1
|
Burgess JK, Gosens R. Mechanotransduction and the extracellular matrix: Key drivers of lung pathologies and drug responsiveness. Biochem Pharmacol 2024; 228:116255. [PMID: 38705536 DOI: 10.1016/j.bcp.2024.116255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
The lung is a biomechanically active organ, with multiscale mechanical forces impacting the organ, tissue and cellular responses within this microenvironment. In chronic lung diseases, such as chronic obstructive pulmonary disease, pulmonary fibrosis and others, the structure of the lung is drastically altered impeding gas exchange. These changes are, in part, reflected in alterations in the composition, amount and organization of the extracellular matrix within the different lung compartments. The transmission of mechanical forces within lung tissue are broadcast by this complex mix of extracellular matrix components, in particular the collagens, elastin and proteoglycans and the crosslinking of these components. At both a macro and a micro level, the mechanical properties of the microenvironment have a key regulatory role in ascertaining cellular responses and the function of the lung. Cells adhere to, and receive signals from, the extracellular matrix through a number of different surface receptors and complexes which are important for mechanotransduction. This review summarizes the multiscale mechanics in the lung and how the mechanical environment changes in lung disease and aging. We then examine the role of mechanotransduction in driving cell signaling events in lung diseases and finish with a future perspective of the need to consider how such forces may impact pharmacological responsiveness in lung diseases.
Collapse
Affiliation(s)
- Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands.
| | - Reinoud Gosens
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands; Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
2
|
Mohamed RH, Abdel Hay NH, Fawzy NM, Tamim YM, Doaa Karem MM, Yehia DAY, Abdel Maksoud OM, Abdelrahim DS. Targeting mevalonate pathway by zoledronate ameliorated pulmonary fibrosis in a rat model: Promising therapy against post-COVID-19 pulmonary fibrosis. Fundam Clin Pharmacol 2024; 38:703-717. [PMID: 38357833 DOI: 10.1111/fcp.12994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Rho kinase (ROCK) pathway plays a critical role in post-COVID-19 pulmonary fibrosis (PCPF) and its intervention with angiotensin-converting enzyme 2 (ACE2) and vascular endothelial growth factor (VEGF) will be a potential therapeutic target. OBJECTIVES The present study was conducted to investigate the efficacy of zoledronate (ZA) on carbon tetrachloride (CCl4) induced pulmonary fibrosis (PF) in rats through targeting ACE2, ROCK, and VEGF signaling pathways. METHODS Fifty male Wistar rats were divided into five groups: control, vehicle-treated, PF, PF-ZA 50, and PF-ZA 100 groups. ZA was given in two different doses 100 and 50 μg/kg/week intraperitoneally. After anesthesia, mean arterial blood pressure (MBP) was measured. After scarification, lung coefficient was calculated. Lung levels of ACE 2, interleukin-1β (IL-1β), transforming growth factor-β (TGF-β), VEGF, glutathione (GSH), and superoxide dismutase (SOD) were measured. Expression of ROCK, phosphorylated myosin phosphatase target subunit 1 (P-MYPT1), and matrix metalloproteinase (MMP-1), along with histopathological changes and immune-histochemical staining for lung α-smooth muscle actin (α-SMA), tumor necrosis factor-alpha (TNFα), and caspase-3, were evaluated. RESULTS ZA significantly prevented the decrease in MBP. ZA significantly increased ACE2, GSH, and SOD and significantly decreased IL-1β, TGF-β, and VEGF in lung in comparison to PF group. ZA prevented the histopathological changes induced by CCl4. ZA inhibited lung expression of ROCK, P-MYPT1, MMP-1, α-SMA, TNFα, and caspase-3 with significant differences favoring the high dose intervention. CONCLUSION ZA in a dose-dependent manner prevented the pathological effect of CCl4 in the lung by targeting mevalonate pathway. It could be promising therapy against PCPF.
Collapse
Affiliation(s)
- Reham Hussein Mohamed
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nesma Hussein Abdel Hay
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nesma Mohamed Fawzy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Yomna M Tamim
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - M M Doaa Karem
- Department of Histology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Omnia M Abdel Maksoud
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Dina S Abdelrahim
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
- Department of Pharmacology, Faculty of Medicine, Modern Technology and Information University, Cairo, Egypt
| |
Collapse
|
3
|
Li P, Wang Y, Cao Y, Shi J, Jiang M, Han X, Jiang L, Bao Y, Wu W, Liu X. Exercise Attenuate Diaphragm Atrophy in COPD Mice via Inhibiting the RhoA/ROCK Signaling. Int J Chron Obstruct Pulmon Dis 2024; 19:1591-1601. [PMID: 39005647 PMCID: PMC11244622 DOI: 10.2147/copd.s460182] [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: 01/18/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Background Exercise is an indispensable component of pulmonary rehabilitation with strong anti-inflammatory effects. However, the mechanisms by which exercise prevents diaphragmatic atrophy in COPD (chronic obstructive pulmonary disease) remain unclear. Methods Forty male C57BL/6 mice were assigned to the control (n=16) and smoke (n=24) groups. Mice in the smoke group were exposed to the cigarette smoke (CS) for six months. They were then divided into model and exercise training groups for 2 months. Histological changes were observed in lung and diaphragms. Subsequently, agonist U46639 and antagonist Y27632 of RhoA/ROCK were subjected to mechanical stretching in LPS-treated C2C12 myoblasts. The expression levels of Atrogin-1, MuRF-1, MyoD, Myf5, IL-1β, TNF-α, and RhoA/ROCK were determined by Western blotting. Results Diaphragmatic atrophy and increased RhoA/ROCK expression were observed in COPD mice. Exercise training attenuated diaphragmatic atrophy, decreased the expression of MuRF-1, and increased MyoD expression in COPD diaphragms. Exercise also affects the upregulation of RhoA/ROCK and inflammation-related proteins. In in vitro experiments with C2C12 myoblasts, LPS remarkably increased the level of inflammation and protein degradation, whereas Y27632 or combined with mechanical stretching prevented this phenomenon considerably. Conclusion RhoA/ROCK plays an important role in the prevention of diaphragmatic atrophy in COPD.
Collapse
Affiliation(s)
- Peijun Li
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People's Republic of China
| | - Yingqi Wang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People's Republic of China
| | - Yuanyuan Cao
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, 200438, People's Republic of China
| | - Jiacheng Shi
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People's Republic of China
| | - Meiling Jiang
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, 200438, People's Republic of China
| | - Xiaoyu Han
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, 200438, People's Republic of China
| | - Linhong Jiang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People's Republic of China
| | - Yidie Bao
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People's Republic of China
| | - Weibing Wu
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, 200438, People's Republic of China
| | - Xiaodan Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, People's Republic of China
- Institute of Rehabilitation Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, People's Republic of China
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai, 201203, People's Republic of China
| |
Collapse
|
4
|
Madsen JJ, Ohkubo YZ. Elucidating the complex membrane binding of a protein with multiple anchoring domains using extHMMM. PLoS Comput Biol 2024; 20:e1011421. [PMID: 38976709 PMCID: PMC11257402 DOI: 10.1371/journal.pcbi.1011421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 07/18/2024] [Accepted: 06/19/2024] [Indexed: 07/10/2024] Open
Abstract
Membrane binding is a crucial mechanism for many proteins, but understanding the specific interactions between proteins and membranes remains a challenging endeavor. Coagulation factor Va (FVa) is a large protein whose membrane interactions are complicated due to the presence of multiple anchoring domains that individually can bind to lipid membranes. Using molecular dynamics simulations, we investigate the membrane binding of FVa and identify the key mechanisms that govern its interaction with membranes. Our results reveal that FVa can either adopt an upright or a tilted molecular orientation upon membrane binding. We further find that the domain organization of FVa deviates (sometimes significantly) from its crystallographic reference structure, and that the molecular orientation of the protein matches with domain reorganization to align the C2 domain toward its favored membrane-normal orientation. We identify specific amino acid residues that exhibit contact preference with phosphatidylserine lipids over phosphatidylcholine lipids, and we observe that mostly electrostatic effects contribute to this preference. The observed lipid-binding process and characteristics, specific to FVa or common among other membrane proteins, in concert with domain reorganization and molecular tilt, elucidate the complex membrane binding dynamics of FVa and provide important insights into the molecular mechanisms of protein-membrane interactions. An updated version of the HMMM model, termed extHMMM, is successfully employed for efficiently observing membrane bindings of systems containing the whole FVa molecule.
Collapse
Affiliation(s)
- Jesper J. Madsen
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
- Center for Global Health and Infectious Diseases Research, Global and Planetary Health, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Y. Zenmei Ohkubo
- Department of Bioinformatics, School of Life and Natural Sciences, Abdullah Gül University, Kayseri, Turkey
| |
Collapse
|
5
|
Qiao J, Tan Y, Liu H, Yang B, Zhang Q, Liu Q, Sun W, Li Z, Wang Q, Feng W, Yang S, Cui L. Histone H3K18 and Ezrin Lactylation Promote Renal Dysfunction in Sepsis-Associated Acute Kidney Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307216. [PMID: 38767134 PMCID: PMC11267308 DOI: 10.1002/advs.202307216] [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: 09/29/2023] [Revised: 04/29/2024] [Indexed: 05/22/2024]
Abstract
Histone lactylation is a metabolic stress-related histone modification. However, the role of histone lactylation in the development of sepsis-associated acute kidney injury (SA-AKI) remains unclear. Here, histone H3K18 lactylation (H3K18la) is elevated in SA-AKI, which is reported in this study. Furthermore, this lactate-dependent histone modification is enriched at the promoter of Ras homolog gene family member A (RhoA) and positively correlated with the transcription. Correction of abnormal lactate levels resulted in a reversal of abnormal histone lactylation at the promoter of RhoA. Examination of related mechanism revealed that histone lactylation promoted the RhoA/Rho-associated protein kinase (ROCK) /Ezrin signaling, the activation of nuclear factor-κB (NF-κB), inflammation, cell apoptosis, and aggravated renal dysfunction. In addition, Ezrin can undergo lactylation modification. Multiple lactylation sites are identified in Ezrin and confirmed that lactylation mainly occurred at the K263 site. The role of histone lactylation is revealed in SA-AKI and reportes a novel post-translational modification in Ezrin. Its potential role in regulating inflammatory metabolic adaptation of renal proximal tubule epithelial cells is also elucidated. The results provide novel insights into the epigenetic regulation of the onset of SA-AKI.
Collapse
Affiliation(s)
- Jiao Qiao
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Yuan Tan
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Hongchao Liu
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Boxin Yang
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Qian Zhang
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Qi Liu
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Wenyuan Sun
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Zhongxin Li
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Qingchen Wang
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Weimin Feng
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Shuo Yang
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Liyan Cui
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| |
Collapse
|
6
|
Wang S, Liu B, He H, Huang J, He F, He Y, Tao A. Cell-in-cell-mediated intercellular communication exacerbates the pro-inflammatory progression in asthma. Biochem Cell Biol 2024; 102:262-274. [PMID: 38567768 DOI: 10.1139/bcb-2023-0228] [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: 06/02/2024] Open
Abstract
Cell-in-cell (CIC) structures have been suggested to mediate intracellular substance transport between cells and have been found widely in inflammatory lung tissue of asthma. The aim of this study was to investigate the significance of CIC structures in inflammatory progress of asthma. CIC structures and related inflammatory pathways were analyzed in asthmatic lung tissue and normal lung tissue of mouse model. In vitro, the activation of inflammatory pathways by CIC-mediated intercellular communication was analyzed by RNA-Seq and verified by Western blotting and immunofluorescence. Results showed that CIC structures of lymphocytes and alveolar epithelial cells in asthmatic lung tissue mediated intercellular substance (such as mitochondria) transfer and promoted pro-inflammation in two phases. At early phase, internal lymphocytes triggered inflammasome-dependent pro-inflammation and cell death of itself. Then, degraded lymphocytes released cellular contents such as mitochondria inside alveolar epithelial cells, further activated multi-pattern-recognition receptors and NF-kappa B signaling pathways of alveolar epithelial cells, and thereby amplified pro-inflammatory response in asthma. Our work supplements the mechanism of asthma pro-inflammation progression from the perspective of CIC structure of lymphocytes and alveolar epithelial cells, and provides a new idea for anti-inflammatory therapy of asthma.
Collapse
Affiliation(s)
- Shan Wang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou 510260, China
| | - Bowen Liu
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou 510260, China
| | - Huiru He
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou 510260, China
| | - Jiahao Huang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou 510260, China
| | - Fangping He
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou 510260, China
| | - Ying He
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou 510260, China
| | - Ailin Tao
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou 510260, China
| |
Collapse
|
7
|
Liang J, Dai W, Xue S, Wu F, Cui E, Pan R. Recent progress in mesenchymal stem cell-based therapy for acute lung injury. Cell Tissue Bank 2024; 25:677-684. [PMID: 38466563 DOI: 10.1007/s10561-024-10129-0] [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: 06/07/2022] [Accepted: 01/24/2024] [Indexed: 03/13/2024]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening diseases in critically ill patients. Although pathophysiology of ALI/ARDS has been investigated in many studies, effective therapeutic strategies are still limited. Mesenchymal stem cell (MSC)-based therapy is emerging as a promising therapeutic intervention for patients with ALI. During the last two decades, researchers have focused on the efficacy and mechanism of MSC application in ALI animal models. MSC derived from variant resources exhibited therapeutic effects in preclinical studies of ALI with different mechanisms. Based on this, clinical studies on MSC treatment in ALI/ARDS has been tried recently, especially in COVID-19 caused lung injury. Emerging clinical trials of MSCs in treating COVID-19-related conditions have been registered in past two years. The advantages and potential of MSCs in the defense against COVID-19-related ALI or ARDS have been confirmed. This review provides a brief overview of recent research progress in MSC-based therapies in preclinical study and clinical trials in ALI treatment, as well as the underlying mechanisms.
