101
|
Dexmedetomidine Ameliorates Lung Injury Induced by Intestinal Ischemia/Reperfusion by Upregulating Cannabinoid Receptor 2, Followed by the Activation of the Phosphatidylinositol 3-Kinase/Akt Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6120194. [PMID: 32655771 PMCID: PMC7327571 DOI: 10.1155/2020/6120194] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/27/2020] [Accepted: 05/05/2020] [Indexed: 02/08/2023]
Abstract
Intestinal ischemia/reperfusion (I/R) is a clinical emergency, which often causes lung injury with high morbidity and mortality. Although dexmedetomidine has been identified to have a protective effect on lung injury caused by intestinal I/R, its specific mechanism is still elucidated. In recent years, the cannabinoid (CB2) receptor pathway has been found to be involved in I/R injury of some organs. In the current study, we investigated whether the CB2 receptor pathway contributes to the protective effect of dexmedetomidine on the intestinal I/R-induced lung injury in rats. Dexmedetomidine treatment upregulated the expression of CB2 receptor and suppressed the I/R-induced increases in lung injury scores, inflammatory cell infiltration, lung wet/dry ratio, MPO activity, MDA level, inflammatory cytokines, and caspase-3 expression while augmenting SOD activity and Bcl-2 expression, indicating attenuation of lung injury. Dexmedetomidine treatment also increased the expression of Akt. The protective effects of dexmedetomidine treatment were reversed by the CB2 receptor antagonist AM630 or the PI3K inhibitor wortmannin. And the CB2 receptor antagonist AM630 also downregulated the expression of Akt. Thus, our findings suggest that treatment with dexmedetomidine provides a protective role against lung injury caused by intestinal I/R in rats, possibly due to the upregulation of the CB2 receptor, followed by the activation of the PI3K/Akt pathway.
Collapse
|
102
|
Dorrello NV, Vunjak-Novakovic G. Bioengineering of Pulmonary Epithelium With Preservation of the Vascular Niche. Front Bioeng Biotechnol 2020; 8:269. [PMID: 32351946 PMCID: PMC7174601 DOI: 10.3389/fbioe.2020.00269] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 03/16/2020] [Indexed: 12/20/2022] Open
Abstract
The shortage of transplantable donor organs directly affects patients with end-stage lung disease, for which transplantation remains the only definitive treatment. With the current acceptance rate of donor lungs of only 20%, rescuing even one half of the rejected donor lungs would increase the number of transplantable lungs threefold, to 60%. We review recent advances in lung bioengineering that have potential to repair the epithelial and vascular compartments of the lung. Our focus is on the long-term support and recovery of the lung ex vivo, and the replacement of defective epithelium with healthy therapeutic cells. To this end, we first review the roles of the lung epithelium and vasculature, with focus on the alveolar-capillary membrane, and then discuss the available and emerging technologies for ex vivo bioengineering of the lung by decellularization and recellularization. While there have been many meritorious advances in these technologies for recovering marginal quality lungs to the levels needed to meet the standards for transplantation – many challenges remain, motivating further studies of the extended ex vivo support and interventions in the lung. We propose that the repair of injured epithelium with preservation of quiescent vasculature will be critical for the immediate blood supply to the lung and the lung survival and function following transplantation.
Collapse
Affiliation(s)
- N Valerio Dorrello
- Department of Pediatrics, Columbia University, New York, NY, United States
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY, United States.,Department of Medicine, Columbia University, New York, NY, United States
| |
Collapse
|
103
|
Ginsenoside Rh3 activates Nrf2 signaling and protects endometrial cells from oxygen and glucose deprivation-reoxygenation. Aging (Albany NY) 2020; 12:6109-6119. [PMID: 32259797 PMCID: PMC7185134 DOI: 10.18632/aging.103009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 02/05/2020] [Indexed: 01/08/2023]
Abstract
Oxygen and glucose deprivation (OGD)-reoxygenation (OGDR) induces oxidative injury to endometrial cells in vitro. We tested the potential effect of ginsenoside Rh3 (GRh3) in the process. Our results show that GRh3 activated Nrf2 signaling in T-HESC cells and primary murine endometrial cells. GRh3 induced Nrf2 Ser-40 phosphorylation and Keap1-Nrf2 disassociation, causing Nrf2 protein stabilization and nuclear translocation, which led to transcription and expression of antioxidant response element-dependent genes (HO1, NQO1 and GCLC). In T-HESC cells and primary murine endometrial cells, GRh3 potently attenuated OGDR-induced reactive oxygen species production, lipid peroxidation and mitochondrial depolarization, as well as cell viability reduction and necrosis. Activation of Nrf2 is required for GRh3-induced anti-OGDR actions in endometrial cells. Nrf2 inhibition, by Nrf2 shRNA, knockout (through CRISPR-Cas9-editing) or S40T mutation, abolished GRh3-induced endometrial cell protection against OGDR. Additionally, forced activation of Nrf2, by Keap1 knockout, mimicked and nullified GRh3-induced anti-OGDR actions in T-HESC cells. Together, we conclude that GRh3 protects endometrial cells from OGDR via activation of Nrf2 signaling.
Collapse
|
104
|
Huwae TECJ, Santoso ARB, Kesuma W, Sujuti H, Ratnawati R, Sukmajaya WP, Hidayat M. Reperfusion Interval as a Prevention of Lung Injury Due to Limb Ischemia-Reperfusion After Application of Tourniquet in Murine Experimental Study. Indian J Orthop 2020; 54:704-710. [PMID: 32850036 PMCID: PMC7429627 DOI: 10.1007/s43465-020-00100-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/26/2020] [Indexed: 02/04/2023]
Abstract
BACKGROUND Tourniquet use is prevalent in the orthopaedic field to achieve a bloodless operating field, but it poses risks of local and systemic complications, including lung injury. This study aims to examine the effect of tourniquet application on the hindlimb of a rat to its lung. MATERIALS AND METHODS This is an experimental study with 48 male Wistar strain rats as samples. The rats were divided into group A (n = 24), killed directly after fracturization and tourniquet application, and group B (n = 24), killed 14 days post-procedure. Each group was divided into four: group A1/B1 (control group, three hours tourniquet application without reperfusion interval), A2/B2 (5-min reperfusion between 2-h and 1-h tourniquet application), A3/B3 (10-min reperfusion), and A4/B4 (15-min reperfusion). The lung tissue was examined histologically within ten high-power fields (400 × magnification). The severity of lung injury was measured using the Lung Injury Score (LIS). The oxidative damage was measured by determining the malondialdehyde (MDA) level, using the TBARS (thiobarbituric acid reactive substance assay) method. RESULTS There was a dose-dependent decrease of LIS and MDA in groups A and B with increasing reperfusion interval. Fifteen-minute reperfusion interval caused a 54.55% and 45.33% LIS reduction in groups A and B, respectively. All pair-wise group comparisons (p < 0.05) showed significant differences. Five-minute interval reduced the MDA level by 16.56% and 30.13% in groups A and B, respectively. All possible pair-wise comparisons in both groups A and B also showed a significant difference (p < 0.05). CONCLUSIONS Reperfusion interval is a possible clinical approach to mitigate the remote organ damage induced by limb ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Thomas Erwin Christian Junus Huwae
- Faculty of Medicine, Universitas Brawijaya, Jl. Veteran, Malang, 65145 Indonesia ,Department of Orthopaedics and Traumatology, Saiful Anwar General Hospital, Malang, Indonesia
| | | | - Wongso Kesuma
- Department of Orthopaedics and Traumatology, Saiful Anwar General Hospital, Malang, Indonesia
| | - Hidayat Sujuti
- Department of Ophthalmology, Saiful Anwar General Hospital, Malang, Indonesia
| | - Retty Ratnawati
- Department of Physiology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | | | - Mohammad Hidayat
- Department of Orthopaedics and Traumatology, Saiful Anwar General Hospital, Malang, Indonesia
| |
Collapse
|
105
|
Malçok ÜA, Aras AB, Şehitoğlu MH, Akman T, Yüksel Y. Therapeutic effects of syringaldehyde on spinal cord ischemia in rabbits. Saudi Med J 2020; 41:341-350. [PMID: 32291420 PMCID: PMC7841612 DOI: 10.15537/smj.2020.4.24993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 02/04/2020] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVES To investigate the effects of syringaldehyde (SA) on the antioxidant and oxidant system in spinal cord ischemia (SCI). METHODS These study and experiments were conducted at Medical Research Center, Çanakkale Onsekiz Mart University, Çanakkale, Turkey, between 2014-2018. Eighteen New Zealand White adult male rabbits were randomly divided into 3 groups (n=6). Superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), myeloperoxidase (MPO) activities, and malondialdehyde (MDA) levels were measured in the spinal cord tissues. Degenerated neurons, hemorrhage and in ammatory cell migration in the spinal cord were investigated histopathologically. Expressions of neuronal nitric oxide synthase (nNOS), caspase-3, and nuclear factor-κB (NF-κB) were evaluated immunohistochemically. Clinically, it was evaluated with modified Tarlov score. RESULTS Biochemically, there was an expected decrease in SOD, CAT, and GPx enzyme activities in ischemia groups, there was also an increase in MPO activity at the same time. When the enzyme activities spinal cord ischemia/ reperfusion (SCI/R)+SA, control and SCI/R groups were compared, the difference was found to be statistically significant (p less than 0.05). Glutathione peroxidase enzyme activity levels were very low in ischemia group compared to the significant increase in the SA group (p less than 0.05). Histopathologically, when SCI/R and SCI/R+SA groups were compared, there were statistically significant differences in the number of degenerative neurons and amount of hemorrhage; this comparison shows the significance of treatment in terms of inflammatory cell migration (p less than 0.05). The expressions of nNOS, caspase-3, and NF-κB were found significantly increased in SCI/R group compared to the control group (p less than 0.05). Syringaldehyde treatment decreased nNOS, caspase-3, and NF-κB expressions immunohistochemically. Clinical evaluation showed improvement in the SA-treated group. CONCLUSION Syringaldehyde therapy administered for protective purposes may reduce oxidative stress, degenerative changes and in ammatory cell migration in the ischemic spinal cord.Saudi Med J 2020; Vol. 41 (4): 341-350doi: 10.15537/smj.2020.4.24993 How to cite this article:Malçok UA, Aras AB, Şehitoğlu MH, Akman T, Yüksel Y. Therapeutic effects of syringaldehyde on spinal cord ischemia in rabbits. Saudi Med J 2020; Vol. 41: 341-350. doi: 10.15537/smj.2020.4.24993.
Collapse
Affiliation(s)
- Ümit A Malçok
- Department of Neurosurgery, School of Medicine, Çanakkale Onsekiz Mart University, Çanakkale, Turkey. E-mail.
| | | | | | | | | |
Collapse
|
106
|
Edaravone attenuates lung injury in a hind limb ischemia-reperfusion rat model: A histological, immunohistochemical and biochemical study. Ann Anat 2020; 228:151433. [DOI: 10.1016/j.aanat.2019.151433] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/29/2019] [Accepted: 10/07/2019] [Indexed: 11/20/2022]
|
107
|
Keap1-targeting microRNA-941 protects endometrial cells from oxygen and glucose deprivation-re-oxygenation via activation of Nrf2 signaling. Cell Commun Signal 2020; 18:32. [PMID: 32102665 PMCID: PMC7045607 DOI: 10.1186/s12964-020-0526-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/29/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Mimicking ischemia-reperfusion injury, oxygen and glucose deprivation (OGD)-re-oxygenation (OGDR) applied to endometrial cells produces significant oxidative stress and programmed necrosis, which can be inhibited by nuclear-factor-E2-related factor 2 (Nrf2) signaling. MicroRNA (miRNA)-induced repression of Keap1, a Nrf2 suppressor protein that facilitates Nrf2 degradation, is novel strategy to activate Nrf2 cascade. METHODS MicroRNA-941 (miR-941) was exogenously expressed in HESC and primary human endometrial cells, and the Nrf2 pathway examined by Western blotting and real-time quantitative PCR analysis. The endometrial cells were treated with OGDR, cell programmed necrosis and apoptosis were tested. RESULTS MiR-941 is a novel Keap1-targeting miRNA that regulates Nrf2 activity. In T-HESC cells and primary human endometrial cells, ectopic overexpression of miR-941 suppressed Keap1 3'-UTR (untranslated region) expression and downregulated its mRNA/protein expression, leading to activation of the Nrf2 cascade. Conversely, inhibition of miR-941 elevated Keap1 expression and activity in endometrial cells, resulting in suppression of Nrf2 activation. MiR-941 overexpression in endometrial cells attenuated OGDR-induced oxidative stress and programmed necrosis, whereas miR-941 inhibition enhanced oxidative stress and programmed necrosis. MiR-941 overexpression and inhibition were completely ineffective in Keap1-/Nrf2-KO T-HESC cells (using CRISPR/Cas9 strategy). Restoring Keap1 expression, using an UTR-depleted Keap1 construct, abolished miR-941-induced anti-OGDR activity in T-HESC cells. Thus Keap1-Nrf2 cascade activation is required for miR-941-induced endometrial cell protection. CONCLUSIONS Targeting Keap1 by miR-941 activates Nrf2 cascade to protect human endometrial cells from OGDR-induced oxidative stress and programmed necrosis. Video Abstract.
