1
|
Wu X, Cap AP, Bynum JA, Chance TC, Darlington DN, Meledeo MA. Prolyl hydroxylase domain inhibitor is an effective pre-hospital pharmaceutical intervention for trauma and hemorrhagic shock. Sci Rep 2024; 14:3874. [PMID: 38365865 PMCID: PMC10873291 DOI: 10.1038/s41598-024-53945-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/07/2024] [Indexed: 02/18/2024] Open
Abstract
Pre-hospital potentially preventable trauma related deaths are mainly due to hypoperfusion-induced tissue hypoxia leading to irreversible organ dysfunction at or near the point of injury or during transportation prior to receiving definitive therapy. The prolyl hydroxylase domain (PHD) is an oxygen sensor that regulates tissue adaptation to hypoxia by stabilizing hypoxia inducible factor (HIF). The benefit of PHD inhibitors (PHDi) in the treatment of anemia and lactatemia arises from HIF stabilization, which stimulates endogenous production of erythropoietin and activates lactate recycling through gluconeogenesis. The results of this study provide insight into the therapeutic roles of MK-8617, a pan-inhibitor of PHD-1, 2, and 3, in the mitigation of lactatemia in anesthetized rats with polytrauma and hemorrhagic shock. Additionally, in an anesthetized rat model of lethal decompensated hemorrhagic shock, acute administration of MK-8617 significantly improves one-hour survival and maintains survival at least until 4 h following limited resuscitation with whole blood (20% EBV) at one hour after hemorrhage. This study suggests that pharmaceutical interventions to inhibit prolyl hydroxylase activity can be used as a potential pre-hospital countermeasure for trauma and hemorrhage at or near the point of injury.
Collapse
Affiliation(s)
- Xiaowu Wu
- Blood and Shock Resuscitation, USA Army Institute of Surgical Research, 3698 Chambers Pass, Bldg 3610, JBSA Fort Sam Houston, TX, 78234-7767, USA.
| | - Andrew P Cap
- Blood and Shock Resuscitation, USA Army Institute of Surgical Research, 3698 Chambers Pass, Bldg 3610, JBSA Fort Sam Houston, TX, 78234-7767, USA
| | - James A Bynum
- Department of Surgery, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Tiffani C Chance
- Department of Health and Human Services, Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Daniel N Darlington
- Blood and Shock Resuscitation, USA Army Institute of Surgical Research, 3698 Chambers Pass, Bldg 3610, JBSA Fort Sam Houston, TX, 78234-7767, USA
| | - Michael A Meledeo
- Blood and Shock Resuscitation, USA Army Institute of Surgical Research, 3698 Chambers Pass, Bldg 3610, JBSA Fort Sam Houston, TX, 78234-7767, USA
| |
Collapse
|
2
|
Alberghina L. The Warburg Effect Explained: Integration of Enhanced Glycolysis with Heterogeneous Mitochondria to Promote Cancer Cell Proliferation. Int J Mol Sci 2023; 24:15787. [PMID: 37958775 PMCID: PMC10648413 DOI: 10.3390/ijms242115787] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
The Warburg effect is the long-standing riddle of cancer biology. How does aerobic glycolysis, inefficient in producing ATP, confer a growth advantage to cancer cells? A new evaluation of a large set of literature findings covering the Warburg effect and its yeast counterpart, the Crabtree effect, led to an innovative working hypothesis presented here. It holds that enhanced glycolysis partially inactivates oxidative phosphorylation to induce functional rewiring of a set of TCA cycle enzymes to generate new non-canonical metabolic pathways that sustain faster growth rates. The hypothesis has been structured by constructing two metabolic maps, one for cancer metabolism and the other for the yeast Crabtree effect. New lines of investigation, suggested by these maps, are discussed as instrumental in leading toward a better understanding of cancer biology in order to allow the development of more efficient metabolism-targeted anticancer drugs.
Collapse
Affiliation(s)
- Lilia Alberghina
- Centre of Systems Biology, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| |
Collapse
|
3
|
Suresh MV, Francis S, Aktay S, Kralovich G, Raghavendran K. Therapeutic potential of curcumin in ARDS and COVID-19. Clin Exp Pharmacol Physiol 2023; 50:267-276. [PMID: 36480131 PMCID: PMC9877870 DOI: 10.1111/1440-1681.13744] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/13/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022]
Abstract
Curcumin is a safe, non-toxic, readily available and naturally occurring compound, an active constituent of Curcuma longa (turmeric). Curcumin could potentially treat diseases, but faces poor physicochemical and pharmacological characteristics. To overcome these limitations, we developed a stable, water-soluble formulation of curcumin called cyclodextrin-complexed curcumin (CDC). We have previously shown that direct delivery of CDC to the lung following lipopolysaccharides exposure reduces acute lung injury (ALI) and effectively reduces lung injury, inflammation and mortality in mice following Klebsiella pneumoniae. Recently, we found that administration of CDC led to a significant reduction in angiotensin-converting enzyme 2 and signal transducer and activator of transcription 3 expression in gene and protein levels following pneumonia, indicating its potential in treating coronavirus disease 2019 (COVID-19). In this review, we consider the clinical features of ALI and acute respiratory distress syndrome (ARDS) and the role of curcumin in modulating the pathogenesis of bacterial/viral-induced ARDS and COVID-19.
Collapse
Affiliation(s)
| | - Sairah Francis
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Sinan Aktay
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Georgia Kralovich
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
4
|
Chen L, Yang J, Zhang M, Fu D, Luo H, Yang X. SPP1 exacerbates ARDS via elevating Th17/Treg and M1/M2 ratios through suppression of ubiquitination-dependent HIF-1α degradation. Cytokine 2023; 164:156107. [PMID: 36773529 DOI: 10.1016/j.cyto.2022.156107] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/24/2022] [Accepted: 12/05/2022] [Indexed: 02/11/2023]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a severe inflammatory pulmonary condition that leads to respiratory failure. The imbalance of Th17/Treg and M1/M2 is implicated in ARDS. A better understanding of the regulation of the balance of Th17/Treg and M1/M2 may provide novel therapeutic targets for ARDS. METHODS Plasma and BALF samples were collected from ARDS patients. Inflammatory cytokines were examined by ELISA. Th17, Treg, M1 and M2 were identified via immunofluorescence staining of RORγt, Foxp3, iNOS and Arg-1. H&E and Masson's trichrome staining were applied for evaluating pulmonary damage and fibrosis. A mouse model of ARDS was established through LPS administration. HIF-1α was immunoprecipitated and subjected to ubiquitination analysis via western blotting. The expression of SPP1, VHL and HIF-1α was examined by RT-qPCR and western blotting. RESULTS ARDS patients showed elevated levels of inflammatory cytokines and ratios of Th17/Treg and M1/M2. SPP1 was upregulated in ARDS mice, and silencing of SPP1 alleviated lung injury and fibrosis. SPP1 inhibited VHL expression to reduce the ubiquitination and degradation of HIF-1α in ARDS. Overexpression of SPP1 facilitated Th17, Treg and M1 polarization but inhibited M2 polarization through upregulation of HIF-1α. CONCLUSION SPP1 elevates Th17/Treg and M1/M2 ratio by suppressing VHL expression and ubiquitination-dependent HIF-1α degradation, thus exacerbating ARDS. Our study provides novel mechanistic insights into ARDS pathogenesis and promising therapeutic targets.
Collapse
Affiliation(s)
- Liang Chen
- Intensive Care Unit, Chongqing General Hospital, Chongqing 401147, PR China.
| | - Jin Yang
- Intensive Care Unit, Chongqing General Hospital, Chongqing 401147, PR China
| | - Meng Zhang
- Intensive Care Unit, Chongqing General Hospital, Chongqing 401147, PR China
| | - Donglin Fu
- Intensive Care Unit, Chongqing General Hospital, Chongqing 401147, PR China
| | - Huan Luo
- Intensive Care Unit, Chongqing General Hospital, Chongqing 401147, PR China
| | - Xiaolei Yang
- Intensive Care Unit, Chongqing General Hospital, Chongqing 401147, PR China
| |
Collapse
|
5
|
Therapeutic Targets for Regulating Oxidative Damage Induced by Ischemia-Reperfusion Injury: A Study from a Pharmacological Perspective. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8624318. [PMID: 35450409 PMCID: PMC9017553 DOI: 10.1155/2022/8624318] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/28/2022] [Accepted: 03/15/2022] [Indexed: 12/22/2022]
Abstract
Ischemia-reperfusion (I-R) injury is damage caused by restoring blood flow into ischemic tissues or organs. This complex and characteristic lesion accelerates cell death induced by signaling pathways such as apoptosis, necrosis, and even ferroptosis. In addition to the direct association between I-R and the release of reactive oxygen species and reactive nitrogen species, it is involved in developing mitochondrial oxidative damage. Thus, its mechanism plays a critical role via reactive species scavenging, calcium overload modulation, electron transport chain blocking, mitochondrial permeability transition pore activation, or noncoding RNA transcription. Other receptors and molecules reduce tissue and organ damage caused by this pathology and other related diseases. These molecular targets have been gradually discovered and have essential roles in I-R resolution. Therefore, the current study is aimed at highlighting the importance of these discoveries. In this review, we inquire about the oxidative damage receptors that are relevant to reducing the damage induced by oxidative stress associated with I-R. Several complications on surgical techniques and pathology interventions do not mitigate the damage caused by I-R. Nevertheless, these therapies developed using alternative targets could work as coadjuvants in tissue transplants or I-R-related pathologies
Collapse
|
6
|
Li LF, Yu CC, Wu HP, Chu CM, Huang CY, Liu PC, Liu YY. Reduction in Ventilation-Induced Diaphragmatic Mitochondrial Injury through Hypoxia-Inducible Factor 1α in a Murine Endotoxemia Model. Int J Mol Sci 2022; 23:ijms23031083. [PMID: 35163007 PMCID: PMC8835058 DOI: 10.3390/ijms23031083] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 11/16/2022] Open
Abstract
Mechanical ventilation (MV) is essential for patients with sepsis-related respiratory failure but can cause ventilator-induced diaphragm dysfunction (VIDD), which involves diaphragmatic myofiber atrophy and contractile inactivity. Mitochondrial DNA, oxidative stress, mitochondrial dynamics, and biogenesis are associated with VIDD. Hypoxia-inducible factor 1α (HIF-1α) is crucial in the modulation of diaphragm immune responses. The mechanism through which HIF-1α and mitochondria affect sepsis-related diaphragm injury is unknown. We hypothesized that MV with or without endotoxin administration would aggravate diaphragmatic and mitochondrial injuries through HIF-1α. C57BL/6 mice, either wild-type or HIF-1α-deficient, were exposed to MV with or without endotoxemia for 8 h. MV with endotoxemia augmented VIDD and mitochondrial damage, which presented as increased oxidative loads, dynamin-related protein 1 level, mitochondrial DNA level, and the expressions of HIF-1α and light chain 3-II. Furthermore, disarrayed myofibrils; disorganized mitochondria; increased autophagosome numbers; and substantially decreased diaphragm contractility, electron transport chain activities, mitofusin 2, mitochondrial transcription factor A, peroxisome proliferator activated receptor-g coactivator-1α, and prolyl hydroxylase domain 2 were observed (p < 0.05). Endotoxin-stimulated VIDD and mitochondrial injuries were alleviated in HIF-1α-deficient mice (p < 0.05). Our data revealed that endotoxin aggravated MV-induced diaphragmatic dysfunction and mitochondrial damages, partially through the HIF-1α signaling pathway.