Collapse
Affiliation(s)
- Jinfeng Liang
- Zhejiang Center for Drug and Cosmetic Evaluation, Hangzhou, China
| | - Weiyou Dai
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Shihang Xue
- Xiangshan First People's Hospital Medical and Health Group, Ningbo, China
| | - Feifei Wu
- Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, No.181 Wuchang Road, Hangzhou, 311122, Zhejiang, People's Republic of China
| | - Enhai Cui
- Huzhou Central Hospital, Zhejiang University Huzhou Hospital, Huzhou, 313000, People's Republic of China.
| | - Ruolang Pan
- Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China.
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, No.181 Wuchang Road, Hangzhou, 311122, Zhejiang, People's Republic of China.
| |
Collapse
|
8
|
Yuan S, Jiang SC, Zhang ZW, Li ZL, Hu J. Substance Addiction Rehabilitation Drugs. Pharmaceuticals (Basel) 2024; 17:615. [PMID: 38794185 PMCID: PMC11124501 DOI: 10.3390/ph17050615] [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: 04/23/2024] [Revised: 05/04/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The relapse rate of substance abusers is high, and addiction rehabilitation adjunct drugs need to be developed urgently. There have been numerous reports on blocking the formation of substance addiction, but studies on drugs that can alleviate withdrawal symptoms are very limited. Both the dopamine transporter (DAT) hypothesis and D3 dopamine receptor (D3R) hypothesis are proposed. DAT activators reduce the extracellular dopamine level, and D3R antagonists reduce the neuron's sensitivity to dopamine, both of which may exacerbate the withdrawal symptoms subsequently. The D3R partial agonist SK608 has biased signaling properties via the G-protein-dependent pathway but did not induce D3R desensitization and, thus, may be a promising drug for the withdrawal symptoms. Drugs for serotoninergic neurons or GABAergic neurons and anti-inflammatory drugs may have auxiliary effects to addiction treatments. Drugs that promote structural synaptic plasticity are also discussed.
Collapse
Affiliation(s)
- Shu Yuan
- College of Resources, Sichuan Agricultural University, Chengdu 611130, China;
| | - Si-Cong Jiang
- Haisco Pharmaceutical Group Comp. Ltd., Chengdu 611138, China;
| | - Zhong-Wei Zhang
- College of Resources, Sichuan Agricultural University, Chengdu 611130, China;
| | - Zi-Lin Li
- Department of Cardiovascular Surgery, Xijing Hospital, Medical University of the Air Force, Xi’an 710032, China;
| | - Jing Hu
- School of Medicine, Northwest University, Xi’an 710069, China;
| |
Collapse
|
9
|
Shan M, Wan H, Ran L, Ye J, Xie W, Lu J, Hu X, Deng S, Zhang W, Chen M, Wang F, Guo Z. Dynasore Alleviates LPS-Induced Acute Lung Injury by Inhibiting NLRP3 Inflammasome-Mediated Pyroptosis. Drug Des Devel Ther 2024; 18:1369-1384. [PMID: 38681210 PMCID: PMC11055558 DOI: 10.2147/dddt.s444408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 04/12/2024] [Indexed: 05/01/2024] Open
Abstract
Background Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are clinically severe respiratory disorders without available pharmacological therapies. Dynasore is a cell-permeable molecule that inhibits GTPase activity and exerts protective effects in several disease models. However, whether dynasore can alleviate lipopolysaccharide (LPS)-induced ALI is unknown. This study investigated the effect of dynasore on macrophage activation and explored its potential mechanisms in LPS-induced ALI in vitro and in vivo. Methods Bone marrow-derived macrophages (BMDMs) were activated classically with LPS or subjected to NLRP3 inflammasome activation with LPS+ATP. A mouse ALI model was established by the intratracheal instillation (i.t.) of LPS. The expression of PYD domains-containing protein 3 (NLRP3), caspase-1, and gasdermin D (GSDMD) protein was detected by Western blots. Inflammatory mediators were analyzed in the cell supernatant, in serum and bronchoalveolar lavage fluid (BALF) by enzyme-linked immunosorbent assays. Morphological changes in lung tissues were evaluated by hematoxylin and eosin staining. F4/80, Caspase-1 and GSDMD distribution in lung tissue was detected by immunofluorescence. Results Dynasore downregulated nuclear factor (NF)-κB signaling and reduced proinflammatory cytokine production in vitro and inhibited the production and release of interleukin (IL)-1β, NLRP3 inflammasome activation, and macrophage pyroptosis through the Drp1/ROS/NLRP3 axis. Dynasore significantly reduced lung injury scores and proinflammatory cytokine levels in both BALF and serum in vivo, including IL-1β and IL-6. Dynasore also downregulated the co-expression of F4/80, caspase-1 and GSDMD in lung tissue. Conclusion Collectively, these findings demonstrated that dynasore could alleviate LPS-induced ALI by regulating macrophage pyroptosis, which might provide a new therapeutic strategy for ALI/ARDS.
Collapse
Affiliation(s)
- Mengtian Shan
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Huimin Wan
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Linyu Ran
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Jihui Ye
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Wang Xie
- Department of Respiratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, People’s Republic of China
| | - Jingjing Lu
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Xueping Hu
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Shengjie Deng
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Wenyu Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Miao Chen
- Department of Emergency, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People’s Republic of China
| | - Feilong Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Zhongliang Guo
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
- Department of Respiratory Medicine, Ji’an Hospital, Shanghai East Hospital, Shanghai, Jiangxi, People’s Republic of China
| |
Collapse
|
10
|
Lv Y, Yu W, Xuan R, Yang Y, Xue X, Ma X. Human Placental Mesenchymal Stem Cells-Exosomes Alleviate Endothelial Barrier Dysfunction via Cytoskeletal Remodeling through hsa-miR-148a-3p/ROCK1 Pathway. Stem Cells Int 2024; 2024:2172632. [PMID: 38681858 PMCID: PMC11055650 DOI: 10.1155/2024/2172632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/18/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024] Open
Abstract
Background Endothelial barrier disruption of human pulmonary vascular endothelial cells (HPVECs) is an important pathogenic factor for acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). Mesenchymal stem cells-exosome (MSCs-Exo) represents an ideal carrier for cell-free therapy. The therapeutic implication and underlying mechanism of human placental MSCs-Exo (HPMSCs-Exo) in ALI/ARDS need to be further explored. Materials and Methods HPMSCs-Exo was extracted from HPMSCs and characterized. Then, the therapeutic effects of exosomes were evaluated in ALI mice and HPVECs. RNA-sequencing was applied to reveal the miRNA profile of HPMSCs-Exo and differentially expressed genes (DEGs) in HPMSCs-Exo-pretreated HPVECs. The targets of miRNAs were predicted by bioinformatics methods and correlated to DEGs. Finally, the role of hsa-miR-148a-3p/ROCK1 pathway in HPVECs has been further discussed. Results The results showed that HPMSCs-Exo could downregulate Rho-associated coiled-coil-containing protein kinase 1 (ROCK1), upregulate the expression of zonula occludens-1 (ZO-1) and F-actin, promote HPVECs migration and tube formation, reduce cytoskeletal disorders and cell permeability, and thus improve ALI/ARDS. RNA-sequencing revealed the DEGs were mainly enriched in cell junction, angiogenesis, inflammation, and energy metabolism. HPMSCs-Exo contains multiple miRNAs which are associated with cytoskeletal function; the expression abundance of hsa-miR-148a-3p is the highest. Bioinformatic analysis identified ROCK1 as a target of hsa-miR-148a-3p. The overexpression of hsa-miR-148a-3p in HPMSCs-Exo promoted the migration and tube formation of HPVECs and reduced ROCK1 expression. However, the overexpression of ROCK1 on HPVECs reduced the therapeutic effect of HPMSCs-Exo. Conclusions HPMSCs-Exo represents a protective regimen against endothelial barrier disruption of HPVECs in ALI/ARDS, and the hsa-miR-148a-3p/ROCK1 pathway plays an important role in this therapeutics implication.
Collapse
Affiliation(s)
- Yuzhen Lv
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
- Ningxia Institute for Human Stem Cell Research, General Hospital of Ningxia Medical University, Yinchuan 750003, China
| | - Wenqin Yu
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
- Ningxia Institute for Human Stem Cell Research, General Hospital of Ningxia Medical University, Yinchuan 750003, China
| | - Ruiui Xuan
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
| | - Yulu Yang
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
| | - Xiaolan Xue
- School of Clinical, Ningxia Medical University, Yinchuan 750003, China
| | - Xiaowei Ma
- Intensive Care Unit, Cardiocerebral Vascular Disease Hospital, General Hospital of Ningxia Medical University, Yinchuan 750003, China
| |
Collapse
|
11
|
Golding R, Braun RK, Miller L, Lasarev M, Hacker TA, Rodgers AC, Staehler A, Eldridge MW, Al-Subu A. Differential changes in expression of inflammatory mRNA and protein after oleic acid-induced acute lung injury. Exp Lung Res 2024; 50:96-105. [PMID: 38625585 DOI: 10.1080/01902148.2024.2341099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 04/03/2024] [Indexed: 04/17/2024]
Abstract
Background: Acute Respiratory Distress syndrome (ARDS) is a clinical syndrome of noncardiac pulmonary edema and inflammation leading to acute respiratory failure. We used the oleic acid infusion pig model of ARDS resembling human disease to explore cytokine changes in white blood cells (WBC) and plasma proteins, comparing baseline to ARDS values. Methods: Nineteen juvenile female swine were included in the study. ARDS defined by a PaO2/FiO2 ratio < 300 was induced by continuous oleic acid infusion. Arterial blood was drawn before and during oleic acid infusion, and when ARDS was established. Cytokine expression in WBC was analyzed by RT-qPCR and plasma protein expression by ELISA. Results: The median concentration of IFN-γ mRNA was estimated to be 59% (p = 0.006) and of IL-6 to be 44.4% (p = 0.003) of the baseline amount. No significant changes were detected for TNF-α, IL-17, and IL-10 mRNA expression. In contrast, the concentrations of plasma IFN-γ and IL-6 were significantly higher (p = 0.004 and p = 0.048 resp.), and TNF-α was significantly lower (p = 0.006) at ARDS compared to baseline. Conclusions: The change of proinflammatory cytokines IFN-γ and IL-6 expression is different comparing mRNA and plasma proteins at oleic acid-induced ARDS compared to baseline. The migration of cells to the lung may be the cause for this discrepancy.
Collapse
Affiliation(s)
- Regina Golding
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Rudolf K Braun
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Lorenzo Miller
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Michael Lasarev
- Department of Biostatistics & Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Timothy A Hacker
- Cardiovascular Physiology Core Facility, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Allison C Rodgers
- Cardiovascular Physiology Core Facility, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ava Staehler
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Marlowe W Eldridge
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Awni Al-Subu
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
12
|
Xu Y, Xin J, Sun Y, Wang X, Sun L, Zhao F, Niu C, Liu S. Mechanisms of Sepsis-Induced Acute Lung Injury and Advancements of Natural Small Molecules in Its Treatment. Pharmaceuticals (Basel) 2024; 17:472. [PMID: 38675431 PMCID: PMC11054595 DOI: 10.3390/ph17040472] [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: 03/13/2024] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis-induced acute lung injury (ALI), characterized by widespread lung dysfunction, is associated with significant morbidity and mortality due to the lack of effective pharmacological treatments available clinically. Small-molecule compounds derived from natural products represent an innovative source and have demonstrated therapeutic potential against sepsis-induced ALI. These natural small molecules may provide a promising alternative treatment option for sepsis-induced ALI. This review aims to summarize the pathogenesis of sepsis and potential therapeutic targets. It assembles critical updates (from 2014 to 2024) on natural small molecules with therapeutic potential against sepsis-induced ALI, detailing their sources, structures, effects, and mechanisms of action.
Collapse
Affiliation(s)
- Yaxi Xu
- School of Pharmacy, Yantai University, Yantai 264005, China; (Y.X.); (Y.S.); (X.W.)
| | - Jianzeng Xin
- School of Life Sciences, Yantai University, Yantai 264005, China;
| | - Yupei Sun
- School of Pharmacy, Yantai University, Yantai 264005, China; (Y.X.); (Y.S.); (X.W.)
| | - Xuyan Wang
- School of Pharmacy, Yantai University, Yantai 264005, China; (Y.X.); (Y.S.); (X.W.)
| | - Lili Sun
- College of Pharmacy, University of Utah, Salt Lake City, UT 84108, USA;
| | - Feng Zhao
- School of Pharmacy, Yantai University, Yantai 264005, China; (Y.X.); (Y.S.); (X.W.)
| | - Changshan Niu
- College of Pharmacy, University of Utah, Salt Lake City, UT 84108, USA;
| | - Sheng Liu
- School of Pharmacy, Yantai University, Yantai 264005, China; (Y.X.); (Y.S.); (X.W.)
| |
Collapse
|
13
|
Cacciapuoti M, Caputo I, Stefanelli LF, Davis PA, Nalesso F, Calò LA. "Every Cloud Has a Silver Lining": How Three Rare Diseases Defend Themselves from COVID-19 and What We Have Learnt from It. Clin Pract 2024; 14:614-618. [PMID: 38666806 PMCID: PMC11048753 DOI: 10.3390/clinpract14020048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/19/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
The process of SARS-CoV-2 infection, responsible for the COVID-19 pandemic, is carried out through different steps, with the interaction between ACE2 and Spike protein (S) being crucial. Besides of that, the acidic environment of endosomes seems to play a relevant role in the virus uptake into cells and its intracellular replication. Patients affected by two rare genetic tubulopathies, Gitelman's and Bartter's Syndromes, and a rare genetic metabolic disease, Fabry Disease, have shown intrinsic protection from SARS-CoV-2 infection and COVID-19 on account of specific intrinsic features that interfere with the virus uptake into cells and its intracellular replication, which will be reported and discussed in this paper, providing interesting insights for present and future research.