Collapse
|
108
|
Wang F, Wang F, Li F, Wang D, Li H, He X, Zhang J. Methane attenuates lung ischemia-reperfusion injury via regulating PI3K-AKT-NFκB signaling pathway. J Recept Signal Transduct Res 2020; 40:209-217. [PMID: 32079441 DOI: 10.1080/10799893.2020.1727925] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objective: This study aims to investigate the protective effects and possible mechanism of methane-rich saline (MS) on lung ischemia-reperfusion injury (LIRI) in rats.Methods: MS (2 ml/kg and 20 ml/kg) was injected intraperitoneally in rats after LIRI. Lung injury was assayed by Hematoxylin-eosin (HE) staining and wet-to-dry weight (W/D). The cells in the bronchoalveolar lavage fluid (BALF) and blood were counted. Oxidative stress was examined by the level of malondialdehyde (MDA) and superoxide dismutase (SOD). Inflammatory factors including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-10 (IL-10) were determined by ELISA. Lung tissue apoptosis was detected by TUNEL staining and western blotting of Bcl-2, Bax, and caspase-3. The expressions of IкBα, p38, PI3K, AKT, and NF-κB were analyzed with Western blotting.Results: MS effectively decreased the lung W/D ratio as well as the lung pathological damage and reduced the localized infiltration of inflammatory cells. Methane suppressed the expression of the PI3K-AKT-NFκB signaling pathway during the lung IR injury, which inhibited the activation of NF-kB and decreased the level of inflammatory cytokines, such as TNF-α, IL-1β, and IL-10. Moreover, we found that MS treatment relieved reactive oxygen species (ROS) damage by downregulating MDA and upregulating SOD. MS treatment also regulated apoptosis-related proteins, such as Bcl-2, Bax, and caspase-3.Conclusions: MS could repair LIRI and reduce the release of oxidative stress, inflammatory cytokines, and cell apoptosis via the PI3K-AKT-NFκB signaling pathway, which may provide a novel and promising strategy for the treatment of LIRI.
Collapse
Affiliation(s)
- Fang Wang
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Feidi Wang
- Hou Zonglian Medical Experimental Class, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fengtao Li
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Dong Wang
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Haopeng Li
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xijing He
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jingyao Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
109
|
Kumar A, Noda K, Philips B, Velayutham M, Stolz DB, Gladwin MT, Shiva S, D'Cunha J. Nitrite attenuates mitochondrial impairment and vascular permeability induced by ischemia-reperfusion injury in the lung. Am J Physiol Lung Cell Mol Physiol 2020; 318:L580-L591. [PMID: 32073901 DOI: 10.1152/ajplung.00367.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Primary graft dysfunction (PGD) is directly related to ischemia-reperfusion (I/R) injury and a major obstacle in lung transplantation (LTx). Nitrite (NO2-), which is reduced in vivo to form nitric oxide (NO), has recently emerged as an intrinsic signaling molecule with a prominent role in cytoprotection against I/R injury. Using a murine model, we provide the evidence that nitrite mitigated I/R-induced injury by diminishing infiltration of immune cells in the alveolar space, reducing pulmonary edema, and improving pulmonary function. Ultrastructural studies support severe mitochondrial impairment in the lung undergoing I/R injury, which was significantly protected by nitrite treatment. Nitrite also abrogated the increased pulmonary vascular permeability caused by I/R. In vitro, hypoxia-reoxygenation (H/R) exacerbated cell death in lung epithelial and microvascular endothelial cells. This contributed to mitochondrial dysfunction as characterized by diminished complex I activity and mitochondrial membrane potential but increased mitochondrial reactive oxygen species (mtROS). Pretreatment of cells with nitrite robustly attenuated mtROS production through modulation of complex I activity. These findings illustrate a potential novel mechanism in which nitrite protects the lung against I/R injury by regulating mitochondrial bioenergetics and vascular permeability.
Collapse
Affiliation(s)
- Ajay Kumar
- Division of Lung Transplantation and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kentaro Noda
- Division of Lung Transplantation and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brian Philips
- Division of Lung Transplantation and Lung Failure, Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Murugesan Velayutham
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania.,Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Donna B Stolz
- Center for Biological Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sruti Shiva
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jonathan D'Cunha
- Department of Cardiothoracic Surgery, Mayo Clinic Arizona, Phoenix, Arizona
| |
Collapse
|
110
|
Luo C, Xie X, Feng X, Lei B, Fang C, Li Y, Cai X, Ling G, Zheng B. Deficiency of Interleukin-36 Receptor Protected Cardiomyocytes from Ischemia-Reperfusion Injury in Cardiopulmonary Bypass. Med Sci Monit 2020; 26:e918933. [PMID: 32048631 PMCID: PMC7034403 DOI: 10.12659/msm.918933] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Interleukin-36 has been demonstrated to be involved in inflammatory responses. Inflammatory responses due to ischemia-reperfusion injury following cardiopulmonary bypass (CPB) can cause heart dysfunction or damage. Material/Methods The CPB models were constructed in IL-36R−/−, IL-36RN−/−, and wild-type SD rats. Ultrasonic cardiography and ELISA were used to evaluate the cardiac function and measuring myocardial biomarker levels in different groups. TUNEL assay was used to evaluate apoptosis. Western blot assays and RT-PCR were performed to measure the expression of chemokines and secondary inflammatory cytokines in the heart. Oxidative stress in tissue and cultured cells was assessed using a DCFH-DA fluorescence probe and quantification of superoxide dismutase activity. Results Improved systolic function and decreased serum levels of myocardial damage biomarkers were found in IL-36R−/− rats compared to WT rats, while worse cardiac function and cardiomyocyte IR injury were observed in IL-36RN−/− rats compared to WT rats. TUNEL staining and Western blot analyses found that cardiomyocyte apoptosis and inflammation were significantly lower in the hearts of IL-36R−/− rats compared with that of WT rats. Oxidative stress was significantly lower in IL-36R−/− rats compared to WT rats. iNOS expression was significantly reduced, while eNOS expression was increased in the hearts of IL-36R−/− rats. Silencing of IL-36R expression in vitro activated SIRT1/FOXO1/p53 signaling in cardiomyocytes. Conclusions IL-36R deficiency in cardiomyocytes repressed infiltration of bone marrow-derived inflammatory cells and oxidative stress dependent on SIRT1-FOXO1 signaling, thus protecting cardiomyocytes and improving cardiac function in CPB model rats.
Collapse
Affiliation(s)
- Cheng Luo
- The First Affiliated Hospital of Guangxi Medical University, Cardiac Surgery Ward, Nanning, Guangxi, China (mainland)
| | - Xiaoyong Xie
- The First Affiliated Hospital of Guangxi Medical University, Cardiac Surgery Ward, Nanning, Guangxi, China (mainland)
| | - Xu Feng
- The First Affiliated Hospital of Guangxi Medical University, Cardiac Surgery Ward, Nanning, Guangxi, China (mainland)
| | - Binfeng Lei
- The First Affiliated Hospital of Guangxi Medical University, Cardiac Surgery Ward, Nanning, Guangxi, China (mainland)
| | - Chen Fang
- The First Affiliated Hospital of Guangxi Medical University, Cardiac Surgery Ward, Nanning, Guangxi, China (mainland)
| | - Yugui Li
- The First Affiliated Hospital of Guangxi Medical University, Cardiac Surgery Ward, Nanning, Guangxi, China (mainland)
| | - Xiongwei Cai
- The First Affiliated Hospital of Guangxi Medical University, Cardiac Surgery Ward, Nanning, Guangxi, China (mainland)
| | - Guoxing Ling
- The First Affiliated Hospital of Guangxi Medical University, Cardiac Surgery Ward, Nanning, Guangxi, China (mainland)
| | - Baoshi Zheng
- The First Affiliated Hospital of Guangxi Medical University, Cardiac Surgery Ward, Nanning, Guangxi, China (mainland)
| |
Collapse
|
111
|
Xu HB, Zheng YF, Wu D, Li Y, Zhou LN, Chen YG. microRNA-1203 targets and silences cyclophilin D to protect human endometrial cells from oxygen and glucose deprivation-re-oxygenation. Aging (Albany NY) 2020; 12:3010-3024. [PMID: 32041924 PMCID: PMC7041737 DOI: 10.18632/aging.102795] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/12/2020] [Indexed: 02/06/2023]
Abstract
Oxygen and glucose deprivation (OGD)-re-oxygenation (OGDR) stimulation to the human endometrial cells mimics ischemia-reperfusion injury. Cyclophilin D (CypD)-dependent programmed necrosis pathway mediates OGDR-induced cytotoxicity to human endometrial cells. We here identified a novel CypD-targeting miRNA, microRNA-1203 (miR-1203). In T-HESC and primary human endometrial cells, ectopic overexpression of miR-1203, using a lentiviral construct, potently downregulated the CypD 3’-untranslated region (3’-UTR) activity and its expression. Both were however upregulated in endometrial cells with forced miR-1203 inhibition by its anti-sense sequence. Functional studies demonstrated that ectopic miR-1203 overexpression in endometrial cells alleviated OGDR-induced programmed necrosis, inhibiting mitochondrial CypD-p53-adenine nucleotide translocator 1 association, mitochondrial depolarization, reactive oxygen species production, and medium lactate dehydrogenase release. Contrarily OGDR-induced programmed necrosis and cytotoxicity were intensified with forced miR-1203 inhibition in endometrial cells. Significantly, ectopic miR-1203 overexpression or inhibition failed to change OGDR-induced cytotoxicity in CypD-knockout T-HESC cells. Furthermore, ectopic miR-1203 overexpression was unable to protect T-HESC endometrial cells from OGDR when CypD was restored by an UTR-depleted CypD construct. Collectively, these results show that miR-1203 targets and silences CypD to protect human endometrial cells from OGDR
Collapse
Affiliation(s)
- Hong-Bin Xu
- Obstetrics and Gynecology Department, The First Affiliated Hospital of Soochow University, Suzhou, China.,Obstetrics and Gynecology Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yu-Fan Zheng
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Di Wu
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Ya Li
- The Central Lab, North District, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Li-Na Zhou
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, China
| | - You-Guo Chen
- Obstetrics and Gynecology Department, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
112
|
Huang W, Xiong Y, Chen Y, Cheng Y, Wang R. NOX2 is involved in CB2-mediated protection against lung ischemia-reperfusion injury in mice. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:277-285. [PMID: 32211110 PMCID: PMC7061802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 01/19/2020] [Indexed: 06/10/2023]
Abstract
Lung ischemia-reperfusion injury (LIRI) can occur in many clinical scenarios. Activation of the cannabinoid 2 (CB2) receptor limits tissue injury in some ischemia-reperfusion (I/R) models. However, whether and how CB2 receptor activation alleviates lung injury induced by I/R remain unclear. In this study, we sought to determine whether JWH133, a selective CB2 receptor agonist, could alleviate lung injury induced by I/R and to examine the role of NOX2 in this process. Here, an I/R model was established using male C57BL/6 mice, by blocking the left pulmonary hilum for 1 h, followed by reperfusion for 2 h. Results showed that pretreatment with JWH133 significantly attenuated I/R-induced lung injury (decreased lung injury scores and wet-to-dry weight ratio and increased oxygenation index), alleviated oxidative stress (increased superoxide dismutase (SOD), and decreased Malondialdehyde (MDA) levels). It also significantly increased CB2 receptor mRNA expression and protein levels and significantly reduced NOX2 mRNA and protein expression. Further, the CB2 receptor antagonist AM630 eliminated these effects mediated by JWH133. Pretreatment with the NOX2 inhibitor, gp91 ds-tat, reduced NOX2 expression, but did not affect CB2 receptor expression and failed to alleviate lung injury and oxidative stress after additional JWH133 treatment. Our study suggests that CB2 receptor activation alleviates LIRI by inhibiting oxidative stress and that NOX2 is involved in CB2-mediated protection against LIRI in mice.
Collapse
Affiliation(s)
- Wei Huang
- Department of Anesthesiology, West China Second University Hospital of Sichuan UniversityChengdu 610041, Sichuan, P. R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital of Sichuan UniversityChengdu 610041, Sichuan, P. R. China
| | - Yaqin Xiong
- Department of Anesthesiology, West China Second University Hospital of Sichuan UniversityChengdu 610041, Sichuan, P. R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital of Sichuan UniversityChengdu 610041, Sichuan, P. R. China
| | - Yali Chen
- Department of Anesthesiology, West China Hospital of Sichuan UniversityChengdu 610041, Sichuan, P. R. China
| | - Yan Cheng
- Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan UniversityChengdu 610041, Sichuan, P. R. China
| | - Rurong Wang
- Department of Anesthesiology, West China Hospital of Sichuan UniversityChengdu 610041, Sichuan, P. R. China
| |
Collapse
|
113
|
Yang C, Yang W, He Z, He H, Yang X, Lu Y, Li H. Kaempferol Improves Lung Ischemia-Reperfusion Injury via Antiinflammation and Antioxidative Stress Regulated by SIRT1/HMGB1/NF-κB Axis. Front Pharmacol 2020; 10:1635. [PMID: 32116668 PMCID: PMC7025570 DOI: 10.3389/fphar.2019.01635] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/16/2019] [Indexed: 12/24/2022] Open
Abstract
Trauma, organ transplantation, and thromboembolism are the main causes of lung ischemia-reperfusion injury (LIRI), and new therapies and drugs are urgent to relieve LIRI. In preliminary experiment, authors found that kaempferol could improve LIRI in rats, and the current study further explored its possible mechanism. The model of rat LIRI was established and appropriate research methods were implemented. Results shown that kaempferol could significantly improve LIRI, inhibit release of inflammatory factors including interleukin (IL) 6 and tumor necrosis factor (TNF) α in bronchoalveolar lavage fluid, and reduce oxidative stress reaction. Western blotting was used to detect protein expression levels and found that kaempferol could up-regulate the protein expressions of phosphorylated (p-) p65 and p65, and down-regulate the protein expression of sirtuin (SIRT) 1. Immunofluorescence was used to localize the expression of high mobility group box (HMGB) 1 and found its higher expression in outside of nucleus. However, the above effects of kaempferol on LIRI markedly attenuated by EX 527, a selective inhibitor of SIRT 1. Taken together, we first reported the protective effect of kaempferol on rat LIRI and confirmed that kaempferol’s antiinflammation and antioxidative stress involving the SIRT1/HMGB1/NF-κB axis.
