1
|
Hu CH, Qian R, Wang YB, Li LD, Miao CX. Dexmedetomidine Blocks the ERK Pathway by Inhibiting MAP3K8 to Achieve a Protective Effect in Lung Ischemia/Reperfusion Injury. Kaohsiung J Med Sci 2025:e70045. [PMID: 40372180 DOI: 10.1002/kjm2.70045] [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: 02/18/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025] Open
Abstract
Lung ischemia/reperfusion injury (LIRI) is a primary contributor to morbidity and mortality following lung transplantation. Dexmedetomidine (DEX) protects the lungs from I/R injury, but the underlying mechanisms remain uncertain. This paper examined the protective effect of DEX in LIRI and elucidated the potential regulation involved. LIRI was induced in mice, followed by the detection of pulmonary arterial pressure, lung compliance, pathological changes, pulmonary vascular permeability, oxidative stress, inflammation, and apoptosis. Mice were infected with overexpression (OE)-mitogen-activated protein kinase kinase kinase 8 (MAP3K8) adenovirus and treated with DEX. MAP3K8 expression was examined in mouse lung tissue and pulmonary microvascular endothelial cells (PMVECs). Cells were infected using OE-MAP3K8 lentivirus and treated with DEX, followed by detection of cell viability and apoptosis, VE-cadherin and α-E-catenin, and pro-inflammatory factors. Rescue experiments were performed by MAP3K8 overexpression and combined extracellular signal-regulated protein kinase (ERK) pathway blocker, PD98059. The results demonstrated that DEX protected mice from LIRI. DEX inhibited MAP3K8 expression. MAP3K8 overexpression increased ERK1/2 phosphorylation and activated the ERK pathway. Upregulation of MAP3K8 impaired the protective effect of DEX in vivo and in vitro, which was reversed by the ERK inhibitor PD98059. Overall, DEX achieved its protective effect against LIRI by inhibiting the MAP3K8-ERK axis.
Collapse
Affiliation(s)
- Chun-Huan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Ru Qian
- Department of Critical Medicine, The Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Yong-Bo Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Lian-Di Li
- Department of Anesthesiology, The Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| | - Chun-Xing Miao
- Department of Chest Surgery, The Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang, People's Republic of China
| |
Collapse
|
2
|
Zhang B, Li Z, Meng C, Zhang G, Kang J, Zhou H. Nrf2/HO-1 Pathway Mediated Protective Effects of Hydrogen in a Model of Lung Transplantation Simulated by Rat Pulmonary Microvascular Endothelial Cells. Cell Biochem Biophys 2025:10.1007/s12013-025-01671-z. [PMID: 39853631 DOI: 10.1007/s12013-025-01671-z] [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] [Accepted: 01/06/2025] [Indexed: 01/26/2025]
Abstract
This study aimed to observe the mechanism of hydrogen (H2) in a lung transplantation model simulated by pulmonary microvascular endothelial cells (PMVECs), which were divided into 5 groups. The blank group was the normal PMVECs. During cold ischemia period, PMVECs in the control, O2, or H2 groups were aerated with no gas, O2, or 3% H2, and 3% H2 after transfected with a small interfering RNA targeting Nrf2 in the H2+si-Nrf2 group. Treatment with O2 and H2 decreased the oxidative stress injury, inflammation, cell apoptosis, and attenuated energy metabolism compared with the control group (P < 0.05). And the H2 group showed a better outcome with the increased protein expression of the Nrf2 and HO-1, which were conversed in the H2+si-Nrf2 group. In conclusion, H2 attenuated inflammation, oxidative stress injury, cell apoptosis, and maintained the balance between energy supply and demand in a rat PMVECs lung transplantation model via Nrf2/HO-1.
Collapse
Affiliation(s)
- Bing Zhang
- Department of Pain, Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Zhe Li
- Department of Pain, Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Department of Anesthesiology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Chao Meng
- Department of Pain Management, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Guangchao Zhang
- Department of Pain, Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Jiyu Kang
- Department of Pain, Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Huacheng Zhou
- Department of Pain, Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
3
|
Ta HQ, Kuppusamy M, Sonkusare SK, Roeser ME, Laubach VE. The endothelium: gatekeeper to lung ischemia-reperfusion injury. Respir Res 2024; 25:172. [PMID: 38637760 PMCID: PMC11027545 DOI: 10.1186/s12931-024-02776-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
The success of lung transplantation is limited by the high rate of primary graft dysfunction due to ischemia-reperfusion injury (IRI). Lung IRI is characterized by a robust inflammatory response, lung dysfunction, endothelial barrier disruption, oxidative stress, vascular permeability, edema, and neutrophil infiltration. These events are dependent on the health of the endothelium, which is a primary target of IRI that results in pulmonary endothelial barrier dysfunction. Over the past 10 years, research has focused more on the endothelium, which is beginning to unravel the multi-factorial pathogenesis and immunologic mechanisms underlying IRI. Many important proteins, receptors, and signaling pathways that are involved in the pathogenesis of endothelial dysfunction after IR are starting to be identified and targeted as prospective therapies for lung IRI. In this review, we highlight the more significant mediators of IRI-induced endothelial dysfunction discovered over the past decade including the extracellular glycocalyx, endothelial ion channels, purinergic receptors, kinases, and integrins. While there are no definitive clinical therapies currently available to prevent lung IRI, we will discuss potential clinical strategies for targeting the endothelium for the treatment or prevention of IRI. The accruing evidence on the essential role the endothelium plays in lung IRI suggests that promising endothelial-directed treatments may be approaching the clinic soon. The application of therapies targeting the pulmonary endothelium may help to halt this rapid and potentially fatal injury.
Collapse
Affiliation(s)
- Huy Q Ta
- Department of Surgery, University of Virginia, P. O. Box 801359, Charlottesville, VA, 22908, USA
| | - Maniselvan Kuppusamy
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - Swapnil K Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Mark E Roeser
- Department of Surgery, University of Virginia, P. O. Box 801359, Charlottesville, VA, 22908, USA
| | - Victor E Laubach
- Department of Surgery, University of Virginia, P. O. Box 801359, Charlottesville, VA, 22908, USA.
| |
Collapse
|
4
|
Bojic D, Aujla T, Sugihara J, Wong A, Keshavjee S, Liu M. Thyroid hormone protects human lung epithelial cells from cold preservation and warm reperfusion-induced injury. J Transl Med 2024; 22:221. [PMID: 38429788 PMCID: PMC10908176 DOI: 10.1186/s12967-024-05024-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/23/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Cellular stress associated with static-cold storage (SCS) and warm reperfusion of donor lungs can contribute to ischemia-reperfusion (IR) injury during transplantation. Adding cytoprotective agents to the preservation solution may be conducive to reducing graft deterioration and improving post-transplant outcomes. METHODS SCS and warm reperfusion were simulated in human lung epithelial cells (BEAS-2B) by exposing cells to low potassium dextran glucose solution at 4 °C for different periods and then switching back to serum-containing culture medium at 37 °C. Transcriptomic analysis was used to explore potential cytoprotective agents. Based on its results, cell viability, caspase activity, cell morphology, mitochondrial function, and inflammatory gene expression were examined under simulated IR conditions with or without thyroid hormones (THs). RESULTS After 18 h SCS followed by 2 h warm reperfusion, genes related to inflammation and cell death were upregulated, and genes related to protein synthesis and metabolism were downregulated in BEAS-2B cells, which closely mirrored gene profiles found in thyroid glands of mice with congenital hypothyroidism. The addition of THs (T3 or T4) to the preservation solution increases cell viability, inhibits activation of caspase 3, 8 and 9, preserves cell morphology, enhances mitochondrial membrane potential, reduces mitochondrial superoxide production, and suppresses inflammatory gene expression. CONCLUSION Adding THs to lung preservation solutions may protect lung cells during SCS by promoting mitochondrial function, reducing apoptosis, and inhibiting pro-inflammatory pathways. Further in vivo testing is warranted to determine the potential clinical application of adding THs as therapeutics in lung preservation solutions.
Collapse
Affiliation(s)
- Dejan Bojic
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tanroop Aujla
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Junichi Sugihara
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Aaron Wong
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
5
|
Lu Z, Xu S, Liao H, Zhang Y, Lu Z, Li Z, Chen Y, Guo F, Tang F, He Z. Identification of signature genes for renal ischemia‒reperfusion injury based on machine learning and WGCNA. Heliyon 2023; 9:e21151. [PMID: 37928383 PMCID: PMC10622618 DOI: 10.1016/j.heliyon.2023.e21151] [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: 05/27/2023] [Revised: 09/04/2023] [Accepted: 10/17/2023] [Indexed: 11/07/2023] Open
Abstract
Background As an inevitable event after kidney transplantation, ischemia‒reperfusion injury (IRI) can lead to a decrease in kidney transplant success. The search for signature genes of renal ischemia‒reperfusion injury (RIRI) is helpful in improving the diagnosis and guiding clinical treatment. Methods We first downloaded 3 datasets from the GEO database. Then, differentially expressed genes (DEGs) were identified and applied for functional enrichment analysis. After that, we performed three machine learning methods, including random forest (RF), Lasso regression analysis, and support vector machine recursive feature elimination (SVM-RFE), to further predict candidate genes. WGCNA was also executed to screen candidate genes from DEGs. Then, we took the intersection of candidate genes to obtain the signature genes of RIRI. Receiver operating characteristic (ROC) analysis was conducted to measure the predictive ability of the signature genes. Kaplan‒Meier analysis was used for association analysis between signature genes and graft survival. Verifying the expression of signature genes in the ischemia cell model. Results A total of 117 DEGs were screened out. Subsequently, RF, Lasso regression analysis, SVM-RFE and WGCNA identified 17, 25, 18 and 74 candidate genes, respectively. Finally, 3 signature genes (DUSP1, FOS, JUN) were screened out through the intersection of candidate genes. ROC analysis suggested that the 3 signature genes could well diagnose and predict RIRI. Kaplan‒Meier analysis indicated that patients with low FOS or JUN expression had a longer OS than those with high FOS or JUN expression. Finally, we validated using the ischemia cell model that compared to the control group, the expression level of JUN increased under hypoxic conditions. Conclusions Three signature genes (DUSP1, FOS, JUN) offer a good prediction for RIRI outcome and may serve as potential therapeutic targets for RIRI intervention, especially JUN. The prediction of graft survival by FOS and JUN may improve graft survival in patients with RIRI.