Collapse
Affiliation(s)
- Martina Cacciapuoti
- Nephrology, Dialysis and Transplantation Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (M.C.); (I.C.); (F.N.)
| | - Ilaria Caputo
- Nephrology, Dialysis and Transplantation Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (M.C.); (I.C.); (F.N.)
| | - Lucia Federica Stefanelli
- Nephrology, Dialysis and Transplantation Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (M.C.); (I.C.); (F.N.)
| | - Paul A. Davis
- Department of Nutrition, University of California, Davis, CA 95616, USA;
| | - Federico Nalesso
- Nephrology, Dialysis and Transplantation Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (M.C.); (I.C.); (F.N.)
| | - Lorenzo A. Calò
- Nephrology, Dialysis and Transplantation Unit, Department of Medicine, University of Padova, 35128 Padova, Italy; (M.C.); (I.C.); (F.N.)
| |
Collapse
|
14
|
Liang Y, Zhang H, Li J, Wang X, Xie J, Li Y, Li J, Qian Y, Zhang H, Wang T, Tang H, Chen X. GLUT1 regulates the release of VEGF-A in the alveolar epithelium of lipopolysaccharide-induced acute lung injury. Cell Biol Int 2024; 48:510-520. [PMID: 38225684 DOI: 10.1002/cbin.12127] [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: 04/02/2023] [Revised: 11/20/2023] [Accepted: 01/01/2024] [Indexed: 01/17/2024]
Abstract
Acute lung injury (ALI) is a severe disease with high mortality and poor prognosis, characterized by excessive and uncontrolled inflammatory response. Vascular endothelial growth factor A (VEGF-A) contributes to the development and progression of ALI. The aim of this study was to evaluate the role of glucose transporter 1 (GLUT1) in alveolar epithelial VEGF-A production in lipopolysaccharide (LPS)-induced ALI. An ALI mouse model was induced by LPS oropharyngeal instillation. Mice were challenged with LPS and then treated with WZB117, a specific antagonist of GLUT1. For the vitro experiments, cultured A549 cells (airway epithelial cell line) were exposed to LPS, with or without the GLUT1 inhibitors WZB117 or BAY876. LPS significantly upregulated of GLUT1 and VEGF-A both in the lung from ALI mice and in cultured A549. In vivo, treatment with WZB117 not only markedly decreased LPS-induced pulmonary edema, injury, neutrophilia, as well as levels of interleukin (IL)-1β, IL-6 and tumor necrosis factor-α in bronchoalveolar lavage fluid (BALF), but also reduced VEGF-A production. Yet, the maximum tolerated concentration of WZB117 failed to suppress LPS-induced VEGF-A overexpression in vitro. While administration of BAY876 inhibited gene and protein expression as well as secretion of VEGF-A in response to LPS in A549. These results illustrated that GLUT1 upregulates VEGF-A production in alveolar epithelia from LPS-induced ALI, and inhibition of GLUT1 alleviates ALI.
Collapse
Affiliation(s)
- Yan Liang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hailing Zhang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiahui Li
- Department of Pulmonary and Critical Care Medicine, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Xilong Wang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianpeng Xie
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yijian Li
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiehong Li
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunyao Qian
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Haiyun Zhang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Tao Wang
- State key Laboratory of Respiratory Diseases, Guangzhou Key Laboratory of Vascular Diseases, Guangzhou Institute of Respiratory Health, The Frist Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haixiong Tang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
15
|
Zhou Y, Chen C, Chen Y, Ding Y, Li S, Wu J, Hong S, Lu B, Liang H, Liu Y, Ouyang Y, Yin W, Hu C. Synthetic steroid of 5α-Androst-3β,5α,6β-Triol alleviates acute lung injury via inhibiting inflammation and oxidative stress. Int Immunopharmacol 2024; 129:111486. [PMID: 38326121 DOI: 10.1016/j.intimp.2024.111486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/19/2023] [Accepted: 01/01/2024] [Indexed: 02/09/2024]
Abstract
Acute lung injury (ALI) is a severe and potentially fatal respiratory condition with limited treatment options. The pathological evolution of ALI is driven by persistent inflammation, destruction of the pulmonary vascular barrier and oxidative stress. Evidence from prior investigations has identified 5α-androst-3β,5α,6β-Triol (TRIOL), a synthetic analogue of the naturally occurring neuroprotective compound cholestane-3β,5α,6β-triol, possesses notable anti-inflammatory and antioxidative properties. However, the precise effects of TRIOL on alleviating lung injury along with the mechanisms, have remained largely unexplored. Here, TRIOL exhibited pronounced inhibitory actions on lipopolysaccharide (LPS)-induced inflammation and oxidative stress damage in both lung epithelial and endothelial cells. This protective effect is achieved by its ability to mitigate oxidative stress and restrain the inflammatory cascade orchestrated by nuclear factor-kappa B (NF-κB), thereby preserving the integrity of the pulmonary epithelial barrier. We further validated that TRIOL can attenuate LPS-induced lung injury in rats and mice by reducing inflammatory cell infiltration and improving pulmonary edema. Furthermore, TRIOL decreased the pro-inflammatory factors and increased of anti-inflammatory factors induced by LPS. In conclusion, our study presents TRIOL as a promising novel candidate for the treatment of ALI.
Collapse
Affiliation(s)
- YuWei Zhou
- Department of Laboratory Medicine, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China; Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Chen Chen
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Department of Molecular Biology and Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - YuPin Chen
- Guangzhou Cellprotek Pharmaceutical Co., Ltd., Guangzhou 510663, China
| | - YuXuan Ding
- Department of Molecular Biology and Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - ShengLong Li
- Department of Molecular Biology and Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - JiaXin Wu
- Department of Molecular Biology and Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - ShiRan Hong
- Department of Molecular Biology and Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - BingZheng Lu
- Guangzhou Cellprotek Pharmaceutical Co., Ltd., Guangzhou 510663, China
| | - HuaFeng Liang
- Guangzhou Cellprotek Pharmaceutical Co., Ltd., Guangzhou 510663, China
| | - Ying Liu
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ying Ouyang
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wei Yin
- Department of Molecular Biology and Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Cheng Hu
- Department of Urology, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China.
| |
Collapse
|
16
|
Kutsuzawa N, Ito Y, Kagawa S, Kohno C, Takiguchi H, Asano K. Dexamethasone restores TNFα-induced epithelial barrier dysfunction in primary rat alveolar epithelial cells. PLoS One 2023; 18:e0295684. [PMID: 38150443 PMCID: PMC10752552 DOI: 10.1371/journal.pone.0295684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 11/27/2023] [Indexed: 12/29/2023] Open
Abstract
Alveolar barrier dysfunction is one of the major pathophysiological changes in acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). In ALI/ARDS, tumor necrosis factor-alpha (TNFα) disrupts the barriers of alveolar epithelium and endothelium. Glucocorticoids (GCs) exert anti-inflammatory effects and ameliorate pulmonary edema in ALI/ARDS. However, the involvement of GCs in the restoration of alveolar epithelial barrier dysfunction has not been extensively studied. Here, we elucidated that dexamethasone (Dex) restored TNFα-induced alveolar epithelial barrier dysfunction in vitro using primary rat alveolar epithelial cells isolated from Sprague-Dawley rats. Moreover, Dex promoted the alveolar epithelial cell barrier integrity by initiating GC receptor-mediated signaling via the downregulation of myosin light chain kinase (MLCK) expression and the dephosphorylation of myosin light chain (MLC) 2. Further investigation revealed that Dex enhanced the expression of zonula occludens-1 (ZO-1), a tight junction-related protein, at intercellular junction sites. These findings suggest that GCs strengthen the integrity of the alveolar epithelial barrier in ALI/ARDS via the GR-MLCK-pMLC2 axis.
Collapse
Affiliation(s)
- Naokata Kutsuzawa
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yoko Ito
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Shizuko Kagawa
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Chinatsu Kohno
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Hiroto Takiguchi
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Koichiro Asano
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| |
Collapse
|
17
|
Wu Y, Zhang Y, Tang X, Ye S, Shao J, Tu L, Pan J, Chen L, Liang G, Yin L. Synergistic anti-oxidant and anti-inflammatory effects of ceria/resatorvid co-decorated nanoparticles for acute lung injury therapy. J Nanobiotechnology 2023; 21:502. [PMID: 38129906 PMCID: PMC10740228 DOI: 10.1186/s12951-023-02237-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/03/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI) is a critical inflammatory response syndrome that rapidly develops into acute respiratory distress syndrome (ARDS). Currently, no effective therapeutic modalities are available for patients with ALI/ARDS. According to recent studies, inhibiting both the release of pro-inflammatory cytokines and the formation of reactive oxygen species (ROS) as early as possible may be a promising therapy for ALI. RESULTS In this study, a ROS-responsive nano-delivery system based on oxidation-sensitive chitosan (Ox-CS) was fabricated for the simultaneous delivery of Ce NPs and RT. The in vitro experiments have shown that the Ox-CS/Ceria-Resatorvid nanoparticles (Ox-CS/CeRT NPs) were rapidly and efficiently internalised by inflammatory endothelial cells. Biological evaluations validated the significant attenuation of ROS-induced oxidative stress and cell apoptosis by Ox-CS/CeRT NPs, while maintaining mitochondrial function. Additionally, Ox-CS/CeRT NPs effectively inhibited the release of pro-inflammatory factors. After intraperitoneal (i.p.) administration, Ox-CS/CeRT NPs passively targeted the lungs of LPS-induced inflamed mice and released the drug activated by the high ROS levels in inflammatory tissues. Finally, Ox-CS/CeRT NPs significantly alleviated LPS-induced lung injury through inhibiting both oxidative stress and pro-inflammatory cytokine expression. CONCLUSIONS The created Ox-CS/CeRT NPs could act as a prospective nano-delivery system for a combination of anti-inflammatory and anti-oxidant therapy of ALI.
Collapse
Affiliation(s)
- Yue Wu
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmacy, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China
| | - Yawen Zhang
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmacy, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Xuanyu Tang
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmacy, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Shuhui Ye
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmacy, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Jingjing Shao
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China
| | - Linglan Tu
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmacy, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Junzhi Pan
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China
| | - Lingfeng Chen
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmacy, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| | - Guang Liang
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmacy, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| | - Lina Yin
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmacy, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, Zhejiang, China.
| |
Collapse
|
18
|
Polopalli S, Saha A, Niri P, Kumar M, Das P, Kamboj DV, Chattopadhyay P. ROCK Inhibitors as an Alternative Therapy for Corneal Grafting: A Systematic Review. J Ocul Pharmacol Ther 2023; 39:585-599. [PMID: 37738326 DOI: 10.1089/jop.2023.0040] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/24/2023] Open
Abstract
Currently, corneal blindness is affecting >10 million individuals worldwide, and there is a significant unmet medical need because only 1.5% of transplantation needs are met globally due to a lack of high-quality grafts. In light of this global health disaster, researchers are developing corneal substitutes that can resemble the human cornea in vivo and replace human donor tissue. Thus, this review examines ROCK (Rho-associated coiled-coil containing protein kinases) inhibitors as a potential corneal wound-healing (CWH) therapy by reviewing the existing clinical and nonclinical findings. The systematic review was done from PubMed, Scopus, Web of Science, and Google Scholar for CWH, corneal injury, corneal endothelial wound healing, ROCK inhibitors, Fasudil, Netarsudil, Ripasudil, Y-27632, clinical trial, clinical study, case series, case reports, preclinical study, in vivo, and in vitro studies. After removing duplicates, all downloaded articles were examined. The literature search included the data till January 2023. This review summarized the results of ROCK inhibitors in clinical and preclinical trials. In a clinical trial, various ROCK inhibitors improved CWH in individuals with open-angle glaucoma, cataract, iris cyst, ocular hypertension, and other ocular diseases. ROCK inhibitors also improved ocular wound healing by increasing cell adhesion, migration, and proliferation in vitro and in vivo. ROCK inhibitors have antifibrotic, antiangiogenic, anti-inflammatory, and antiapoptotic characteristics in CWH, according to the existing research. ROCK inhibitors were effective topical treatments for corneal infections. Ripasudil, Y-27632, H-1152, Y-39983, and AMA0526 are a few new ROCK inhibitors that may help CWH and replace human donor tissue.