Collapse
Affiliation(s)
- Chunli Yang
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Wenkai Yang
- Department of Cardiovascular Surgery, Affiliated Central People's Hospital of Zhanjiang of Guangdong Medical University, Zhanjiang, China
| | - Zhaohui He
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Huiwei He
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Xiaogang Yang
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Yuanhua Lu
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Hongbo Li
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| |
Collapse
|
114
|
Giorgio M, Dellino GI, Gambino V, Roda N, Pelicci PG. On the epigenetic role of guanosine oxidation. Redox Biol 2020; 29:101398. [PMID: 31926624 PMCID: PMC6926346 DOI: 10.1016/j.redox.2019.101398] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/23/2019] [Accepted: 12/02/2019] [Indexed: 01/14/2023] Open
Abstract
Chemical modifications of DNA and RNA regulate genome functions or trigger mutagenesis resulting in aging or cancer. Oxidations of macromolecules, including DNA, are common reactions in biological systems and often part of regulatory circuits rather than accidental events. DNA alterations are particularly relevant since the unique role of nuclear and mitochondrial genome is coding enduring and inheritable information. Therefore, an alteration in DNA may represent a relevant problem given its transmission to daughter cells. At the same time, the regulation of gene expression allows cells to continuously adapt to the environmental changes that occur throughout the life of the organism to ultimately maintain cellular homeostasis. Here we review the multiple ways that lead to DNA oxidation and the regulation of mechanisms activated by cells to repair this damage. Moreover, we present the recent evidence suggesting that DNA damage caused by physiological metabolism acts as epigenetic signal for regulation of gene expression. In particular, the predisposition of guanine to oxidation might reflect an adaptation to improve the genome plasticity to redox changes.
Collapse
Affiliation(s)
- Marco Giorgio
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Via Adamello 16, 20139, Milano, Italy; Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padova, Italy.
| | - Gaetano Ivan Dellino
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Via Adamello 16, 20139, Milano, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Valentina Gambino
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Via Adamello 16, 20139, Milano, Italy
| | - Niccolo' Roda
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Via Adamello 16, 20139, Milano, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Via Adamello 16, 20139, Milano, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
115
|
Zeng J, Li X, Cheng Y, Ke B, Wang R. Activation of cannabinoid receptor type 2 reduces lung ischemia reperfusion injury through PI3K/Akt pathway. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:4096-4105. [PMID: 31933805 PMCID: PMC6949786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 10/24/2019] [Indexed: 06/10/2023]
Abstract
Cannabinoid receptor-2 activation plays a protective role against ischemic reperfusion injury (IRI) in various organs, and exerts a protective effect against paraquat-induced acute lung injury, while the role of CB2 in lung IRI remains unclear. Hence, the present study was designed to explore the role of CB2 in lung IRI, and whether the PI3K pathway was involved. C57BL/6 mice were subjected to lung ischemia by clamping the left hilum for 1 hour, followed by 2 hours' reperfusion. Mice were pretreated with vehicle, CB2 agonist JWH133, or antagonist AM630 followed by JWH133. Arterial blood and left lung tissues were collected to detect the PaO2/FiO2 ratio, lung wet-to-dry weight ratio, lung pathologic scoring, pro-inflammatory cytokines, MDA, and SOD. Secondly, mice were pretreated with vehicle, JWH133, or both PI3K-inhibitor LY294002 and JWH133. Arterial blood and left lung tissues were collected for the above studies and protein expression of CB2 receptor, p-AKT, and AKT. After mice were pretreated with JWH133, IR-induced lung edema and lung histopathologic changes were significantly attenuated. Pretreatment with JWH133 improved PaO2/FiO2 ratio, decreased lung TNF-α, IL-6, MDA levels and MPO activities, and increased SOD activity. By contrast, the protective effect of JWH133 was blocked by pretreatment with CB2 antagonist AM630. Similarly, pretreatment with PI3K-inhibitor weakened the protection induced by JWH133, and downregulated the expression of p-AKT without altering CB2 expression. The study suggested that activation of CB2 receptor plays a protective role against IR-induced lung injury through reducing inflammation in mice. The PI3K/Akt pathway might be involved in the protective effect of CB2 receptors in lung IRI.
Collapse
Affiliation(s)
- Jieting Zeng
- Department of Anesthesiology, Laboratory of Anesthesia and Intensive Care Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Xuehan Li
- Department of Anesthesiology, Laboratory of Anesthesia and Intensive Care Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Yan Cheng
- Department of Anesthesiology, Laboratory of Anesthesia and Intensive Care Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Bowen Ke
- Department of Anesthesiology, Laboratory of Anesthesia and Intensive Care Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Rurong Wang
- Department of Anesthesiology, Laboratory of Anesthesia and Intensive Care Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| |
Collapse
|
116
|
Evaluation with endothelial nitric oxide synthase (eNOS) immunoreactivity of the protective role of astaxanthin on hepatorenal injury of remote organs caused by ischaemia reperfusion of the lower extremities. GASTROENTEROLOGY REVIEW 2019; 15:161-172. [PMID: 32550950 PMCID: PMC7294969 DOI: 10.5114/pg.2019.88620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/09/2019] [Indexed: 02/07/2023]
Abstract
Introduction Ischemia and following reperfusion triggers local and systemic damage with the involvement of free oxygen radicals and inflammatory mediators. Although blood flow saves extremity from necrosis,multi organ dysfunction may progress and cause death of the patient. Aim The study aims to examine the effect of astaxanthin (AST) on the prevention of remote tissue injury resulting from lower extremity ischaemia–reperfusion (I/R). To elucidate the potential hepatoprotective and renoprotective effects of AST, in addition to histopathological findings, the intrahepatic and intrarenal kinetics of endothelial nitric oxide synthase (eNOS) during I/R were determined by using the immunohistochemical method. Material and methods Twenty-eight male Wistar albino rats were divided into four groups. For the control group, only the anaesthesia procedure (2 h) was conducted without I/R. In the I/R group, 2 h of reperfusion was conducted following ischaemia under anaesthesia. For the I/R group + AST, 7 days prior to ischaemia, 125 mg/kg AST was given with gavage, and 2 h of ischaemia and 2 h of reperfusion were conducted under anaesthesia. Following necropsy, liver and kidney tissue samples were fixed in 10% buffered formalin for 48 h for histopathological and immunohistochemical investigation. Results The histological analysis revealed that severe I/R hepatorenal injury such as inflammatory cell infiltration, dilatation in sinusoids and lumen of tubuli, congestion in glomerular capillaries, degeneration in hepatocyte and epithelial cells of tubuli, and necrosis was ameliorated by AST. Immunohistochemical studies showed that the I/R-induced elevation in eNOS expression was reduced by AST treatment. Conclusions In the case of acute lower extremity I/R, AST decreased the ischaemic injury in liver and renal tissues by protecting the microcirculation and providing a cytoprotective effect with vasodilatation.
Collapse
|
117
|
Xu W, Zhou J, You M, Lu C, Yang W, Gong Y, Dong X. Pre-B-cell colony enhancing factor regulates the alveolar epithelial sodium-water transport system through the ERK and AKT pathways. Am J Transl Res 2019; 11:5824-5835. [PMID: 31632551 PMCID: PMC6789215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 08/14/2019] [Indexed: 06/10/2023]
Abstract
The sodium-water transport system is crucial for alveolar fluid clearance. The pulmonary edema caused by extracorporeal circulation is mainly due to increased alveolar capillary permeability and reduced fluid clearance. We previously demonstrated that pre-B-cell colony enhancing factor (PBEF) increases alveolar capillary permeability and inhibits the sodium-water transport system. However, the specific mechanism by which PBEF inhibits the sodium-water transport system is unclear. In this study, we used HPAEpiC (alveolar type II epithelial cells) to construct an anoxia-reoxygenation model and simulate the extracorporeal circulation microenvironment. The impact of PBEF on the expression of genes and proteins implicated in sodium transport and its effect on the activation status of the ERK, P38, and AKT signaling pathways were explored in HPAEpiC by real-time fluorescent PCR and western blotting. Specific inhibitors were employed to verify the role of the three signaling pathways in the regulation of the sodium-water transport system. PBEF was substantially non-toxic to alveolar epithelial cells, inhibited the expression of ENaC, NKA, and AQP1, and affected the ERK, P38, and AKT signaling pathways. ERK pathway inhibitors attenuated PBEF-induced downregulation of EnaC, NKA, and AQP1, and increased NKA activity. P38 pathway inhibitors only attenuated PBEF-induced suppression of NKA expression. AKT pathway inhibitors potentiated the inhibitory effects of PBEF, reducing EnaC, AQP1, and NKA expression, as well as NKA activity. In conclusion, PBEF inhibited the sodium-water transport system by activation of ERK and suppression of AKT signaling.
Collapse
Affiliation(s)
- Weichang Xu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| | - Jianliang Zhou
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| | - Miaomiao You
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| | - Chao Lu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| | - Wei Yang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| | - Yi Gong
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| | - Xiao Dong
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| |
Collapse
|
118
|
Temporal changes in physiological and molecular markers in various brain regions following transient global ischemia in rats. Mol Biol Rep 2019; 46:6215-6230. [PMID: 31576510 DOI: 10.1007/s11033-019-05060-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 08/31/2019] [Indexed: 12/11/2022]
Abstract
Several mechanisms are involved in the loss of cellular integrity and tissue destructions in various brain regions during ischemic insult. The affected brain employs various self-repair mechanisms during the poststroke recovery. Therefore, the current study involves time course changes in different brain regions following ischemia in terms of inflammation, oxidative stress and apoptosis for which a bilateral common carotid arteries occlusion model was chosen. The development of oxidative stress was seen with a marked increase in ROS and NO levels with concomitant decrease in GSH levels and also the activities of anti-oxidant enzymes. These alterations were accompanied with decreased levels of neurotransmitters and motor and cognitive deficits at various time points. Increased expressions of various pro-inflammatory cytokines and a decline in BDNF levels in hippocampal regions on 7th day post ischemia, suggesting their role in its pathogenesis. The restoration of BDNF and neurotransmitter levels along with significant decline in inflammatory cytokine levels 14th day onwards following ischemia in hippocampus suggested poststroke recovery. The extent of neuronal damage was found to be increased significantly on 7th day post ischemia as indicated by TUNEL assay and hematoxylin and eosin staining depicting enhanced number of pyknotic neurons in cortical and hippocampal regions. Cortical regions of the ischemic brains were severely affected while hippocampal regions showed significant poststroke recovery, which might attributed to the normalization of BDNF and pro-inflammatory cytokine levels. In conclusion, the present study established the central role of BDNF and pro-inflammatory cytokines in the poststroke recovery. Also, the cortical and hippocampal regions were found to be more susceptible for ischemic injury. As our results indicated, full recovery after ischemic injury in different brain regions was not achieved, therefore further studies with long-term recovery time are required to be conducted.
Collapse
|
119
|
Peng CK, Huang KL, Wu CP, Wu YK, Tzeng IS, Lan CC. Phosphodiesterase-4 Inhibitor Roflumilast Attenuates Pulmonary Air Emboli-Induced Lung Injury. J Surg Res 2019; 241:24-30. [PMID: 31004869 DOI: 10.1016/j.jss.2019.03.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 01/26/2019] [Accepted: 03/22/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Pulmonary air embolism (PAE)-induced acute lung injury (ALI) can be caused by massive air entry into the lung circulation. PAE can occur during diving, aviation, and some iatrogenic invasive procedures. PAE-induced ALI presents with severe inflammation, hypoxia, and pulmonary hypertension, and it is a serious complication resulting in significant morbidity and mortality. Phosphodiesterase-4 (PDE4) inhibitors can regulate inflammation and are therefore expected to have a therapeutic effect on ALI. However, the effect of the PDE4 inhibitor roflumilast on PAE-induced ALI is unknown. METHODS The PAE model was undertaken in isolated-perfused rat lungs. Four groups (n = 6 in each group) were defined as follows: control, PAE, PAE + roflumilast 2.5 mg/kg, and PAE + roflumilast 5 mg/kg. Induction of PAE-induced ALI was achieved via the infusion of 0.7 cc air through the pulmonary artery. Roflumilast was administered via perfusate. All groups were assessed for pulmonary microvascular permeability, lung histopathology changes, pulmonary edema (lung weight/body weight, lung wet/dry weight ratio), tumor necrosis factor alpha (TNF-α), interleukin-1β (IL-1β), IL-6, IL-17, nuclear factor-kappa B (NF-κB), and inhibitor of NF-κB alpha (IκB-α). RESULTS After the induction of air, PAE-induced ALI presented with pulmonary edema, pulmonary microvascular hyperpermeability, and lung inflammation with neutrophilic sequestration. The PAE-induced ALI also presented with increased expressions of IL-1β, IL-6, IL-8, IL-17, TNF-α, and NF-κB and decreased expression of IκB-α. The administration of roflumilast decreased pulmonary edema, inflammation, cytokines, NF-κB, and restored IκB-α level. CONCLUSIONS PAE-induced ALI presents with lung inflammation with neutrophilic sequestration, pulmonary edema, hyperpermeability, increased cytokine levels, and activation of the NF-κB pathway. Roflumilast attenuates lung edema and inflammation and downregulates the NF-κB pathway and cytokines.