Collapse
Affiliation(s)
- Li-Fu Li
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chung-Chieh Yu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Huang-Pin Wu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Chien-Ming Chu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Chih-Yu Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ping-Chi Liu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yung-Yang Liu
- Chest Department, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Correspondence: ; Tel.: 886-2-28712121 (ext. 3071); Fax: 886-2-28757858
| |
Collapse
|
7
|
Suresh MV, Yalamanchili G, Rao TC, Aktay S, Kralovich A, Shah YM, Raghavendran K. Hypoxia‐inducible factor (HIF)‐1α‐induced regulation of lung injury in pulmonary aspiration is mediated through NF‐kB. FASEB Bioadv 2022; 4:309-328. [PMID: 35520392 PMCID: PMC9065579 DOI: 10.1096/fba.2021-00132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/11/2022] Open
Abstract
Aspiration‐induced lung injury is a common grievance encountered in the intensive care unit (ICU). It is a significant risk factor for improving ventilator‐associated pneumonia (VAP) and acute respiratory distress syndrome (ARDS). Hypoxia‐inducible factor (HIF)‐1α is one of the primary transcription factors responsible for regulating the cellular response to changes in oxygen tension. Here, we sought to determine the role of HIF‐1α and specifically the role of type 2 alveolar epithelial cells in generating the acute inflammatory response following acid and particles (CASP) aspiration. Previous studies show HIF‐1 α is involved in regulating the hypoxia‐stimulated expression of MCP‐1 in mice and humans. The CASP was induced in C57BL/6, ODD‐Luc, HIF‐1α (+/+) control, and HIF‐1α conditional knockout (HIF‐1α (−/−) mice). Following an injury in ODD mice, explanted organs were subjected to IVIS imaging to measure the degree of hypoxia. HIF‐1α expression, BAL albumin, cytokines, and histology were measured following CASP. In C57BL/6 mice, the level of HIF‐1α was increased at 1 h after CASP. There were significantly increased levels of albumin and cytokines in C57BL/6 and ODD‐Luc mice lungs following CASP. HIF‐1α (+/+) mice given CASP demonstrated a synergistic increase in albumin leakage, increased pro‐inflammatory cytokines, and worse injury. MCP‐1 antibody neutralized HIF‐1α (+/+) mice showed reduced granuloma formation. The NF‐κB expression was increased substantially in the HIF‐1α (+/+) mice following CASP compared to HIF‐1α (−/−) mice. Our data collectively identify that HIF‐1α upregulation of the acute inflammatory response depends on NF‐κB following CASP.
Collapse
Affiliation(s)
| | | | - Tejeshwar C. Rao
- Department of Cell, Developmental, and Integrative Biology The University of Alabama at Birmingham Birmingham UK
| | - Sinan Aktay
- Department of Surgery University of Michigan Ann Arbor Michigan USA
| | - Alex Kralovich
- Department of Surgery University of Michigan Ann Arbor Michigan USA
| | - Yatrik M. Shah
- Molecular & Integrative Physiology University of Michigan Ann Arbor Michigan USA
| | | |
Collapse
|
8
|
Hypoxia-Inducible Factor Signaling in Inflammatory Lung Injury and Repair. Cells 2022; 11:cells11020183. [PMID: 35053299 PMCID: PMC8774273 DOI: 10.3390/cells11020183] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 01/27/2023] Open
Abstract
Inflammatory lung injury is characterized by lung endothelial cell (LEC) death, alveolar epithelial cell (AEC) death, LEC-LEC junction weakening, and leukocyte infiltration, which together disrupt nutrient and oxygen transport. Subsequently, lung vascular repair is characterized by LEC and AEC regeneration and LEC-LEC junction re-annealing, which restores nutrient and oxygen delivery to the injured tissue. Pulmonary hypoxia is a characteristic feature of several inflammatory lung conditions, including acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and severe coronavirus disease 2019 (COVID-19). The vascular response to hypoxia is controlled primarily by the hypoxia-inducible transcription factors (HIFs) 1 and 2. These transcription factors control the expression of a wide variety of target genes, which in turn mediate key pathophysiological processes including cell survival, differentiation, migration, and proliferation. HIF signaling in pulmonary cell types such as LECs and AECs, as well as infiltrating leukocytes, tightly regulates inflammatory lung injury and repair, in a manner that is dependent upon HIF isoform, cell type, and injury stimulus. The aim of this review is to describe the HIF-dependent regulation of inflammatory lung injury and vascular repair. The review will also discuss potential areas for future study and highlight putative targets for inflammatory lung conditions such as ALI/ARDS and severe COVID-19. In the development of HIF-targeted therapies to reduce inflammatory lung injury and/or enhance pulmonary vascular repair, it will be vital to consider HIF isoform- and cell-specificity, off-target side-effects, and the timing and delivery strategy of the therapeutic intervention.
Collapse
|
9
|
Li LF, Yu CC, Huang HY, Wu HP, Chu CM, Huang CY, Liu PC, Liu YY. Suppression of Hypoxia-Inducible Factor 1α by Low-Molecular-Weight Heparin Mitigates Ventilation-Induced Diaphragm Dysfunction in a Murine Endotoxemia Model. Int J Mol Sci 2021; 22:ijms22041702. [PMID: 33567713 PMCID: PMC7914863 DOI: 10.3390/ijms22041702] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/29/2022] Open
Abstract
Mechanical ventilation (MV) is required to maintain life for patients with sepsis-related acute lung injury but can cause diaphragmatic myotrauma with muscle damage and weakness, known as ventilator-induced diaphragm dysfunction (VIDD). Hypoxia-inducible factor 1α (HIF-1α) plays a crucial role in inducing inflammation and apoptosis. Low-molecular-weight heparin (LMWH) was proven to have anti-inflammatory properties. However, HIF-1α and LMWH affect sepsis-related diaphragm injury has not been investigated. We hypothesized that LMWH would reduce endotoxin-augmented VIDD through HIF-1α. C57BL/6 mice, either wild-type or HIF-1α–deficient, were exposed to MV with or without endotoxemia for 8 h. Enoxaparin (4 mg/kg) was administered subcutaneously 30 min before MV. MV with endotoxemia aggravated VIDD, as demonstrated by increased interleukin-6 and macrophage inflammatory protein-2 levels, oxidative loads, and the expression of HIF-1α, calpain, caspase-3, atrogin-1, muscle ring finger-1, and microtubule-associated protein light chain 3-II. Disorganized myofibrils, disrupted mitochondria, increased numbers of autophagic and apoptotic mediators, substantial apoptosis of diaphragm muscle fibers, and decreased diaphragm function were also observed (p < 0.05). Endotoxin-exacerbated VIDD and myonuclear apoptosis were attenuated by pharmacologic inhibition by LMWH and in HIF-1α–deficient mice (p < 0.05). Our data indicate that enoxaparin reduces endotoxin-augmented MV-induced diaphragmatic injury, partially through HIF-1α pathway inhibition.
Collapse
Affiliation(s)
- Li-Fu Li
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chung-Chieh Yu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Hung-Yu Huang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Huang-Pin Wu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Chien-Ming Chu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Chih-Yu Huang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ping-Chi Liu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yung-Yang Liu
- Chest Department, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan
- Correspondence: ; Tel.: +(886)-2-28712121 (ext. 3071); Fax: +(886)-2-28757858
| |
Collapse
|
10
|
Sherif IO, Al-Shaalan NH. Alleviation of remote lung injury following liver ischemia/reperfusion: Possible protective role of vildagliptin. Int Immunopharmacol 2021; 91:107305. [PMID: 33388732 DOI: 10.1016/j.intimp.2020.107305] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/21/2022]
Abstract
Lung injury is a serious condition encountered following hepatic ischemia/reperfusion (IR). This study aimed to explore whether a dipeptidyl peptidase-4 inhibitor agent vildagliptin (V) could alleviate the lung injury caused by hepatic IR in a rat model and if so elucidate its molecular protective mechanism. Three groups of rats were used. Sham group: received normal saline and exposed to a sham operation, IR group: received normal saline and subjected to the operation of hepatic I (45 min)/ R (180 min), V+IR group: received for 10 days intraperitoneal injection of V (10 mg/kg/day). After reperfusion, liver and lung were collected for biochemical and histological evaluation. Hepatic IR exhibited significant elevation in serum alanine aminotransferase (ALT), alkaline phosphatase (ALP), and lactate dehydrogenase (LDH) enzyme levels, serum and lung malondialdehyde (MDA) and tumor necrosis factor-alpha (TNF-α) in addition to lung nitric oxide (NO) levels, hypoxia-inducible factor 1-alpha (HIF-1α) mRNA and protein levels, hepatocyte growth factor (HGF) mRNA expression, and inducible nitric oxide synthase (iNOS) mRNA and protein expressions in lung tissue along with a marked reduction in the serum and lung content of catalase in comparison to the sham group. Moreover, liver and lung injury in the IR group was detected by histopathological examination. Vildagliptin ameliorated markedly the biochemical changes as well as liver and lung architecture in comparison to the IR group. Vildagliptin mitigated the induced lung injury by hepatic IR via suppression of oxidative stress markers, pro-inflammatory cytokine TNF-α as well as the HIF1-α/iNOS/HGF expressions in lung tissue.