Collapse
Affiliation(s)
- Subramanyam Polopalli
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, India
- Department of Chemical Technology, University of Calcutta, Kolkata, India
| | - Achintya Saha
- Department of Chemical Technology, University of Calcutta, Kolkata, India
| | - Pakter Niri
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, India
- Department of Chemical Technology, University of Calcutta, Kolkata, India
| | - Mohit Kumar
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, India
| | - Parikshit Das
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, India
| | - Dev Vrat Kamboj
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, India
| | - Pronobesh Chattopadhyay
- Division of Pharmaceutical Technology, Defence Research Laboratory (DRL), Defence Research and Development Organisation (DRDO), Tezpur, India
| |
Collapse
|
19
|
Yan C, Chen J, Tang H, Deng C, Zhang Q, Wang X. IgG immune complex-induced acute lung injury is ameliorated by cAMP via down-regulation of C/EBP- and AP-1-mediated transcriptions. J Inflamm (Lond) 2023; 20:34. [PMID: 37864223 PMCID: PMC10588139 DOI: 10.1186/s12950-023-00359-6] [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: 02/23/2022] [Accepted: 09/28/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS) are life threatening pulmonary diseases, and we are now lack of effective therapeutic methods. Inflammatory responses are essential for initiating ALI/ARDS. Thus, ameliorating inflammatory reaction might be beneficial for treatment of the disease. There are increasing data that phosphodiesterase-4 (PDE4)-selective inhibitors, which may elevate cellular cyclic adenosine 3', 5'-monophosphate (cAMP) level, could suppress inflammation. However, whether they could be used to treat IgG immune complex (IgG-IC)-associated ALI has not been determined. METHODS ALI is induced by treating mice with airway deposition of IgG immune complexes. Cellular cAMP concentrations are elevated by treating mice or macrophages with Rolipram/Roflumilast. The degree of pulmonary injury is reflected by lung permeability, leukocyte accumulation, histological change and expressions of pro-inflammatory mediators. 6-Bnz-cAMP and H-89 are used to regulate protein kinase A (PKA) activity, and 8-pCPT-2'-O-Me-cAMP is applied to activate exchange proteins directly activated by cAMP (Epac). Gene expressions are analyzed by real-time PCR, ELISA or Western blot. CCAAT/enhancer binding protein (C/EBP) and activation protein 1 (AP-1) transcription activities are estimated by measuring the luciferase productions. RESULTS IgG-IC-induced ALI is attenuated by the PDE4-selective inhibitor, which is due to reduced expressions of cytokine and chemokines. Interestingly, we find that cAMP downstream effector molecule PKA but not Epac is involved in negative regulation of IgG-IC-mediated pro-inflammatory mediators' productions. Mechanistically, activation of cAMP-PKA signal axis leads to inactivation of MAPK pathway, resulting in a decrease in C/EBP- and AP-1-mediated transcriptions of pro-inflammatory mediators. CONCLUSIONS Our data demonstrate, for the first time, that cAMP-PKA signal is involved in down-regulation of IgG-IC-associated inflammatory responses via down-regulating MAPK activation, which is critical for transcriptional activities of C/EBP and AP-1. Collectively, our experiments provide theoretical base for the potential application of PDE4-selective inhibitor to clinic for treatment of IgG-IC-related acute lung injury.
Collapse
Affiliation(s)
- Chunguang Yan
- Department of Pathogenic Biology and Immunology, Medical School of Southeast University, Nanjing, 210009, China.
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital of Southeast University, Nanjing, 210009, China.
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine for Acute Abdominal Diseases, Integrated Chinese and Western Medicine Hospital, Tianjin University, Tianjin, 300100, China.
| | - Jing Chen
- Department of Pathogenic Biology and Immunology, Medical School of Southeast University, Nanjing, 210009, China.
| | - Huifang Tang
- Zhejiang Respiratory Drugs Research Laboratory of the State Food and Drug Administration of China, School of Medicine, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Chunmin Deng
- Department of Clinical Laboratory Medicine, Suzhou Science and Technology Town Hospital, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, 215153, China
| | - Qi Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine for Acute Abdominal Diseases, Integrated Chinese and Western Medicine Hospital, Tianjin University, Tianjin, 300100, China
| | - Ximo Wang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine for Acute Abdominal Diseases, Integrated Chinese and Western Medicine Hospital, Tianjin University, Tianjin, 300100, China.
| |
Collapse
|
20
|
Liu P, Xiao Z, Lu X, Zhang X, Huang J, Li C. Fasudil and SR1001 synergistically protect against sepsis-associated pancreatic injury by inhibiting RhoA/ROCK pathway and Th17/IL-17 response. Heliyon 2023; 9:e20118. [PMID: 37809525 PMCID: PMC10559842 DOI: 10.1016/j.heliyon.2023.e20118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
Sepsis is defined as a dysregulated host response to infection that can result in organ dysfunction and high mortality, which needs more effective treatment urgently. Pancreas is one of the most vulnerable organs in sepsis, resulting in sepsis-associated pancreatic injury, which is a fatal complication of sepsis. The aim of this study was to investigate the effect of combination of fasudil and SR1001 on sepsis-associated pancreatic injury and to explore the underlying mechanisms. The model of sepsis-associated pancreatic injury was induced by cecal ligation and puncture. Pancreatic injury was evaluated by HE staining, histopathological scores and amylase activity. The frequency of Th17 cells was analyzed by flow cytometry. Serum IL-17 level was determined by ELISA. Protein levels of RORγt, p-STAT3, GEF-H1, RhoA and ROCK1 were determined by Western blot. The apoptosis of pancreatic cells was examined by TUNEL analysis and Hoechst33342/PI staining. Compared to the sham group, the model group showed significant pathological injury including edema, hyperemia, vacuolization and necrosis. After treatment with fasudil, model mice showed an obvious reduction of Th17 cells and IL-17. SR1001 significantly reduced the expressions of GEF-H1, RhoA and ROCK1 in the model mice. The combination treatment with fasudil and SR1001 significantly inhibited the differentiation of Th17 cells, expressions of IL-17, GEF-H1, RhoA and ROCK1, which were more effective than each mono-treatment. In addition, our data revealed a remarkable decrease of apoptosis in pancreatic acinar cells culturing with fasudil or SR1001, which was further inhibited by their combination culture. Lipopolysaccharide remarkably upregulated the differentiation of Th17 cells in vitro, which could be significantly downregulated by fasudil or SR1001, and further downregulated by their combination treatment. Taken together, the combination of fasudil with SR1001 has a synergistic effect on protecting against sepsis-associated pancreatic injury in C57BL/6 mice.
Collapse
Affiliation(s)
- Pingping Liu
- Department of Emergency, Key Laboratory of Pediatric Emergency Medicine of Hunan Province, Hunan Children's Hospital, Changsha, 410007, Hunan, PR China
| | - Zhenghui Xiao
- Department of Emergency, Key Laboratory of Pediatric Emergency Medicine of Hunan Province, Hunan Children's Hospital, Changsha, 410007, Hunan, PR China
| | - Xiulan Lu
- Department of Emergency, Key Laboratory of Pediatric Emergency Medicine of Hunan Province, Hunan Children's Hospital, Changsha, 410007, Hunan, PR China
| | - Xinping Zhang
- Department of Emergency, Key Laboratory of Pediatric Emergency Medicine of Hunan Province, Hunan Children's Hospital, Changsha, 410007, Hunan, PR China
| | - Jiaotian Huang
- Department of Emergency, Key Laboratory of Pediatric Emergency Medicine of Hunan Province, Hunan Children's Hospital, Changsha, 410007, Hunan, PR China
| | - Cheng Li
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, PR China
| |
Collapse
|
21
|
Yu H, Lv M, Zhang S, Zou K, Qian Y, Lv S. Combination therapy with budesonide and acetylcysteine alleviates LPS-induced acute lung injury via the miR-381/NLRP3 molecular axis. PLoS One 2023; 18:e0289818. [PMID: 37556466 PMCID: PMC10411794 DOI: 10.1371/journal.pone.0289818] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI) usually has a high morbidity and mortality rate, but the current treatment is relatively scarce. Both budesonide (Bud) and N-acetylcysteine (NAC) exhibit protective effects in ALI, so we further investigated whether they have a synergistic effect on ALI when used together. METHODS Establishment of a rat model of ALI with Lipopolysaccharide (LPS). Bud and NAC were administered by nebulized inhalation alone or in combination. Subsequently, HE staining was performed to observe the pathological changes in lungs of rat. Evans blue staining was implemented to assess alveolar permeability, and the pulmonary edema was assessed by measuring the ratio of wet to dry weight of the lung. Moreover, a TUNEL kit was served to test apoptosis in lung tissues. Western blot and immunohistochemistry were analyzed for expression of scorch-related proteins and NLRP3 in lung tissue, respectively. ELISA was implemented to detect inflammatory factor levels in BALF. and RT-qPCR was utilized to assess the expression level of miR-381. After stable transfection of miR-381 inhibitor or OE-NLRP3 in BEAS-2B treated with LPS, Bud and NAC, miR-381 expression was assessed by RT-qPCR, scorch death-related protein expression was measured by western blot, cell proliferation/viability was assayed by CCK-8, apoptosis was measured by flow cytometry, and ELISA was implemented to assess inflammatory factor levels. Furthermore, the Dual-luciferase assay was used to verify the targeting relationship. RESULTS Bud and NAC treatment alone or in combination with nebulized inhalation attenuated the increased alveolar permeability, pulmonary edema, inflammatory response and scorching in LPS-induced ALI rats, and combined treatment with Bud and NAC was the most effective. In addition, combined treatment with Bud and NAC upregulated miR-381 expression and inhibited NLRP3 expression in cellular models and LPS-induced ALI rats. Transfection of the miR-381 inhibitor and OE-NLRP3 partially reversed the protective effects of Bud and NAC combination treatment on BEAS-2B cell proliferation inhibition, apoptosis, focal death and the inflammatory response. CONCLUSION Combined Bud and NAC nebulization therapy alleviates LPS-induced ALI by modulating the miR-381/NLRP3 molecular axis.
Collapse
Affiliation(s)
- Huimin Yu
- Department of Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Meifen Lv
- Department of Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Shiying Zhang
- Department of Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Kaiwen Zou
- Department of Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Yan Qian
- Department of Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| | - Shaokun Lv
- Department of Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan, China
| |
Collapse
|
22
|
Ni J, Li G, Dai N, Quan Z, Tong H, Liu Y. Esculin alleviates LPS-induced acute lung injury via inhibiting neutrophil recruitment and migration. Int Immunopharmacol 2023; 119:110177. [PMID: 37068336 PMCID: PMC10105132 DOI: 10.1016/j.intimp.2023.110177] [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: 02/13/2023] [Revised: 03/25/2023] [Accepted: 04/09/2023] [Indexed: 04/19/2023]
Abstract
OBJECTIVES Acute lung injury (ALI) poses a serious threat to human health globally, particularly with the Coronavirus 2019 (COVID-19) pandemic. Excessive recruitment and infiltration of neutrophils is the major etiopathogenesis of ALI. Esculin, also known as 6,7-dihydroxycoumarin, is a remarkable compound derived from traditional Chinese medicine Cortex fraxini. Accumulated evidence indicates that esculin has potent anti-inflammatory effects, but its pharmaceutical effect against ALI and potential mechanisms are still unclear. METHODS This study evaluated the protective effect of esculin against ALI by histopathological observation and biochemical analysis of lung tissues and bronchoalveolar lavage fluid (BALF) in lipopolysaccharide (LPS)-challenged ALI mice in vivo. The effects of esculin on N-formyl-met-leu-phe (fMLP)-induced neutrophil migration and chemotaxis were quantitatively assessed using a Transwell assay and an automated cell imaging system equipped with a Zigmond chamber, respectively. The drug affinity responsive target stability (DARTS) assay, in vitro protein binding assay and molecular docking were performed to identify the potential therapeutic target of esculin and the potential binding sites and pattern. RESULTS Esculin significantly attenuated LPS-induced lung pathological injury, reduced the levels of pro-inflammatory cytokines in both BALF and lung, and suppressed the activation of NF-κB signaling. Esculin also significantly reduced the number of total cells and neutrophils as well as myeloperoxidase (MPO) activity in the BALF. Esculin impaired neutrophil migration and chemotaxis as evidenced by the reduced migration distance and velocity. Furthermore, esculin remarkably inhibited Vav1 phosphorylation, suppressed Rac1 activation and the PAK1/LIMK1/cofilin signaling axis. Mechanistically, esculin could interact with β2 integrin and then diminish its ligand affinity with intercellular adhesion molecule-1 (ICAM-1). CONCLUSIONS Esculin inhibits β2 integrin-dependent neutrophil migration and chemotaxis, blocks the cytoskeletal remodeling process required for neutrophil recruitment, thereby contributing to its protective effect against ALI. This study demonstrates the new therapeutic potential of esculin as a novel lead compound.
Collapse
Affiliation(s)
- Jiangwei Ni
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Ge Li
- Department of Thoracic Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, PR China
| | - Ningfeng Dai
- Department of Thoracic Surgery, The Affiliated Cangnan Hospital of Wenzhou Medical University, Wenzhou 325800, PR China
| | - Zijiao Quan
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, PR China
| | - Haibin Tong
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325000, PR China.
| | - Yu Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China.
| |
Collapse
|
23
|
Dave BP, Shah KC, Shah MB, Chorawala MR, Patel VN, Shah PA, Shah GB, Dhameliya TM. Unveiling the modulation of Nogo receptor in neuroregeneration and plasticity: Novel aspects and future horizon in a new frontier. Biochem Pharmacol 2023; 210:115461. [PMID: 36828272 DOI: 10.1016/j.bcp.2023.115461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023]
Abstract
Neurodegenerative diseases (NDs) such as Alzheimer's, Parkinson's, Multiple Sclerosis, Hereditary Spastic Paraplegia, and Amyotrophic Lateral Sclerosis have emerged as the most dreaded diseases due to a lack of precise diagnostic tools and efficient therapies. Despite the fact that the contributing factors of NDs are still unidentified, mounting evidence indicates the possibility that genetic and cellular changes may lead to the significant production of abnormally misfolded proteins. These misfolded proteins lead to damaging effects thereby causing neurodegeneration. The association between Neurite outgrowth factor (Nogo) with neurological diseases and other peripheral diseases is coming into play. Three isoforms of Nogo have been identified Nogo-A, Nogo-B and Nogo-C. Among these, Nogo-A is mainly responsible for neurological diseases as it is localized in the CNS (Central Nervous System), whereas Nogo-B and Nogo-C are responsible for other diseases such as colitis, lung, intestinal injury, etc. Nogo-A, a membrane protein, had first been described as a CNS-specific inhibitor of axonal regeneration. Several recent studies have revealed the role of Nogo-A proteins and their receptors in modulating neurite outgrowth, branching, and precursor migration during nervous system development. It may also modulate or affect the inhibition of growth during the developmental processes of the CNS. Information about the effects of other ligands of Nogo protein on the CNS are yet to be discovered however several pieces of evidence have suggested that it may also influence the neuronal maturation of CNS and targeting Nogo-A could prove to be beneficial in several neurodegenerative diseases.