Collapse
Affiliation(s)
- Chung-Kan Peng
- Division of Pulmonary Medicine, Tri-Service General Hospital, Institute of Undersea and Hyperbaric Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Division of Pulmonary Medicine, Tri-Service General Hospital, Institute of Undersea and Hyperbaric Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Chin-Pyng Wu
- Department of Critical Care Medicine, Li-Shin Hospital, Tao-Yuan County, Taiwan
| | - Yao-Kuang Wu
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, School of Medicine, Tzu-Chi University, Hualien, Taiwan
| | - I-Shiang Tzeng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Chou-Chin Lan
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, School of Medicine, Tzu-Chi University, Hualien, Taiwan.
| |
Collapse
|
120
|
Apocynin alleviates lung injury by suppressing NLRP3 inflammasome activation and NF-κB signaling in acute pancreatitis. Int Immunopharmacol 2019; 75:105821. [PMID: 31437787 DOI: 10.1016/j.intimp.2019.105821] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 07/31/2019] [Accepted: 08/11/2019] [Indexed: 02/08/2023]
Abstract
Mounting evidence has demonstrated that acute pancreatitis (AP) is one of the causes of multiple organ damage. NADPH (nicotinamide adenine dinucleotide phosphate) act as a substrate of NADPH oxidase (NOX) to generate reactive oxygen species (ROS), but the role NADPH oxidase signaling pathway plays in AP-induced acute lung injury remains unclear. Apocynin, an inhibitor of NOX, is highly effective in suppressing the production of ROS. Here, we used rat model of severe acute pancreatitis (SAP) to explore whether the NOX inhibitor apocynin produced protective effects in against SAP-induced lung injury via inhibition of inflammation and oxidation. We observed that apocynin significantly attenuated severe acute pancreatitis-induced increase of NOX2, NOX4 and ROS expressions in lung tissues. In addition, the phosphorylation and degradation of IκBα, and the nuclear localization of NF-κB p65 in SAP-induced lung injury were also inhibited after using apocynin. Simultaneously, down-regulation of NOX suppressed the levels of inflammasome proteins including NLRP3, ASC, pro-Caspase-1 and cleaved-Caspase-1 in the lung. Serum levels of TNF-α, interleukin (IL)-1β and IL-6 were also reduced. Our findings suggest that beyond anti-oxidative effects, apocynin may also have anti-inflammatory effects by suppressing NLRP3 inflammasome activation and NF-κB signaling in acute pancreatitis. Therefore, apocynin may have therapeutic potential in the treatment of SAP and SAP-induced lung injury.
Collapse
|
121
|
Tan YB, Pastukh VM, Gorodnya OM, Mulekar MS, Simmons JD, Machuca TN, Beaver TM, Wilson GL, Gillespie MN. Enhanced Mitochondrial DNA Repair Resuscitates Transplantable Lungs Donated After Circulatory Death. J Surg Res 2019; 245:273-280. [PMID: 31421373 DOI: 10.1016/j.jss.2019.07.057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 06/18/2019] [Accepted: 07/18/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Transplantation of lungs procured after donation after circulatory death (DCD) is challenging because postmortem metabolic degradation may engender susceptibility to ischemia-reperfusion (IR) injury. Because oxidative mitochondrial DNA (mtDNA) damage has been linked to endothelial barrier disruption in other models of IR injury, here we used a fusion protein construct targeting the DNA repair 8-oxoguanine DNA glycosylase-1 (OGG1) to mitochondria (mtOGG1) to determine if enhanced repair of mtDNA damage attenuates endothelial barrier dysfunction after IR injury in a rat model of lung procurement after DCD. MATERIALS AND METHODS Lungs excised from donor rats 1 h after cardiac death were cold stored for 2 h after which they were perfused ex vivo in the absence and presence of mt-OGG1 or an inactive mt-OGG1 mutant. Lung endothelial barrier function and mtDNA integrity were determined during and at the end of perfusion, respectively. RESULTS AND CONCLUSIONS Mitochondria-targeted OGG1 attenuated indices of lung endothelial dysfunction incurred after a 1h post-mortem period. Oxidative lung tissue mtDNA damage as well as accumulation of proinflammatory mtDNA fragments in lung perfusate, but not nuclear DNA fragments, also were reduced by mitochondria-targeted OGG1. A repair-deficient mt-OGG1 mutant failed to protect lungs from the adverse effects of DCD procurement. CONCLUSIONS These findings suggest that endothelial barrier dysfunction in lungs procured after DCD is driven by mtDNA damage and point to strategies to enhance mtDNA repair in concert with EVLP as a means of alleviating DCD-related lung IR injury.
Collapse
Affiliation(s)
- Yong B Tan
- Department of Surgery, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Viktor M Pastukh
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Olena M Gorodnya
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Madhuri S Mulekar
- Department of Mathematics and Statistics, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Jon D Simmons
- Department of Surgery, College of Medicine, University of South Alabama, Mobile, Alabama; Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Tiago N Machuca
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida
| | - Thomas M Beaver
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida
| | | | - Mark N Gillespie
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, Alabama.
| |
Collapse
|
122
|
Mesenchymal Stem Cell Therapy Facilitates Donor Lung Preservation by Reducing Oxidative Damage during Ischemia. Stem Cells Int 2019; 2019:8089215. [PMID: 31481974 PMCID: PMC6701419 DOI: 10.1155/2019/8089215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/28/2019] [Accepted: 07/09/2019] [Indexed: 12/16/2022] Open
Abstract
Lung transplantation is a lifesaving therapy for people living with severe, life-threatening lung disease. The high mortality rate among patients awaiting transplantation is mainly due to the low percentage of lungs that are deemed acceptable for implantation. Thus, the current shortage of lung donors may be significantly reduced by implementing different therapeutic strategies which facilitate both organ preservation and recovery. Here, we studied whether the anti-inflammatory effect of human umbilical cord-derived mesenchymal stem cells (HUCPVCs) increases lung availability by improving organ preservation. We developed a lung preservation rat model that mimics the different stages by which donor organs must undergo before implantation. The therapeutic schema was as follows: cardiac arrest, warm ischemia (2 h at room temperature), cold ischemia (1.5 h at 4°C, with Perfadex), and normothermic lung perfusion with ventilation (Steen solution, 1 h). After 1 h of warm ischemia, HUCPVCs (1 × 106 cells) or vehicle was infused via the pulmonary artery. Physiologic data (pressure-volume curves) were acquired right after the cardiac arrest and at the end of the perfusion. Interestingly, although lung edema did not change among groups, lung compliance dropped to 34% in the HUCPVC-treated group, while the vehicle group showed a stronger reduction (69%, p < 0.0001). Histologic assessment demonstrated less overall inflammation in the HUCPVC-treated lungs. In addition, MPO activity, a neutrophil marker, was reduced by 41% compared with vehicle (p < 0.01). MSC therapy significantly decreased tissue oxidative damage by controlling reactive oxygen species production. Accordingly, catalase and superoxide dismutase enzyme activities remained at baseline levels. In conclusion, these results demonstrate that the anti-inflammatory effect of MSCs protects donor lungs against ischemic injury and postulates MSC therapy as a novel tool for organ preservation.
Collapse
|
123
|
Alda-1 Prevents Pulmonary Epithelial Barrier Dysfunction following Severe Hemorrhagic Shock through Clearance of Reactive Aldehydes. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2476252. [PMID: 31467875 PMCID: PMC6699483 DOI: 10.1155/2019/2476252] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/09/2019] [Indexed: 01/11/2023]
Abstract
Severe hemorrhagic shock and resuscitation (HS/R) can lead to lung injury, resulting in respiratory insufficiency. We investigated whether treatment with Alda-1, an ALDH2 activator, decreased lung injury induced by severe HS/R in a rat model. Male Sprague-Dawley rats were randomized into three groups, hemorrhagic shock + placebo, hemorrhagic shock + Alda-1, and sham. All animals were heparinized, and then 50% of the total calculated blood volume was collected over 60 minutes. After 40 minutes of hemorrhagic shock, animals were reinfused with the shed blood over 40 minutes and then observed for an additional 2 hours. Concentrations of 4-HNE, TNF-α, IL-6, and ALDH2 activity were detected; lung injury and lung wet-to-dry weight ratios were assessed. Expression of occludin and ZO-1 proteins in lung tissues was also determined. At 2 hours after resuscitation, lung injury was significantly reduced and the wet-to-dry weight ratio was notably decreased in the Alda-1 group compared with placebo (P<0.05). Alda-1 treatment also significantly increased the activity of ALDH2 and decreased the levels of toxic 4-HNE (P<0.05). In the Alda-1 group, IL-6 and TNF-α were dramatically decreased compared with placebo-treated animals (P<0.05). Expression of occludin and ZO-1 proteins was significantly decreased in the placebo group compared with the Alda-1 group (P<0.05). Thus, in a rat model of severe HS/R, treatment with Alda-1 increased the activity of ALDH2, significantly accelerated the clearance of reactive aldehydes, and concomitantly alleviated lung injury through improvement of pulmonary epithelial barrier integrity resulting in decreased alveolar epithelial tissue permeability, lung edema, and diffuse infiltration of inflammatory cells.
Collapse
|
124
|
Chen J, Jiang Z, Zhou X, Sun X, Cao J, Liu Y, Wang X. Dexmedetomidine Preconditioning Protects Cardiomyocytes Against Hypoxia/Reoxygenation-Induced Necroptosis by Inhibiting HMGB1-Mediated Inflammation. Cardiovasc Drugs Ther 2019; 33:45-54. [PMID: 30675709 DOI: 10.1007/s10557-019-06857-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a serious threat to the health of people around the world. Recent evidence has indicated that high-mobility group box-1 (HMGB1) is involved in I/R-induced inflammation, and inflammation can cause necroptosis of cells. Interestingly, dexmedetomidine (DEX) has anti-inflammatory properties. Therefore, we speculated that DEX preconditioning may suppress H/R-induced necroptosis by inhibiting expression of HMGB1 in cardiomyocytes. We found that hypoxia/reoxygenation (H/R) significantly increased cellular damage, as measured by cell viability (100 ± 3.26% vs. 53.33 ± 3.29, p < 0.01), CK-MB (1 vs. 3.25 ± 0.26, p < 0.01), cTnI (1 vs. 2.69 ± 0.31, p < 0.01), inflammation as indicated by TNF-α (1 ± 0.09 vs. 2.57 ± 0.12, p < 0.01), IL-1β (1 ± 0.33 vs. 3.87 ± 0.41, p < 0.01) and IL-6 (1 ± 0.36 vs. 3.60 ± 0.45, p < 0.01), and necroptosis, which were accompanied by significantly increased protein levels of HMGB1. These changes [cellular damage as measured by cell viability (53.33 ± 3.29% vs. 67.59 ± 2.69%, p < 0.01), CK-MB (3.25 ± 0.26 vs. 2.27 ± 0.22, p < 0.01), cTnI (2.69 ± 0.31 vs. 1.90 ± 0.25, p < 0.01), inflammation as indicated by TNF-α (2.57 ± 0.12 vs. 1.75 ± 0.15, p < 0.01), IL-1β (3.87 ± 0.41 vs. 2.09 ± 0.36, p < 0.01) and IL-6 (3.60 ± 0.45 vs. 2.21 ± 0.39, p < 0.01), and necroptosis proteins] were inhibited by DEX preconditioning. We also found that silencing expression of HMGB1 reinforced the protective effects of DEX preconditioning and overexpression of HMGB1 counteracted the protective effects of DEX preconditioning. Thus, we concluded that DEX preconditioning inhibits H/R-induced necroptosis by inhibiting expression of HMGB1 in cardiomyocytes.