Collapse
Affiliation(s)
- Iman O Sherif
- Emergency Hospital, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Nora H Al-Shaalan
- Chemistry Department, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| |
Collapse
|
11
|
Vishnupriya S, Priya Dharshini LC, Sakthivel KM, Rasmi RR. Autophagy markers as mediators of lung injury-implication for therapeutic intervention. Life Sci 2020; 260:118308. [PMID: 32828942 PMCID: PMC7442051 DOI: 10.1016/j.lfs.2020.118308] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
Abstract
Lung injury is characterized by inflammatory processes demonstrated as loss of function of the pulmonary capillary endothelial and alveolar epithelial cells. Autophagy is an intracellular digestion system that work as an inducible adaptive response to lung injury which is a resultant of exposure to various stress agents like hypoxia, ischemia-reperfusion and xenobiotics which may be manifested as acute lung injury (ALI), acute respiratory distress syndrome (ARDS), chronic lung injury (CLI), bronchopulmonary dysplasia (BPD), chronic obstructive pulmonary disease (COPD), asthma, ventilator-induced lung injury (VILI), ventilator-associated lung injury (VALI), pulmonary fibrosis (PF), cystic fibrosis (CF) and radiation-induced lung injury (RILI). Numerous regulators like LC3B-II, Beclin 1, p62, HIF1/BNIP3 and mTOR play pivotal role in autophagy induction during lung injury possibly for progression/inhibition of the disease state. The present review focuses on the critical autophagic mediators and their potential cross talk with the lung injury pathophysiology thereby bringing to limelight the possible therapeutic interventions.
Collapse
Affiliation(s)
- Selvaraj Vishnupriya
- Department of Biotechnology, PSG College of Arts and Science, Civil Aerodrome Post, Coimbatore 641 014, Tamil Nadu, India
| | | | - Kunnathur Murugesan Sakthivel
- Department of Biochemistry, PSG College of Arts and Science, Civil Aerodrome Post, Coimbatore 641 014, Tamil Nadu, India
| | - Rajan Radha Rasmi
- Department of Biotechnology, PSG College of Arts and Science, Civil Aerodrome Post, Coimbatore 641 014, Tamil Nadu, India.
| |
Collapse
|
12
|
Zhou F, Liu Z, Cai H, Miao Z, Wei F, Song C. Role of microRNA-15a-5p/TNFAIP3-interacting protein 2 axis in acute lung injury induced by traumatic hemorrhagic shock. Exp Ther Med 2020; 20:2. [PMID: 32934667 PMCID: PMC7471858 DOI: 10.3892/etm.2020.9130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 06/17/2020] [Indexed: 12/15/2022] Open
Abstract
The present study aimed to investigate the role of microRNA (miR)-15a-5p in the pathogenesis of acute lung injury induced by traumatic hemorrhagic shock (THS), and to explore the underlying molecular mechanism. The expression level of miR-15a-5p was detected using reverse transcription-quantitative (RT-qPCR) and the association between miR-15a-5p and TNFAIP3-interacting protein 2 (TNIP2) was revealed using TargetScan and dual luciferase reporter assays. To investigate the effect of miR-15a-5p on THS-induced acute lung injury, a THS rat model was established. Lung capillary permeability and lung edema were then determined. Moreover, proinflammatory factors in the bronchoalveolar lavage fluid (BALF) and serum of the THS rat model were detected using ELISA. In addition, protein levels in the current study were measured via western blotting. It was revealed that miR-15a-5p was significantly upregulated in both patients with THS and samples from the THS rat model. TNIP2 represents a direct target of miR-15a-5p, and it was downregulated in both patients with THS and the THS rat model. Further analyses indicated that downregulation of miR-15a-5p significantly relieved acute lung injury induced by THS, evidenced by a decreased ratio of Evan's blue dye (EBD) in the BALF to EBD in plasma of THS rats, decreased lung permeability index and reduced lung wet/dry ratio. Inhibition of miR-15a-5p also decreased THS-induced upregulation of pro-inflammatory factors. Furthermore, the data revealed that THS-induced NF-κB activation in the lung tissues of rats was inhibited by miR-15a-5p knockdown. Moreover, it was demonstrated that all the effects of miR-15a-5p on THS rats were ablated following TNIP2 silencing. Taken together, the data of the current study indicate that miR-15a-5p downregulation serves a protective role in THS-induced acute lung injury via directly targeting TNIP2.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Zhizhen Liu
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Huazhong Cai
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Zhenjun Miao
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Faxing Wei
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Chao Song
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| |
Collapse
|
13
|
Li X, Shan C, Wu Z, Yu H, Yang A, Tan B. Emodin alleviated pulmonary inflammation in rats with LPS-induced acute lung injury through inhibiting the mTOR/HIF-1α/VEGF signaling pathway. Inflamm Res 2020; 69:365-373. [PMID: 32130427 DOI: 10.1007/s00011-020-01331-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/10/2020] [Accepted: 02/25/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE AND DESIGN This study aimed to investigate the anti-pulmonary inflammation effect of emodin on Wistar rats with lipopolysaccharide (LPS)-induced acute lung injury (ALI) and RAW264.7 cells through the mammalian target of rapamycin (mTOR)/hypoxia-inducible factor-1α (HIF-1α)/vascular endothelial growth factor (VEGF) signaling pathway. SUBJECTS Wistar rats and RAW264.7 cells were studied. TREATMENT LPS was used to induce inflammation in rats or RAW264.7 cells and emodin was given once a day before LPS stimulation and continued for a certain number of days. METHODS Lung tissues and bronchoalveolar lavage fluid (BALF) were collected for the in vivo experiment, while cells and supernatant were collected for the in vitro experiment. Pathological changes in the lung tissues were assessed by hematoxylin and eosin staining. The levels of inflammatory factors, including TNF-α, IL-1β, and IL-6, were determined by enzyme-linked immunosorbent assay. The expression levels of p-mTOR, HIF-1α, and VEGF proteins were measured by Western blot analysis and immunohistochemistry. The mRNA levels of p70S6K, eIF4E-BP1, and eIF4E were measured by quantitative polymerase chain reaction. RESULTS Emodin ameliorated pathological changes and infiltrated inflammatory cells in LPS-induced ALI. It also significantly reduced the expression of inflammatory factors, including TNF-α, IL-1β, and IL-6, in BALF and downregulated the expression of p-mTOR, HIF-1α, and VEGF proteins in the lung tissues. Similar anti-inflammatory effects and the downregulation of the mTOR/HIF-1α/VEGF signaling pathway were found in RAW264.7 cells. The mRNA levels of p70S6K, eIF4E-BP1, and eIF4E also decreased in the macrophages. CONCLUSION Emodin alleviated LPS-induced pulmonary inflammation in rat lung tissues and RAW264.7 cells through inhibiting the mTOR/HIF-1α/VEGF signaling pathway, which accounted for the therapeutic effects of emodin on ALI.
Collapse
Affiliation(s)
- Xiaoqian Li
- Research Centre on Application of Classical Prescriptions, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Department of Febrile Disease, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Cong Shan
- Research Centre on Application of Classical Prescriptions, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Department of Febrile Disease, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhonghua Wu
- Research Centre on Application of Classical Prescriptions, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Department of Febrile Disease, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hongji Yu
- Research Centre on Application of Classical Prescriptions, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Department of Febrile Disease, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Aidong Yang
- Research Centre on Application of Classical Prescriptions, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China. .,Department of Febrile Disease, Basic Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Bo Tan
- Clinical Pharmacokinetic Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
14
|
Activation of Hypoxia-Inducible Factor-1α Via Succinate Dehydrogenase Pathway During Acute Lung Injury Induced by Trauma/Hemorrhagic Shock. Shock 2020; 53:208-216. [DOI: 10.1097/shk.0000000000001347] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
15
|
Rao TW, Shen YH, Zhao XG, Jiang SY. Effect of oxygen supplement during targeted temperature management on acute lung injury in the early stage of traumatic hemorrhagic shock. EUR J INFLAMM 2020. [DOI: 10.1177/2058739220930448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Ideal concentrations of inhaled oxygen with regard to lung protection during early traumatic hemorrhagic shock (THS) remain unknown especially in the era of targeted temperature management (TTM). We speculated that a significant increase in oxygen supply in early stage of THS would magnify the protecting role of hypothermia in acute lung injury. Forty male New Zealand rabbits were randomly divided into four groups (n = 10): sham group, control group, group 1, and group 2. Except for sham group, all other animals were submitted to 30 min of uncontrolled THS and received limited fluid resuscitation for 60 min. During resuscitation, in addition to 34°C of TTM, animals in group 1 inhaled 21% oxygen while animals in group 2 inhaled 50% oxygen. Animals in control group inhaled room air and were kept normothermia. Oxidative stress, inflammation, and apoptosis parameters in the lung tissues were determined. THS induced higher expression of malondialdehyde, surfactant protein A, nuclear factor kappa B, and caspase 3 as well as lower expression of Bcl-2 mRNA and superoxide dismutase activity. Compared with inhalation of 21% oxygen, inhalation of 50% oxygen during TTM significantly improves oxidative stress, inflammation, apoptosis, and acute lung injury. Oxygen supplement during TTM therapy alleviated acute lung injury in the early stage of THS. Further studies are required to explore the ideal combination forms of TTM and oxygen supplement with the purpose of maximizing therapeutic effect while minimizing adverse effects.