Collapse
Affiliation(s)
- Bhavarth P Dave
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Kashvi C Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Maitri B Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India.
| | - Vishvas N Patel
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Palak A Shah
- Department of Pharmacology, K. B. Institute of Pharmaceutical Education and Research, Gandhinagar 380023, Gujarat, India
| | - Gaurang B Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Tejas M Dhameliya
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad-382481, Gujarat, India
| |
Collapse
|
24
|
Tang Y, Chen K, Xiao Z, Hong G, Hu L, Cai J, Lu Z. A novel mechanism of Dimethyl ester of Alpha-ketoglutarate in suppressing Paraquat-induced BEAS-2B cell injury by alleviating GSDME dependent pyroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154698. [PMID: 36773430 DOI: 10.1016/j.phymed.2023.154698] [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: 10/11/2022] [Revised: 01/09/2023] [Accepted: 02/02/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Acute lung injury (ALI) induced by paraquat (PQ) progresses rapidly, leading to high mortality; however, there is no specific antidote. Our limited knowledge of the pathogenic toxicological mechanisms of PQ has hindered the development of treatments against PQ exposure. PURPOSE Pyroptosis is a form of programmed cell death recently identified as a novel molecular mechanism adopted by chemotherapeutic drugs for cancer therapy. However, the involvement of pyroptosis in PQ-induced lung injury has not been reported. Therefore, we investigated the effects of PQ on the lung tissues to elucidate the molecular mechanisms underlying its toxicity, especially its ability to induce pyroptosis. METHODS To observe the morphological changes of BEAS-2B cells exposed to PQ, the plasma membrane damage of the cells was detected by LDH release assay, mitochondrial function and cell metabolism were detected by energy metabolism analysis. Western blotting was used to detect the protein levels of GSDMD, C-GSDMD, GSDME and N-GSDME in BEAS-2B cells. Metabolites of TCA cycle were detected by metabolomics, and the changes of TCA cycle metabolic enzymes in cells were detected by Western blotting. RESULTS We observed that PQ induced proteolytic cleavage of gasdermin E (GSDME) with concomitant cleavage of caspase 3 in BEAS-2B cells. Knockout of GSDME attenuated PQ-induced cell death. Additionally, PQ induced ROS accumulation, mitochondrial depolarisation, and mitochondrial dysfunction in these cells. PQ activated the caspase 3/GSDME pathway and damaged the cytoplasmic membrane in cells, leading to pyroptosis. We demonstrated that DMK suppressed PQ-induced pyroptosis by blocking PQ-induced caspase 3/GSDME pathway activation, reducing cellular ROS levels, and improving mitochondrial function. CONCLUSION These findings provide novel insights into the previously unrecognized mechanism of GSDME-dependent pyroptosis in PQ poisoning.
Collapse
Affiliation(s)
- Yahui Tang
- Department of Emergency Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325000, China.
| | - Kaiyuan Chen
- Department of Emergency Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325000, China
| | - Zhong Xiao
- Department of Emergency Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325000, China
| | - Guangliang Hong
- Department of Emergency Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325000, China
| | - Lufeng Hu
- Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325000, China; Department of Pharmacy, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianping Cai
- Ministry of Health Key Laboratory of Geriatrics (J.-P.C.), Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, China
| | - Zhongqiu Lu
- Department of Emergency Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325000, China.
| |
Collapse
|
25
|
Ru Q, Wang Y, Zhou E, Chen L, Wu Y. The potential therapeutic roles of Rho GTPases in substance dependence. Front Mol Neurosci 2023; 16:1125277. [PMID: 37063367 PMCID: PMC10097952 DOI: 10.3389/fnmol.2023.1125277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/14/2023] [Indexed: 04/03/2023] Open
Abstract
Rho GTPases family are considered to be molecular switches that regulate various cellular processes, including cytoskeleton remodeling, cell polarity, synaptic development and maintenance. Accumulating evidence shows that Rho GTPases are involved in neuronal development and brain diseases, including substance dependence. However, the functions of Rho GTPases in substance dependence are divergent and cerebral nuclei-dependent. Thereby, comprehensive integration of their roles and correlated mechanisms are urgently needed. In this review, the molecular functions and regulatory mechanisms of Rho GTPases and their regulators such as GTPase-activating proteins (GAPs) and guanine nucleotide exchange factors (GEFs) in substance dependence have been reviewed, and this is of great significance for understanding their spatiotemporal roles in addictions induced by different addictive substances and in different stages of substance dependence.
Collapse
Affiliation(s)
| | | | | | - Lin Chen
- *Correspondence: Lin Chen, ; Yuxiang Wu,
| | - Yuxiang Wu
- *Correspondence: Lin Chen, ; Yuxiang Wu,
| |
Collapse
|
26
|
Pan T, Cao G, Tang E, Zhao Y, Penaloza-MacMaster P, Fang Y, Huang J. A single-cell atlas reveals shared and distinct immune responses and metabolic profiles in SARS-CoV-2 and HIV-1 infections. Front Genet 2023; 14:1105673. [PMID: 36992700 PMCID: PMC10040851 DOI: 10.3389/fgene.2023.1105673] [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: 11/22/2022] [Accepted: 03/01/2023] [Indexed: 03/16/2023] Open
Abstract
Introduction: Within the inflammatory immune response to viral infection, the distribution and cell type-specific profiles of immune cell populations and the immune-mediated viral clearance pathways vary according to the specific virus. Uncovering the immunological similarities and differences between viral infections is critical to understanding disease progression and developing effective vaccines and therapies. Insight into COVID-19 disease progression has been bolstered by the integration of single-cell (sc)RNA-seq data from COVID-19 patients with data from related viruses to compare immune responses. Expanding this concept, we propose that a high-resolution, systematic comparison between immune cells from SARS-CoV-2 infection and an inflammatory infectious disease with a different pathophysiology will provide a more comprehensive picture of the viral clearance pathways that underscore immunological and clinical differences between infections. Methods: Using a novel consensus single-cell annotation method, we integrate previously published scRNA-seq data from 111,566 single PBMCs from 7 COVID-19, 10 HIV-1+, and 3 healthy patients into a unified cellular atlas. We compare in detail the phenotypic features and regulatory pathways in the major immune cell clusters. Results: While immune cells in both COVID-19 and HIV-1+ cohorts show shared inflammation and disrupted mitochondrial function, COVID-19 patients exhibit stronger humoral immunity, broader IFN-I signaling, elevated Rho GTPase and mTOR pathway activity, and downregulated mitophagy. Discussion: Our results indicate that differential IFN-I signaling regulates the distinct immune responses in the two diseases, revealing insight into fundamental disease biology and potential therapeutic candidates.
Collapse
Affiliation(s)
- Tony Pan
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Guoshuai Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Erting Tang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Yu Zhao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | | | - Yun Fang
- Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - Jun Huang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| |
Collapse
|
27
|
Zhao G, Ren Y, Yan J, Zhang T, Lu P, Lei J, Rao H, Kang X, Cao Z, Peng F, Peng C, Rao C, Li Y. Neoprzewaquinone A Inhibits Breast Cancer Cell Migration and Promotes Smooth Muscle Relaxation by Targeting PIM1 to Block ROCK2/STAT3 Pathway. Int J Mol Sci 2023; 24:ijms24065464. [PMID: 36982538 PMCID: PMC10051292 DOI: 10.3390/ijms24065464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Salvia miltiorrhiza Bunge (Danshen) has been widely used to treat cancer and cardiovascular diseases in Chinese traditional medicine. Here, we found that Neoprzewaquinone A (NEO), an active component of S. miltiorrhiza, selectively inhibits PIM1. We showed that NEO potently inhibits PIM1 kinase at nanomolar concentrations and significantly suppresses the growth, migration, and Epithelial-Mesenchymal Transition (EMT) in the triple-negative breast cancer cell line, MDA-MB-231 in vitro. Molecular docking simulations revealed that NEO enters the PIM1 pocket, thereby triggering multiple interaction effects. Western blot analysis revealed that both NEO and SGI-1776 (a specific PIM1 inhibitor), inhibited ROCK2/STAT3 signaling in MDA-MB-231 cells, indicating that PIM1 kinase modulates cell migration and EMT via ROCK2 signaling. Recent studies indicated that ROCK2 plays a key role in smooth muscle contraction, and that ROCK2 inhibitors effectively control the symptoms of high intraocular pressure (IOP) in glaucoma patients. Here, we showed that NEO and SGI-1776 significantly reduce IOP in normal rabbits and relax pre-restrained thoracic aortic rings in rats. Taken together, our findings indicated that NEO inhibits TNBC cell migration and relaxes smooth muscles mainly by targeting PIM1 and inhibiting ROCK2/STAT3 signaling, and that PIM1 may be an effective target for IOP and other circulatory diseases.
Collapse
Affiliation(s)
- Guiying Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yali Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jie Yan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tingrui Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Peng Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jieting Lei
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Huanan Rao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xin Kang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhixing Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Fu Peng
- West China School of Pharmacy, Sichuan University, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chaolong Rao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica with Medical and Edible Values, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Correspondence: (C.R.); (Y.L.)
| | - Yuzhi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Correspondence: (C.R.); (Y.L.)
| |
Collapse
|
28
|
Deng Y, Huang X, Hu Y, Zhong W, Zhang H, Mo C, Wang H, Ding BS, Wang C. Deficiency of endothelial FGFR1 signaling via upregulation of ROCK2 activity aggravated ALI/ARDS. Front Immunol 2023; 14:1041533. [PMID: 36969192 PMCID: PMC10036754 DOI: 10.3389/fimmu.2023.1041533] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/13/2023] [Indexed: 03/12/2023] Open
Abstract
Vascular leakage and inflammation are pathological hallmarks of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). Endothelial cells (ECs) serve as a semipermeable barrier and play a key role in disease progression. It is well known that fibroblast growth factor receptor 1 (FGFR1) is required for maintaining vascular integrity. However, how endothelial FGFR1 functions in ALI/ARDS remains obscure. Here, we revealed that conditional deletion of endothelial FGFR1 aggravated LPS-induced lung injury, including inflammation and vascular leakage. Inhibition of its downstream Rho-associated coiled-coil–forming protein kinase 2 (ROCK2) by AAV Vec-tie-shROCK2 or its selective inhibitor TDI01 effectively attenuated inflammation and vascular leakage in a mouse model. In vitro, TNFα-stimulated human umbilical vein endothelial cells (HUVECs) showed decreased FGFR1 expression and increased ROCK2 activity. Furthermore, knockdown of FGFR1 activated ROCK2 and thus promoted higher adhesive properties to inflammatory cells and higher permeability in HUVECs. TDI01 effectively suppressed ROCK2 activity and rescued the endothelial dysfunction. These data demonstrated that the loss of endothelial FGFR1 signaling mediated an increase in ROCK2 activity, which led to an inflammatory response and vascular leakage in vivo and in vitro. Moreover, inhibition of ROCK2 activity by TDI01 provided great value and shed light on clinical translation.
Collapse
Affiliation(s)
- Yue Deng
- Peking University China–Japan Friendship School of Clinical Medicine, Beijing, China
- National Center for Respiratory Medicine, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China
| | - Xingming Huang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yan Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Weiting Zhong
- Beijing Tide Pharmaceutical Co., Ltd., Beijing, China
| | - Hua Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Chunheng Mo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hongjun Wang
- Beijing Tide Pharmaceutical Co., Ltd., Beijing, China
- *Correspondence: Chen Wang, ; Bi-Sen Ding, ; Hongjun Wang,
| | - Bi-Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- *Correspondence: Chen Wang, ; Bi-Sen Ding, ; Hongjun Wang,
| | - Chen Wang
- Peking University China–Japan Friendship School of Clinical Medicine, Beijing, China
- National Center for Respiratory Medicine, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- *Correspondence: Chen Wang, ; Bi-Sen Ding, ; Hongjun Wang,
| |
Collapse
|
29
|
Yang J, Wang M, Xu Y, Liao J, Li X, Zhou Y, Dai J, Li X, Chen P, Chen G, Cho WJ, Chattipakorn N, Samorodov AV, Pavlov VN, Wang Y, Liang G, Tang Q. Discovery of 4-oxo-N-phenyl-1,4-dihydroquinoline-3-carboxamide derivatives as novel anti-inflammatory agents for the treatment of acute lung injury and sepsis. Eur J Med Chem 2023; 249:115144. [PMID: 36708679 DOI: 10.1016/j.ejmech.2023.115144] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 01/09/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023]
Abstract
Acute lung injury (ALI) and sepsis, characterized by systemic inflammatory response syndrome, remain the major causes of death in severe patients. Inhibiting the release of proinflammatory cytokines is considered to be a promising method for the treatment of inflammation-related diseases. In this study, a total of 28 4-oxo-N-phenyl-1,4-dihydroquinoline-3-carboxamide derivatives were designed and synthesized and their anti-inflammatory activities in J774A.1 were evaluated. Among them, derivative 13a was found to significantly inhibit lipopolysaccharide (LPS)-induced expression of the proinflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) on J774A.1, THP-1 and LX-2 cells, and inhibited the activation of the NF-κB pathway. Furthermore, administration of 13ain vivo significantly improved the symptoms in LPS-induced ALI mice, including alleviation of pathological changes in the lung tissue, reduction of pulmonary edema, and inhibition of macrophage infiltration. Moreover, the administration of 13ain vivo significantly promoted survival in LPS-induced sepsis mice. 13a demonstrated favorable pharmacokinetic properties with T1/2 value of 11.8 h and F value of 36.3%. Therefore, this study has identified a novel 4-oxo-N-phenyl-1,4-dihydroquinoline-3-carboxamide derivative, 13a, which is an effective anti-inflammatory agent. The findings have laid a foundation for the further development of agents to treat ALI and sepsis.