Collapse
Affiliation(s)
- Jingyi Chen
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
- Institute of Anesthesiology, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
| | - Zhenzhen Jiang
- Department of Anesthesiology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Xing Zhou
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
- Institute of Anesthesiology, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Xingxing Sun
- Department of Ultrasonography Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Jianwei Cao
- Department of Orthopedics, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Yongpan Liu
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
- Institute of Anesthesiology, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Xianyu Wang
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
- Institute of Anesthesiology, Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| |
Collapse
|
125
|
Peng CK, Wu CP, Lin JY, Peng SC, Lee CH, Huang KL, Shen CH. Gas6/Axl signaling attenuates alveolar inflammation in ischemia-reperfusion-induced acute lung injury by up-regulating SOCS3-mediated pathway. PLoS One 2019; 14:e0219788. [PMID: 31318922 PMCID: PMC6638944 DOI: 10.1371/journal.pone.0219788] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 07/01/2019] [Indexed: 01/17/2023] Open
Abstract
Background Axl is a cell surface receptor tyrosine kinase, and activation of the Axl attenuates inflammation induced by various stimuli. Growth arrest-specific 6 (Gas6) has high affinity for Axl receptor. The role of Gas6/Axl signaling in ischemia-reperfusion-induced acute lung injury (IR-ALI) has not been explored previously. We hypothesized that Gas6/Axl signaling regulates IR-induced alveolar inflammation via a pathway mediated by suppressor of cytokine signaling 3 (SOCS3). Methods IR-ALI was induced by producing 30 min of ischemia followed by 90 min of reperfusion in situ in an isolated and perfused rat lung model. The rats were randomly allotted to a control group and IR groups, which were treated with three different doses of Gas6. Mouse alveolar epithelium MLE-12 cells were cultured in control and hypoxia-reoxygenation (HR) conditions with or without Gas6 and Axl inhibitor R428 pretreatment. Results We found that Gas6 attenuated IR-induced lung edema, the production of proinflammatory cytokines in perfusates, and the severity of ALI ex vivo. IR down-regulated SOCS3 expression and up-regulated NF-κB, and Gas6 restored this process. In the model of MLE-12 cells with HR, Gas6 suppressed the activation of TRAF6 and NF-κB by up-regulating SOCS3. Axl expression of alveolar epithelium was suppressed in IR-ALI but Gas6 restored phosphorylation of Axl. The anti-inflammatory effect of Gas6 was antagonized by R428, which highlighted that phosphorylation of Axl mediated the protective role of Gas6 in IR-ALI. Conclusions Gas6 up-regulates phosphorylation of Axl on alveolar epithelium in IR-ALI. The Gas6/Axl signaling activates the SOCS3-mediated pathway and attenuates IR-related inflammation and injury.
Collapse
Affiliation(s)
- Chung-Kan Peng
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chin-Pyng Wu
- Department of Critical Care Medicine, Landseed Hospital, Taoyuan, Taiwan
| | - Jr-Yu Lin
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Chi Peng
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Hsing Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Hao Shen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
126
|
Zhu J, Ye Q, Xu S, Chang YX, Liu X, Ma Y, Zhu Y, Hua S. Shengmai injection alleviates H 2O 2‑induced oxidative stress through activation of AKT and inhibition of ERK pathways in neonatal rat cardiomyocytes. JOURNAL OF ETHNOPHARMACOLOGY 2019; 239:111677. [PMID: 30615921 DOI: 10.1016/j.jep.2019.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 12/07/2018] [Accepted: 01/02/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shengmai injection (SMI) is a classical traditional Chinese medicine (TCM) officially recorded in Pharmacopoeia of the People's Republic of China (version 2015) and has long been used to treat heart failure in China. However scientific evidence for the anti-oxidative stress potential of SMI used in traditional medicine is lacking. AIM OF STUDY The present study aimed to evaluate the efficacy of SMI in alleviating H2O2‑induced Oxidative Stress the underlying mechanisms MATERIALS AND METHODS: H2O2-induced oxidative stress model of cardiomyocytes was established with primary cultured neonatal rat cardiomyocytes. CCK8 cell viability assay and lacatate dehydrogenase cytotoxicity assay were performed to ensure the safety dose and lowest effective dose for the mode employing CCK-8 cell viability assay kit and lactate dehydrogenase cytotoxicity assay kit. ROS levels were determined using CM-H2DCFDA fluorescent probe in cardiomyocytes with H2O2-induced oxidative stress. The change of NAD(P)H level in cardiomyocytes was evaluated during the process of oxidative stress. The content of myocardial cytosolic Ca2+ and Ca2+ was determined using Fura-2/AM and Rhod 2-AM fluorescent probe in mitochondrial in the process of oxidative stress. Annexin V-FITC/PI double staining was applied to examine the apoptotic cells in cardiomyocytes with oxidative stress. To identify the apoptosis after oxidative stress myocardial cells with the application of Annexin V-FITC/PI double staining apoptosis detection kit. Quantitative polymerase chain reaction (RT-PCR) was applied to measure the expression of antioxidant enzymes: catalase (CAT), superoxide dismutase (SOD), glutathione reductase (GSR). Western blot was performed to observe the phosphorylation of AKT and ERK1/2. RESULTS SMI was shown to significantly attenuate oxidative stress-induced cell proliferation arrest and apoptosis in neonatal rat cardiomyocytes. In addition, SMI treatment could decrease the production of reactive oxygen species (ROS), nicotinamide adenine dinucleotide (NADH) and malondialdehyde (MDA), and reduce the overloads of cytoplasmic Ca2+ and mitochondrial Ca2+ induced by H2O2. SMI could also restore the mRNA expression and activities of SOD, GSR, and CAT suppressed by H2O2. Mechanistically, SMI upregulated intracellular AKT phosphorylation and downregulate ERK1/2 phosphorylation in H2O2-treated cardiomyocytes. Pretreatment with LY294002, an AKT phosphorylation inhibitor, suppressed the protective role of SMI in cardiomyocytes, while pretreatment with PD98059, an ERK1/2 phosphorylation inhibitor, enhanced the effect of SMI. CONCLUSIONS In conclusion, SMI may attenuate oxidative stress-induced damage in cardiomyocytes potentially through the AKT and ERK1/2 pathway and can function as a promising injectable traditional Chinese medicine to treat oxidative stress-induced injury.
Collapse
Affiliation(s)
- Jinqiang Zhu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Qiaofeng Ye
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 314 An Shan Xi Road, Nan Kai District, Tianjin 300193, China
| | - Yan-Xu Chang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin Key Laboratory of Phytochemistry and Pharmaceutical Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xuan Liu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yan Ma
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300150, China
| | - Yan Zhu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, Tianjin 300457, China
| | - Shengyu Hua
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; College of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| |
Collapse
|
127
|
Cameli P, Carleo A, Bergantini L, Landi C, Prasse A, Bargagli E. Oxidant/Antioxidant Disequilibrium in Idiopathic Pulmonary Fibrosis Pathogenesis. Inflammation 2019; 43:1-7. [DOI: 10.1007/s10753-019-01059-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
128
|
Baicalin ameliorates lipopolysaccharide-induced acute lung injury in mice by suppressing oxidative stress and inflammation via the activation of the Nrf2-mediated HO-1 signaling pathway. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:1421-1433. [DOI: 10.1007/s00210-019-01680-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 06/19/2019] [Indexed: 12/22/2022]
|
129
|
Hung KY, Liao WI, Pao HP, Wu SY, Huang KL, Chu SJ. Targeting F-Box Protein Fbxo3 Attenuates Lung Injury Induced by Ischemia-Reperfusion in Rats. Front Pharmacol 2019; 10:583. [PMID: 31178737 PMCID: PMC6544082 DOI: 10.3389/fphar.2019.00583] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/06/2019] [Indexed: 01/12/2023] Open
Abstract
Background: Increasing evidence suggests that Fbxo3 signaling has an important impact on the pathophysiology of the inflammatory process. Fbxo3 protein inhibition has reduced cytokine-driven inflammation and improved disease severity in animal model of Pseudomonas-induced lung injury. However, it remains unclear whether inhibition of Fbxo3 protein provides protection in acute lung injury induced by ischemia-reperfusion (I/R). In this study, we investigated the protective effects of BC-1215 administration, a Fbxo3 inhibitor, on acute lung injury induced by I/R in rats. Methods: Lung I/R injury was induced by ischemia (40 min) followed by reperfusion (60 min). The rats were randomly assigned into one of six experimental groups (n = 6 rats/group): the control group, control + BC-1215 (Fbxo3 inhibitor, 0.5 mg/kg) group, I/R group, or I/R + BC-1215 (0.1, 0.25, 0.5 mg/kg) groups. The effects of BC-1215 on human alveolar epithelial cells subjected to hypoxia-reoxygenation (H/R) were also examined. Results: BC-1215 significantly attenuated I/R-induced lung edema, indicated by a reduced vascular filtration coefficient, wet/dry weight ratio, lung injury scores, and protein levels in bronchoalveolar lavage fluid (BALF). Oxidative stress and the level of inflammatory cytokines in BALF were also significantly reduced following administration of BC-1215. Additionally, BC-1215 mitigated I/R-stimulated apoptosis, NF-κB, and mitogen-activated protein kinase activation in the injured lung tissue. BC-1215 increased Fbxl2 protein expression and suppressed Fbxo3 and TNFR associated factor (TRAF)1–6 protein expression. BC-1215 also inhibited IL-8 production and NF-κB activation in vitro in experiments with alveolar epithelial cells exposed to H/R. Conclusions: Our findings demonstrated that Fbxo3 inhibition may represent a novel therapeutic approach for I/R-induced lung injury, with beneficial effects due to destabilizing TRAF proteins.
Collapse
Affiliation(s)
- Kuei-Yi Hung
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Wen-I Liao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shi-Jye Chu
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
130
|
Protective Effect of Thymosin β4 against Abdominal Aortic Ischemia-Reperfusion-Induced Acute Lung Injury in Rats. ACTA ACUST UNITED AC 2019; 55:medicina55050187. [PMID: 31121838 PMCID: PMC6572620 DOI: 10.3390/medicina55050187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/25/2019] [Accepted: 05/17/2019] [Indexed: 12/29/2022]
Abstract
Background and objectives: Ischemia-reperfusion (IR) caused by infrarenal abdominal aorta cross-clamping is an important factor in the development of ischemia-reperfusion injury in various distant organs. Materials and Methods: We investigated potential antioxidant/anti-inflammatory effects of thymosin beta 4 (Tβ4) in a rat model of abdominal aortic surgery-induced IR. Tβ4 (10 mg/kg, intravenous (i.v.)) was administered to rats with IR (90-min ischemia, 180-min reperfusion) at two different periods. One group received Tβ4 1 h before ischemia, and the other received 15 min before the reperfusion period. Results: Results were compared to control and non-Tβ4-treated rats with IR. Serum, bronchoalveolar lavage fluid and lung tissue levels of oxidant parameters were higher, while antioxidant levels were lower in the IR group compared to control. IR also increased inflammatory cytokine levels. Tβ4 reverted these parameters in both Tβ4-treated groups compared to the untreated IR group. Conclusions: Since there is no statistical difference between the prescribed results of both Tβ4-treated groups, our study demonstrates that Tβ4 reduced lung oxidative stress and inflammation following IR and prevented lung tissue injury regardless of timing of administration.
Collapse
|
131
|
Yu H, Kalogeris T, Korthuis RJ. Reactive species-induced microvascular dysfunction in ischemia/reperfusion. Free Radic Biol Med 2019; 135:182-197. [PMID: 30849489 PMCID: PMC6503659 DOI: 10.1016/j.freeradbiomed.2019.02.031] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/26/2019] [Accepted: 02/26/2019] [Indexed: 12/13/2022]
Abstract
Vascular endothelial cells line the inner surface of the entire cardiovascular system as a single layer and are involved in an impressive array of functions, ranging from the regulation of vascular tone in resistance arteries and arterioles, modulation of microvascular barrier function in capillaries and postcapillary venules, and control of proinflammatory and prothrombotic processes, which occur in all segments of the vascular tree but can be especially prominent in postcapillary venules. When tissues are subjected to ischemia/reperfusion (I/R), the endothelium of resistance arteries and arterioles, capillaries, and postcapillary venules become dysfunctional, resulting in impaired endothelium-dependent vasodilator and enhanced endothelium-dependent vasoconstrictor responses along with increased vulnerability to thrombus formation, enhanced fluid filtration and protein extravasation, and increased blood-to-interstitium trafficking of leukocytes in these functionally distinct segments of the microcirculation. The number of capillaries open to flow upon reperfusion also declines as a result of I/R, which impairs nutritive perfusion. All of these pathologic microvascular events involve the formation of reactive species (RS) derived from molecular oxygen and/or nitric oxide. In addition to these effects, I/R-induced RS activate NLRP3 inflammasomes, alter connexin/pannexin signaling, provoke mitochondrial fission, and cause release of microvesicles in endothelial cells, resulting in deranged function in arterioles, capillaries, and venules. It is now apparent that this microvascular dysfunction is an important determinant of the severity of injury sustained by parenchymal cells in ischemic tissues, as well as being predictive of clinical outcome after reperfusion therapy. On the other hand, RS production at signaling levels promotes ischemic angiogenesis, mediates flow-induced dilation in patients with coronary artery disease, and instigates the activation of cell survival programs by conditioning stimuli that render tissues resistant to the deleterious effects of prolonged I/R. These topics will be reviewed in this article.