Collapse
Affiliation(s)
- Tai-Wen Rao
- Department of Emergency Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Research Institute of Emergency Medicine, Zhejiang University, Hangzhou, China
| | - Ye-Hua Shen
- Department of Radiology, The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiao-Gang Zhao
- Department of Emergency Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Research Institute of Emergency Medicine, Zhejiang University, Hangzhou, China
| | - Shou-Yin Jiang
- Department of Emergency Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Research Institute of Emergency Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
16
|
Wu B, Miao X, Ye J, Pu X. The Protective Effects of Protease Inhibitor MG-132 on Sepsis-Induced Acute Lung Rats and Its Possible Mechanisms. Med Sci Monit 2019; 25:5690-5699. [PMID: 31366881 PMCID: PMC6688517 DOI: 10.12659/msm.915743] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Background The aim of the present study was to investigate the protective effects of protease inhibitor MG-132 on sepsis-induced acute lung injury rats. Material/Methods Sprague Dawley rats were employed to induce sepsis by cecal ligation and puncture (CLP) method. Rats were divided into 4 groups: control, sham, model (CLP), and MG-132. Histopathology observation was detected by hematoxylin and eosin staining. The ratio of wet lung to dry lung (W/D) was calculated. In addition, the levels of inflammatory factors in bronchoalveolar lavage fluid (BALF) were measured by enzyme-linked immunosorbent assay (ELISA). Also, superoxide dismutase (SOD) and malondialdehyde (MDA) levels were evaluated. Western blotting was performed to measure the expression of hypoxia-inducible factor-1 α (HIF-1α). In order to assess the role of HIF-1α, YC-1, the inhibitor of HIF-1α, was used to treat the rats. The expression of phosphor-mTOR (p-mTOR), p-4EBP1, and p-EIF4E were evaluated by western blotting. Results Obvious pathological injury and increasing ratio of W/D in the model group were observed. Both pathological injury and W/D were improved in the MG-132 group, and the greatest improvement could be seen in the YC-1+MG-132 group. Furthermore, the MDA levels in the MG-132 group was decreased, accompanied by an increase in SOD levels. The level of HIF-1α was increased in the model group while a decreased was detected in the MG-132 group. The levels of inflammatory factors were high in the model group, whereas the opposite result was found in the MG-132 group, and the lowest in were in the YC-1+MG-132 group. Furthermore, the expression levels of p-mTOR, p-4EBP1, and p-EIF4E proteins were downregulated in the MG-132 group compared to the model group, and the lowest was in the YC-1+MG-132 group. Conclusions Our study suggested that MG-132 was able to protect against acute lung injury via inhibition of HIF-1α mediated mTOR/4EBP1/EIF4E pathway.
Collapse
Affiliation(s)
- Bingbing Wu
- Department of Intensive Care Unit, Taizhou People's Hospital, Taizhou, Jiangsu, China (mainland)
| | - Xiaoli Miao
- Department of Intensive Care Unit, Taizhou People's Hospital, Taizhou, Jiangsu, China (mainland)
| | - Jilu Ye
- Department of Intensive Care Unit, Taizhou People's Hospital, Taizhou, Jiangsu, China (mainland)
| | - Xuehua Pu
- Department of Intensive Care Unit, Taizhou People's Hospital, Taizhou, Jiangsu, China (mainland)
| |
Collapse
|
17
|
Molecular Characterization of Hypoxic Alveolar Epithelial Cells After Lung Contusion Indicates an Important Role for HIF-1α. Ann Surg 2019; 267:382-391. [PMID: 27811509 DOI: 10.1097/sla.0000000000002070] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To understand the fate and regulation of hypoxic type II alveolar epithelial cells (AECs) after lung contusion (LC). BACKGROUND LC due to thoracic trauma is a major risk factor for the development of acute respiratory distress syndrome. AECs have recently been implicated as a primary driver of inflammation in LC. The main pathological consequence of LC is hypoxia, and a key mediator of adaptation to hypoxia is hypoxia-inducible factor (HIF)-1. We have recently published that HIF-1α is a major driver of acute inflammation after LC through type II AEC. METHODS LC was induced in wild-type mice (C57BL/6), luciferase-based hypoxia reporter mice (ODD-Luc), and HIF-1α conditional knockout mice. The degree of hypoxia was assessed using hypoxyprobe and in vivo imaging system. The fate of hypoxic AEC was evaluated by luciferase dual staining with caspases-3 and Ki-67, terminal deoxynucleotidyl transferase dUTP nick end labeling, and flow cytometry with ApoStat. NLRP-3 expression was determined by western blot. Laser capture microdissection was used to isolate AECs in vivo, and collected RNA was analyzed by Q-PCR for HIF-related pathways. RESULTS Global hypoxia was present after LC, but hypoxic foci were not uniform. Hypoxic AECs preferentially undergo apoptosis. There were significant reductions in NLRP-3 in HIF-1α conditional knockout mice. The expression of proteins involved in HIF-related pathways and inflammasome activation were significantly increased in hypoxic AECs. CONCLUSIONS These are the first in vivo data to identify, isolate, and characterize hypoxic AECs. HIF-1α regulation through hypoxic AECs is critical to the initiation of acute inflammation after LC.
Collapse
|
18
|
Pu J, Zhu S, Zhou D, Zhao L, Yin M, Wang Z, Hong J. Propofol Alleviates Apoptosis Induced by Chronic High Glucose Exposure via Regulation of HIF-1 α in H9c2 Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4824035. [PMID: 31093315 PMCID: PMC6481038 DOI: 10.1155/2019/4824035] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 01/26/2019] [Accepted: 02/04/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND The sedative anesthetic, propofol, is a cardioprotective agent for hyperglycemia-induced myocardial hypertrophy and dysfunction in rats. However, the specific protective mechanism has not been clarified. METHODS AND RESULTS In this experiment, we used H9c2 cells subjected to 22 mM glucose lasting for 72 hours as an in vitro model of cardiomyocyte injury by hyperglycemia and investigated the potential mechanism of propofol against hyperglycemic stress in cells. Propofol (5, 10, or 20 μM) was added to the cell cultures before and during the high glucose culture phases. Cell viability and levels of ROS were measured. The levels of proinflammatory cytokines were tested by ELISA. The levels of SIRT3, SOD2, PHD2, HIF-1α, Bcl-2, P53, and cleaved caspase-3 proteins were detected by western blotting. Our data showed that propofol attenuated high glucose-induced cell apoptosis accompanied by a decrease in the level of reactive oxygen species (ROS) and proinflammatory cytokines. Meanwhile, propofol decreased the apoptosis of H9c2 cells via increasing the expression of Bcl-2, SIRT3, SOD2, and PHD2 proteins and decreasing the expression of cleaved caspase-3, P53, and HIF-1α. Real-time PCR analysis showed that propofol did not significantly change the HIF-1α but increase PHD2 at mRNA level. HIF-1α silence significantly decreased apoptosis and inflammation in H9c2 cell during high glucose stress. Pretreatment of IOX2 (the inhibitor of PHD2) inhibited cell viability until the concentration reached 200 μM during high glucose stress. However, 50 μM TYP (the inhibitor of SIRT3) significantly inhibited cell viability during high glucose stress. Delayed IOX2 treatment for 6 hours significantly inhibited cell viability during high glucose stress. CONCLUSIONS Propofol might alleviate cell apoptosis via SIRT3-HIF-1α axis during high glucose stress.
Collapse
Affiliation(s)
- Jinjun Pu
- 1Department of Internal and Emergency Medicine, Shanghai General Hospital (Originally Named Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- 2Department of Emergency Medicine, Putuo Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shun Zhu
- 1Department of Internal and Emergency Medicine, Shanghai General Hospital (Originally Named Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dandan Zhou
- 1Department of Internal and Emergency Medicine, Shanghai General Hospital (Originally Named Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lidong Zhao
- 1Department of Internal and Emergency Medicine, Shanghai General Hospital (Originally Named Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Yin
- 3School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Zejian Wang
- 3School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Jiang Hong
- 1Department of Internal and Emergency Medicine, Shanghai General Hospital (Originally Named Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Ketamine induces neuronal apoptosis and cognitive disorder via miR-199a-5p/HIF-1α in neonatal rats. Mol Cell Toxicol 2017. [DOI: 10.1007/s13273-017-0044-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
20
|
Ji Z, Liu H, Fang L, Yu Y, Zhou Z. Use of immunoproteomics to identify immunogenic proteins in a rat model of acute respiratory distress syndrome. Mol Med Rep 2017; 16:7625-7632. [PMID: 28944852 DOI: 10.3892/mmr.2017.7557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/17/2017] [Indexed: 11/06/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common and life‑threatening clinical syndrome, and seeking biomarkers of ARDS has been an area of continuing research. The present study hypothesized that alterations to certain immunogenic substances occur in injured lungs and are able to specifically bind with corresponding proteins in the blood, and that these proteins may be readily detected. To investigate this hypothesis, a rat model of ARDS was established by cecal ligation and puncture surgery, and an immunoproteomics approach, using serum as the primary antibody in a western blot analysis, was used with the aim of identifying immunogenic proteins in the injured lungs. Ingenuity Pathway Analysis (IPA) was used for bioinformatics analysis, and mass spectrometric analysis was used to identify a total of 38 differentially expressed immunogenic proteins. Bioinformatics analysis revealed that the top canonical pathways in which the identified proteins may be involved were gluconeogenesis I, glycolysis I, choline degradation I, NADH repair and heme degradation. IPA Biomarker Filter analysis with the terms 'acute respiratory distress syndrome/acute lung injury' was used to screen 13 proteins as candidate biomarkers. These proteins were described as antigens, and suggested that paired antibodies may be detected in the plasma of patients at high risk of ARDS. Analysis of these identified proteins may provide novel insights into the potential pathological mechanisms of ARDS.