Collapse
Affiliation(s)
- Jun Yang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Minxiu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Yulan Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Jing Liao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Xiang Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Ying Zhou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Jintian Dai
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325024, Zhejiang, China
| | - Xiaobo Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Pan Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; College of Pharmacy, Chonnam National University, Gwangju, 61186, South Korea
| | - Gaozhi Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China
| | - Won-Jea Cho
- College of Pharmacy, Chonnam National University, Gwangju, 61186, South Korea
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Aleksandr V Samorodov
- Department of Pharmacology, Bashkir State Medical University, Ufa City 450005, Russia
| | - Valentin N Pavlov
- Department of Pharmacology, Bashkir State Medical University, Ufa City 450005, Russia
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325024, Zhejiang, China; School of Pharmacy, Hangzhou Medical College, Hangzhou 311399, Zhejiang, China.
| | - Qidong Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325024, Zhejiang, China.
| |
Collapse
|
30
|
Lazrak A, Song W, Yu Z, Zhang S, Nellore A, Hoopes CW, Woodworth BA, Matalon S. Low molecular weight hyaluronan inhibits lung epithelial ion channels by activating the calcium-sensing receptor. Matrix Biol 2023; 116:67-84. [PMID: 36758905 PMCID: PMC10012407 DOI: 10.1016/j.matbio.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
Herein, we tested the hypothesis that low molecular weight hyaluronan (LMW-HA) inhibits lung epithelial ions transport in-vivo, ex-vivo, and in-vitro by activating the calcium-sensing receptor (CaSR). Twenty-four hours post intranasal instillation of 50-150 µg/ml LMW-HA to C57BL/6 mice, there was a 75% inhibition of alveolar fluid clearance (AFC), a threefold increase in the epithelial lining fluid (ELF) depth, and a 20% increase in lung wet/dry (W/D) ratio. Incubation of human and mouse precision cut lung slices with 150 µg/ml LMW-HA reduced the activity and the open probability (Po) of epithelial sodium channel (ENaC) in alveolar epithelial type 2 (ATII) cells, and in mouse tracheal epithelial cells (MTEC) monolayers as early as 4 h. The Cl- current through cystic fibrosis transmembrane conductance regulator (CFTR) and the activity of Na,K-ATPase were both inhibited by more than 66% at 24 h. The inhibitory effects of LMW-HA on ion channels were reversed by 1 µM NPS-2143, or 150 µg/ml high molecular weight hyaluronan (HMW-HA). In HEK-293 cells expressing the calcium-sensitive Cl- channel TMEM16-A, CaSR was required for the activation of the Cl- current by LMW-HA. This is the first demonstration of lung ions and water transport inhibition by LMW-HA, and its mediation through the activation of CaSR.
Collapse
Affiliation(s)
- Ahmed Lazrak
- Department of Anesthesiology and Perioperative Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Division of Molecular and Translational Biomedicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Pulmonary Injury and Repair Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Gregory Fleming James Cystic Fibrosis Research Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA.
| | - Weifeng Song
- Department of Anesthesiology and Perioperative Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Division of Molecular and Translational Biomedicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Zhihong Yu
- Department of Anesthesiology and Perioperative Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Division of Molecular and Translational Biomedicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Pulmonary Injury and Repair Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Shaoyan Zhang
- Department of Otolaryngology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Gregory Fleming James Cystic Fibrosis Research Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Anoma Nellore
- Department of Medicine, Division of Infectious Diseases, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Charles W Hoopes
- Division of Cardiothoracic Surgery, Heersink School of Medicine, University of Alabama at Birmingham, AL 35295, USA
| | - Bradford A Woodworth
- Department of Otolaryngology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Gregory Fleming James Cystic Fibrosis Research Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Division of Molecular and Translational Biomedicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Pulmonary Injury and Repair Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| |
Collapse
|
31
|
Kan W, Li Q, Li P, Ren L, Mu W, Lin L, Wen J, Ge F, Hou M, Hui S, He P, Liang L, Xu Y, Li X, Xu G, Xiao X, Bai Z. Glycyrrhiza uralensis polysaccharides ameliorate acute lung injury by inhibiting the activation of multiple inflammasomes. J Funct Foods 2023. [DOI: 10.1016/j.jff.2022.105386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
32
|
Chang J, Zhang W. Remifentanil modulates the TLR4‑mediated MMP‑9/TIMP1 balance and NF‑κB/STAT3 signaling in LPS‑induced A549 cells. Exp Ther Med 2022; 25:79. [PMID: 36684659 PMCID: PMC9842940 DOI: 10.3892/etm.2022.11778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/28/2022] [Indexed: 01/01/2023] Open
Abstract
Remifentanil is a widely used in general anesthetic that has been found to suppress the inflammatory response in aortic endothelial cells. Therefore, it was hypothesized that remifentanil can inhibit inflammatory dysfunction in lung epithelial cells to alleviate acute lung injury (ALI). The present study aimed to examine the effects of remifentanil on inflammatory injury, MMP-9/tissue inhibitor of metalloproteinase 1 (TIMP1) balance and the potential associated regulatory pathways in A549 cells. Lipopolysaccharide (LPS) was used to treat A549 cells to establish ALI models. The possible roles of different concentrations of remifentanil in cell viability was then determined by CCK-8 and Lactate dehydrogenase release assay. Apoptosis was assessed by flow cytometry analysis and western blotting. Inflammation and oxidative stress were measured by ELISA and corresponding kits respectively. Subsequently, the effects of remifentanil on Toll-like receptor 4 (TLR4) expression and the MMP-9/TIMP1 balance were assessed by western blotting and ELISA. In addition, the effects of remifentanil on NF-κB/STAT3 signaling were evaluated by measuring the protein expression levels of associated pathway components and the degree of NF-κB nuclear translocation using western blotting and immunofluorescence respectively. Remifentanil was found to increase cell viability whilst reducing apoptosis, inflammation and oxidative stress in the LPS-treated cells. In addition, TLR4 inhibitor CLI-095 suppressed MMP-9 expression and secretion while potentiating TIMP1 expression and secretion in LPS-challenged cells. Remifentanil treatment was able to modulate TLR4 to mediate LPS-induced MMP-9/TIMP1 imbalance and suppress the phosphorylation of NF-κB/STAT3 signaling components, in addition to inhibiting NF-κB nuclear translocation. Taken together, remifentanil downregulated TLR4 to reduce MMP-9/TIMP1 imbalance to inhibit inflammatory dysfunction in LPS-treated A549 cells, by regulating NF-κB/STAT3 signaling.
Collapse
Affiliation(s)
- Jun Chang
- Department of Anesthesiology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, Shanxi 030029, P.R. China
| | - Wei Zhang
- Department of Thoracic Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, Shanxi 030029, P.R. China,Correspondence to: Dr Wei Zhang, Department of Thoracic Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, 3 Zhigongxin Street, Taiyuan, Shanxi 030029, P.R. China
| |
Collapse
|
33
|
Jeon JH, Im S, Kim HS, Lee D, Jeong K, Ku JM, Nam TG. Chemical Chaperones to Inhibit Endoplasmic Reticulum Stress: Implications in Diseases. Drug Des Devel Ther 2022; 16:4385-4397. [PMID: 36583112 PMCID: PMC9793730 DOI: 10.2147/dddt.s393816] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022] Open
Abstract
The endoplasmic reticulum (ER) is responsible for structural transformation or folding of de novo proteins for transport to the Golgi. When the folding capacity of the ER is exceeded or excessive accumulation of misfolded proteins occurs, the ER enters a stressed condition (ER stress) and unfolded protein responses (UPR) are triggered in order to rescue cells from the stress. Recovery of ER proceeds toward either survival or cell apoptosis. ER stress is implicated in many pathologies, such as diabetes, cardiovascular diseases, inflammatory diseases, neurodegeneration, and lysosomal storage diseases. As a survival or adaptation mechanism, chaperone molecules are upregulated to manage ER stress. Chemical versions of chaperone have been developed in search of drug candidates for ER stress-related diseases. In this review, synthetic or semi-synthetic chemical chaperones are categorized according to potential therapeutic area and listed along with their chemical structure and activity. Although only a few chemical chaperones have been approved as pharmaceutical drugs, a dramatic increase in literatures over the recent decades indicates enormous amount of efforts paid by many researchers. The efforts warrant clearer understanding of ER stress and the related diseases and consequently will offer a promising drug discovery platform with chaperone activity.
Collapse
Affiliation(s)
- Jae-Ho Jeon
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University ERICA campus, Ansan, Gyeonggi-do, 15588, Republic of Korea
| | - Somyoung Im
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University ERICA campus, Ansan, Gyeonggi-do, 15588, Republic of Korea
| | - Hyo Shin Kim
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University ERICA campus, Ansan, Gyeonggi-do, 15588, Republic of Korea
| | - Dongyun Lee
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University ERICA campus, Ansan, Gyeonggi-do, 15588, Republic of Korea
| | - Kwiwan Jeong
- Gyeonggi Bio-Center, Gyeonggido Business and Science Accelerator, Suwon, Gyeonggi-do, 16229, Republic of Korea
| | - Jin-Mo Ku
- Gyeonggi Bio-Center, Gyeonggido Business and Science Accelerator, Suwon, Gyeonggi-do, 16229, Republic of Korea
| | - Tae-Gyu Nam
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University ERICA campus, Ansan, Gyeonggi-do, 15588, Republic of Korea,Correspondence: Tae-Gyu Nam, Tel +82-31-400-5807, Fax +82-31-400-5958, Email
| |
Collapse
|
34
|
Elsholtzia bodinieri Vaniot Ameliorated Acute Lung Injury by NQO1, BCL2 and PTGS2 In Silico and In Vitro Analyses. Int J Mol Sci 2022; 23:ijms232415651. [PMID: 36555290 PMCID: PMC9779453 DOI: 10.3390/ijms232415651] [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: 10/03/2022] [Revised: 11/29/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Acute lung injury (ALI) is a clinical respiratory disease caused by various factors, which lacks effective pharmacotherapy to reduce the mortality rate. Elsholtzia bodinieri Vaniot is an annual herbaceous plant used as a traditional herbal tea and folk medicine. Here we used bioinformatic databases and software to explore and analyze the potential key genes in ALI regulated by E. bodinieri Vaniot, including B cell leukemia/lymphoma 2 (Bcl2), prostaglandin-endoperoxide synthase 2 (Ptgs2) and NAD(P)H dehydrogenase, quinone 1 (Nqo1). In an inflammatory cells model, we verified bioinformatics results, and further mechanistic analysis showed that methanol extract of E. bodinieri Vaniot (EBE) could alleviate oxidative stress by upregulating the expression of NQO1, suppress pyroptosis by upregulating the expression of BCL2, and attenuate inflammation by downregulating the expression of PTGS2. In sum, our results demonstrated that EBE treatment could alleviate oxidative stress, suppress pyroptosis and attenuate inflammation by regulating NQO1, BCL2 and PTGS2 in a cells model, and E. bodinieri Vaniot might be a promising source for functional food or as a therapeutic agent.
Collapse
|
35
|
Signaling pathways involved in paraquat-induced pulmonary toxicity: Molecular mechanisms and potential therapeutic drugs. Int Immunopharmacol 2022; 113:109301. [DOI: 10.1016/j.intimp.2022.109301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/19/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022]
|
36
|
Wang B, Lin Y, Zhou M, Fu S, Zhu B, Chen Y, Ding Z, Zhou F. Polysaccharides from Tetrastigma Hemsleyanum Diels et Gilg attenuate LPS-induced acute lung injury by modulating TLR4/COX-2/NF-κB signaling pathway. Biomed Pharmacother 2022; 155:113755. [DOI: 10.1016/j.biopha.2022.113755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/02/2022] Open
|
37
|
Rayees S, Joshi JC, Joshi B, Vellingiri V, Banerjee S, Mehta D. Protease-activated receptor 2 promotes clearance of Pseudomonas aeruginosa infection by inducing cAMP-Rac1 signaling in alveolar macrophages. Front Pharmacol 2022; 13:874197. [PMID: 36204227 PMCID: PMC9530345 DOI: 10.3389/fphar.2022.874197] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
Efficient phagocytosis of pathogens by the innate immune system during infectious injury is vital for restoring tissue integrity. Impaired phagocytosis, such as in the case of infection with Pseudomonas aeruginosa, a broad-spectrum antibiotic-resistant Gram-negative bacterium, can lead to a life threatening lung disorder, acute lung injury (ALI). Evidence indicates that loss of protease-activated receptor 2 (PAR2) impaired Pseudomonas aeruginosa clearance leading to non-resolvable ALI, but the mechanism remains unclear. Here, we focused on the alveolar macrophages (AMs), the predominant population of lung-resident macrophages involved in sensing bacteria, to understand their role in PAR2-mediated phagocytosis of Pseudomonas aeruginosa. We found that upon binding Pseudomonas aeruginosa, PAR2-expressing but not PAR2-null AMs had increased cAMP levels, which activated Rac1 through protein kinase A. Activated Rac1 increased actin-rich protrusions to augment the phagocytosis of Pseudomonas aeruginosa. Administration of liposomes containing constitutively active Rac1 into PAR2-null mice lungs rescued phagocytosis and enhanced the survival of PAR2-null mice from pneumonia. These studies showed that PAR2 drives the cAMP-Rac1 signaling cascade that activates Pseudomonas aeruginosa phagocytosis in AMs, thereby preventing death from bacterial pneumonia.