Collapse
Affiliation(s)
- Hong Yu
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA
| | - Ted Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA
| | - Ronald J Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA; Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Drive, Columbia, MO 65211, USA.
| |
Collapse
|
132
|
Mariscal A, Caldarone L, Tikkanen J, Nakajima D, Chen M, Yeung J, Cypel M, Liu M, Keshavjee S. Pig lung transplant survival model. Nat Protoc 2019; 13:1814-1828. [PMID: 30072720 DOI: 10.1038/s41596-018-0019-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although lung transplant is a life-saving therapy for some patients, primary graft dysfunction (PGD) is a leading cause of mortality and morbidity soon after a transplant. Ischemia reperfusion injury is known to be one of the most critical factors in PGD development. PGD is by definition an acute lung injury syndrome that occurs during the first 3 d following lung transplantation. To successfully translate laboratory discoveries to clinical practice, a reliable and practical large animal model is critical. This protocol describes a surgical technique for swine lung transplantation and postoperative management for a further 3 d post transplant. The protocol includes the background and rationale, required supplies, and a detailed description of the donor operation, transplant surgery, postoperative care, and sacrifice surgery. A pig lung transplant model is reliably produced in which the recipients survive for 3 d post transplant. This 3-d survival model can be used by lung transplant researchers to assess the development of PGD and to test therapeutic strategies targeting PGD. In total, the protocol requires 5 h for the surgeries, plus ~2 h in total for the postoperative care.
Collapse
Affiliation(s)
- Andrea Mariscal
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada.,Toronto Lung Transplant Program, Department of Thoracic Surgery, University Health Network, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Lindsay Caldarone
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Jussi Tikkanen
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada.,Toronto Lung Transplant Program, Department of Thoracic Surgery, University Health Network, Toronto, ON, Canada
| | - Daisuke Nakajima
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada.,Toronto Lung Transplant Program, Department of Thoracic Surgery, University Health Network, Toronto, ON, Canada
| | - Manyin Chen
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada
| | - Jonathan Yeung
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada.,Toronto Lung Transplant Program, Department of Thoracic Surgery, University Health Network, Toronto, ON, Canada
| | - Marcelo Cypel
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada.,Toronto Lung Transplant Program, Department of Thoracic Surgery, University Health Network, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Mingyao Liu
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
| | - Shaf Keshavjee
- Department of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, Toronto, ON, Canada. .,Toronto Lung Transplant Program, Department of Thoracic Surgery, University Health Network, Toronto, ON, Canada. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
133
|
Wang ML, Wei CH, Wang WD, Wang JS, Zhang J, Wang JJ. Melatonin attenuates lung ischaemia-reperfusion injury via inhibition of oxidative stress and inflammation. Interact Cardiovasc Thorac Surg 2019; 26:761-767. [PMID: 29346581 DOI: 10.1093/icvts/ivx440] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/22/2017] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES Lung ischaemia-reperfusion injury is a complex pathophysiological process due to the production of reactive oxygen species and the generation of inflammatory reaction. We investigated the protective effects and the corresponding mechanism of melatonin (MT), a potent free-radical scavenger, on lung injury induced by ischaemia-reperfusion in a mouse model. METHODS Adult male C57BL/6J mice (n = 30) were randomly and equally allocated into 5 groups: sham controls, IR, IR + 10 mg/kg MT, IR + 20 mg/kg MT and IR + 30 mg/kg MT. Lung ischaemia-reperfusion injury was induced by thoracotomy followed by clamping of the left hilum for 1 h and subsequent reperfusion for 2 h. RESULTS Histological scoring analysis showed that lung parenchymal damage was ameliorated in the melatonin pretreatment groups when compared with the IR group, with the IR + 20 mg/kg MT group showing the strongest effect among the melatonin pretreatment groups. Wet-to-dry weight ratio, detection of malondialdehyde, protein expressions of inflammatory factors (tumour necrosis factor-α, interleukin-1β, NF-κB and IKK-γ) and apoptotic factors (cleaved caspase-3 and Bax/Bcl-2), as well as TUNEL assay showed changes similar to those of the lung injury scores in all groups. In contrast, the examination of superoxide dismutase showed a pattern contrary to that of the lung injury score in all groups. In addition, immunohistochemistry staining showed that the expressions of the antioxidants glutathione peroxidase and glutathione reductase were increased in the melatonin pretreatment groups. CONCLUSIONS This study demonstrated that melatonin pretreatment attenuated lung ischaemia-reperfusion injury via inhibition of oxidative stress, inflammation and apoptosis.
Collapse
Affiliation(s)
- Ming-Liang Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun-Hua Wei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-Dong Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Shun Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Zhang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Jun Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
134
|
Moon DH, Kang DY, Haam SJ, Yumoto T, Tsukahara K, Yamada T, Nakao A, Lee S. Hydrogen gas inhalation ameliorates lung injury after hemorrhagic shock and resuscitation. J Thorac Dis 2019; 11:1519-1527. [PMID: 31179095 DOI: 10.21037/jtd.2019.03.23] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background Hemorrhagic shock and resuscitation (HSR) is known to cause inflammatory reactions in the lung parenchyma and acute lung injury, increasing the risk of complications that can lead to death. Hydrogen gas has shown to inhibit the formation and eliminate reactive oxygen species (ROS), which are known to cause reperfusion injury. Hence, the purpose of this study was to investigate the protective effect of 2% inhaled hydrogen gas on post-HSR lung injury. Methods Rats weighing 300-500 g were divided into three groups: sham, HSR, and hydrogen (H2)/HSR groups. In the latter two groups, HSR was induced via femoral vein cannulation. Gas containing 2% hydrogen gas was inhaled only by those in the H2/HSR group. Lung tissue and abdominal aorta blood were obtained for histologic examination and arterial blood gas analyses, respectively. Neutrophil infiltration and proinflammatory mediators were also measured. Results PO2 was lower in the HSR and H2/HSR groups than in the sham group. Blood lactate level was not significantly different between the sham and H2/HSR groups, but it was significantly higher in the HSR group. Infiltration of inflammatory cells into the lung tissues was more frequent in the HSR group. Myeloperoxidase (MPO) activity was significantly different among the three groups (highest in the HSR group). All proinflammatory mediators, except IL-6, showed a significant difference among the three groups (highest in the HSR group). Conclusions Inhalation of 2% hydrogen gas after HSR minimized the extent of lung injury by decreasing MPO activity and reducing infiltration of inflammatory cells into lung tissue.
Collapse
Affiliation(s)
- Duk Hwan Moon
- Department of Thoracic and Cardiovascular Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Du-Young Kang
- Department of Cardiovascular and Thoracic Surgery, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seok Jin Haam
- Department of thoracic and cardiovascular surgery, Ajou university hospital, Suwon, Republic of Korea
| | - Tetsuya Yumoto
- Department of Emergency, Critical Care and Disaster Medicine, Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kohei Tsukahara
- Department of Emergency, Critical Care and Disaster Medicine, Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Taihei Yamada
- Department of Emergency, Critical Care and Disaster Medicine, Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Atsunori Nakao
- Department of Emergency, Critical Care and Disaster Medicine, Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Sungsoo Lee
- Department of Thoracic and Cardiovascular Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
135
|
Ferrari RS, Thomaz LDGR, Simoneti LEL, Ulbrich JM, Andrade CF. Effect of vaporized perfluorocarbon on oxidative stress during the cold ischemia phase of lung graft preservation. ACTA ACUST UNITED AC 2019; 45:e20170288. [PMID: 30942284 PMCID: PMC6733723 DOI: 10.1590/1806-3713/e20170288] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 08/12/2018] [Indexed: 01/24/2023]
Abstract
Liquid perfluorocarbon (PFC) instillation has been studied experimentally as an adjuvant therapy in the preservation of lung grafts during cold ischemia. The objective of this study was to evaluate whether vaporized PFC is also protective of lung grafts at different cold ischemia times. We performed histological analysis of and measured oxidative stress in the lungs of animals that received only preservation solution with low-potassium dextran (LPD) or vaporized PFC together with LPD. We conclude that vaporized PFC reduces the production of free radicals and the number of pulmonary structural changes resulting from cold ischemia.
Collapse
Affiliation(s)
- Renata Salatti Ferrari
- . Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul - UFRGS - Porto Alegre (RS) Brasil
| | | | - Lucas Elias Lise Simoneti
- . Laboratório de Vias Aéreas e Pulmão, Hospital de Clínicas de Porto Alegre - HCPA - Porto Alegre (RS) Brasil
| | - Jane Maria Ulbrich
- . Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul - UFRGS - Porto Alegre (RS) Brasil.,. Hospital de Clínicas de Porto Alegre - HCPA - Porto Alegre (RS) Brasil
| | - Cristiano Feijó Andrade
- . Programa de Pós-Graduação em Ciências Pneumológicas, Universidade Federal do Rio Grande do Sul - UFRGS - Porto Alegre (RS) Brasil.,. Hospital de Clínicas de Porto Alegre - HCPA - Porto Alegre (RS) Brasil
| |
Collapse
|
136
|
Liu WC, Chen SB, Liu S, Ling X, Xu QR, Yu BT, Tang J. Inhibition of mitochondrial autophagy protects donor lungs for lung transplantation against ischaemia-reperfusion injury in rats via the mTOR pathway. J Cell Mol Med 2019; 23:3190-3201. [PMID: 30887674 PMCID: PMC6484325 DOI: 10.1111/jcmm.14177] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/05/2018] [Accepted: 01/02/2019] [Indexed: 12/12/2022] Open
Abstract
Impaired mitochondrial function is a key factor attributing to lung ischaemia‐reperfusion (IR) injury, which contributes to major post‐transplant complications. Thus, the current study was performed to investigate the role of mitochondrial autophagy in lung I/R injury and the involvement of the mTOR pathway. We established rat models of orthotopic left lung transplantation to investigate the role of mitochondrial autophagy in I/R injury following lung transplantation. Next, we treated the donor lungs with 3‐MA and Rapamycin to evaluate mitochondrial autophagy, lung function and cell apoptosis with different time intervals of cold ischaemia preservation and reperfusion. In addition, mitochondrial autophagy, and cell proliferation and apoptosis of pulmonary microvascular endothelial cells (PMVECs) exposed to hypoxia‐reoxygenation (H/R) were monitored after 3‐MA administration or Rapamycin treatment. The cell apoptosis could be inhibited by mitochondrial autophagy at the beginning of lung ischaemia, but was rendered out of control when mitochondrial autophagy reached normal levels. After I/R of donor lung, the mitochondrial autophagy was increased until 6 hours after reperfusion and then gradually decreased. The elevation of mitochondrial autophagy was accompanied by promoted apoptosis, aggravated lung injury and deteriorated lung function. Moreover, the suppression of mitochondrial autophagy by 3‐MA inhibited cell apoptosis of donor lung to alleviate I/R‐induced lung injury as well as inhibited H/R‐induced PMVEC apoptosis, and enhanced its proliferation. Finally, mTOR pathway participated in I/R‐ and H/R‐mediated mitochondrial autophagy in regulation of cell apoptosis. Inhibition of I/R‐induced mitochondrial autophagy alleviated lung injury via the mTOR pathway, suggesting a potential therapeutic strategy for lung I/R injury.
Collapse
Affiliation(s)
- Wei-Cheng Liu
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Shi-Biao Chen
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Sheng Liu
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Xiang Ling
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Qi-Rong Xu
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Ben-Tong Yu
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Jian Tang
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| |
Collapse
|
137
|
Nethi SK, Barui AK, Mukherjee S, Patra CR. Engineered Nanoparticles for Effective Redox Signaling During Angiogenic and Antiangiogenic Therapy. Antioxid Redox Signal 2019; 30:786-809. [PMID: 29943661 DOI: 10.1089/ars.2017.7383] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Redox signaling plays a vital role in regulating various cellular signaling pathways and disease biology. Recently, nanomedicine (application of nanotechnology in biology and medicine) has been demonstrated to regulate angiogenesis through redox signaling. A complete understanding of redox signaling pathways influenced angiogenesis/antiangiogenesis triggered by therapeutic nanoparticles is extensively reviewed in this article. Recent Advances: In recent times, nanomedicines are regarded as the Trojan horses that could be employed for successful drug delivery, gene delivery, peptide delivery, disease diagnosis, and others, conquering barriers associated with conventional theranostic approaches. CRITICAL ISSUES Physiological angiogenesis is a tightly regulated process maintaining a balance between proangiogenic and antiangiogenic factors. The redox signaling is one of the main factors that contribute to this physiological balance. An aberrant redox signaling cascade can be caused by several exogenous and endogenous factors and leads to reduced or augmented angiogenesis that ultimately results in several disease conditions. FUTURE DIRECTIONS Redox signaling-based nanomedicine approach has emerged as a new platform for angiogenesis-related disease therapy, where nanoparticles promote angiogenesis via controlled reactive oxygen species (ROS) production and antiangiogenesis by triggering excessive ROS formation. Recently, investigators have identified different efficient nano-candidates, which modulate angiogenesis by controlling intracellular redox molecules. Considering the importance of angiogenesis in health care a thorough understanding of nanomedicine-regulated redox signaling would inspire researchers to design and develop more novel nanomaterials that could be used as an alternative strategy for the treatment of various diseases, where angiogenesis plays a vital role.