Collapse
Affiliation(s)
- Zongshu Ji
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Haiyan Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Linsen Fang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Youxin Yu
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Zheng Zhou
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
21
|
Sun HD, Liu YJ, Chen J, Chen MY, Ouyang B, Guan XD. The pivotal role of HIF-1α in lung inflammatory injury induced by septic mesenteric lymph. Biomed Pharmacother 2017; 91:476-484. [DOI: 10.1016/j.biopha.2017.04.103] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 04/13/2017] [Accepted: 04/23/2017] [Indexed: 11/16/2022] Open
|
22
|
Sordi R, Nandra KK, Chiazza F, Johnson FL, Cabrera CP, Torrance HD, Yamada N, Patel NSA, Barnes MR, Brohi K, Collino M, Thiemermann C. Artesunate Protects Against the Organ Injury and Dysfunction Induced by Severe Hemorrhage and Resuscitation. Ann Surg 2017; 265:408-417. [PMID: 28059970 DOI: 10.1097/sla.0000000000001664] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To evaluate the effects of artesunate on organ injury and dysfunction associated with hemorrhagic shock (HS) in the rat. BACKGROUND HS is still a common cause of death in severely injured patients and is characterized by impairment of organ perfusion, systemic inflammatory response, and multiple organ failure. There is no specific therapy that reduces organ injury/dysfunction. Artesunate exhibits pharmacological actions beyond its antimalarial activity, such as anticancer, antiviral, and anti-inflammatory effects. METHODS Rats were submitted to HS. Mean arterial pressure was reduced to 30 mm Hg for 90 minutes, followed by resuscitation. Rats were randomly treated with artesunate (2.4 or 4.8 mg/kg i.v.) or vehicle upon resuscitation. Four hours later, parameters of organ injury and dysfunction were assessed. RESULTS Artesunate attenuated the multiple organ injury and dysfunction caused by HS. Pathway analysis of RNA sequencing provided good evidence to support an effect of artesunate on the Akt-survival pathway, leading to downregulation of interleukin-1 receptor-associated kinase 1. Using Western blot analysis, we confirmed that treatment of HS rats with artesunate enhanced the phosphorylation (activation) of Protein kinase B (Akt) and endothelial nitric oxide synthase and the phosphorylation (inhibition) of glycogen synthase kinase-3β (GSK-3β). Moreover, artesunate attenuated the HS-induced activation of nuclear factor kappa B and reduced the expression of proinflammatory proteins (inducible nitric oxide synthase, tumor necrosis factor-α, and interleukin 6). CONCLUSIONS Artesunate attenuated the organ injury/dysfunction associated with HS by a mechanism that involves the activation of the Akt-endothelial nitric oxide synthase survival pathway, and the inhibition of glycogen synthase kinase-3β and nuclear factor kappa B. A phase II clinical trial evaluating the effects of good manufacturing practice-artesunate in patients with trauma and severe hemorrhage is planned.
Collapse
Affiliation(s)
- Regina Sordi
- *Centre for Translational Medicine and Therapeutics, Queen Mary University of London, William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, London, UK †Department of Drug Science and Technology, University of Turin, Turin, Italy ‡Department of Clinical Pharmacology, Queen Mary University of London, William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, London, UK §Centre for Trauma Sciences, Queen Mary University of London, Blizard Institute, Barts and The London School of Medicine & Dentistry, London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Liu X, Zhang J, Han W, Wang Y, Liu Y, Zhang Y, Zhou D, Xiang L. Inhibition of BTK protects lungs from trauma-hemorrhagic shock-induced injury in rats. Mol Med Rep 2017; 16:192-200. [PMID: 28487990 PMCID: PMC5482099 DOI: 10.3892/mmr.2017.6553] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 01/31/2017] [Indexed: 01/05/2023] Open
Abstract
The present study aimed to investigate the role of Bruton's tyrosine kinase (BTK) in the pathogenesis of lung injury induced by trauma‑hemorrhagic shock (THS), and to examine the pulmonary protective effects of BTK inhibition. Male Sprague‑Dawley rats were divided into four groups (n=12/group): i) A Sham group, which received surgery without induced trauma; ii) a THS‑induced injury group; iii) a THS‑induced injury group that also received treatment with the BTK inhibitor LFM‑A13 prior to trauma induction; and iv) a Sham group that was pretreated with LFM‑A13 prior to surgery but did not receive induced trauma. The expression of phosphorylated‑BTK protein in the lungs was measured by immunohistochemistry and western blot analysis. The bronchoalveolar lavage fluid (BALF) protein concentration, total leukocyte and eosinophil numbers, and the expression levels of peripheral blood proinflammatory factors were measured. Morphological alterations in the lungs were detected by hematoxylin and eosin staining. Pulmonary nitric oxide (NO) concentration and inducible NO synthase (iNOS) expression were also assessed. Activities of the nuclear factor (NF)‑κB and mitogen‑activated protein kinase (MAPK) signaling pathways were determined by western blotting or electrophoretic mobility shift assay. BTK was notably activated in lungs of THS rats. BALF protein concentration, total leukocytes and eosinophils, peripheral blood expression levels of tumor necrosis factor‑α, interleukin (IL)‑1β, IL‑6 and monocyte chemotactic protein 1 were significantly upregulated after THS induction, and each exhibited decreased expression upon LFM‑A13 treatment. THS‑induced interstitial hyperplasia, edema and neutrophilic infiltration in lungs were improved by the inhibition of BTK. In addition, THS‑induced NO release, iNOS overexpression, and NF‑κB and MAPK signaling were suppressed by BTK inhibition. Results from the present study demonstrate that BTK may serve a pivotal role in the pathogenesis of THS‑related lung injury, and the inhibition of BTK may significantly alleviate THS‑induced lung damage. These results provide a potential therapeutic application for the treatment of THS‑induced lung injury.
Collapse
Affiliation(s)
- Xinwei Liu
- Department of Orthopaedic Surgery, Rescue Center for Severe Wound and Trauma of Chinese PLA, The General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Jingdong Zhang
- Department of Orthopaedic Surgery, Rescue Center for Severe Wound and Trauma of Chinese PLA, The General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Wenfeng Han
- Department of Orthopaedic Surgery, Rescue Center for Severe Wound and Trauma of Chinese PLA, The General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Yu Wang
- Department of Orthopaedic Surgery, Rescue Center for Severe Wound and Trauma of Chinese PLA, The General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Yunen Liu
- Laboratory of Severe and War‑Related Trauma Center, The General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Yubiao Zhang
- Laboratory of Severe and War‑Related Trauma Center, The General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Dapeng Zhou
- Department of Orthopaedic Surgery, Rescue Center for Severe Wound and Trauma of Chinese PLA, The General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| | - Liangbi Xiang
- Department of Orthopaedic Surgery, Rescue Center for Severe Wound and Trauma of Chinese PLA, The General Hospital of Shenyang Military Area Command, Shenyang, Liaoning 110016, P.R. China
| |
Collapse
|
24
|
Hua S, Liu X, Lv S, Wang Z. Protective Effects of Cucurbitacin B on Acute Lung Injury Induced by Sepsis in Rats. Med Sci Monit 2017; 23:1355-1362. [PMID: 28315572 PMCID: PMC5367850 DOI: 10.12659/msm.900523] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background The aim of this study was to investigate the protective effects of cucurbitacin B (CuB) on sepsis-induced acute lung injury (ALI) in rats. Material/Methods An ALI model was made by cecal ligation and puncture (CLP) in SD rats. Rats were randomly divided into 5 groups (n=15 per group): animals undergoing a sham CLP (sham group); animals undergoing CLP (CLP control group); animals undergoing CLP and treated with CuB at 1 mg/kg of body weight (bw) (low-dose CuB [L-CuB] group), animals undergoing CuB at 2 mg/kg of bw (mid-dose CuB [M-CuB] group); and animals undergoing CuB at 5 mg/kg of bw (high-dose CuB [H-CuB] group). Samples of blood and lung tissue were harvested at different time points (6, 12, and 24 hour post-CLP surgery) for the detection of indicators which represented ALI. Five rats were respectively sacrificed at each time point. Pathological changes of lung tissue were observed by H&E staining. Another 50 rats were distributed into the same five groups to record the 72 hour survival rates. Results Treatment with CuB significantly increased the blood gas PaO2 levels and decreased lung wet/dry (W/D) ratio (p<0.05). It significantly reduced protein concentration, accumulation of the inflammatory cells, and levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), (p<0.05), in the bronchoalveolar lavage fluid (BALF). Pulmonary pathological damage and survival rates at 72 hours were found to be effectively improved by CuB. In addition, CuB performed its pulmonary protection effects in a dose-depended manner. Conclusions CuB can effectively improve the pulmonary gas exchange function, reduce pulmonary edema, and inhibit the inflammatory response in the lung, revealing that CuB may serve as a potential therapeutic strategy for sepsis-induced ALI.