Collapse
|
38
|
Yang M, Chen K, Hou C, Yang Y, Zhai X, Chen K, Wei X, Bai Y, Li M. RHOA inhibits chondrogenic differentiation of mesenchymal stem cells in adolescent idiopathic scoliosis. Connect Tissue Res 2022; 63:475-484. [PMID: 35019797 DOI: 10.1080/03008207.2021.2019247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE The etiology of adolescent idiopathic scoliosis (AIS) remains unclear. The chondrogenic differentiation of mesenchymal stem cells (MSCs) is important in AIS, and the Ras homolog gene family member A (RHOA) is associated with chondrogenesis. The purpose of this study was to explore the effect of RHOA on the chondrogenic differentiation of MSCs in AIS. METHODS We isolated MSCs from patients with AIS (AIS MSCs) and individuals without AIS (control MSCs). The inhibitor Y27632 was used to inhibit the function of RHOA/ROCK signaling, and plasmid-based overexpression and siRNA-mediated knockdown were used to manipulate RHOA expression. CCK-8 was used to detect cell viability. The phosphorylation levels of LIMK1, MLC2 and cofilin were detected by Western blotting. The mRNA expression of aggrecan, SOX9, and COL2A1 were confirmed using RT-PCR. Immunofluorescence was used to analyze F-actin and collagen II. Alcian blue staining was performed to assess the secretion of glycosaminoglycans (GAGs). RESULTS We found that RHOA was significantly upregulated in AIS MSCs, and the phosphorylation levels of LIMK1, MLC2, and cofilin were increased. The mRNA expressions of aggrecan, SOX9, and COL2A1 were notably reduced in AIS MSCs. However, these effects were abolished by Y27632 treatment and RHOA knockdown in AIS MSCs. In addition, RHOA knockdown in AIS MSCs increased the content of collagen II and GAGs. RHOA overexpression in the control MSCs markedly activated the RHOA/ROCK signaling and decreased the expression of aggrecan, SOX9, and COL2A1, F-actin, and GAGs. CONCLUSION RHOA regulates the chondrogenic differentiation ability of MSCs in AIS via the RHOA/ROCK signaling pathway and this regulation may involve SOX9.
Collapse
Affiliation(s)
- Mingyuan Yang
- Department of Orthopedics, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Kai Chen
- Department of Orthopedics, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Canglong Hou
- Department of Orthopedics, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Yilin Yang
- Department of Orthopedics, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Xiao Zhai
- Department of Orthopedics, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Kai Chen
- Department of Orthopedics, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Xianzhao Wei
- Department of Orthopedics, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Yushu Bai
- Department of Orthopedics, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| | - Ming Li
- Department of Orthopedics, Changhai Hospital, Navy Medical University, Shanghai, People's Republic of China
| |
Collapse
|
39
|
Lee CY, Chen SP, Huang-Liu R, Gau SY, Li YC, Chen CJ, Chen WY, Wu CN, Kuan YH. Fucoxanthin decreases lipopolysaccharide-induced acute lung injury through the inhibition of RhoA activation and the NF-κB pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:2214-2222. [PMID: 35616142 DOI: 10.1002/tox.23587] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/03/2022] [Accepted: 05/14/2022] [Indexed: 06/15/2023]
Abstract
Fucoxanthin is a natural pigment widely distributed in macroalgae and microalgae. An orange-colored xanthophyll, it has several bioactive effects, including anticancer, anti-obesity, oxidative stress reduction, and anti-inflammation. Acute lung injury (ALI) caused by acute infections or injurious stimuli to the lung tissues is a severe pulmonary inflammatory disease. To date, no evidence has shown ALI to be reduced by fucoxanthin through activation of Ras homolog family member A (RhoA) and the nuclear factor (NF)-κB pathway in lipopolysaccharide (LPS)-treated mice. Pretreatment with fucoxanthin inhibited histopathological changes in lung tissues and neutrophil infiltration into bronchoalveolar lavage fluid induced by LPS in ALI mice. Moreover, LPS-induced proinflammatory cytokine expression and neutrophil infiltration were inhibited by fucoxanthin in a concentration-dependent manner. Pretreatment of mice with fucoxanthin inhibited NF-κB phosphorylation and IκB degradation in the lungs of mice with LPS-induced ALI. We further found that phosphorylation of Akt and p38 mitogen-activated protein KINASE (MAPK) was inhibited by fucoxanthin. By contrast, the phosphorylation of extracellular signal-regulated kinase and c-Jun N-terminal kinase was not inhibited by fucoxanthin. Furthermore, we found that the activation of RhoA was inhibited by fucoxanthin in LPS-induced ALI. On the basis of these results, we propose that fucoxanthin disrupts the RhoA activation-mediated phosphorylation of Akt and p38 MAPK, leading to NF-κB activation in mice with LPS-induced ALI.
Collapse
Affiliation(s)
- Chien-Ying Lee
- Department of Pharmacology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shih-Pin Chen
- Department of Internal Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Rosa Huang-Liu
- School of Nutrition, Chung Shan Medical University, Taichung, Taiwan
| | - Shuo-Yan Gau
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Ching Li
- Department of Pharmacology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chun-Jung Chen
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chun-Nan Wu
- Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yu-Hsiang Kuan
- Department of Pharmacology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
40
|
Zeng M, Zhang B, Ren Y, Wang S, Guo P, Liu M, Zhang Q, Jia J, Li J, Zheng X, Feng W. A sesquiterpene isolated from the stems and leaves of Dioscorea opposita thunb. Transforms the composition of immune cells through ERβ in a mouse model of LPS-induced lung injury. Heliyon 2022; 8:e10500. [PMID: 36105471 PMCID: PMC9465438 DOI: 10.1016/j.heliyon.2022.e10500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/22/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
Abstract
Acute lung injury (ALI) is a common critical disease with a high mortality rate. Natural products have marked efficacy in the prevention and treatment of ALI, in addition, estrogen and its receptors are involved in the pathogenesis and development of lung injury. Our previous research shows that sesquiterpenes isolated from the stems and leaves of Dioscorea opposita Thunb. have anti-inflammatory and estrogenic-like activity. In the present study, sesquiterpene (A1) is a natural extract from the stems and leaves of Dioscorea opposita Thunb. with a view to determining whether A1 can improve lung function in a mouse model of LPS-induced ALI and exploring the involvement of the estrogen receptor β (ERβ) pathway. A1 (20 or 40 mg/kg, i. g., 2 times/day) was administered for 3 d, followed by the induction of ALI via an intratracheal LPS drip (5 mg/kg/2 h). The lung function and levels of inflammation, immune cells, apoptosis, and ERβ expression were examined. The antagonistic activity of specific ERβ blocker (THC, 1 μM) against A1 (20 μM) in co-cultured BEAS-2B cells and splenic lymphocytes induced with LPS (1 μg/mL, 24 h) was also investigated to assess whether the observed effects of A1 were mediated by ERβ. A1 improved lung function, regulated the immune system, and decreased inflammation and apoptosis. Moreover, A1 increased the expression of ERβ in LPS-induced mice, and antagonism of ERβ decreased the protective effects of A1 in a co-culture system. A1 had anti-ALI effects that might partially mediated through ERβ signaling. Our data provide molecular justification for the use of A1 in the treatment of ALI.
Collapse
Affiliation(s)
- Mengnan Zeng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Beibei Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Yingjie Ren
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Shengchao Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Pengli Guo
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Meng Liu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Qinqin Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Jufang Jia
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Jinyue Li
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, China
- Corresponding author.
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou, China
- Corresponding author.
| |
Collapse
|
41
|
Cheng H, Zhu Y, Chen L, Wang Y. Nesfatin-1 alleviated lipopolysaccharide-induced acute lung injury through regulating inflammatory response associated with macrophages modulation. J Cardiothorac Surg 2022; 17:206. [PMID: 36008865 PMCID: PMC9413923 DOI: 10.1186/s13019-022-01952-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/17/2022] [Indexed: 11/21/2022] Open
Abstract
Acute lung injury (ALI) is a continuum of lung changes associated with uncontrolled excessive lung inflammation. However, the pathogenesis of ALI is still complicated and effective clinical pharmacological management is required. Various signaling pathways are involved in the inflammatory responses of ALI. Here, we aimed to explore the role of nesfatin-1, an amino-acid peptide with anti-inflammatory action, in an LPS-induced ALI mice model, and its role in regulating macrophages in response to LPS stimulation in vitro. This was to clarify the underlying mechanisms of regulating the inflammatory response in the development of ALI. The results show that nesfatin-1 expression was downregulated in the lung tissues of ALI mice compared to control mice. Nesfatin-1 treatment ameliorated the inflammatory response and lung tissue damage in LPS-induced ALI in mice. In vitro studies showed that nesfatin-1 attenuated the generation and release of proinflammatory cytokines interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α) in LPS-induced RAW 264.7 cells. Nesfatin-1 also inhibited reactive oxygen species production and improved superoxide dismutase (SOD) activity in LPS-induced RAW 264.7 cells. These findings suggest that nesfatin-1 exerted a crucial role in regulating the LPS-mediated activation of M1 macrophages. Further mechanism investigations indicated that nesfatin-1 inhibited the activation of p38 MAPK/c-Jun and NF-κB pathways in LPS-induced RAW 264.7 cells, as evidenced by decreased expression levels of p-p38, p-c-Fos, and p-p65. Overall, nesfatin-1 alleviated LPS-induced ALI, which might be attributed to regulating inflammatory response through macrophages modulation.
Collapse
Affiliation(s)
- Hongbing Cheng
- Xiantao First People's Hospital Affiliated to Yangtze University, No. 29, Middle Section of Mianzhou Avenue, Nancheng New District, Xiantao City, 433099, Hubei Province, China
| | - Yanfang Zhu
- Xiantao First People's Hospital Affiliated to Yangtze University, No. 29, Middle Section of Mianzhou Avenue, Nancheng New District, Xiantao City, 433099, Hubei Province, China
| | - Liangji Chen
- Xiantao First People's Hospital Affiliated to Yangtze University, No. 29, Middle Section of Mianzhou Avenue, Nancheng New District, Xiantao City, 433099, Hubei Province, China
| | - Yalan Wang
- Xiantao First People's Hospital Affiliated to Yangtze University, No. 29, Middle Section of Mianzhou Avenue, Nancheng New District, Xiantao City, 433099, Hubei Province, China.
| |
Collapse
|
42
|
Wang JF, Shi CY, Ying HZ. Cephalosporins-induced intestinal dysbiosis exacerbated pulmonary endothelial barrier disruption in streptococcus pneumoniae-infected mice. Front Cell Infect Microbiol 2022; 12:997368. [PMID: 36093187 PMCID: PMC9449322 DOI: 10.3389/fcimb.2022.997368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/02/2022] [Indexed: 11/19/2022] Open
Abstract
Antibiotic abuse is growing more severe in clinic, and even short-term antibiotic treatment can cause long-term gut dysbiosis, which may promote the development and aggravation of diseases. Cephalosporins as the broad-spectrum antibiotics are widely used for prevention and treatment of community-acquired respiratory tract infection in children. However, their potential consequences in health and disease have not been fully elaborated. In this study, the effects of cefaclor, cefdinir and cefixime on intestinal microbiota and lung injury were investigated in Streptococcus pneumoniae (Spn)-infected mice. The results showed that the proportion of coccus and bacillus in intestinal microbiota were changed after oral administration with cefaclor, cefdinir and cefixime twice for 10 days, respectively. Compared with the Spn-infected group, the proportion of Bifidobacterium and Lactobacillus in intestine were significantly reduced, while Enterococcus and Candida was increased after cephalosporin treatment. Furthermore, 3 cephalosporins could obviously increase the number of total cells, neutrophils and lymphocytes in BALF as well as the serum levels of endotoxin, IL-2, IL-1β, IL-6 and TNF-α. Mechanically, cephalosporins accelerated Spn-induced pulmonary barrier dysfunction via mediating the mRNA expressions of endothelial barrier-related proteins (Claudin 5, Occludin, and ZO-1) and inflammation-related proteins (TLR4, p38 and NF-κB). However, all of those consequences could be partly reversed by Bifidobacterium bifidum treatment, which was closely related to the elevated acetate production, indicating the protective effects of probiotic against antibiotic-induced intestinal dysbiosis. Therefore, the present study demonstrated that oral administration with cephalosporins not only disrupted intestinal microecological homeostasis, but also increased the risk of Spn infection, resulting in severer respiratory inflammation and higher bacterial loads in mice.
Collapse
|
43
|
Differential expression profile of plasma exosomal microRNAs in acute type A aortic dissection with acute lung injury. Sci Rep 2022; 12:11667. [PMID: 35804020 PMCID: PMC9270349 DOI: 10.1038/s41598-022-15859-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/30/2022] [Indexed: 11/08/2022] Open
Abstract
MicroRNAs (miRNAs) packaged into exosomes mediate cell communication and contribute to the pathogenesis of acute type A aortic dissection (ATAAD) with acute lung injury (ALI). The expression profile of plasma exosomal miRNAs in ATAAD patients with ALI hasn’t been identified. We performed a miRNA-sequencing to analyze the differentially expressed miRNAs (DE-miRNAs) of circulating exosomes in ATAAD patients with ALI compared to patients without ALI, founding 283 specific miRNAs in two groups. We respectively selected the top 10 downregulated and upregulated DE-miRNAs for further studies. The predicted transcription factors (TFs) of these DE-miRNAs were SMAD2, SRSF1, USF1, etc. The Gene Ontology (GO) and Kyoto Encyclopedia Genes and Genomes (KEGG) analysis predicted their target genes mainly involved acute inflammatory response, cell junction, cytoskeleton, NF-κB signaling pathway, etc. Construction and analysis of the PPI network revealed that RHOA and INSR were considered hub genes with the highest connectivity degrees. Moreover, we confirmed two exosomal miRNAs (hsa-miR-485-5p and hsa-miR-206) by real-time quantitative polymerase chain reaction (RT-qPCR) in a validation cohort. Our study identified a plasma exosomal miRNAs signature related to ATAAD with ALI. Certain DE-miRNAs may contribute to the progression of this disease, which help us better understand the pathogenesis of ATAAD with ALI.