Collapse
Affiliation(s)
- Zechao Lu
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Senkai Xu
- The Sixth Clinical College of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Haiqin Liao
- The Second Clinical College of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Yixin Zhang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, China
| | - Zeguang Lu
- The Second Clinical College of Guangzhou Medical University, Guangzhou, Guangdong, 511436, China
| | - Zhibiao Li
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Yushu Chen
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Feng Guo
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Fucai Tang
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Zhaohui He
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| |
Collapse
|
6
|
Mechanisms of pulmonary microvascular endothelial cells barrier dysfunction induced by LPS: The roles of ceramides and the Txnip/NLRP3 inflammasome. Microvasc Res 2023; 147:104491. [PMID: 36709858 DOI: 10.1016/j.mvr.2023.104491] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/12/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) are characterized by pulmonary microvascular endothelial cells (PMVECs) barrier dysfunction and proinflammatory cytokine influx into lung tissue, resulting in pulmonary oedema. Ceramide overproduction is an important mediator of pulmonary hyperinflammation and pulmonary oedema in Acute lung injury (ALI). Ceramides induce NLRP3 inflammasome activation are essential for the hyperinflammatory response. However, the roles and specific mechanisms of ceramide-induced NLRP3 inflammasome activation, proinflammatory cytokine manufacturing and PMVECs barrier dysfunction in ALI are unclear. Herein, pretreatment with the acid sphingomyelinase (ASMase) inhibitor imipramine (but not a neutral sphingomyelinase (NSMase) inhibitor or de novo pathway inhibitor) significantly inhibited ceramide early production in rats with lipopolysaccharide (LPS)-induced ALI; Furthermore, the Txnip/NLRP3 inflammasome activation, proinflammatory cytokine release, increased PMVECs permeability and lung injury were significantly decreased. Verapamil, a Txnip inhibitor, substantially inhibited Txnip/NLRP3 inflammasome activation, proinflammatory cytokine release, increased PMVECs permeability and lung injury in rats with C8-ceramide-induced ALI. In vitro, short hairpin RNA-mediated Txnip silencing significantly inhibited C8-ceramide-induced Txnip/NLRP3 inflammasome activation in NR8383 alveolar macrophages (AMs) and early secretion of the proinflammatory cytokines IL-1β (4-12 h) as well as IL-6 and TNF-α at subsequent times (later than 12 h). However, C8-ceramide significantly increased the early secretion (within 8 h) of the proinflammatory cytokines IL-1β, IL-6 and TNF-α in a co-culture model of NR8383 AMs and PMVECs, and Txnip silencing of NR8383 AMs inhibited the secretion of pro-inflammatory cytokines and reduced cytoskeletal rearrangements, intercellular connection breakage and hyperpermeability in PMVECs. Overall, our results suggest that in LPS-induced ALI, ceramide-mediated Txnip/NLRP3 inflammasome activation in NR8383 AMs leads to early IL-1β release, subsequently inducing PMVECs injury and release of the proinflammatory cytokines IL-6 and TNF-α, ultimately leading to PMVECs barrier dysfunction and ALI.
Collapse
|
7
|
Keskinidou C, Vassiliou AG, Dimopoulou I, Kotanidou A, Orfanos SE. Mechanistic Understanding of Lung Inflammation: Recent Advances and Emerging Techniques. J Inflamm Res 2022; 15:3501-3546. [PMID: 35734098 PMCID: PMC9207257 DOI: 10.2147/jir.s282695] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening lung injury characterized by an acute inflammatory response in the lung parenchyma. Hence, it is considered as the most appropriate clinical syndrome to study pathogenic mechanisms of lung inflammation. ARDS is associated with increased morbidity and mortality in the intensive care unit (ICU), while no effective pharmacological treatment exists. It is very important therefore to fully characterize the underlying pathobiology and the related mechanisms, in order to develop novel therapeutic approaches. In vivo and in vitro models are important pre-clinical tools in biological and medical research in the mechanistic and pathological understanding of the majority of diseases. In this review, we will present data from selected experimental models of lung injury/acute lung inflammation, which have been based on clinical disorders that can lead to the development of ARDS and related inflammatory lung processes in humans, including ventilation-induced lung injury (VILI), sepsis, ischemia/reperfusion, smoke, acid aspiration, radiation, transfusion-related acute lung injury (TRALI), influenza, Streptococcus (S.) pneumoniae and coronaviruses infection. Data from the corresponding clinical conditions will also be presented. The mechanisms related to lung inflammation that will be covered are oxidative stress, neutrophil extracellular traps, mitogen-activated protein kinase (MAPK) pathways, surfactant, and water and ion channels. Finally, we will present a brief overview of emerging techniques in the field of omics research that have been applied to ARDS research, encompassing genomics, transcriptomics, proteomics, and metabolomics, which may recognize factors to help stratify ICU patients at risk, predict their prognosis, and possibly, serve as more specific therapeutic targets.
Collapse
Affiliation(s)
- Chrysi Keskinidou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Alice G Vassiliou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Ioanna Dimopoulou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Anastasia Kotanidou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Stylianos E Orfanos
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| |
Collapse
|
8
|
Han J, Liu X, Wang L. Dexmedetomidine protects against acute lung injury in mice via the DUSP1/MAPK/NF-κB axis by inhibiting miR-152-3p. Pulm Pharmacol Ther 2022:102131. [PMID: 35551994 DOI: 10.1016/j.pupt.2022.102131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 04/07/2022] [Accepted: 05/01/2022] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Acute lung injury (ALI) is a debilitating condition in clinics. Dexmedetomidine (Dex) is known for its anti-apoptotic and anti-inflammatory properties. This study attempted to investigate the protective mechanism of Dex in ALI mice. METHODS Mice were pretreated with Dex before model establishment by tracheal injection of lipopolysaccharide (LPS). Pulmonary function indexes and wet-to-dry (W/D) ratio were measured. Pulmonary pathological changes were observed through HE staining, CD31+-positive mouse pulmonary microvascular endothelial cells (MPMVECs) were counted through immunofluorescence staining, and apoptosis was detected through TUNEL staining. miR-152-3p mimic, sh-DUSP1, or p38 MAPK inhibitor was delivered into MPMVECs, followed by combined treatment of Dex and LPS. miR-152-3p expression, apoptosis, levels of apoptosis- and MAPK/NF-κB pathway-associated proteins, and inflammatory factors were measured through RT-qPCR, flow cytometry, Western blot, and ELISA. The binding relationship of miR-152-3p and DUSP1 was verified through bioinformatics software and dual-luciferase assay. ALI mouse model was established after injection of miR-152-3p antagomir. RESULTS Dex improved ALI mouse pulmonary function and mitigated injury in mice and MPMVECs. miR-125-3p overexpression or sh-DUSP1 partially abolished the protection of Dex on MPMVECs. miR-152-3p targeted DUSP1. sh-DUSP1 partially averted the protection of Dex on MPMVECs. Dex inhibited the activation of the MAPK/NF-κB pathway in MPMVECs mediated by LPS, which was partially reversed by sh-DUSP1. The p38 MAPK inhibitor SB203580 antagonized the protective effect of Dex on MPMVECs mediated by sh-DUSP1. Similarly, downregulation of miR-152-3p mitigated ALI via the DUSP1/MAPK/NF-κB axis in vivo. CONCLUSION Dex relieved ALI in mice via the DUSP1/MAPK/NF-κB axis by down-regulating miR-152-3p.