Collapse
Affiliation(s)
- Shu Hua
- Department of Pediatrics, Yantai Hospital of Traditional of Chinese Medicine, Yantai, Shandong, China (mainland)
| | - Xing Liu
- Intensive Care Unit, Yantai Hospital of Traditional of Chinese Medicine, Yantai, Shandong, China (mainland)
| | - Shuguang Lv
- Department of Heart Disease, Yantai Hospital of Traditional of Chinese Medicine, Yantai, Shandong, China (mainland)
| | - Zhifang Wang
- Intensive Care Unit, Yantai Hospital of Traditional of Chinese Medicine, Yantai, Shandong, China (mainland)
| |
Collapse
|
25
|
Wu C, Wang X, Jiang T, Li C, Zhang L, Gao X, Tian F, Li N, Li J. Partial Enteral Nutrition Mitigated Ischemia/Reperfusion-Induced Damage of Rat Small Intestinal Barrier. Nutrients 2016; 8:nu8080502. [PMID: 27548209 PMCID: PMC4997415 DOI: 10.3390/nu8080502] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/07/2016] [Accepted: 08/11/2016] [Indexed: 01/03/2023] Open
Abstract
Background and Aims: This study was designed to investigate a relatively optimum dose of partial enteral nutrition (PEN) which effectively attenuates intestinal barrier dysfunction initiated by ischemia/reperfusion injury (IRI). Methods: In experiment 1, 60 male Sprague-Dawley (SD) rats were subjected to intestinal IRI and assigned to six groups according to the different proportion of EN administrations: namely total parenteral nutrition (TPN or 0%EN), 10%EN, 20%EN, 40%EN, 60%EN, and total enteral nutrition (TEN or 100%) groups, the deficits of intraluminal calorie were supplemented by PN. In experiment 2, 50 male SD rats were subjected to intestinal IRI and divided into five groups based on the results of experiment 1: TPN, TEN, 20%EN, TPN plus pretreatment with NF-κB antagonist 30 min before IRI (TPN+PDTC), and TPN plus pretreatment with HIF-1α antagonist 30 min before IRI (TPN+YC-1) groups. Results: In experiment 1, previous IRI combined with subsequent EN shortage disrupted the structure of intestinal epithelial cell and tight junctions (TJs). While 20% dose of EN had an obviously protective effect on these detrimental consequences. In experiment 2, compared with TPN only, 20%EN exerted a significant protection of barrier function of intestinal epithelium. Analogous results were observed when TPN combined with specific NF-κB/HIF-1α inhibitors (PDTC and YC-1). Meanwhile, the expression of NF-κB/HIF-1α had a similar trend among the groups. Conclusions: Our findings indicate that 20%EN is the minimally effective dosage of EN which promotes the recovery of intestinal barrier function after IRI in a rat model. Furthermore, we discreetly speculate that this benefit is, at least partly, related to NF-κB/HIF-1α pathway expression.
Collapse
Affiliation(s)
- Chao Wu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Xinying Wang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Tingting Jiang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Chaojun Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University and Model Animal Research Center, National Resource Center for Mutant Mice, Nanjing 210093, China.
| | - Li Zhang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Xuejin Gao
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Feng Tian
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Ning Li
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Jieshou Li
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| |
Collapse
|
26
|
Jin Y, Zhao X, Zhang H, Li Q, Lu G, Zhao X. Modulatory effect of silymarin on pulmonary vascular dysfunction through HIF-1α-iNOS following rat lung ischemia-reperfusion injury. Exp Ther Med 2016; 12:1135-1140. [PMID: 27446333 DOI: 10.3892/etm.2016.3370] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 11/25/2015] [Indexed: 12/20/2022] Open
Abstract
Silymarin is a traditional therapeutic used to protect the liver, acting to oppose lipid peroxidation, to enhance liver regeneration and functioning as an antioxidant. However, the effects of silymarin on pulmonary vascular dysfunction have not been investigated. In the present study, the modulatory effects of silymarin on pulmonary vascular dysfunction and the underlying mechanisms behind this were investigated in a lung ischemia-reperfusion (I/R) injury rat model. Male Sprague Dawley rats were randomly divided into 3 groups, including: i) A control group (n=10); ii) an I/R group (n=10); and iii) a silymarin-treated group (n=10). All experimental rats received 250 mg/kg/day of silymarin for 8 days. Silymarin was demonstrated to markedly improve lung I/R-induced pulmonary vascular dysfunction and lung moisture. Following silymarin treatment, inflammation and oxidative stress in the lung I/R-injury rats were demonstrably suppressed. Treatment with silymarin also inhibited the activation of caspase-3 and -9, and hypoxia inducible factor-1α (HIF-1α) and inducible nitric oxide synthase (iNOS) protein expression in the lung I/R-injury rats. Silymarin was concluded to impact upon pulmonary vascular dysfunction through the HIF-1α-iNOS pathway in the lung I/R injury rat model.
Collapse
Affiliation(s)
- Yanwu Jin
- Department of Anesthesiology II, Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Xin Zhao
- Department of Anesthesiology II, Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - He Zhang
- Department of Anesthesiology II, Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Qingsong Li
- Department of Anesthesiology II, Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Guodong Lu
- Department of Anesthesiology II, Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Xiaogang Zhao
- Department of Thoracic Surgery, Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
27
|
Xiao Z, Wilson C, Robertson HL, Roberts DJ, Ball CG, Jenne CN, Kirkpatrick AW. Inflammatory mediators in intra-abdominal sepsis or injury - a scoping review. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2015; 19:373. [PMID: 26502877 PMCID: PMC4623902 DOI: 10.1186/s13054-015-1093-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 10/07/2015] [Indexed: 12/16/2022]
Abstract
Introduction Inflammatory and protein mediators (cytokine, chemokine, acute phase proteins) play an important, but still not completely understood, role in the morbidity and mortality of intra-abdominal sepsis/injury. We therefore systematically reviewed preclinical and clinical studies of mediators in intra-abdominal sepsis/injury in order to evaluate their ability to: (1) function as diagnostic/prognostic biomarkers; (2) serve as therapeutic targets; and (3) illuminate the pathogenesis mechanisms of sepsis or injury-related organ dysfunction. Methods We searched MEDLINE, PubMed, EMBASE and the Cochrane Library. Two investigators independently reviewed all identified abstracts and selected articles for full-text review. We included original studies assessing mediators in intra-abdominal sepsis/injury. Results Among 2437 citations, we selected 182 studies in the scoping review, including 79 preclinical and 103 clinical studies. Serum procalcitonin and C-reactive protein appear to be useful to rule out infection or monitor therapy; however, the diagnostic and prognostic value of mediators for complications/outcomes of sepsis or injury remains to be established. Peritoneal mediator levels are substantially higher than systemic levels after intra-abdominal infection/trauma. Common limitations of current studies included small sample sizes and lack of uniformity in study design and outcome measures. To date, targeted therapies against mediators remain experimental. Conclusions Whereas preclinical data suggests mediators play a critical role in intra-abdominal sepsis or injury, there is no consensus on the clinical use of mediators in diagnosing or managing intra-abdominal sepsis or injury. Measurement of peritoneal mediators should be further investigated as a more sensitive determinant of intra-abdominal inflammatory response. High-quality clinical trials are needed to better understand the role of inflammatory mediators. Electronic supplementary material The online version of this article (doi:10.1186/s13054-015-1093-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhengwen Xiao
- Regional Trauma Services, Foothills Medical Centre, 1403 - 29 Street NW, T2N 2T9, Calgary, AB, Canada.
| | - Crystal Wilson
- Regional Trauma Services, Foothills Medical Centre, 1403 - 29 Street NW, T2N 2T9, Calgary, AB, Canada.
| | - Helen Lee Robertson
- Health Sciences Library, Health Sciences Centre, University of Calgary, 3330 Hospital Drive NW, T2N 4N1, Calgary, AB, Canada.
| | - Derek J Roberts
- Department of Surgery, Foothills Medical Centre, University of Calgary, 1403 - 29 Street NW, T2N 2T9, Calgary, AB, Canada. .,Department of Community Health Sciences (Division of Epidemiology), University of Calgary, 3280 Hospital Drive Northwest, T2N 4Z6, Calgary, AB, Canada.
| | - Chad G Ball
- Regional Trauma Services, Foothills Medical Centre, 1403 - 29 Street NW, T2N 2T9, Calgary, AB, Canada. .,Department of Surgery, Foothills Medical Centre, University of Calgary, 1403 - 29 Street NW, T2N 2T9, Calgary, AB, Canada.
| | - Craig N Jenne
- Department of Critical Care Medicine, Foothills Medical Centre, University of Calgary, 3134 Hospital Drive NW, T2N 5A1, Calgary, AB, Canada. .,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, 3280 Hospital Drive NW, T2N 4N1, Calgary, AB, Canada.
| | - Andrew W Kirkpatrick
- Regional Trauma Services, Foothills Medical Centre, 1403 - 29 Street NW, T2N 2T9, Calgary, AB, Canada. .,Department of Surgery, Foothills Medical Centre, University of Calgary, 1403 - 29 Street NW, T2N 2T9, Calgary, AB, Canada. .,Department of Critical Care Medicine, Foothills Medical Centre, University of Calgary, 3134 Hospital Drive NW, T2N 5A1, Calgary, AB, Canada.
