rat COPD model induced by smoking and lipopolysaccharide to explore andrographolide’s
regulation on lung inflammation and Th17/Treg in COPD rats. By contrast, the study found that normal rats, COPD rats forced expiratory volume of 0.3 seconds (FEV0.3), FEV0.3/forced vital capacity (FVC), and peak expiratory flow (PEF) levels decreased. In addition,
the levels of IL-8, TNF-α, IL-17, and IL-6 in alveolar lavage fluid increased, and the level of IL-10 decreased. Concurrently, the total number of white blood cells, monocytes and macrophages, neutrophils, and lymphocytes increased. Meanwhile, the contents of CD25, CD4, and Foxp3 in
lung tissue all increased, and the protein levels of HMGB1, TLR4, and p65 increased. After treatment with andrographolide, the levels of FEV0.3, FEV0.3/FVC, and PEF increased, proving the increase was positively correlated with the concentration of andrographolide. The
levels of IL-8, TNF-α, IL-17, and IL-6 in rat alveolar lavage fluid decreased, and the level of IL-10 sequentially. The total number of white blood cells, the number of monocytes and macrophages, the number of lymphocytes, and the neutral Granulocytes decreased significantly. And the
contents of CD25, CD4, and Foxp3 in lung tissue significantly decreased, and the protein levels of HMGB1, TLR4, and p65 significantly decreased. The above results indicate that andrographolide might be a potential COPD treatment approach. Andrographolide improves the lung function of rats
with COPD, reduces lung inflammation, regulates Th17/Treg balance, and its mechanism may be related to HMGB1/TLR4/NF-кB signaling.
Collapse
Affiliation(s)
- Hong Li
- Department of Respiratory Medicine, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 201306, PR China
| | - Shuang Song
- Department of Respiratory Medicine, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 201306, PR China
| | - Zhibin Kong
- Department of Respiratory Medicine, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 201306, PR China
| | - Zhen Zhu
- Department of Respiratory Medicine, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 201306, PR China
| | - Yi Liu
- Department of Respiratory Medicine, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 201306, PR China
| | - Sheng Zuo
- Department of Geriatrics, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 201306, PR China
| | - Shaojun Yin
- Department of Respiratory Medicine, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 201306, PR China
| |
Collapse
|
38
|
Chen S, Tan S, Yang S, Chen G, Zhu L, Sun Z, Li H, Yao S. Nicotine induces apoptosis through exacerbation of blocked alveolar macrophage autophagic degradation in silicosis. Toxicol Lett 2020; 334:94-101. [PMID: 33010382 DOI: 10.1016/j.toxlet.2020.09.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 08/25/2020] [Accepted: 09/25/2020] [Indexed: 12/25/2022]
Abstract
Silica dust mainly attacks alveolar macrophages (AMs). The apoptosis of AMs is correlated with the progress of silicosis. Our previous study showed that autophagic degradation was blocked in AMs from silicosis patients. However, the effects of nicotine on AM autophagy and apoptosis in silicosis are unknown. In this study, we collected AMs from twenty male workers exposed to silica and divided them into observer and silicosis patient groups, according to the tuberous pathological changes observed by X-ray. The AMs from both groups were exposed to nicotine. We found increased levels of LC3, p62, and cleaved caspase-3, decreased levels of LAMP2, and damaged lysosomes after nicotine stimulation of the AMs from both groups. We also found that the autophagy inhibitor 3-methyladenine (3MA) inhibited nicotine-induced apoptosis in the AMs. Furthermore, 3MA reversed both the nicotine-induced decrease in Bcl-2 and the increase in Bax in both groups. These results suggest that nicotine may induce apoptosis by blocking AM autophagic degradation in human silicosis.
Collapse
Affiliation(s)
- Shi Chen
- School of Medicine, Hunan Normal University, Changsha, Hunan Province 410013, PR China
| | - Shiyi Tan
- School of Medicine, Hunan Normal University, Changsha, Hunan Province 410013, PR China
| | - Shang Yang
- School of Medicine, Hunan Normal University, Changsha, Hunan Province 410013, PR China
| | - Gang Chen
- Department of Pneumoconiosis, Beidaihe Sanitarium for China Coal Miners, Qinhuangdao, Hebei Province 066104, PR China
| | - Li Zhu
- Department of Pneumoconiosis, Beidaihe Sanitarium for China Coal Miners, Qinhuangdao, Hebei Province 066104, PR China
| | - Zhiqian Sun
- Department of Pneumoconiosis, Beidaihe Sanitarium for China Coal Miners, Qinhuangdao, Hebei Province 066104, PR China
| | - Haibin Li
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Sanqiao Yao
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China.