Collapse
Affiliation(s)
- Jieran Han
- Department of Anesthesiology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Xiao Liu
- Department of Anesthesiology, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, 471003, China
| | - Ling Wang
- Department of Anesthesiology, 989 Hospital of JOINT Logistic Support Force of PLA, Luoyang, 471031, China.
| |
Collapse
|
9
|
Knockdown of circRNA-Memo1 Reduces Hypoxia/Reoxygenation Injury in Human Brain Endothelial Cells Through miRNA-17-5p/SOS1 Axis. Mol Neurobiol 2022; 59:2085-2097. [PMID: 35041140 DOI: 10.1007/s12035-022-02743-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/09/2022] [Indexed: 12/23/2022]
Abstract
Circ-Memo1 has been proved to be upregulated in ischemia-reperfusion induced acute injury of kidney tissues. However, the potential role of circ-Memo1 in cerebral hypoxia/reoxygenation (H/R) injury is still unclear.Blood samples were collected from 25 ischemic stroke patients and 25 healthy controls. To construct the H/R model, human brain microvascular endothelial cells (HBMVECs) were cultured under the hypoxic condition, followed by reoxygenation. Cell viability was analyzed by MTT assay. Flow cytometry was carried out to examine cell apoptosis. The level of malondialdehyde (MDA) and the activity of superoxide dismutase (SOD) were measured by MDA and SOD assay kits, respectively. The levels of TNF-α, IL-1β, and IL-6 were determined by enzyme-linked immunosorbent assay (ELISA). Dual-luciferase reporter gene detection was employed to verify the binding relationships between circ-Memo1, miR-17-5p, and SOS1.Circ-Memo1 and SOS1 expressions were increased, and miR-17-5p expression was reduced in ischemic stroke patients. Circ-Memo1 silencing promoted cell viability, inhibited the activation of ERK/NF-κB signaling pathway, reduced oxidative stress and inflammatory response, and inhibited cell apoptosis. Moreover, miR-17-5p functioned as the sponge of circ-Memo1, and SOS1 was identified as the target of miR-17-5p. The protective effect of circ-Memo1 knockdown on cell injury after H/R treatment was weakened by miR-17-5p inhibition.Knockdown of circ-Memo1 alleviated H/R injury of HBMVEC cells by regulating the miR-17-5p/SOS1 axis, indicating that circ-Memo1 might be a potential treatment target for cerebral H/R injury.
Collapse
|
10
|
Jiang T, Liu T, Deng X, Ding W, Yue Z, Yang W, Lv X, Li W. Adiponectin ameliorates lung ischemia-reperfusion injury through SIRT1-PINK1 signaling-mediated mitophagy in type 2 diabetic rats. Respir Res 2021; 22:258. [PMID: 34602075 PMCID: PMC8489101 DOI: 10.1186/s12931-021-01855-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/29/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is a key contributing factor to poor survival in lung transplantation recipients. Mitochondrial dysfunction is recognized as a critical mediator in the pathogenesis of diabetic lung ischemia-reperfusion (IR) injury. The protective effects of adiponectin have been demonstrated in our previous study, but the underlying mechanism remains unclear. Here we demonstrated an important role of mitophagy in the protective effect of adiponectin during diabetic lung IR injury. METHODS High-fat diet-fed streptozotocin-induced type 2 diabetic rats were exposed to adiponectin with or without administration of the SIRT1 inhibitor EX527 following lung transplantation. To determine the mechanisms underlying the action of adiponectin, rat pulmonary microvascular endothelial cells were transfected with SIRT1 small-interfering RNA or PINK1 small-interfering RNA and then subjected to in vitro diabetic lung IR injury. RESULTS Mitophagy was impaired in diabetic lungs subjected to IR injury, which was accompanied by increased oxidative stress, inflammation, apoptosis, and mitochondrial dysfunction. Adiponectin induced mitophagy and attenuated subsequent diabetic lung IR injury by improving lung functional recovery, suppressing oxidative damage, diminishing inflammation, decreasing cell apoptosis, and preserving mitochondrial function. However, either administration of 3-methyladenine (3-MA), an autophagy antagonist or knockdown of PINK1 reduced the protective action of adiponectin. Furthermore, we demonstrated that APN affected PINK1 stabilization via the SIRT1 signaling pathway, and knockdown of SIRT1 suppressed PINK1 expression and compromised the protective effect of adiponectin. CONCLUSION These data demonstrated that adiponectin attenuated reperfusion-induced oxidative stress, inflammation, apoptosis and mitochondrial dysfunction via activation of SIRT1- PINK1 signaling-mediated mitophagy in diabetic lung IR injury.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Anesthesiology (Hei Long Jiang Province Key Lab of Research On Anesthesiology and Critical Care Medicine), The Second Affiliated Hospital, Harbin Medical University, No.194, XueFu Road, NanGang District, Harbin, China
| | - Tianhua Liu
- Department of Anesthesiology (Hei Long Jiang Province Key Lab of Research On Anesthesiology and Critical Care Medicine), The Second Affiliated Hospital, Harbin Medical University, No.194, XueFu Road, NanGang District, Harbin, China
| | - Xijin Deng
- Department of Anesthesiology (Hei Long Jiang Province Key Lab of Research On Anesthesiology and Critical Care Medicine), The Second Affiliated Hospital, Harbin Medical University, No.194, XueFu Road, NanGang District, Harbin, China
| | - Wengang Ding
- Department of Anesthesiology (Hei Long Jiang Province Key Lab of Research On Anesthesiology and Critical Care Medicine), The Second Affiliated Hospital, Harbin Medical University, No.194, XueFu Road, NanGang District, Harbin, China
| | - Ziyong Yue
- Department of Anesthesiology (Hei Long Jiang Province Key Lab of Research On Anesthesiology and Critical Care Medicine), The Second Affiliated Hospital, Harbin Medical University, No.194, XueFu Road, NanGang District, Harbin, China
| | - Wanchao Yang
- Department of Anesthesiology (Hei Long Jiang Province Key Lab of Research On Anesthesiology and Critical Care Medicine), The Second Affiliated Hospital, Harbin Medical University, No.194, XueFu Road, NanGang District, Harbin, China
| | - Xiangqi Lv
- Department of Anesthesiology (Hei Long Jiang Province Key Lab of Research On Anesthesiology and Critical Care Medicine), The Second Affiliated Hospital, Harbin Medical University, No.194, XueFu Road, NanGang District, Harbin, China
| | - Wenzhi Li
- Department of Anesthesiology (Hei Long Jiang Province Key Lab of Research On Anesthesiology and Critical Care Medicine), The Second Affiliated Hospital, Harbin Medical University, No.194, XueFu Road, NanGang District, Harbin, China.
| |
Collapse
|
11
|
Al-Hassan S, Attia H, Alomar H, Arafa M, Ali RA. The inhibitory mechanisms of losartan and vitamin D on amiodarone-induced lung inflammation in rats: Role of mitogen-activated protein kinases/activator protein-1. J Biochem Mol Toxicol 2021; 35:e22923. [PMID: 34590760 DOI: 10.1002/jbt.22923] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 08/22/2021] [Accepted: 09/20/2021] [Indexed: 11/09/2022]
Abstract
Amiodarone (AMD), an antiarrhythmic drug, is used cautiously due to its lung toxicity that is characterized by alveolar inflammation followed by fatal fibrosis. AMD induces lung inflammation via increasing the alveolar macrophages and disturbing the balance of T-helper-1 (Th1) and Th2 cells cytokines. In this study, the role of the mitogen-activated protein kinases (MAPKs)/activator protein-1 (AP-1) pathway in AMD-induced lung inflammation was evaluated. Also, the anti-inflammatory and antifibrotic effects of losartan and/or vitamin D were investigated following 7, 14, and 28 days of AMD administration. AMD resulted in lung injury, inflammatory infiltration, and increased pulmonary levels of inflammatory cytokines starting from Week 1 of exposure. A significant increase in serum levels of interleukin-4 along with a significant reduction of interferon-gamma, in addition to strong expression of CD68, were reported after 14 and 28 days of AMD administration reflecting Th1/Th2 cytokines imbalance and the accumulation of alveolar macrophages, respectively. The phosphorylation of MAPKs (ERK1/2, JNK, p38) and AP-1 was significantly enhanced starting from Week 1 of exposure. Marked expression of transforming growth factor beta-1 and massive deposition of collagen were detected after 28 days reflecting late fibrosis. All these abnormalities were significantly mitigated by vitamin D and its combination with losartan. Losartan alone has less prominent anti-inflammatory effects particularly after 28 days; however, it efficiently prevented late fibrosis. This study concludes that MAPKs/AP-1 pathway is involved in AMD-induced lung inflammation and that vitamin D and/or losartan could be used as a prophylactic agent to prevent AMD-induced lung toxicity.