| |
Collapse
|
28
|
Wagner K, Gröger M, McCook O, Scheuerle A, Asfar P, Stahl B, Huber-Lang M, Ignatius A, Jung B, Duechs M, Möller P, Georgieff M, Calzia E, Radermacher P, Wagner F. Blunt Chest Trauma in Mice after Cigarette Smoke-Exposure: Effects of Mechanical Ventilation with 100% O2. PLoS One 2015. [PMID: 26225825 PMCID: PMC4520521 DOI: 10.1371/journal.pone.0132810] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cigarette smoking (CS) aggravates post-traumatic acute lung injury and increases ventilator-induced lung injury due to more severe tissue inflammation and apoptosis. Hyper-inflammation after chest trauma is due to the physical damage, the drop in alveolar PO2, and the consecutive hypoxemia and tissue hypoxia. Therefore, we tested the hypotheses that 1) CS exposure prior to blunt chest trauma causes more severe post-traumatic inflammation and thereby aggravates lung injury, and that 2) hyperoxia may attenuate this effect. Immediately after blast wave-induced blunt chest trauma, mice (n=32) with or without 3-4 weeks of CS exposure underwent 4 hours of pressure-controlled, thoraco-pulmonary compliance-titrated, lung-protective mechanical ventilation with air or 100 % O2. Hemodynamics, lung mechanics, gas exchange, and acid-base status were measured together with blood and tissue cytokine and chemokine concentrations, heme oxygenase-1 (HO-1), activated caspase-3, and hypoxia-inducible factor 1-α (HIF-1α) expression, nuclear factor-κB (NF-κB) activation, nitrotyrosine formation, purinergic receptor 2X4 (P2XR4) and 2X7 (P2XR7) expression, and histological scoring. CS exposure prior to chest trauma lead to higher pulmonary compliance and lower PaO2 and Horovitz-index, associated with increased tissue IL-18 and blood MCP-1 concentrations, a 2-4-fold higher inflammatory cell infiltration, and more pronounced alveolar membrane thickening. This effect coincided with increased activated caspase-3, nitrotyrosine, P2XR4, and P2XR7 expression, NF-κB activation, and reduced HIF-1α expression. Hyperoxia did not further affect lung mechanics, gas exchange, pulmonary and systemic cytokine and chemokine concentrations, or histological scoring, except for some patchy alveolar edema in CS exposed mice. However, hyperoxia attenuated tissue HIF-1α, nitrotyrosine, P2XR7, and P2XR4 expression, while it increased HO-1 formation in CS exposed mice. Overall, CS exposure aggravated post-traumatic inflammation, nitrosative stress and thereby organ dysfunction and injury; short-term, lung-protective, hyperoxic mechanical ventilation have no major beneficial effect despite attenuation of nitrosative stress, possibly due to compensation of by regional alveolar hypoxia and/or consecutive hypoxemia, resulting in down-regulation of HIF-1α expression.
Collapse
MESH Headings
- Acute Lung Injury/etiology
- Acute Lung Injury/physiopathology
- Acute Lung Injury/therapy
- Animals
- Disease Models, Animal
- Female
- Hyperoxia/complications
- Hyperoxia/pathology
- Hyperoxia/physiopathology
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Lung/pathology
- Lung/physiopathology
- Male
- Mice
- Mice, Inbred C57BL
- Oxidative Stress
- Pulmonary Disease, Chronic Obstructive/etiology
- Pulmonary Disease, Chronic Obstructive/physiopathology
- Pulmonary Disease, Chronic Obstructive/therapy
- Reactive Nitrogen Species/metabolism
- Receptors, Purinergic P2X/metabolism
- Respiration, Artificial/adverse effects
- Smoking/adverse effects
- Thoracic Injuries/complications
- Thoracic Injuries/physiopathology
- Thoracic Injuries/therapy
- Wounds, Nonpenetrating/complications
- Wounds, Nonpenetrating/physiopathology
- Wounds, Nonpenetrating/therapy
Collapse
Affiliation(s)
- Katja Wagner
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Ulm, Germany
- Klinik für Anästhesiologie, Universitätsklinikum, Ulm, Germany
| | - Michael Gröger
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Ulm, Germany
| | - Oscar McCook
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Ulm, Germany
| | | | - Pierre Asfar
- Laboratoire HIFIH, UPRES EA 3859, PRES l’UNAM, IFR 132, CNRS UMR 6214, INSERM U1083, Université Angers, Département de Réanimation Médicale et de Médecine Hyperbare, Centre Hospitalier Universitaire, Angers, France
| | - Bettina Stahl
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Ulm, Germany
| | - Markus Huber-Lang
- Klinik für Unfall-, Hand-, Plastische und Wiederherstellungschirurgie, Universitätsklinikum, Ulm, Germany
| | - Anita Ignatius
- Institut für Unfallchirurgische Forschung und Biomechanik, Universitätsklinikum, Ulm, Germany
| | - Birgit Jung
- Abteilung Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
| | - Matthias Duechs
- Abteilung Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
| | - Peter Möller
- Institut für Pathologie, Universitätsklinikum, Ulm, Germany
| | | | - Enrico Calzia
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Ulm, Germany
| | - Peter Radermacher
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Ulm, Germany
- * E-mail:
| | - Florian Wagner
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Ulm, Germany
- Klinik für Anästhesiologie, Universitätsklinikum, Ulm, Germany
| |
Collapse
|
29
|
Sordi R, Chiazza F, Johnson FL, Patel NSA, Brohi K, Collino M, Thiemermann C. Inhibition of IκB Kinase Attenuates the Organ Injury and Dysfunction Associated with Hemorrhagic Shock. Mol Med 2015; 21:563-75. [PMID: 26101953 DOI: 10.2119/molmed.2015.00049] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/16/2015] [Indexed: 12/21/2022] Open
Abstract
Nuclear factor-kappa B (NF-κB) activation is widely implicated in multiple organ failure (MOF); however, a direct inhibitor of IκB kinase (IKK), which plays a pivotal role in the activation of NF-κB, has not been investigated in shock. Thus, the aim of the present work was to investigate the effects of an IKK inhibitor on the MOF associated with hemorrhagic shock (HS). Therefore, rats were subjected to HS and were resuscitated with the shed blood. Rats were treated with the inhibitor of IKK or vehicle at resuscitation. Four hours later, blood and organs were assessed for organ injury and signaling events involved in the activation of NF-κB. Additionally, survival following serum deprivation was assessed in HK-2 cells treated with the inhibitor of IKK. HS resulted in renal dysfunction, lung, liver and muscular injury, and increases in serum inflammatory cytokines. Kidney and liver tissue from HS rats revealed increases in phosphorylation of IKKαβ and IκBα, nuclear translocation of NF-κB and expression of inducible isoform of nitric oxide synthase (iNOS). IKK16 treatment upon resuscitation attenuated NF-κB activation and activated the Akt survival pathway, leading to a significant attenuation of all of the above parameters. Furthermore, IKK16 exhibited cytoprotective effects in human kidney cells. In conclusion, the inhibitor of IKK complex attenuated the MOF associated with HS. This effect may be due to the inhibition of the NF-κB pathway and activation of the survival kinase Akt. Thus, the inhibition of the IKK complex might be an effective strategy for the prevention of MOF associated with HS.
Collapse
Affiliation(s)
- Regina Sordi
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom.,Capes Foundation, Ministry of Education of Brazil, Brasilia, DF, Brazil
| | - Fausto Chiazza
- University of Turin, Department of Drug Science and Technology, Turin, Italy
| | - Florence L Johnson
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Nimesh S A Patel
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Karim Brohi
- Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Massimo Collino
- University of Turin, Department of Drug Science and Technology, Turin, Italy
| | - Christoph Thiemermann
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
30
|
Sordi R, Chiazza F, Patel NSA, Doyle RA, Collino M, Thiemermann C. 'Preconditioning' with low dose lipopolysaccharide aggravates the organ injury / dysfunction caused by hemorrhagic shock in rats. PLoS One 2015; 10:e0122096. [PMID: 25830444 PMCID: PMC4382161 DOI: 10.1371/journal.pone.0122096] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 02/17/2015] [Indexed: 01/08/2023] Open
Abstract
Methods Male rats were ‘pretreated’ with phosphate-buffered saline (PBS; i.p.) or LPS (1 mg/kg; i.p.) 24 h prior to HS. Mean arterial pressure (MAP) was maintained at 30 ± 2 mmHg for 90 min or until 25% of the shed blood had to be re-injected to sustain MAP. This was followed by resuscitation with the remaining shed blood. Four hours after resuscitation, parameters of organ dysfunction and systemic inflammation were assessed. Results HS resulted in renal dysfunction, and liver and muscular injury. At a first glance, LPS preconditioning attenuated organ dysfunction. However, we discovered that HS-rats that had been preconditioned with LPS (a) were not able to sustain a MAP at 30 mmHg for more than 50 min and (b) the volume of blood withdrawn in these animals was significantly less than in the PBS-control group. This effect was associated with an enhanced formation of the nitric oxide (NO) derived from inducible NO synthase (iNOS). Thus, a further control group in which all animals were resuscitated after 50 min of hemorrhage was performed. Then, LPS preconditioning aggravated both circulatory failure and organ dysfunction. Most notably, HS-rats pretreated with LPS exhibited a dramatic increase in NF-κB activation and pro-inflammatory cytokines. Conclusion In conclusion, LPS preconditioning predisposed animals to an earlier vascular decompensation, which may be mediated by an excess of NO production secondary to induction of iNOS and activation of NF-κB. Moreover, LPS preconditioning increased the formation of pro-inflammatory cytokines, which is likely to have contributed to the observed aggravation of organ injury/dysfunction caused by HS.