Collapse
|
44
|
Mechanistic and therapeutic perspectives of baicalin and baicalein on pulmonary hypertension: A comprehensive review. Biomed Pharmacother 2022; 151:113191. [PMID: 35643068 DOI: 10.1016/j.biopha.2022.113191] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 11/20/2022] Open
Abstract
Pulmonary hypertension (PH) is a chronic and fatal disease, for which new therapeutic drugs and approaches are needed urgently. Baicalein and baicalin, the active compounds of the traditional Chinese medicine, Scutellaria baicalensis Georgi, exhibit a wide range of pharmacological activities. Numerous studies involving in vitro and in vivo models of PH have revealed that the treatment with baicalin and baicalein may be effective. This review summarizes the potential mechanisms driving the beneficial effects of baicalin and baicalein treatment on PH, including anti-inflammatory response, inhibition of pulmonary smooth muscle cell proliferation and endothelial-to-mesenchymal transformation, stabilization of the extracellular matrix, and mitigation of oxidative stress. The pharmacokinetics of these compounds have also been reviewed. The therapeutic potential of baicalin and baicalein warrants their continued study as natural treatments for PH.
Collapse
|
45
|
Chen B, Gong S, Li M, Liu Y, Nie J, Zheng J, Zheng X, Li J, Gan Y, Su Z, Chen J, Li Y, Xie Q, Yan F. Protective effect of oxyberberine against acute lung injury in mice via inhibiting RhoA/ROCK signaling pathway. Biomed Pharmacother 2022; 153:113307. [PMID: 35753262 DOI: 10.1016/j.biopha.2022.113307] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
Acute lung injury (ALI), hallmarked with alveolar epithelial barrier impairment and pulmonary edema induced by acute inflammation, presents a severe health burden to the public, due to the limited available interventions. Oxyberberine (OBB), having improved anti-inflammatory activity and safety, is a representative component with various activities derived from berberine, whereas its role against ALI with alveolar epithelial barrier injury remains uncertain. To investigate the influence and underlying mechanisms of OBB on ALI, we induced acute inflammation in mice and A549 cells by using lipopolysaccharide (LPS). Changes in alveolar permeability were assessed by analyzing lung histopathology, measuring the dry/wet weight ratio of the lungs, and altering proinflammatory cytokines and neutrophils levels in the bronchoalveolar lavage fluid (BALF). Parameters of pulmonary permeability were assessed through ELISA, western blotting, quantitative real-time PCR, and immunofluorescence analysis. U46619, the agonist of RhoA/ROCK, was employed to further investigate the mechanism of OBB on ALI. Unexpectedly, we found OBB mitigated lung impairment, pulmonary edema, inflammatory reactions in BALF and lung tissue, reduction in ZO-1, and addition of connexin-43. Besides, OBB markedly reduced the expression of RhoA in association with its downstream factors, which are linked to the intercellular junctions and permeability both in vivo and in vitro. Nevertheless, U46619 abolished the benefits obtained from OBB in A549 cells. In conclusion, these outcomes indicated that OBB exerted RhoA/ROCK inhibitor-like effect to moderate alveolar epithelial barrier impairment and permeability, ultimately preventing ALI progression.
Collapse
Affiliation(s)
- Baoyi Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Shiting Gong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Minhua Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yanlu Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Juan Nie
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Medical School, Hubei Minzu University, Hubei 445000, China
| | - Jingna Zheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiaohong Zheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jincan Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yuxuan Gan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; South China Hospital of Shenzhen University, Shenzhen 518116, China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qingfeng Xie
- Li Ke and Qi Yu-ru Academic Experience Inheritance Studio, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510006, China; The Second Clinical College Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Fang Yan
- Li Ke and Qi Yu-ru Academic Experience Inheritance Studio, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510006, China; The Second Clinical College Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
46
|
Wang Z, Li F, Liu J, Luo Y, Guo H, Yang Q, Xu C, Ma S, Chen H. Intestinal Microbiota - An Unmissable Bridge to Severe Acute Pancreatitis-Associated Acute Lung Injury. Front Immunol 2022; 13:913178. [PMID: 35774796 PMCID: PMC9237221 DOI: 10.3389/fimmu.2022.913178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022] Open
Abstract
Severe acute pancreatitis (SAP), one of the most serious abdominal emergencies in general surgery, is characterized by acute and rapid onset as well as high mortality, which often leads to multiple organ failure (MOF). Acute lung injury (ALI), the earliest accompanied organ dysfunction, is the most common cause of death in patients following the SAP onset. The exact pathogenesis of ALI during SAP, however, remains unclear. In recent years, advances in the microbiota-gut-lung axis have led to a better understanding of SAP-associated lung injury (PALI). In addition, the bidirectional communications between intestinal microbes and the lung are becoming more apparent. This paper aims to review the mechanisms of an imbalanced intestinal microbiota contributing to the development of PALI, which is mediated by the disruption of physical, chemical, and immune barriers in the intestine, promotes bacterial translocation, and results in the activation of abnormal immune responses in severe pancreatitis. The pathogen-associated molecular patterns (PAMPs) mediated immunol mechanisms in the occurrence of PALI via binding with pattern recognition receptors (PRRs) through the microbiota-gut-lung axis are focused in this study. Moreover, the potential therapeutic strategies for alleviating PALI by regulating the composition or the function of the intestinal microbiota are discussed in this review. The aim of this study is to provide new ideas and therapeutic tools for PALI patients.
Collapse
Affiliation(s)
- Zhengjian Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fan Li
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jin Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yalan Luo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Haoya Guo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qi Yang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Caiming Xu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Shurong Ma
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- *Correspondence: Shurong Ma, ; Hailong Chen,
| | - Hailong Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- *Correspondence: Shurong Ma, ; Hailong Chen,
| |
Collapse
|
47
|
Tang D, Cao F, Yan C, Fang K, Ma J, Gao L, Sun B, Wang G. Extracellular Vesicle/Macrophage Axis: Potential Targets for Inflammatory Disease Intervention. Front Immunol 2022; 13:705472. [PMID: 35769456 PMCID: PMC9234271 DOI: 10.3389/fimmu.2022.705472] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Extracellular vesicles (EVs) can regulate the polarization of macrophages in a variety of inflammatory diseases by mediating intercellular signal transduction and affecting the occurrence and development of diseases. After macrophages are regulated by EVs, they mainly show two phenotypes: the proinflammatory M1 type and the anti-inflammatory M2 type. A large number of studies have shown that in diseases such as mastitis, inflammatory bowel disease, Acute lung injury, and idiopathic pulmonary fibrosis, EVs promote the progression of the disease by inducing the M1-like polarization of macrophages. In diseases such as liver injury, asthma, and myocardial infarction, EVs can induce M2-like polarization of macrophages, inhibit the inflammatory response, and reduce the severity of the disease, thus indicating new pathways for treating inflammatory diseases. The EV/macrophage axis has become a potential target for inflammatory disease pathogenesis and comprehensive treatment. This article reviews the structure and function of the EV/macrophage axis and summarizes its biological functions in inflammatory diseases to provide insights for the diagnosis and treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Desheng Tang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Feng Cao
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Changsheng Yan
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kun Fang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiamin Ma
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Gao
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Central Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Gang Wang,
| |
Collapse
|
48
|
Wang D, Cao Q. Shp2 in Alveolar Macrophages Regulates Macrophage I Phenotype in Acute Lung Injury. Int J Toxicol 2022; 41:412-419. [PMID: 35652623 DOI: 10.1177/10915818221105227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Macrophage play important role in acute lung injury (ALI). This study aims to explore the possible role of Shp2 in regulating macrophage 1 (M1) in ALI progression. ALI was induced in rats by intravenous injection of lipopolysacharide (LPS). Lentivirus was used to knock down Shp2 expression. Lungs from LPS-induced ALI rats were evaluated by H&E staining and wet/dry lung weight ratio (W/D ratio) measurement. The expression of inflammatory cytokines IL-1β, TNF-α and IL-6 in bronchoalveolar lavage fluid were detected by ELISA. The expressions of M1 biomarker (iNOS) and macrophage 2 (M2) biomarker (Arg-1) in lung tissues and macrophages were measured by immunofluorescence and western blot. The ratio of M2/M1 was detected by flow cytometry. Inflammatory cytokines were highly expressed in ALI rat models, in which elevated expression of iNOS and decreased Arg-1 expression were detected. Shp2 was found to be highly expressed in lung tissues of ALI rat models. LPS treatment in NR8383 cells lead to increased M1 phenotype and elevated expression of Shp2. Suppression on Shp2 expression can counteract the LPS-induced effect and further attenuate ALI progression. Evidence collected from ALI rat and cell models showed that suppression Shp2 expression in macrophages can inhibit M1 phenotype to attenuate ALI progression.
Collapse
Affiliation(s)
- Dawei Wang
- Department of General Medicine, 117902Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Qiumei Cao
- Department of Emergency, 117902Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
49
|
Wang W, Liu Y, Pan P, Huang Y, Chen T, Yuan T, Ma Y, Han G, Li J, Jin Y, Xie F. Pulmonary delivery of resveratrol- β-cyclodextrin inclusion complexes for the prevention of zinc chloride smoke-induced acute lung injury. Drug Deliv 2022; 29:1122-1131. [PMID: 35380089 PMCID: PMC8986301 DOI: 10.1080/10717544.2022.2048135] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Smoke bombs are often used in military/fire training, which can produce a large amount of zinc chloride (ZnCl2) smoke. Inhalation of ZnCl2 smoke usually causes acute lung injury (ALI) that would likely develop to acute respiratory distress syndrome (ARDS). However, there is no effective prevention or treatment strategy for the smoke-induced ALI. Resveratrol (RES) is a natural polyphenol with good anti-inflammatory and anti-apoptotic activities, but its low solubility, stability, and bioavailability restrict its clinical application. In this study, an inhalable RES formulation composed of RES-β-cyclodextrin inclusion complexes (RES-β-CD) was prepared for the prevention of ZnCl2 smoke-induced ALI. RES-β-CD powders had a small mass median aerodynamic diameter of 3.61 μm and a high fine particle fraction of 38.84%, suitable for pulmonary inhalation. RES-β-CD exhibited low BEAS-2B cytotoxicity. Pulmonary delivery of RES-β-CD to mice remarkably prevented the smoke-induced ALI with downregulation of TNF-α, IL-1β, STAT3, and GATA3, and upregulation of T-bet and Foxp3. RES-β-CD protected the respiratory function, percutaneous oxygen saturation, physical activity, lung capillary integrity, and lung liquid balance, alleviating inflammation and apoptosis. Pulmonary delivery of the positive drug, budesonide (BUD), also alleviated the smoke-induced ALI by reduction of inflammation and cell apoptosis. RES-β-CD exhibited the regulation of the Th1/Th2 and Treg/Th17 balances, while BUD did not show any effect on immune balances. In conclusion, pulmonary delivery of RES-β-CD is a promising anti-inflammatory and anti-apoptosis strategy for the prevention of ZnCl2 smoke-induced ALI by direct lung drug distribution and regulation of immune balance.
Collapse
Affiliation(s)
- Wanmei Wang
- Pharmaceutical College of Henan University, Kaifeng, China.,Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yan Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Pan Pan
- Respiratory Intensive Care Unit, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yueqi Huang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Ting Chen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Tianyu Yuan
- Pharmaceutical College of Henan University, Kaifeng, China.,Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yulong Ma
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Guang Han
- Pharmaceutical College of Henan University, Kaifeng, China
| | - Jiahuan Li
- Pharmaceutical College of Henan University, Kaifeng, China
| | - Yiguang Jin
- Pharmaceutical College of Henan University, Kaifeng, China.,Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Fei Xie
- Respiratory Intensive Care Unit, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
50
|
Zhang H, Zhu K, Zhang X, Ding Y, Zhu B, Meng W, Zhang F. Rutaecarpine ameliorates lipopolysaccharide‑induced BEAS‑2B cell injury through inhibition of endoplasmic reticulum stress via activation of the AMPK/SIRT1 signaling pathway. Exp Ther Med 2022; 23:373. [PMID: 35495603 PMCID: PMC9019775 DOI: 10.3892/etm.2022.11300] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/01/2022] [Indexed: 11/05/2022] Open
Abstract
Rutaecarpine (RUT) is an alkaloid isolated from Tetradium ruticarpum, which has been reported to protect against several inflammatory diseases. However, to the best of our knowledge, the role of RUT in acute lung injury (ALI) and the specific molecular mechanism remain unknown. In the present study, an in vitro model of ALI was established in BEAS-2B cells by lipopolysaccharide (LPS) administration. Cell viability following RUT treatment with or without LPS stimulation was evaluated using a Cell Counting Kit-8 assay. The inflammatory response and oxidative stress were detected using ELISA kits and commercially available kits, respectively. TUNEL assay and western blotting were performed to assess cell apoptosis. The expression levels of endoplasmic reticulum (ER) stress-related proteins and AMP-activated protein kinase (AMPK)/sirtuin 1 (SIRT1) signaling pathway-related proteins were measured by western blotting. The results revealed that RUT markedly improved cell viability after LPS treatment in a dose-dependent manner. In addition, RUT inhibited the LPS-induced inflammatory response and oxidative stress in BEAS-2B cells, and suppressed the LPS-induced apoptosis of BEAS-2B cells. Mechanistically, RUT alleviated ER stress by inhibiting the production of CHOP, glucose-regulated protein-78, caspase-12 and activating transcription factor 6. Additionally, western blotting demonstrated that RUT activated the phosphorylation of AMPK and SIRT1, which indicated the involvement of the AMPK/SIRT1 signaling pathway in the protective effect of RUT against LPS-induced lung injury. In conclusion, these results demonstrated that RUT mitigated LPS-induced lung cell injury by inhibiting ER stress via the activation of the AMPK/SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Hao Zhang
- Emergency Department, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Kun Zhu
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Qiqihar Medical College, Qiqihar, Heilongjiang 161000, P.R. China
| | - Xuefeng Zhang
- Department of Cardiothoracic Surgery, The Affiliated Heilongjiang Provincial Hospital of Harbin Institute of Technology, Harbin, Heilongjiang 150036, P.R. China
| | - Yihui Ding
- Department of Cardiothoracic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Bing Zhu
- Department of Cardiothoracic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Wen Meng
- Department of Cardiothoracic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Fan Zhang
- Department of Cardiothoracic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|