Collapse
Affiliation(s)
- Sara Al-Hassan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Department of Pricing and Pharmacoeconomics, Drug Sector, Saudi Food and Drug Authority, Riyadh, Saudi Arabia
| | - Hala Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Department of Biochemistry, College of Pharmacy, Mansours University, Mansoura, Egypt
| | - Hatun Alomar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Maha Arafa
- Department of Pathology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Rehab A Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
12
|
Wang T, Liu C, Pan LH, Liu Z, Li CL, Lin JY, He Y, Xiao JY, Wu S, Qin Y, Li Z, Lin F. Inhibition of p38 MAPK Mitigates Lung Ischemia Reperfusion Injury by Reducing Blood-Air Barrier Hyperpermeability. Front Pharmacol 2020; 11:569251. [PMID: 33362540 PMCID: PMC7759682 DOI: 10.3389/fphar.2020.569251] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/29/2020] [Indexed: 01/18/2023] Open
Abstract
Background: Lung ischemia reperfusion injury (LIRI) is a complex pathophysiological process activated by lung transplantation and acute lung injury. The p38 mitogen-activated protein kinase (MAPK) is involved in breakdown of the endothelial barrier during LIRI, but the mechanism is still unclear. Therefore, we investigated the function of p38 MAPK in LIRI in vivo and in vitro. Methods: Sprague–Dawley rats were subjected to ischemia reperfusion with or without pretreatment with a p38 MAPK inhibitor. Lung injury was assessed using hematoxylin and eosin staining, and pulmonary blood–air barrier permeability was evaluated using Evans blue staining. A rat pulmonary microvascular endothelial cell line was infected with lentiviral expressing short hairpin (sh)RNA targeting p38 MAPK and then cells were subjected to oxygen/glucose deprivation and reoxygenation (OGD/R). Markers of endothelial destruction were measured by western blot and immunofluorescence. Results:In vivo LIRI models showed structural changes indicative of lung injury and hyperpermeability of the blood–air barrier. Inhibiting p38 MAPK mitigated these effects. Oxygen/glucose deprivation and reoxygenation promoted hyperpermeability of the endothelial barrier in vitro, but knockdown of p38 MAPK attenuated cell injury; maintained endothelial barrier integrity; and partially reversed injury-induced downregulation of permeability protein AQP1, endothelial protective protein eNOS, and junction proteins ZO-1 and VE-cadherin while downregulating ICAM-1, a protein involved in destroying the endothelial barrier, and ET-1, a protein involved in endothelial dysfunction. Conclusion: Inhibition of p38 MAPK alleviates LIRI by decreasing blood–air hyperpermeability. Blocking p38 MAPK may be an effective treatment against acute lung injury.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Chunxia Liu
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ling-Hui Pan
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Zhen Liu
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Chang-Long Li
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jin-Yuan Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yi He
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jing-Yuan Xiao
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Siyi Wu
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yi Qin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Zhao Li
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fei Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
13
|
Ozeki N, Chen-Yoshikawa TF. Commentary: Constant dripping wears a stone away. J Thorac Cardiovasc Surg 2020; 162:e159-e160. [PMID: 32690414 DOI: 10.1016/j.jtcvs.2020.04.136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 11/18/2022]
Affiliation(s)
- Naoki Ozeki
- Department of Thoracic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | |
Collapse
|
14
|
Blum K, Downs BW, Bagchi M, Kushner S, Morrison BS, Galvin J, Randsdorp K, Randsdorp J, Badgaiyan RD, Braverman ER, Bagchi D. Induction of homeostatic biological parameters in reward deficiency as a function of an iron-free multi-nutrient complex: Promoting hemoglobinization, aerobic metabolism, viral immuno-competence, and neuroinflammatory regulation. JOURNAL OF SYSTEMS AND INTEGRATIVE NEUROSCIENCE 2020; 7:10.15761/JSIN.1000234. [PMID: 35096420 PMCID: PMC8793786 DOI: 10.15761/jsin.1000234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND A common neurological condition worldwide is Reward Deficiency Syndrome (RDS) leading to both substance and non-substance addictive behaviors, that must be combatted by integrating both central nervous system and peripheral nervous system biological approaches. Integrity of hemoglobin is a crucial determining factor for the overall health functions. Nutrient repletion therapy should be a fundamental strategy to restore the healthy properties of blood. A unique patent-pending iron-free VMP35 formulation was engineered by our laboratory to restore iron-dependent hemoglobin in anemic cells using a proprietary Prodosome® absorption technology. This formulation, containing an array of nano-emulsified botanical ingredients rich in bioflavonoids, strengthens the structural integrity of connective tissues, and potentiates immune competence, cellular aerobic metabolism, and enhances efficient regulation of inflammatory events. We discuss the intricate aspects of strong vs. fragile immunity and consequential inflammatory responses to convey a deeper understanding of the varied and overly complex sequela of immunological behaviors and events. The effect of the VMP35 is mediated through highly absorbable nutritional/nutrigenomic repletion enabling improvements in the systemic set of functional behaviors. In fact, the iron-free VMP35 facilitates a "Systems Biology Approach" which restores hemoglobin status, reverses anaerobic hypoxia, improves competent immune responsivity, and regulates appropriate and controlled activation of general and neuro-inflammatory sequela. Under these pathogenic circumstances, iron-deficiency anemia has been misconceptualized, and a new nosological term, Chronic Anemia Syndrome, is proposed. The comparative therapeutic rationale of Reductionist vs. Systems Biology approaches is also explained in detail. METHODS The efficacy of the novel therapeutic iron-free VMP35 liquid nutraceutical is detailed in restoring iron-dependent hemoglobin to RBCs and boosting cellular morphology, viability, and immune competence, thereby reducing the need for prolonging inflammatory sequela. RESULTS This was demonstrated in a previous IRB approved multi-subject human study. In addition, two recent case studies report dramatic restorative benefits of nutrient repletion therapy of the VMP35 on subjects having experienced near-fatal events, which confirmed the findings explained in this manuscript. CONCLUSIONS This novel iron-free VMP35 modulates an array of homeostatic biological parameters such as enhanced hemoglobinization, aerobic metabolism, viral immuno-competence, and inflammatory regulation. Further research, examining mechanistic and beneficial effects in athletic performance, is in progress. Importantly, during these troubled immune challenging times, modulating an array of homeostatic immunological and inflammatory dysfunctions are tantamount to improved population outcomes. TRIAL REGISTRATION The Clinical investigation in a total of 38 subjects was conducted under an Institutional Review Board (IRB) from the Path Foundation in New York, NY (#13-009 April 25, 2013). The two case studies were done at Lancaster General Hospital, Lancaster, PA, and Jefferson University Hospital, Philadelphia, PA, USA. Both studies were retrospectively registered.
Collapse
Affiliation(s)
- Kenneth Blum
- Graduate College, Western University, Health Sciences, Pomona, CA, USA
| | - Bernard W Downs
- Victory Nutrition International, Inc., Department of R&D, Lederach, PA USA
| | | | | | | | - Jeffrey Galvin
- Vitality Medical Wellness Institute, PLLC, Charlotte, NC USA
| | | | | | - Rajendra D Badgaiyan
- Department of Psychiatry, ICHAN School of Medicine, Mount Sinai, New York, NY, USA
- Department of Psychiatry, South Texas Veteran Health Care System, Audie L. Murphy Memorial VA Hospital, San Antonio, TX, USA
- Long School of Medicine, University of Texas Medical Center, San Antonio, TX, USA
| | | | - Debasis Bagchi
- Victory Nutrition International, Inc., Department of R&D, Lederach, PA USA
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston college of Pharmacy, Houston, TX, USA
| |
Collapse
|
15
|
D'Cunha J. Commentary: A tale of two isoforms in lung ischemia reperfusion injury: One is bad, two is good. J Thorac Cardiovasc Surg 2020; 162:e158-e159. [PMID: 32561194 DOI: 10.1016/j.jtcvs.2020.04.145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 04/26/2020] [Accepted: 04/29/2020] [Indexed: 11/26/2022]
Affiliation(s)
- Jonathan D'Cunha
- Department of Cardiothoracic Surgery, Mayo Clinic Arizona, Phoenix, Ariz.
| |
Collapse
|
16
|
Tan J, Gao W, Yang W, Zeng X, Wang L, Cui X. Isoform-specific functions of c-Jun N-terminal kinase 1 and 2 in lung ischemia-reperfusion injury through the c-Jun/activator protein-1 pathway. J Thorac Cardiovasc Surg 2020; 162:e143-e156. [PMID: 32414595 DOI: 10.1016/j.jtcvs.2020.03.083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND c-Jun N-terminal kinase 1 (JNK1) and JNK2 regulate distinct pathological processes in lung diseases. Here we discriminated the respective roles of these kinases in lung transplantation-induced ischemia-reperfusion injury (IRI). METHODS Rat pulmonary microvascular endothelial cells were transfected with JNK1 small-interfering RNA (siRNA) and JNK2 siRNA and then subjected to in vitro IRI. For the isoform confirmed to aggravate IRI, the delivery of short-hairpin RNA (shRNA) plasmid was performed by intratracheal administration 48 hours before transplantation into donor rats. After a 3-hour reperfusion, the samples were collected. RESULTS JNK1 siRNA decreased but JNK2 siRNA increased JNK phosphorylation and activity, phosphorylated and total c-Jun, and activator protein-1 activity. Although JNK1 siRNA decreased apoptosis and the levels of malondialdehyde, interleukin (IL)-1, IL-6, and tumor necrosis factor (TNF-α), it increased the levels of superoxide dismutase, S-phase percentage, and cyclin D1; JNK2 siRNA had a converse effect. JNK1 siRNA decreased the level of lactate dehydrogenase and increased the levels of VE-cadherin, nitric oxide, phosphorylated nitric oxide synthase, and cell viability; JNK2 si RNA had a converse effect. Compared with the control group, the JNK1 shRNA group exhibited a higher lung oxygenation index and lower lung apoptosis index, injury score, wet weight:dry weight ratio, and levels of IL-1, IL-6, and TNF-α. CONCLUSIONS JNK1 aggravated, but JNK2 alleviated, IRI through differential regulation of the JNK1 pathway in in vitro ischemia-reperfusion. JNK1 silence attenuated lung graft dysfunction by inhibiting inflammation and apoptosis. These findings provide a theoretical basis for devising therapeutic strategies against IRI after lung transplantation.