| |
Collapse
|
39
|
|
40
|
Glycyrrhizin: An alternative drug for the treatment of COVID-19 infection and the associated respiratory syndrome? Pharmacol Ther 2020; 214:107618. [PMID: 32592716 PMCID: PMC7311916 DOI: 10.1016/j.pharmthera.2020.107618] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023]
Abstract
Safe and efficient drugs to combat the current COVID-19 pandemic are urgently needed. In this context, we have analyzed the anti-coronavirus potential of the natural product glycyrrhizic acid (GLR), a drug used to treat liver diseases (including viral hepatitis) and specific cutaneous inflammation (such as atopic dermatitis) in some countries. The properties of GLR and its primary active metabolite glycyrrhetinic acid are presented and discussed. GLR has shown activities against different viruses, including SARS-associated Human and animal coronaviruses. GLR is a non-hemolytic saponin and a potent immuno-active anti-inflammatory agent which displays both cytoplasmic and membrane effects. At the membrane level, GLR induces cholesterol-dependent disorganization of lipid rafts which are important for the entry of coronavirus into cells. At the intracellular and circulating levels, GLR can trap the high mobility group box 1 protein and thus blocks the alarmin functions of HMGB1. We used molecular docking to characterize further and discuss both the cholesterol- and HMG box-binding functions of GLR. The membrane and cytoplasmic effects of GLR, coupled with its long-established medical use as a relatively safe drug, make GLR a good candidate to be tested against the SARS-CoV-2 coronavirus, alone and in combination with other drugs. The rational supporting combinations with (hydroxy)chloroquine and tenofovir (two drugs active against SARS-CoV-2) is also discussed. Based on this analysis, we conclude that GLR should be further considered and rapidly evaluated for the treatment of patients with COVID-19.
Collapse
|
41
|
Le Y, Wang Y, Zhou L, Xiong J, Tian J, Yang X, Gai X, Sun Y. Cigarette smoke-induced HMGB1 translocation and release contribute to migration and NF-κB activation through inducing autophagy in lung macrophages. J Cell Mol Med 2020; 24:1319-1331. [PMID: 31769590 PMCID: PMC6991703 DOI: 10.1111/jcmm.14789] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 09/25/2019] [Accepted: 10/04/2019] [Indexed: 12/17/2022] Open
Abstract
High-mobility group box 1 (HMGB1) shows pro-inflammatory activity in various inflammatory diseases and has been found up-regulated in chronic obstructive pulmonary disease (COPD). Lung macrophages play an important role in airway inflammation and lung destruction in COPD, yet whether HMGB1 is involved in cigarette smoke (CS)-induced lung macrophage dysfunction is unknown. We sought to evaluate the intracellular localization and release of HMGB1 in lung macrophages from COPD patients and CS-exposed mice, and to investigate the role of HMGB1 in regulating autophagy in CS extract (CSE)-treated lung macrophages (MH-S cells). Our results showed that HMGB1 was highly expressed in lung tissues and sera of COPD patients and CS-exposed mice, along with predominantly cytoplasmic exporting from nuclei in lung macrophages. In vitro experiments revealed that CSE promoted the expression, nucleocytoplasmic translocation and release of HMGB1 partly via the nicotinic acetylcholine receptor (nAChR). Blockade of HMGB1 with chicken anti-HMGB1 polyclonal antibody (anti-HMGB1) or glycyrrhizin (Gly) attenuated the increase of LC3B-II and Beclin1, migration and p65 phosphorylation, suggesting the involvement of HMGB1 in autophagy, migration and NF-κB activation of lung macrophages. Hydroxychloroquine (CQ), an autophagy inhibitor, enhanced the increase of LC3B-II but not Beclin1 in CSE or rHMGB1-treated MH-S cells, and inhibition of autophagy by CQ and 3-methyladenine (3-MA) abrogated the migration and p65 phosphorylation of CSE-treated cells. These results indicate that CS-induced HMGB1 translocation and release contribute to migration and NF-κB activation through inducing autophagy in lung macrophages, providing novel evidence for HMGB1 as a potential target of intervention in COPD.
Collapse
Affiliation(s)
- Yanqing Le
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| | - Yanhong Wang
- Department of Respiratory MedicineZhongshan City People's HospitalZhongshanChina
| | - Lu Zhou
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| | - Jing Xiong
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| | - Jieyu Tian
- Hematology Oncology CenterBeijing Children's HospitalCapital Medical UniversityBeijingChina
| | - Xia Yang
- Department of Respiratory MedicineTianjin Medical University General HospitalTianjingChina
| | - Xiaoyan Gai
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| | - Yongchang Sun
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| |
Collapse
|