Collapse
Affiliation(s)
- Regina Sordi
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
- Capes Foundation, Ministry of Education of Brazil, Brasilia/DF, Brazil
| | - Fausto Chiazza
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Nimesh S. A. Patel
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Rachel A. Doyle
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
| | - Massimo Collino
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Christoph Thiemermann
- The William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, United Kingdom
- * E-mail:
| |
Collapse
|
31
|
Pérez S, Pereda J, Sabater L, Sastre J. Pancreatic ascites hemoglobin contributes to the systemic response in acute pancreatitis. Free Radic Biol Med 2015; 81:145-55. [PMID: 25157787 DOI: 10.1016/j.freeradbiomed.2014.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/31/2014] [Accepted: 08/03/2014] [Indexed: 02/08/2023]
Abstract
Upon hemolysis extracellular hemoglobin causes oxidative stress and cytotoxicity due to its peroxidase activity. Extracellular hemoglobin may release free hemin, which increases vascular permeability, leukocyte recruitment, and adhesion molecule expression. Pancreatitis-associated ascitic fluid is reddish and may contain extracellular hemoglobin. Our aim has been to determine the role of extracellular hemoglobin in the local and systemic inflammatory response during severe acute pancreatitis in rats. To this end we studied taurocholate-induced necrotizing pancreatitis in rats. First, extracellular hemoglobin in ascites and plasma was quantified and the hemolytic action of ascitic fluid was tested. Second, we assessed whether peritoneal lavage prevented the increase in extracellular hemoglobin in plasma during pancreatitis. Third, hemoglobin was purified from rat erythrocytes and administered intraperitoneally to assess the local and systemic effects of ascitic-associated extracellular hemoglobin during acute pancreatitis. Extracellular hemoglobin and hemin levels markedly increased in ascitic fluid and plasma during necrotizing pancreatitis. Peroxidase activity was very high in ascites. The peritoneal lavage abrogated the increase in extracellular hemoglobin in plasma. The administration of extracellular hemoglobin enhanced ascites; dramatically increased abdominal fat necrosis; upregulated tumor necrosis factor-α, interleukin-1β, and interleukin-6 gene expression; and decreased expression of interleukin-10 in abdominal adipose tissue during pancreatitis. Extracellular hemoglobin enhanced the gene expression and protein levels of vascular endothelial growth factor (VEGF) and other hypoxia-inducible factor-related genes in the lung. Extracellular hemoglobin also increased myeloperoxidase activity in the lung. In conclusion, extracellular hemoglobin contributes to the inflammatory response in severe acute pancreatitis through abdominal fat necrosis and inflammation and by increasing VEGF and leukocyte infiltration into the lung.
Collapse
Affiliation(s)
- Salvador Pérez
- Department of Physiology, School of Pharmacy, University of Valencia, 46100 Burjasot, Valencia, Spain
| | - Javier Pereda
- Department of Physiology, School of Pharmacy, University of Valencia, 46100 Burjasot, Valencia, Spain
| | - Luis Sabater
- Department of Surgery, University of Valencia, University Clinic Hospital, 46010 Valencia, Spain
| | - Juan Sastre
- Department of Physiology, School of Pharmacy, University of Valencia, 46100 Burjasot, Valencia, Spain.
| |
Collapse
|
32
|
Suresh MV, Ramakrishnan SK, Thomas B, Machado-Aranda D, Bi Y, Talarico N, Anderson E, Yatrik SM, Raghavendran K. Activation of hypoxia-inducible factor-1α in type 2 alveolar epithelial cell is a major driver of acute inflammation following lung contusion. Crit Care Med 2014; 42:e642-53. [PMID: 25014067 DOI: 10.1097/ccm.0000000000000488] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Lung contusion is a major risk factor for the development of acute respiratory distress syndrome. Hypoxia-inducible factor-1α is the primary transcription factor that is responsible for regulating the cellular response to changes in oxygen tension. We set to determine if hypoxia-inducible factor-1α plays a role in the pathogenesis of acute inflammatory response and injury in lung contusion. DESIGN Nonlethal closed-chest unilateral lung contusion was induced in a hypoxia reporter mouse model and type 2 cell-specific hypoxia-inducible factor-1α conditional knockout mice. The mice were killed at 5-, 24-, 48-, and 72-hour time points, and the extent of systemic and tissue hypoxia was assessed. In addition, injury and inflammation were assessed by measuring bronchoalveolar lavage cells (flow cytometry and cytospin), albumin (permeability injury), and cytokines (inflammation). Isolated type 2 cells from the hypoxia-inducible factor-1α conditional knockout mice were isolated and evaluated for proinflammatory cytokines following lung contusion. Finally, the role of nuclear factor-κB and interleukin-1β as intermediates in this interaction was studied. RESULTS Lung contusion induced profound global hypoxia rapidly. Increased expression of hypoxia-inducible factor-1α from lung samples was observed as early as 60 minutes, following the insult. The extent of lung injury following lung contusion was significantly reduced in conditional knockout mice at all the time points, when compared with the wild-type littermate mice. Release of proinflammatory cytokines, such as interleukin-1β, interleukin-6, macrophage inflammatory protein-2, and keratinocyte chemoattractant, was significantly lower in conditional knockout mice. These actions are in part mediated through nuclear factor-κB. Hypoxia-inducible factor-1α in lung epithelial cells was shown to regulate interleukin-1β promoter activity. CONCLUSION Activation of hypoxia-inducible factor-1α in type 2 cell is a major driver of acute inflammation following lung contusion.
Collapse
Affiliation(s)
- Madathilparambil V Suresh
- 1Department of Surgery, University of Michigan Medical School, Ann Arbor, MI. 2Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI. 3Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Critical Role of Toll-Like Receptor 4 in Hypoxia-Inducible Factor 1α Activation During Trauma/Hemorrhagic Shock–Induced Acute Lung Injury After Lymph Infusion in Mice. Shock 2014; 42:271-8. [DOI: 10.1097/shk.0000000000000212] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
34
|
Liu C, Zhang X, Zhou JX, Wei W, Liu DH, Ke P, Zhang GF, Cai GJ, Su DF. The protective action of ketanserin against lipopolysaccharide-induced shock in mice is mediated by inhibiting inducible NO synthase expression via the MEK/ERK pathway. Free Radic Biol Med 2013; 65:658-666. [PMID: 23954471 DOI: 10.1016/j.freeradbiomed.2013.07.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 07/26/2013] [Accepted: 07/31/2013] [Indexed: 12/20/2022]
Abstract
Nitric oxide (NO) plays an important role in the pathogenesis of endotoxic shock. This work tested the hypothesis that ketanserin could attenuate endotoxic shock by inhibiting the expression of inducible NO synthase (iNOS). The results demonstrated that ketanserin could inhibit iNOS expression in the heart, lungs, liver, and kidneys and nitrate production in the serum upon endotoxic shock in mice. In RAW264.7 cells, ketanserin significantly inhibited the expression of iNOS and decreased the production of NO, TNFα, IL-6, and reactive oxygen species upon lipopolysaccharide (LPS) challenge. Ketanserin also increased the level of ATP and mitochondrial membrane potential in RAW264.7 cells upon LPS exposure. LPS-induced iNOS expression was inhibited by the 5-HT2A receptor antagonist ritanserin and not the α1 receptor antagonist prazosin. Knockdown of 5-HT2A receptor by siRNA abolished the inhibitory effect of ketanserin on the expression of iNOS. These results indicated that the inhibitory effect of ketanserin on the expression of iNOS is mediated by blocking the 5-HT2A receptor. Furthermore, ketanserin significantly inhibited the activation of ERK1/2 and NF-κB signal. Pretreatment with PD184352, a specific inhibitor of ERK1/2, blocked the inhibitory effect of ketanserin on the expression of iNOS and NO production, indicating a critical role for the MEK/ERK1/2 signaling pathway. Collectively, our findings indicate that inhibition of the expression of iNOS via the MEK/ERK pathway mediates the protective effects of ketanserin against LPS-induced shock in mice.
Collapse
Affiliation(s)
- Chong Liu
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China.
| | - Xin Zhang
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China
| | - Jv-Xiang Zhou
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China
| | - Wei Wei
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China
| | - Dian-Hua Liu
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China
| | - Ping Ke
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China
| | - Gu-Fang Zhang
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China
| | - Guo-Jun Cai
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China.
| | - Ding-Feng Su
- Department of Pharmacology, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
35
|
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: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 09/29/2013] [Indexed: 01/06/2023]
|
36
|
Yip HK, Chang YC, Wallace CG, Chang LT, Tsai TH, Chen YL, Chang HW, Leu S, Zhen YY, Tsai CY, Yeh KH, Sun CK, Yen CH. Melatonin treatment improves adipose-derived mesenchymal stem cell therapy for acute lung ischemia-reperfusion injury. J Pineal Res 2013; 54:207-21. [PMID: 23110436 DOI: 10.1111/jpi.12020] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Accepted: 09/21/2012] [Indexed: 12/21/2022]
Abstract
This study investigated whether melatonin-treated adipose-derived mesenchymal stem cells (ADMSC) offered superior protection against acute lung ischemia-reperfusion (IR) injury. Adult male Sprague-Dawley rats (n = 30) were randomized equally into five groups: sham controls, lung IR-saline, lung IR-melatonin, lung IR-melatonin-normal ADMSC, and lung IR-melatonin-apoptotic ADMSC. Arterial oxygen saturation was lowest in lung IR-saline; lower in lung IR-melatonin than sham controls, lung IR-melatonin-normal ADMSC, and lung IR-melatonin-apoptotic ADMSC; lower in lung IR-melatonin-normal ADMSC than sham controls and lung IR-melatonin-apoptotic ADMSC; lower in lung IR-melatonin-apoptotic ADMSC than sham controls (P < 0.0001 in each case). Right ventricular systolic blood pressure (RVSBP) showed a reversed pattern among all groups (all P < 0.0001). Changes in histological scoring of lung parenchymal damage and CD68+ cells showed a similar pattern compared with RVSBP in all groups (all P < 0.001). Changes in inflammatory protein expressions such as VCAM-1, ICAM-1, oxidative stress, TNF-α, NF-κB, PDGF, and angiotensin II receptor, and changes in apoptotic protein expressions of cleaved caspase 3 and PARP, and mitochondrial Bax, displayed identical patterns compared with RVSBP in all groups (all P < 0.001). Numbers of antioxidant (GR+, GPx+, NQO-1+) and endothelial cell biomarkers (CD31+ and vWF+) were lower in sham controls, lung IR-saline, and lung IR-melatonin than lung IR-melatonin-normal ADMSC and lung IR-melatonin-apoptotic ADMSC, and lower in lung IR-melatonin-normal ADMSC than lung IR-melatonin-apoptotic ADMSC (P < 0.001 in each case). In conclusion, when the animals were treated with melatonin, the apoptotic ADMSC were superior to normal ADMSC for protection of lung from acute IR injury.
Collapse
Affiliation(s)
- Hon-Kan Yip
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|