Collapse
Affiliation(s)
- Jing Tan
- Department of Anesthesiology, Hei Long Jiang Province Key Lab of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Wei Gao
- Department of Anesthesiology, Hei Long Jiang Province Key Lab of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Wanchao Yang
- Department of Anesthesiology, Hei Long Jiang Province Key Lab of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xianzhang Zeng
- Department of Anesthesiology, Hei Long Jiang Province Key Lab of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Linlin Wang
- Department of Anesthesiology, Hei Long Jiang Province Key Lab of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xiaoguang Cui
- Department of Anesthesiology, Hei Long Jiang Province Key Lab of Research on Anesthesiology and Critical Care Medicine, Second Affiliated Hospital, Harbin Medical University, Harbin, China; Department of Anesthesiology, First Affiliated Hospital, Hainan Medical University, Hainan, China.
| |
Collapse
|
17
|
Luo F, Xu R, Song G, Lu H, He X, Xia Y. The δ-Opioid Receptor Differentially Regulates MAPKs and Anti-inflammatory Cytokines in Rat Kidney Epithelial Cells Under Hypoxia. Front Physiol 2020; 10:1572. [PMID: 32038276 PMCID: PMC6985288 DOI: 10.3389/fphys.2019.01572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/16/2019] [Indexed: 11/13/2022] Open
Abstract
Hypoxic injury is one of the most important factors in progressive kidney disorders. Since we have found that δ-opioid receptor (DOR) is neuroprotective against hypoxic stress through a differential regulation of mitogen-activated protein kinases (MAPKs) and anti-inflammatory cytokines, we asked if DOR that is highly expressed in the kidney can modulate renal MAPKs and anti-inflammatory cytokines under hypoxia. We exposed cultured rat kidney epithelial cells (NRK-52E) to prolonged hypoxia (1% O2) with applications of specific DOR agonist or/and antagonist to examine if DOR affects hypoxia-induced changes in MAPKs and anti-inflammatory cytokines. The results showed that endogenous DOR expression remained unchanged under hypoxia, while DOR activation with UFP-512 (a specific DOR agonist) reversed the hypoxia-induced up-regulation of ERK1/2 and p38 phosphorylation. DOR inhibition with naltrindole had no appreciable effect on the hypoxia-induced changes in ERK1/2 phosphorylation, but increased p38 phosphorylation. DOR inhibition with naltrindole attenuated the effects of DOR activation on the changes in ERK1/2 and p38 phosphorylation in hypoxia. Moreover, DOR activation/inhibition differentially affected the expression of transcriptional repressor B-cell lymphoma 6 (Bcl-6), anti-inflammatory cytokines tristetraprolin (TTP), and interleukin-10 (IL-10). Taken together, our novel data suggest that DOR activation differentially regulates ERK1/2, p38, Bcl-6, TTP, and IL-10 in the renal cells under hypoxia.
Collapse
Affiliation(s)
- Fengbao Luo
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Renfang Xu
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Guanglai Song
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Hao Lu
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaozhou He
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ying Xia
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Prophylactic supplementation of 20-HETE ameliorates hypoxia/reoxygenation injury in pulmonary vascular endothelial cells by inhibiting apoptosis. Acta Histochem 2020; 122:151461. [PMID: 31706620 DOI: 10.1016/j.acthis.2019.151461] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/21/2019] [Accepted: 10/21/2019] [Indexed: 12/26/2022]
Abstract
Hypoxia reoxygenation (HR) injury perturbs structural and functional syncytium in lung tissues. It is commonly implicated in conditions such as stroke, lung transplant or severe pneumonia. In the present study, we investigated the cytoprotective action of 20-hydroxyeicosatetraenoic acid (20-HETE) on pulmonary vascular endothelial cells (PMVECs) under normoxic and hypoxic niche followed by HR. 20-HETE pretreatment showed a protective effect at a concentration of 1μM as there was a marked increase (20%) in the cell viability compared to control and HR groups. Pretreatment of 20-HETE in HR induced injury decreased ROS production dictated its antioxidant property. Similarly, SOD and ATP levels were also downregulated by 20-HETE pretreatment. Cell apoptosis was detected by TUNEL assay, Acridine orange, and procaspase-3 cleavage, caspase-3 activity assay, respectively. JC-1 mitochondrial membrane potential assay and protein expression pattern of BCL-2, and BAD phosphorylation status were examined. The results showed that HR induced significant increase of apoptotic PMVECs, while 20-HETE pretreatment attenuated the effects. Further, 20-HETE pretreatment activated PI3K/Akt and HIF-1α signaling pathway to exhibit its protective effects against HR-induced oxidative stress and apoptosis. Overall, the results concluded the potent antioxidant role of 20-HETE in aiding cytoprotection upon HR injury.
Collapse
|
19
|
Yang L, Wang W, Wang X, Zhao J, Xiao L, Gui W, Fan H, Xia J, Li Z, Yan J, Alasbahi A, Zhu Q, Hou X. Creg in Hepatocytes Ameliorates Liver Ischemia/Reperfusion Injury in a TAK1-Dependent Manner in Mice. Hepatology 2019; 69:294-313. [PMID: 30076625 DOI: 10.1002/hep.30203] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 06/26/2018] [Indexed: 12/16/2022]
Abstract
Hepatic ischemia/reperfusion (I/R) is a major challenge for liver surgery and specific severe conditions of chronic liver disease. Current surgical and pharmacological strategies are limited to improve liver function after hepatic I/R injury. Thus, an in-depth understanding of the liver I/R mechanism is pivotal to develop new therapeutic methods. The cellular repressor of E1A-stimulated genes (Creg), a key regulator of cellular proliferation, exerts protective roles in cardiovascular diseases and participates in lipid accumulation and inflammatory response in the liver. However, the role of Creg in hepatic I/R remains largely unknown. A genetic engineering technique was used to explore the function of Creg in hepatic I/R injury. Hepatocyte-specific Creg knockout (CregΔHep ) and transgenic mice were generated and subjected to hepatic I/R injury, as were the controls. Creg in hepatocytes prevented against liver I/R injury by suppressing cell death and inflammation. In vitro studies were performed using primary hepatocytes isolated from CregΔHep that were challenged by hypoxia/reoxygenation insult. These cells exhibited more cell death and inflammatory cytokines production similar to observations in vivo. Moreover, further molecular experiments showed that Creg suppressed mitogen-activated protein kinase (MAPK) signaling by inhibiting TAK1 (TGF-β-activated kinase 1) phosphorylation. Inhibiting TAK1 by 5Z-7-ox or mutating the TAK1-binding domain of Creg abolished the protective role of Creg indicating that Creg binding to TAK1 was required for prevention against hepatic I/R injury. Conclusion: These data demonstrate that Creg prevents hepatocytes from liver I/R injury. The Creg-TAK1 interaction inhibited the phosphorylation of TAK1 and the activation of MAPK signaling, which protected against cell death and inflammation during hepatic I/R injury.
Collapse
Affiliation(s)
- Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijun Wang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaozhan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jinfang Zhao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Xiao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenfang Gui
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiqian Fan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Xia
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhonglin Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Yan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Afnan Alasbahi
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Li S, Zhong M, Yuan Y, Zhang L. Differential roles of p38 MAPK and ERK1/2 in angiopoietin-2-mediated rat pulmonary microvascular endothelial cell apoptosis induced by lipopolysaccharide. Exp Ther Med 2018; 16:4729-4736. [PMID: 30546397 DOI: 10.3892/etm.2018.6810] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 02/07/2017] [Indexed: 11/06/2022] Open
Abstract
Angiopoietin-2 (Ang-2) is a Tie-2 ligand that destabilizes vascular structures, enhances vascular permeability and induces vascular regression and endothelial cell apoptosis. Although there is evidence for the involvement of the Ang/Tie2 axis in acute lung injury (ALI), the underlying mechanisms involved in Ang-2-induced cell apoptosis are not well understood. In this study, whether Ang-2 contributes to microvascular endothelial cell injury and mediates lipopolysaccharide (LPS)-induced endothelial cell apoptosis and its associated signaling pathways was investigated. Exposure of rat pulmonary microvascular endothelial cells (RPMVECs) to LPS, Ang-2 and related inhibitors was performed to measure the expression levels of Ang-2, the activation of mitogen-activated protein kinases (MAPKs), the phosphorylation of extracellular signal-regulated kinase (ERK)1/2, and expression of the apoptosis-related proteins Bax and Bcl-2 using western blotting, reverse transcription-quantitative polymerase chain reaction, flow cytometry and fluorescence microscopy. The expression of Ang-2 in the RPMVECs was increased by LPS independent of time. The phosphorylation of p38 MAPK and ERK1/2 was significantly upregulated and the activation of apoptosis-related proteins Bax and Bcl was mediated by Ang-2. In addition, inhibition of the p38 pathway by SB203580 attenuated the Ang-2-mediated cell apoptosis, but inhibition of the ERK1/2 pathway by PD98059 exerted an anti-apoptotic effect against Ang-2. In conclusion, LPS-induced apoptosis is partly mediated via stimulation of p38 and ERK1/2 signaling pathways, where Ang-2 acts an inflammation-related factor to participate in the course of cell apoptosis in RPMVECs.