Collapse
Affiliation(s)
- Susheel Kumar Nethi
- 1 Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,2 Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Ayan Kumar Barui
- 1 Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,2 Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Sudip Mukherjee
- 1 Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,2 Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Chitta Ranjan Patra
- 1 Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,2 Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| |
Collapse
|
138
|
Xu HC, Lv W, Wang LM, Ye P, Hu J. Early Protection by Resveratrol in Rat Lung Transplantation. Med Sci Monit 2019; 25:760-770. [PMID: 30684444 PMCID: PMC6359885 DOI: 10.12659/msm.912345] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Resveratrol is a multifunctional bioactive substance that has effects in anti-inflammation and prevention of ischemia-reperfusion injury. This study compared the inflammation and expression of related proteins during the early stages after transplantation to explore the effects and mechanisms of resveratrol on transplanted lung. MATERIAL AND METHODS Sprague-Dawley rats were randomized to receive pretreatment of resveratrol suspension (60 mg/kg; RES group), dexamethasone (1 mg/kg; DEM group), or normal saline solution (2 mL/kg; control group) 1 h before lung transplantation. The cytokine concentration in the serum and bronchoalveolar lavage fluid (BALF) of the recipients was determined 24 h after transplantation. Histopathologic evaluation, including lung injury score, and the expression of necroptosis-associated proteins was assessed. RESULTS Histopathologic evaluation showed pneumocyte damage and endothelialitis associated with hemorrhage in the alveoli in the control group, the severity of which was greater than that in the other 2 groups. The levels of interleukin-6 and tumor necrosis factor-a in the serum and BALF of the RES and DEM groups were lower than those in the control group. The expression of necroptosis-associated proteins in the RES group was lower than that in the control group, and was inversely proportional to lung injury. CONCLUSIONS Pretreatment with resveratrol protected rat lung in the early stages after transplantation. We determined a relationship between necroptosis-associated proteins and transplanted lung injury, which suggests that the mechanism of lung transplantation-associated ischemia-reperfusion injury may be related to necroptosis.
Collapse
Affiliation(s)
- Hai-Chao Xu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Wang Lv
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Lu-Ming Wang
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Peng Ye
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Jian Hu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
139
|
The Nox1/Nox4 inhibitor attenuates acute lung injury induced by ischemia-reperfusion in mice. PLoS One 2018; 13:e0209444. [PMID: 30571757 PMCID: PMC6301701 DOI: 10.1371/journal.pone.0209444] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/27/2018] [Indexed: 02/05/2023] Open
Abstract
Lung ischemia and reperfusion injury (LIRI) were mediated by several processes including over-production of reactive oxygen species (ROS) and inflammatory activation. ROS generated by nicotinamide adenine dinucletide phosphate (NADPH) oxidase (Nox) may play a pivotal role in pathophysiological changes in a range of disease. However, it was poorly understood in LIRI. Thus, the purpose of our study was to explore whether GKT137831, as a special dual inhibitor of Nox1 and 4, could alleviate LIRI in mice model and explore the minimal dose. According to the protocol, this study was divided into two parts. The first part was to determine the minimal dose of Nox1/4 inhibitor in attenuating LIRI via histopathology and apoptosis analysis. Eighteen C57BL/6J male wild-type mice were randomly divided in to sham, 2.5Nox+sham, 5.0Nox+sham, IR, 2.5Nox+IR and 5.0Nox+IR groups. According to the different group, mice were pretreated with corresponding dose of Nox1/4 inhibitors or normal saline. After LIRI, the results showed 5.0mg/kg Nox1/4 inhibitor could be considered as the minimal dose to alleviate injury by decreasing of lung injury score and the number of TUNEL-positive cells. The second part was to further verify the benefit of 5.0mg/kg Nox1/4 inhibitor in lung protective effects. Thirty-seven C57BL/6J male wild-type mice were divided in to sham, IR and 5.0Nox+IR groups randomly. The results showed that expressions of inflammatory, autophagy cytokines were markedly elevated and PH value was declined after LIRI. However, 5.0 mg/kg Nox1/4 inhibitor significantly attenuated cytokine production as reflected by immunohistochemistry, western blotting and Q-PCR analysis. In conclusion, our findings suggested that 5.0mg/kg Nox1/4 inhibitor contributed to protect lung tissue damage after LIRI via the suppression of inflammatory and autophagy activation.
Collapse
|
140
|
Abreu MDM, Almeida FMD, Santos KBD, Assis EACPD, Hamada RKF, Jatene FB, Pêgo-Fernandes PM, Pazetti R. Does methylene blue attenuate inflammation in nonischemic lungs after lung transplantation? J Bras Pneumol 2018; 44:378-382. [PMID: 30517338 PMCID: PMC6467587 DOI: 10.1590/s1806-37562017000000172] [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: 05/28/2017] [Accepted: 12/07/2017] [Indexed: 11/22/2022] Open
Abstract
Objective: To evaluate whether methylene blue (MB) could minimize the effects of ischemia-reperfusion injury in the nonischemic lung on a lung transplantation rodent model. Methods: Forty female Sprague-Dawley rats were divided into 20 donors and 20 recipients. The 20 recipient rats were divided into two groups (n = 10) according to the treatment (0.9% saline vs. 1% MB solutions). All animals underwent unilateral lung transplantation. Recipients received 2 mL of saline or MB intraperitoneally prior to transplantation. After 2 h of reperfusion, the animals were euthanized and histopathological and immunohistochemical analyses were performed in the nonischemic lung. Results: There was a significant decrease in inflammation-neutrophil count and intercellular adhesion molecule-1 (ICAM-1) expression in lung parenchyma were higher in the saline group in comparison with the MB group-and in apoptosis-caspase-3 expression was higher in the saline group and Bcl-2 expression was higher in MB group. Conclusions: MB is an effective drug for the protection of nonischemic lungs against inflammation and apoptosis following unilateral lung transplantation in rats.
Collapse
Affiliation(s)
- Marcus da Matta Abreu
- . Departamento de Cardiopneumologia, Disciplina de Cirurgia Torácica, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Francine Maria de Almeida
- . Departamento de Cardiopneumologia, Disciplina de Cirurgia Torácica, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Kelli Borges Dos Santos
- . Núcleo de Pesquisa em Transplante, Faculdade de Enfermagem, Universidade Federal de Juiz de Fora, Juiz de Fora (MG) Brasil
| | | | | | - Fabio Biscegli Jatene
- . Departamento de Cardiopneumologia, Disciplina de Cirurgia Torácica, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Paulo Manuel Pêgo-Fernandes
- . Departamento de Cardiopneumologia, Disciplina de Cirurgia Torácica, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Rogerio Pazetti
- . Departamento de Cardiopneumologia, Disciplina de Cirurgia Torácica, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| |
Collapse
|
141
|
Wei J, Wang P, Li Y, Dou Q, Lin J, Tao W, Lin J, Fu X, Huang Z, Zhang W. Inhibition of RHO Kinase by Fasudil Attenuates Ischemic Lung Injury After Cardiac Arrest in Rats. Shock 2018; 50:706-713. [DOI: 10.1097/shk.0000000000001097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
142
|
Parekh D, Dancer RCA, Scott A, D'Souza VK, Howells PA, Mahida RY, Tang JCY, Cooper MS, Fraser WD, Tan L, Gao F, Martineau AR, Tucker O, Perkins GD, Thickett DR. Vitamin D to Prevent Lung Injury Following Esophagectomy-A Randomized, Placebo-Controlled Trial. Crit Care Med 2018; 46:e1128-e1135. [PMID: 30222631 PMCID: PMC6250246 DOI: 10.1097/ccm.0000000000003405] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Observational studies suggest an association between vitamin D deficiency and adverse outcomes of critical illness and identify it as a potential risk factor for the development of lung injury. To determine whether preoperative administration of oral high-dose cholecalciferol ameliorates early acute lung injury postoperatively in adults undergoing elective esophagectomy. DESIGN A double-blind, randomized, placebo-controlled trial. SETTING Three large U.K. university hospitals. PATIENTS Seventy-nine adult patients undergoing elective esophagectomy were randomized. INTERVENTIONS A single oral preoperative (3-14 d) dose of 7.5 mg (300,000 IU; 15 mL) cholecalciferol or matched placebo. MEASUREMENTS AND MAIN RESULTS Primary outcome was change in extravascular lung water index at the end of esophagectomy. Secondary outcomes included PaO2:FIO2 ratio, development of lung injury, ventilator and organ-failure free days, 28 and 90 day survival, safety of cholecalciferol supplementation, plasma vitamin D status (25(OH)D, 1,25(OH)2D, and vitamin D-binding protein), pulmonary vascular permeability index, and extravascular lung water index day 1 postoperatively. An exploratory study measured biomarkers of alveolar-capillary inflammation and injury. Forty patients were randomized to cholecalciferol and 39 to placebo. There was no significant change in extravascular lung water index at the end of the operation between treatment groups (placebo median 1.0 [interquartile range, 0.4-1.8] vs cholecalciferol median 0.4 mL/kg [interquartile range, 0.4-1.2 mL/kg]; p = 0.059). Median pulmonary vascular permeability index values were significantly lower in the cholecalciferol treatment group (placebo 0.4 [interquartile range, 0-0.7] vs cholecalciferol 0.1 [interquartile range, -0.15 to -0.35]; p = 0.027). Cholecalciferol treatment effectively increased 25(OH)D concentrations, but surgery resulted in a decrease in 25(OH)D concentrations at day 3 in both arms. There was no difference in clinical outcomes. CONCLUSIONS High-dose preoperative treatment with oral cholecalciferol was effective at increasing 25(OH)D concentrations and reduced changes in postoperative pulmonary vascular permeability index, but not extravascular lung water index.
Collapse
Affiliation(s)
- Dhruv Parekh
- Warwick Clinical Trials Unit, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Rachel C A Dancer
- Birmingham Acute Care Research Group, Institute of Inflammation and Aging, University of Birmingham, Birmingham, United Kingdom
- Academic Department of Anaesthesia, Critical Care, Resuscitation and Pain, Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Aaron Scott
- Birmingham Acute Care Research Group, Institute of Inflammation and Aging, University of Birmingham, Birmingham, United Kingdom
| | - Vijay K D'Souza
- Birmingham Acute Care Research Group, Institute of Inflammation and Aging, University of Birmingham, Birmingham, United Kingdom
| | - Phillip A Howells
- Birmingham Acute Care Research Group, Institute of Inflammation and Aging, University of Birmingham, Birmingham, United Kingdom
| | - Rahul Y Mahida
- Birmingham Acute Care Research Group, Institute of Inflammation and Aging, University of Birmingham, Birmingham, United Kingdom
| | - Jonathan C Y Tang
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Mark S Cooper
- Discipline of Medicine, Concord Clinical School, University of Sydney, NSW, Australia
| | - William D Fraser
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - LamChin Tan
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, United Kingdom
| | - Fang Gao
- Birmingham Acute Care Research Group, Institute of Inflammation and Aging, University of Birmingham, Birmingham, United Kingdom
- Academic Department of Anaesthesia, Critical Care, Resuscitation and Pain, Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Adrian R Martineau
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Olga Tucker
- Birmingham Acute Care Research Group, Institute of Inflammation and Aging, University of Birmingham, Birmingham, United Kingdom
- Academic Department of Anaesthesia, Critical Care, Resuscitation and Pain, Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Gavin D Perkins
- Warwick Clinical Trials Unit, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- Academic Department of Anaesthesia, Critical Care, Resuscitation and Pain, Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - David R Thickett
- Birmingham Acute Care Research Group, Institute of Inflammation and Aging, University of Birmingham, Birmingham, United Kingdom
- Queen Elizabeth Hospital University Hospitals, Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
143
|
Maltesen RG, Buggeskov KB, Andersen CB, Plovsing R, Wimmer R, Ravn HB, Rasmussen BS. Lung Protection Strategies during Cardiopulmonary Bypass Affect the Composition of Bronchoalveolar Fluid and Lung Tissue in Cardiac Surgery Patients. Metabolites 2018; 8:metabo8040054. [PMID: 30241409 PMCID: PMC6316472 DOI: 10.3390/metabo8040054] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/31/2018] [Accepted: 09/19/2018] [Indexed: 11/18/2022] Open
Abstract
Pulmonary dysfunction is among the most frequent complications to cardiac surgeries. Exposure of blood to the cardiopulmonary bypass (CPB) circuit with subsequent lung ischemia-reperfusion leads to the production of inflammatory mediators and increases in microvascular permeability. The study aimed to elucidate histological, cellular, and metabolite changes following two lung protective regimens during CPB with Histidine-Tryptophan-Ketoglutarate (HTK) enriched or warm oxygenated blood pulmonary perfusion compared to standard regimen with no pulmonary perfusion. A total of 90 patients undergoing CPB were randomized to receiving HTK, oxygenated blood or standard regimen. Of these, bronchoalveolar lavage fluid (BALF) and lung tissue biopsies were obtained before and after CPB from 47 and 25 patients, respectively. Histopathological scores, BALF cell counts and metabolite screening were assessed. Multivariate and univariate analyses were performed. Profound histological, cellular, and metabolic changes were identified in all patients after CPB. Histological and cellular changes were similar in the three groups; however, some metabolite profiles were different in the HTK patients. While all patients presented an increase in inflammatory cells, metabolic acidosis, protease activity and oxidative stress, HTK patients seemed to be protected against severe acidosis, excessive fatty acid oxidation, and inflammation during ischemia-reperfusion. Additional studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Raluca G Maltesen
- Department of Anesthesia and Intensive Care Medicine, Aalborg University Hospital, 9000 Aalborg, Denmark.
| | - Katrine B Buggeskov
- Department of Cardiothoracic Anesthesia, Heart Centre, Rigshospitalet, 2100 Copenhagen, Denmark.
| | - Claus B Andersen
- Department of Forensic Medicine, University of Copenhagen, 2100 Copenhagen, Denmark.
| | - Ronni Plovsing
- Department of Intensive Care, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark.