Collapse
Affiliation(s)
- Shi Li
- ICU, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230061, P.R. China
| | - Mingmei Zhong
- ICU, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230061, P.R. China
| | - Yuan Yuan
- The Central Laboratory of Binhu Hospital, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230061, P.R. China
| | - Lin Zhang
- ICU, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230061, P.R. China
| |
Collapse
|
21
|
Human Mesenchymal Stem Cell Secretome from Bone Marrow or Adipose-Derived Tissue Sources for Treatment of Hypoxia-Induced Pulmonary Epithelial Injury. Int J Mol Sci 2018; 19:ijms19102996. [PMID: 30274394 PMCID: PMC6212866 DOI: 10.3390/ijms19102996] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/20/2018] [Accepted: 09/27/2018] [Indexed: 02/07/2023] Open
Abstract
Alveolar epithelial dysfunction induced by hypoxic stress plays a significant role in the pathological process of lung ischemia-reperfusion injury (IRI). Mesenchymal stem cell (MSC) therapies have demonstrated efficacy in exerting protective immunomodulatory effects, thereby reducing airway inflammation in several pulmonary diseases. Aim: This study assesses the protective effects of MSC secretome from different cell sources, human bone marrow (BMSC) and adipose tissue (ADSC), in attenuating hypoxia-induced cellular stress and inflammation in pulmonary epithelial cells. Methods: Pulmonary epithelial cells, primary rat alveolar epithelial cells (AEC) and A549 cell line were pre-treated with BMSC, or ADSC conditioned medium (CM) and subjected to hypoxia for 24 h. Results: Both MSC-CM improved cell viability, reduced secretion of pro-inflammatory mediators and enhanced IL-10 anti-inflammatory cytokine production in hypoxic injured primary rat AECs. ADSC-CM reduced hypoxic cellular injury by mechanisms which include: inhibition of p38 MAPK phosphorylation and nuclear translocation of subunits in primary AECs. Both MSC-CM enhanced translocation of Bcl-2 to the nucleus, expression of cytoprotective glucose-regulated proteins (GRP) and restored matrix metalloproteinases (MMP) function, thereby promoting repair and cellular homeostasis, whereas inhibition of GRP chaperones was detrimental to cell survival. Conclusions: Elucidation of the protective mechanisms exerted by the MSC secretome is an essential step for maximizing the therapeutic effects, in addition to developing therapeutic targets-specific strategies for various pulmonary syndromes.
Collapse
|
22
|
The Anti-inflammatory Effect of Hydrogen on Lung Transplantation Model of Pulmonary Microvascular Endothelial Cells During Cold Storage Period. Transplantation 2018; 102:1253-1261. [DOI: 10.1097/tp.0000000000002276] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
23
|
Effects of Hypercapnia on Acute Cellular Rejection after Lung Transplantation in Rats. Anesthesiology 2017; 128:130-139. [PMID: 29023354 DOI: 10.1097/aln.0000000000001908] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Hypercapnia alleviates pulmonary ischemia-reperfusion injury, regulates T lymphocytes, and inhibits immune reaction. This study aimed to evaluate the effect of hypercapnia on acute cellular rejection in a rat lung transplantation model. METHODS Recipient rats in sham-operated (Wistar), isograft (Wistar to Wistar), and allograft (Sprague-Dawley to Wistar) groups were ventilated with 50% oxygen, whereas rats in the hypercapnia (Sprague-Dawley to Wistar) group were administered 50% oxygen and 8% carbon dioxide for 90 min during reperfusion (n = 8). Recipients were euthanized 7 days after transplantation. RESULTS The hypercapnia group showed a higher oxygenation index (413 ± 78 vs. 223 ± 24), lower wet weight-to-dry weight ratio (4.23 ± 0.54 vs. 7.04 ± 0.80), lower rejection scores (2 ± 1 vs. 4 ± 1), and lower apoptosis index (31 ± 6 vs. 57 ± 4) as compared with the allograft group. The hypercapnia group showed lower CD8 (17 ± 4 vs. 31 ± 3) and CD68 (24 ± 3 vs. 43 ± 2), lower CD8 T cells (12 ± 2 vs. 35 ± 6), and higher CD4/CD8 ratio (2.2 ± 0.6 vs. 1.1 ± 0.4) compared to the allograft group. Tumor necrosis factor-α (208 ± 40 vs. 292 ± 49), interleukin-2 (30.6 ± 6.7 vs. 52.7 ± 8.3), and interferon-γ (28.1 ± 4.9 vs. 62.7 ± 10.1) levels in the hypercapnia group were lower than those in allograft group. CD4, CD4 T cells, and interleukin-10 levels were similar between groups. CONCLUSIONS Hypercapnia ameliorated acute cellular rejection in a rat lung transplantation model.
Collapse
|
24
|
Wang J, Tan J, Liu Y, Song L, Li D, Cui X. Amelioration of lung ischemia‑reperfusion injury by JNK and p38 small interfering RNAs in rat pulmonary microvascular endothelial cells in an ischemia‑reperfusion injury lung transplantation model. Mol Med Rep 2017; 17:1228-1234. [PMID: 29115603 DOI: 10.3892/mmr.2017.7985] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 05/25/2017] [Indexed: 11/05/2022] Open
Abstract
The inhibition of mitogen‑activated protein kinases (MAPKs), including c‑Jun NH2‑terminal protein kinase (JNK), p38 MAPK (p38) and extracellular signal‑regulated protein kinase 1/2 (ERK1/2), have an important effect on lung ischemia‑reperfusion injury (IRI) during lung transplantation (LT). However, the way in which combined MAPK inhibition exerts optimal protective effects on lung IRI remains to be elucidated. Therefore, the present study evaluated the therapeutic efficacy of the inhibition of MAPKs in rat pulmonary microvascular endothelial cells (PMVECs) in an IRI model of LT. The rat PMVECs were transfected with small interfering RNAs (siRNAs) against JNK, p38 or ERK1/2. Cotransfection was performed with siRNAs against JNK and p38 in the J+p group, JNK and ERK1/2 in the J+E group, p38 and ERK1/2 in the p+E group, or all three in the J+p+E group. Non‑targeting (NT) siRNA was used as a control. The PMVECs were then treated to induce IRI, and the levels of inflammation, apoptosis and oxidative stress were detected. Differences between compared groups were determined using Tukey's honest significant difference test. In all groups, silencing of the MAPKs was shown to attenuate inflammation, apoptosis and oxidative stress to differing extents, compared with the NT group. The J+p and J+p+E groups showed lower levels of interleukin (IL)‑1β, IL‑6 and malondialdehyde, a lower percentage of early‑apoptotic cells, and higher superoxide dismutase (SOD) activity, compared with the other groups. No significant differences were observed in the inflammatory response, SOD activity or early apoptosis between the J+p and J+p+E groups. These findings suggested that the dual inhibition of JNK and p38 led to maximal amelioration of lung IRI in the PMVECs of the IRI model of LT, which occurred through anti‑inflammatory, anti‑oxidative and anti‑apoptotic mechanisms.
Collapse
Affiliation(s)
- Juan Wang
- Department of Anesthesiology, The Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jing Tan
- Department of Anesthesiology, The Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yanhong Liu
- Department of Anesthesiology, The Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Linlin Song
- Department of Anesthesiology, The Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Di Li
- Department of Anesthesiology, The Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaoguang Cui
- Department of Anesthesiology, The Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
25
|
Pharmacological Reconditioning of Marginal Donor Rat Lungs Using Inhibitors of Peroxynitrite and Poly (ADP-ribose) Polymerase During Ex Vivo Lung Perfusion. Transplantation 2017; 100:1465-73. [PMID: 27331361 DOI: 10.1097/tp.0000000000001183] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Donor lungs obtained after prolonged warm ischemia (WI) may be unsuitable for transplantation due to the risk of reperfusion injury, but could be reconditioned using ex-vivo lung perfusion (EVLP). Key processes of reperfusion injury include the formation of reactive oxygen species (ROS)/nitrogen species (RNS) and the activation of poly(adenosine diphosphate-ribose) polymerase (PARP). We explored whether rat lungs obtained after WI could be reconditioned during EVLP using the ROS/RNS scavenger Mn(III)-tetrakis (4-benzoic acid) porphyrin chloride (MnTBAP) or the PARP inhibitor 3-aminobenzamide (3-AB). METHODS Rat lungs obtained after 3 hours cold ischemia (CI group, control), or 1 hour WI plus 2 hours CI (WI group) were placed in an EVLP circuit for normothermic perfusion for 3 hours. Lungs retrieved after WI were treated or not with 3-AB (1 mg/mL) or MnTBAP (0.3 mg/mL), added to the perfusate. Measurements included physiological variables (lung compliance, vascular resistance, oxygenation capacity), lung weight gain, levels of proteins, lactate dehydrogenase, protein carbonyl (marker of ROS), 3-nitrotyrosine (marker of RNS), poly(adenosine diphosphate-ribose) (PAR, marker of PARP activation) and IL-6, in the bronchoalveolar lavage or the lung tissue, and histology. RESULTS In comparison to the CI group, the lungs from the WI group displayed higher protein carbonyls, 3-nitrotyrosine, PAR, lactate dehydrogenase and proteins in bronchoalveolar lavage, lung weight gain, perivascular edema, as well as reduced static compliance, but similar oxygenation. All these alterations were markedly attenuated by 3-AB and MnTBAP. CONCLUSIONS After EVLP, lungs obtained after WI exhibit oxidative stress, PARP activation, and tissue injury, which are suppressed by pharmacological inhibitors of ROS/RNS and PARP.
Collapse
|
26
|
Wang D, Liu Y, Zhong G, Wang Y, Zhang T, Zhao Z, Yan X, Liu Q. Compatibility of Tanshinone IIA and Astragaloside IV in attenuating hypoxia-induced cardiomyocytes injury. JOURNAL OF ETHNOPHARMACOLOGY 2017; 204:67-76. [PMID: 28389356 DOI: 10.1016/j.jep.2017.03.053] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/24/2017] [Accepted: 03/31/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Herbal medicines including Tanshinone IIA (TanIIA) and Astragaloside IV (AsIV) are widely used in Asia as therapeutic agents for cardiovascular diseases, due to their complementary roles and shared properties based on the theory of traditional Chinese medicine and pharmacological researches. However, the underlying pathological mechanisms for their efficacy are still unclear. In addition, the compatibility or incompatibility of the herbal medicines when administered with other herbal remedies or with prescription drugs is unknown. AIM OF THE STUDY We aimed to investigate the compatibility of TanIIA and AsIV in protecting cardiomyocytes against hypoxia-induced injury. MATERIALS AND METHODS Cultured cardiomyocytes were stimulated in hypoxia condition, in the absence or presence of the two herbal compounds, TanIIA and AsIV. Indicators were determined by cytotoxicity assay, quantitative PCR, ELISA, flow cytometry assay, immunofluorescence staining and western blot. RESULTS Either TanIIA alone or the combined herbal compounds inhibited hypoxia-triggered chemokines production including CCL2/5/19, CXCL2 and Transwell assay-indicated monocyte/macrophage recruitment, cytokines production including TNF-α and IL-6. While AsIV alone or the combined herbal compounds attenuated hypoxia-induced cell apoptosis indicated by decreased Annexin V+ cells and the ratio of Bax/Bcl-2, but no significant effect of the herbal compounds was observed in modulating cell apoptosis following both hypoxia and TNF-α stimulation. As an anti-apoptotic factor, stress granule formation was further enhanced by AsIV alone or the combined herbal compounds in hypoxia or heat shock stress. Moreover, immunoblotting analysis indicated that stress-responsive mitogen-activated protein kinases (MAPK) pathways including the phosphorylation of ERK1/2, p38 and JNK were inhibited while the phosphorylation of Akt in phosphatidylinositol 3-kinase (PI3K) -Akt pathway for cell survival was restored by the herbal compounds. Among these results, the combination of TanIIA and AsIV comprised most of the beneficial properties tested, although their combination did not improve the maximal effects achieved by any of the compounds alone. CONCLUSIONS Taken together, these data suggest a compatibility of TanIIA and AsIV in protecting cardiomyocyte against hypoxia-induced injury.
Collapse
Affiliation(s)
- Dawei Wang
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Emergency Department, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510405, China
| | - Yuntao Liu
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Emergency Department, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510405, China
| | - Guofu Zhong
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yuanyuan Wang
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Tong Zhang
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhen Zhao
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xia Yan
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qing Liu
- The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
27
|
Song L, Li D, Wang J, Meng C, Cui X. Effects of p38 mitogen-activated protein kinase on lung ischemia-reperfusion injury in diabetic rats. J Surg Res 2017; 216:9-17. [PMID: 28807219 DOI: 10.1016/j.jss.2017.03.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/02/2017] [Accepted: 03/23/2017] [Indexed: 11/20/2022]
Abstract
BACKGROUND Lung ischemia-reperfusion injury (LIRI) is a pathologic process that is observed in several clinical conditions, and p38 mitogen-activated protein kinase (MAPK) is involved. Diabetes mellitus (DM) results in an increased incidence of ischemia-induced organ damage. The aims of this study were to examine the effects of DM on LIRI in a rat model of DM and to explore the possible mechanisms in relation to the p38 MAPK pathway. METHODS Forty rats were randomly divided into the following five groups (n = 8 each): a control + sham group, a control + IR group (CIR), a DM + sham group, a DM + IR group (DIR), and a DM + IR + SB203580 group. The control and streptozotocin-induced diabetic rats underwent a sham operation or left hilum occlusion for 90 min followed by reperfusion for 4 h. SB203580 was used to inhibit the p38 MAPK pathway. The pulmonary oxygenation index, inflammatory cytokines in the serum, lung edema, histopathology, oxidant stress, apoptosis, and phosphorylated/total-p38 MAPK protein levels were measured. RESULTS The DIR group displayed greater concentrations of tumor necrosis factor-α, interleukin-6, and intercellular adhesion molecule-1 and increases in the wet weight-to-dry weight ratio, lung injury scores, malondialdehyde levels, and cellular apoptosis, and these effects were accompanied by lower pulmonary oxygenation compared with the CIR group (P < 0.05). In the DIR group, the expression levels of p38 MAPK protein were significantly upregulated compared with those of the CIR group. Additionally, all of these alterations were attenuated in the DM + IR + SB203580 group compared with the DIR group. CONCLUSIONS Diabetes exacerbates LIRI by activating the p38 MAPK pathway.
Collapse
Affiliation(s)
- Linlin Song
- Department of Anesthesiology (the Hei Long Jiang Province Key Lab of Research on Anesthesiology and Critical Care Medicine), the Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Di Li
- Department of Anesthesiology (the Hei Long Jiang Province Key Lab of Research on Anesthesiology and Critical Care Medicine), the Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Juan Wang
- Department of Anesthesiology (the Hei Long Jiang Province Key Lab of Research on Anesthesiology and Critical Care Medicine), the Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Chao Meng
- Department of Anesthesiology (the Hei Long Jiang Province Key Lab of Research on Anesthesiology and Critical Care Medicine), the Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xiaoguang Cui
- Department of Anesthesiology (the Hei Long Jiang Province Key Lab of Research on Anesthesiology and Critical Care Medicine), the Second Affiliated Hospital, Harbin Medical University, Harbin, China.
| |
Collapse
|
28
|
Li P, Lv B, Jiang X, Wang T, Ma X, Chang N, Wang X, Gao X. Identification of NF-κB inhibitors following Shenfu injection and bioactivity-integrated UPLC/Q-TOF-MS and screening for related anti-inflammatory targets in vitro and in silico. JOURNAL OF ETHNOPHARMACOLOGY 2016; 194:658-667. [PMID: 27771457 DOI: 10.1016/j.jep.2016.10.052] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 08/11/2016] [Accepted: 10/18/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shenfu injection (SFI) is a commercial medicinal product approved by the China Food and Drug Administration that is widely used in the treatment of stroke and coronary heart disease. However, the material basis and the mechanism of SFI are not fully understood. AIM OF THE STUDY With network pharmacology analysis, our research committed to identify the anti-inflammatory ingredients and mechanism of SFI by combining high-throughput screening. MATERIALS AND METHODS We developed a bioactivity-based UPLC/Q-TOF-MS method followed by network pharmacology and identified the anti-inflammatory active ingredients of SFI from two different perspectives of network computing and high throughput screening. Then we verified the anti-inflammatory effect of SFI in vitro with endothelial cells. After detecting the cell viability, the expression of interleukin-6 (IL-6), inhibitor of nuclear factor kappa-B kinase (IKK), phosphorylated IKK, phosphorylated NF-κB and phosphorylated IκB-α from the supernatant were determined. RESULTS SFI could significantly suppress inflammatory responses, and the mechanism may be via an NF-κB-dependent pathway. The results of high throughput screening (HTS) revealed that protopanaxadiol glycosides (ginsenosides Rb1, Rb2, Rb3, Rc and Rd), protopanaxatriol glycosides (ginsenosides Rg1, Rg2, Re, Rf and F1), diester-type alkaloids (fuziline and neoline) and aconine derivatives (mesaconine and benzoyl-mesaconine) have anti-NF-κB activity. The three compounds (including benzoyl-mesaconine, fuziline and neoline) are the first reported SFI compounds to have NF-κB inhibitor activity. CONCLUSIONS SFI may play a critical role in counteracting inflammation through the NF-κB signaling pathway. The active ingredients are protopanaxadiol glycosides, protopanaxatriol glycosides, diester-type alkaloids and aconine derivatives.