- Department of Anesthesiology, Hvidovre Hospital, University of Copenhagen, 2650 Hvidovre, Denmark.
| | - Reinhard Wimmer
- Department of Chemistry and Bioscience, Aalborg University, 9220 Aalborg, Denmark.
| | - Hanne B Ravn
- Department of Cardiothoracic Anesthesia, Heart Centre, Rigshospitalet, 2100 Copenhagen, Denmark.
| | - Bodil S Rasmussen
- Department of Anesthesia and Intensive Care Medicine, Aalborg University Hospital, 9000 Aalborg, Denmark.
- Department of Clinical Medicine, School of Medicine and Health, Aalborg University, 9220 Aalborg, Denmark.
| |
Collapse
|
144
|
Ju M, Liu B, He H, Gu Z, Liu Y, Su Y, Zhu D, Cang J, Luo Z. MicroRNA-27a alleviates LPS-induced acute lung injury in mice via inhibiting inflammation and apoptosis through modulating TLR4/MyD88/NF-κB pathway. Cell Cycle 2018; 17:2001-2018. [PMID: 30231673 DOI: 10.1080/15384101.2018.1509635] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Acute lung injury (ALI) is a critical clinical condition with a high mortality rate, characterized with excessive uncontrolled inflammation and apoptosis. Recently, microRNAs (miRNAs) have been found to play crucial roles in the amelioration of various inflammation-induced diseases, including ALI. However, it remains unknown the biological function and regulatory mechanisms of miRNAs in the regulation of inflammation and apoptosis in ALI. The aim of this study is to identify and evaluate the potential role of miRNAs in ALI and reveal the underlying molecular mechanisms of their effects. Here, we analyzed microRNA expression profiles in lung tissues from LPS-challenged mice using miRNA microarray. Because microRNA-27a (miR-27a) was one of the miRNAs being most significantly downregulated, which has an important role in regulation of inflammation, we investigated its function. Overexpression of miR-27a by agomir-27a improved lung injury, as evidenced by the reduced histopathological changes, lung wet/dry (W/D) ratio, lung microvascular permeability and apoptosis in the lung tissues, as well as ameliorative survival of ALI mice. This was accompanied by the alleviating of inflammation, such as the reduced total BALF cell and neutrophil counts, decreased levels of tumor necrosis factor alpha (TNF-α), interleukin-1 (IL-6) interleukin-1β (IL-1β) and myeloperoxidase (MPO) activity in BAL fluid. Toll-like receptor 4 (TLR4), an important regulator of the nuclear factor kappa-B (NF-κB) signaling pathway, was identified as a novel target of miR-27a in RAW264.7 cells. Furthermore, our results showed that LPS stimulation increased the expression of MyD88 and NF-κB p65 (p-p65), but inhibited the expression of inhibitor of nuclear factor-κB-α (IκB-α), suggesting the activation of NF-κB signaling pathway. Further investigations revealed that agomir-miR-27a reversed the promoting effect of LPS on NF-κB signaling pathway. The results here suggested that miR-27a alleviates LPS-induced ALI in mice via reducing inflammation and apoptosis through blocking TLR4/MyD88/NF-κB activation.
Collapse
Affiliation(s)
- MinJie Ju
- a Department of Critial Care Medicine , Zhongshan Hospital, Fudan University , Shanghai China
| | - BoFei Liu
- b Department of Intensive Care Medicine , 1st People Hospital , ZhangjiaGang , China
| | - HongYu He
- a Department of Critial Care Medicine , Zhongshan Hospital, Fudan University , Shanghai China
| | - ZhunYong Gu
- a Department of Critial Care Medicine , Zhongshan Hospital, Fudan University , Shanghai China
| | - YiMei Liu
- a Department of Critial Care Medicine , Zhongshan Hospital, Fudan University , Shanghai China
| | - Ying Su
- a Department of Critial Care Medicine , Zhongshan Hospital, Fudan University , Shanghai China
| | - DuMing Zhu
- a Department of Critial Care Medicine , Zhongshan Hospital, Fudan University , Shanghai China
| | - Jing Cang
- c Department of Anesthesiology , Zhongshan Hospital, Fudan University , Shanghai , China
| | - Zhe Luo
- a Department of Critial Care Medicine , Zhongshan Hospital, Fudan University , Shanghai China
| |
Collapse
|
145
|
The Anti-inflammatory Effect of Hydrogen on Lung Transplantation Model of Pulmonary Microvascular Endothelial Cells During Cold Storage Period. Transplantation 2018; 102:1253-1261. [DOI: 10.1097/tp.0000000000002276] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
146
|
Wang Z, Zhang A, Meng W, Wang T, Li D, Liu Z, Liu H. Ozone protects the rat lung from ischemia-reperfusion injury by attenuating NLRP3-mediated inflammation, enhancing Nrf2 antioxidant activity and inhibiting apoptosis. Eur J Pharmacol 2018; 835:82-93. [PMID: 30075224 DOI: 10.1016/j.ejphar.2018.07.059] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 12/12/2022]
Abstract
Ischemia-reperfusion injury (IRI) is a major cause of lung dysfunction during cardiovascular surgery, heart transplantation and cardiopulmonary bypass procedures, and the inflammatory response, oxidative stress, and apoptosis play key and allegedly maladaptive roles in its pathogenesis. The aim of this study was to initially elucidate whether ozone induces oxidative preconditioning by activating nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and secondly to determine whether ozone oxidative preconditioning (OzoneOP) protects the lung from IRI by attenuating nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3)-mediated inflammation, enhancing the antioxidant activity of Nrf2 and inhibiting apoptosis. Rats treated with or without OzoneOP (2 ml containing 100 µg/kg/day) were subjected to 1 h of lung ischemia followed by 2 h of reperfusion for 10 days. Lung damage, antioxidant capacity, inflammation and apoptosis were evaluated and compared among different groups after reperfusion. OzoneOP significantly ameliorated changes in lung morphology and protected the lung from IRI by attenuating oxidative stress, inflammation-induced injury and lung apoptosis. Moreover, OzoneOP increased the expression of Nrf2 and decreased the levels of NLRP3, apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC), un-cleavable cysteine-requiring aspartate protease-1 (procaspase-1), cysteine-requiring aspartate protease-1 (caspase-1) and interleukin-1β (IL-1β) in the rat lungs. In summary, these results provide new insights into the molecular events modulated by ozone and suggest that ozone therapy may be an integrative support for patients with lung IRI.
Collapse
Affiliation(s)
- Zhiwen Wang
- Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang 150001, China
| | - Ai Zhang
- General Hospital of Heilongjiang Province Land Reclamation Bureau, 235 Hashuang Road, Harbin, Heilongjiang 150088, China
| | - Weixin Meng
- Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang 150001, China
| | - Tingting Wang
- Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang 150001, China
| | - Dandan Li
- Institute of Keshan Disease, Harbin Medical University, 157 Baojian Road, Harbin, Heilongjiang 150081, China
| | - Zonghong Liu
- Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang 150001, China
| | - Hongyu Liu
- Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
147
|
Shi X, Liu HY, Li SP, Xu HB. Keratinocyte growth factor protects endometrial cells from oxygen glucose deprivation/re-oxygenation via activating Nrf2 signaling. Biochem Biophys Res Commun 2018; 501:178-185. [DOI: 10.1016/j.bbrc.2018.04.208] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 04/26/2018] [Indexed: 12/20/2022]
|
148
|
Xu C, Liang C, Sun W, Chen J, Chen X. Glycyrrhizic acid ameliorates myocardial ischemic injury by the regulation of inflammation and oxidative state. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:1311-1319. [PMID: 29849452 PMCID: PMC5965375 DOI: 10.2147/dddt.s165225] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background Glycyrrhizic acid (GA), a bioactive triterpenoid saponin isolated from the roots of licorice plants (Glycyrrhiza glabra), has been shown to exert a variety of pharmacological activities and is considered to have potential therapeutic applications. The purpose of the present study was to investigate the cardioprotective effect of GA on myocardial ischemia (MI) injury rats induced by isoproterenol (ISO), and explore the potential mechanisms underlying these effects. Materials and methods The rats were randomized into five groups: control, ISO, ISO+diltiazem (10 mg/kg), ISO+GA (10 mg/kg), and ISO+GA (20 mg/kg). Electrocardiogram and histopathological examination were performed. Markers of cardiac marker enzymes (creatine kinase-MB, lactate dehydrogenase), oxidative stress (superoxide dismutase, malondialdehyde [MDA]), and inflammation (TNF-α, IL-1β, and IL-6) were also measured in each group. Proteins involved in NF-κB and Nrf-2/HO-1 pathway were detected by Western blot. Results GA decreased the ST elevation induced by MI, decreased serum levels of creatine kinase, lactate dehydrogenase, malondialdehyde, IL-6, IL-1β, and TNF-α, and increased serum superoxide dismutase and malondialdehyde activities. Furthermore, GA increased the protein levels of Nrf-2 and HO-1 and downregulated the phosphorylation of IκB, and NF-κB p65 in ISO-induced MI. Conclusion These observations indicated that GA has cardioprotective effects against MI, and these effects might be related to the activation of Nrf-2/HO-1 and inhibition of NF-κB signaling pathway in the myocardium.
Collapse
Affiliation(s)
- Chongli Xu
- Nanjing University of Chinese Medicine, Nanjing 210029, People's Republic of China.,Jiangnin Hospital of Nanjing, Nanjing 211100, People's Republic of China
| | - Caihong Liang
- Jiangnin Hospital of Nanjing, Nanjing 211100, People's Republic of China
| | - Weixin Sun
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Jiandong Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Xiaohu Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| |
Collapse
|
149
|
Thirupathi A, Pinho RA. Effects of reactive oxygen species and interplay of antioxidants during physical exercise in skeletal muscles. J Physiol Biochem 2018; 74:359-367. [PMID: 29713940 DOI: 10.1007/s13105-018-0633-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 04/23/2018] [Indexed: 12/11/2022]
Abstract
A large number of researches have led to a substantial growth of knowledge about exercise and oxidative stress. Initial investigations reported that physical exercise generates free radical-mediated damages to cells; however, in recent years, studies have shown that regular exercise can upregulate endogenous antioxidants and reduce oxidative damage. Yet, strenuous exercise perturbs the antioxidant system by increasing the reactive oxygen species (ROS) content. These alterations in the cellular environment seem to occur in an exercise type-dependent manner. The source of ROS generation during exercise is debatable, but now it is well established that both contracting and relaxing skeletal muscles generate reactive oxygen species and reactive nitrogen species. In particular, exercises of higher intensity and longer duration can cause oxidative damage to lipids, proteins, and nucleotides in myocytes. In this review, we summarize the ROS effects and interplay of antioxidants in skeletal muscle during physical exercise. Additionally, we discuss how ROS-mediated signaling influences physical exercise in antioxidant system.
Collapse
Affiliation(s)
- Anand Thirupathi
- Laboratory of Exercise Biochemistry and Physiology, Graduate Program in Health Sciences, Health Sciences Unit, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105 Bairro Universitário, Criciúma, Santa Catarina, 88806-000, Brazil.
| | - Ricardo A Pinho
- Laboratory of Exercise Biochemistry and Physiology, Graduate Program in Health Sciences, Health Sciences Unit, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105 Bairro Universitário, Criciúma, Santa Catarina, 88806-000, Brazil
| |
Collapse
|
150
|
Pak O, Sydykov A, Kosanovic D, Schermuly RT, Dietrich A, Schröder K, Brandes RP, Gudermann T, Sommer N, Weissmann N. Lung Ischaemia-Reperfusion Injury: The Role of Reactive Oxygen Species. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:195-225. [PMID: 29047088 DOI: 10.1007/978-3-319-63245-2_12] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Lung ischaemia-reperfusion injury (LIRI) occurs in many lung diseases and during surgical procedures such as lung transplantation. The re-establishment of blood flow and oxygen delivery into the previously ischaemic lung exacerbates the ischaemic injury and leads to increased microvascular permeability and pulmonary vascular resistance as well as to vigorous activation of the immune response. These events initiate the irreversible damage of the lung with subsequent oedema formation that can result in systemic hypoxaemia and multi-organ failure. Alterations in the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS) have been suggested as crucial mediators of such responses during ischaemia-reperfusion in the lung. Among numerous potential sources of ROS/RNS within cells, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, xanthine oxidases, nitric oxide synthases and mitochondria have been investigated during LIRI. Against this background, we aim to review here the extensive literature about the ROS-mediated cellular signalling during LIRI, as well as the effectiveness of antioxidants as treatment option for LIRI.
Collapse
Affiliation(s)
- Oleg Pak
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Akylbek Sydykov
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Djuro Kosanovic
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Ralph T Schermuly
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Alexander Dietrich
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, Goethestraße 33, 80336, Munich, Germany
| | - Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Thomas Gudermann
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, Goethestraße 33, 80336, Munich, Germany
| | - Natascha Sommer
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardio-pulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Aulweg 130, 35392, Giessen, Germany.
| |
Collapse
|