Collapse
Affiliation(s)
- Pan Li
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Bin Lv
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Xiaoqing Jiang
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Ting Wang
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Xianghui Ma
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Nianwei Chang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Xiaoying Wang
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Xiumei Gao
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| |
Collapse
|
29
|
Takahashi M, Chen-Yoshikawa TF, Menju T, Ohata K, Kondo T, Motoyama H, Hijiya K, Aoyama A, Date H. Inhibition of Toll-like receptor 4 signaling ameliorates lung ischemia-reperfusion injury in acute hyperglycemic conditions. J Heart Lung Transplant 2016; 35:815-22. [PMID: 26922276 DOI: 10.1016/j.healun.2015.12.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 10/26/2015] [Accepted: 12/15/2015] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Recent lung transplantation studies have shown that peri-operative hyperglycemia is an important factor affecting recipient survival; however, its underlying mechanisms are not well understood. We hypothesized that acute hyperglycemia exacerbates lung ischemia-reperfusion injury (IRI) through up-regulation of Toll-like receptor 4 (TLR4) signaling pathways. METHODS C57BL/6Ncr mice were divided into 3 treatment groups: sham; IRI; and IRI under acute hyperglycemic conditions (IRI+HG). Mice in the IRI and IRI+HG groups were exposed to IRI via clamping the left hilum for 1 hour, followed by reperfusion for 2 hours. Acute hyperglycemia was established by glucose injection. The severity of lung injury and TLR4 signaling pathway activity were assessed. Further, we performed a pharmacologic blockade of TLR4 signaling to determine the effect of TLR4 signaling inhibition on lung injury. RESULTS Compared with normoglycemic mice, hyperglycemic mice had 2-fold higher blood glucose levels (p < 0.001). Pulmonary compliance was significantly lower, and airway resistance was significantly higher, in the IRI+HG group than in the IRI group (p < 0.05). Levels of inflammatory cytokines in bronchoalveolar lavage fluid were significantly higher in the IRI+HG group than in the IRI group. Correspondingly, TLR4 signaling pathways were up-regulated in the IRI+HG group. Moreover, pharmacologic inhibition of TLR4 signaling significantly decreased lung injury markers under hyperglycemic conditions. CONCLUSIONS Acute hyperglycemia exacerbated lung IRI and was associated with up-regulation of TLR4 signaling pathways. Pharmacologic inhibition of TLR4 signaling ameliorated lung IRI with acute hyperglycemia. Targeting TLR4 appears to be a promising approach to managing coexisting pathologies in lung transplant recipients.
Collapse
Affiliation(s)
- Mamoru Takahashi
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Toshi Menju
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Keiji Ohata
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Kondo
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hideki Motoyama
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kyoko Hijiya
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akihiro Aoyama
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| |
Collapse
|
30
|
Zhang P, Fang L, Wu H, Ding P, Shen Q, Liu R. Down-regulation of GRα expression and inhibition of its nuclear translocation by hypoxia. Life Sci 2016; 146:92-9. [PMID: 26767627 DOI: 10.1016/j.lfs.2015.12.059] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 12/08/2015] [Accepted: 12/31/2015] [Indexed: 02/03/2023]
Abstract
AIMS Glucocorticoids are the most effective anti-inflammatory agent in treating pulmonary diseases typically accompanied by hypoxia. Our previous study has demonstrated that glucocorticoid receptor α (GRα) expression is reduced in hypoxia but the underlying mechanism remains elusive. In this study we aim to identify the signaling pathway involved in hypoxia-induced down-regulation of GRα, and whether hypoxia affects nuclear translocation of GRα. MAIN METHODS Female C57BL/6 mice were sensitized with saline or ovalbumin (OVA) as the in vivo model. Mice were divided into control and OVA groups, and their lung histology and the expression of hypoxia inducible factor (HIF-1) and GRα were examined. A549 cells were exposed to chemical hypoxia as the in vitro model, where mitogen-activated protein kinases (MAPKs) were inhibited specifically by SB203580. Next, under normal or hypoxic conditions, the expression of GRα, MAPKs and HIF-1 signal protein were determined by Western blot analysis, and GRα translocation were observed through live-cell imaging. KEY FINDINGS In OVA challenged mice the expression of GRα was down-regulated whereas HIF-1 was up-regulated. Hypoxia caused a time-dependent decrease of GRα expression, and activated multiple signaling pathways including MAPKs and HIF-1. Moreover, GRα expression increased with MAPK inhibition. Interestingly, only MAPK inhibitor SB203580, but not JNK inhibitor SP600125 or ERK inhibitor U0126 improved the expression of GRα under hypoxic condition. GRα nuclear translocation was also significantly inhibited by hypoxia. SIGNIFICANCE Hypoxia down-regulated the expression of GRα through p38 signaling pathway, as well as inhibited GRα nuclear translocation significantly.
Collapse
Affiliation(s)
- Pei Zhang
- Department of Pulmonary, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University
| | - Lei Fang
- Department of Pulmonary, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University
| | - HuiMei Wu
- Department of Pulmonary, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University
| | - Peishan Ding
- Department of Pulmonary, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University
| | - QiYing Shen
- Department of Pulmonary, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University
| | - Rongyu Liu
- Department of Pulmonary, Anhui Geriatric Institute, The First Affiliated Hospital of Anhui Medical University
| |
Collapse
|
31
|
DING WENGANG, GUO YUEPING, CUI XIAOGUANG, ZHANG BING, LI DONGMEI, LI WENZHI. Morphine-induced delayed pre-conditioning against anoxia/reoxygenation injury in pulmonary artery endothelial cells: The role of mitochondrial KATP channels. Mol Med Rep 2015; 13:1047-53. [DOI: 10.3892/mmr.2015.4629] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 11/06/2015] [Indexed: 11/05/2022] Open
|
32
|
Zhao G, Seng J, Beagle J, Syrkina O, Hales CA. Heparin reduces overcirculation-induced pulmonary artery remodeling through p38 MAPK in piglet. Ann Thorac Surg 2015; 99:1677-84. [PMID: 25818573 DOI: 10.1016/j.athoracsur.2014.12.061] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 12/08/2014] [Accepted: 12/16/2014] [Indexed: 10/23/2022]
Abstract
BACKGROUND Artery remodeling is the principal change of pulmonary artery hypertension. Heparin has been shown to inhibit vascular smooth muscle cell proliferation. We hypothesized that heparin may modulate vascular remodeling in pulmonary artery hypertension, and explored the mechanism. METHODS A localized overcirculation-induced artery remodeling was created in piglets by anastomosing the left lower lobe pulmonary artery (LLLPA) to the thoracic aortic artery. Piglets were treated with heparin or saline for 4 weeks. Hemodynamic data were collected, and histology of the lung was assessed. We investigated the expressions of several candidate genes in lung and further observed the involvement of P38 mitogen-activated protein kinases (MAPK). The effects of heparin on the growth of cultured pulmonary arterial vascular smooth muscle cell and P38 MAPK expression were further determined under various conditions. RESULTS Four weeks after the shunt setup, overcirculation caused significant LLLPA remodeling, pressure increase, and pulmonary vascular resistance increase, and LLLPA flow reduction compared with that immediately after the shunt setup. Heparin reduced the LLLPA remodeling, pressure, and pulmonary vascular resistance, and increased the LLLPA flow compared with that not heparin treated. Shunt and heparin treatment did not change the piglet's systemic hemodynamics. Shunt increased the expression of P38 MAPK and heparin decreased its expression in the shunted LLLPA. Both heparin and P38 MAPK inhibitor suppressed VSMC growth and P38 MAPK expression in the cultured VSMC, but they did not present additive effects when the two treatments were combined. CONCLUSIONS Heparin reduces overcirculation-induced pulmonary artery remodeling through a P38 MAPK-dependent pathway.
Collapse
Affiliation(s)
- Gaofeng Zhao
- Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Department of Surgery, First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Jingjing Seng
- Department of Surgery, First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - John Beagle
- Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Olga Syrkina
- Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Charles A Hales
- Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
33
|
CB2 receptor activation ameliorates the proinflammatory activity in acute lung injury induced by paraquat. BIOMED RESEARCH INTERNATIONAL 2014; 2014:971750. [PMID: 24963491 PMCID: PMC4054852 DOI: 10.1155/2014/971750] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 05/08/2014] [Indexed: 01/21/2023]
Abstract
Paraquat, a widely used herbicide, is well known to exhibit oxidative stress and lung injury. In the present study, we investigated the possible underlying mechanisms of cannabinoid receptor-2 (CB2) activation to ameliorate the proinflammatory activity induced by PQ in rats. JWH133, a CB2 agonist, was administered by intraperitoneal injection 1 h prior to PQ exposure. After PQ exposure for 4, 8, 24, and 72 h, the bronchoalveolar lavage fluid was collected to determine levels of TNF-α and IL-1β, and the arterial blood samples were collected for detection of PaO2 level. At 72 h after PQ exposure, lung tissues were collected to determine the lung wet-to-dry weight ratios, myeloperoxidase activity, lung histopathology, the protein expression level of CB2, MAPKs (ERK1/2, p38MAPK, and JNK1/2), and NF-κBp65. After rats were pretreated with JWH133, PQ-induced lung edema and lung histopathological changes were significantly attenuated. PQ-induced TNF-α and IL-1β secretion in BALF, increases of PaO2 in arterial blood, and MPO levels in the lung tissue were significantly reduced. JWH133 could efficiently activate CB2, while inhibiting MAPKs and NF-κB activation. The results suggested that activating CB2 receptor exerted protective activity against PQ-induced ALI, and it potentially contributed to the suppression of the activation of MAPKs and NF-κB pathways.
Collapse
|
34
|
Lim SK, Jeong YW, Kim DI, Park MJ, Choi JH, Kim SU, Kang SS, Han HJ, Park SH. Activation of PRMT1 and PRMT5 mediates hypoxia- and ischemia-induced apoptosis in human lung epithelial cells and the lung of miniature pigs: The role of p38 and JNK mitogen-activated protein kinases. Biochem Biophys Res Commun 2013; 440:707-13. [DOI: 10.1016/j.bbrc.2013.09.136] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 09/29/2013] [Indexed: 01/06/2023]
|