1
|
Yadav SS, Nair RR. Ethyl Pyruvate as a Potential Therapeutic Agent for Endometriosis: A Perspective. Reprod Sci 2025:10.1007/s43032-025-01875-x. [PMID: 40360949 DOI: 10.1007/s43032-025-01875-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025]
Abstract
Endometriosis is a disease where vascularised tissue similar to endometrium (the lining of the uterus) grows outside of the uterus. Its pathogenesis involves a complex interplay of inflammation, angiogenesis, cellular proliferation, reactive oxygen species (ROS) production, altered energy metabolism, and epithelial-to-mesenchymal transition (EMT).Even though endometriosis was described more than 150 years ago, we have been unable to find its effective therapy. Conservative treatment approaches like non-steroidal anti-inflammatory drugs or hormone therapy are available to date for the treatment of endometriosis. Anti-angiogenic inhibitors and immunomodulators like IFN-α, β, and TNF-α inhibitors are also potential treatment options. These treatments are inadequate as they either affect the symptoms only of endometriosis or target only one pathological pathway involved. Surgical excision of the endometriotic lesion is also possible, however, recurrence of the disease is reported in several cases. A single therapeutic agent targeting several pathological processes in endometriosis would always be a better option. Here we present our perspective on the pharmacological potential of Ethyl pyruvate and also propose it as a promising therapeutic agent for endometriosis as it inhibits inflammation, cell proliferation, angiogenesis, aerobic glycolysis, EMT, and ROS activity together.
Collapse
Affiliation(s)
- Suresh Singh Yadav
- Department of Molecular Biology & Biochemistry, Guru Nanak Dev University, Amritsar, 143005, Punjab, India
| | - Rohini Ravindran Nair
- Department of Medical Biotechnology, Gujrat Biotechnology University, Gandhinagar, 382355, Gujrat, India.
| |
Collapse
|
2
|
Lv X, Li J, Wei R, Meng L, Kong X, Wei K, Tang M, Liu K, Liu C. Ethyl pyruvate alleviates pulmonary arterial hypertension via PI3K-Akt signaling. Mol Cell Biochem 2025; 480:1045-1054. [PMID: 38740701 DOI: 10.1007/s11010-024-05020-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/24/2024] [Indexed: 05/16/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a pathophysiological syndrome that is extremely difficult to manage, and there is currently no effective treatment. We want to elucidate the therapeutic effect of ethyl pyruvate (EP) on PAH and its possible mechanism. Pulmonary artery endothelial cells (PAECs) were cultured in conventional low-oxygen environments, and cellular proliferation was monitored after treatment with EP. Expression of p-PI3K/Akt, LC3-II, and Beclin-1 was detected by Western blot. After hyperkinetic PAH rabbits' models were treated with EP, hemodynamic data were collected. Right ventricular hypertrophy and pulmonary vascular remodeling were evaluated. Expression of p-PI3K/Akt, LC3-II, and Beclin-1 protein was also detected after using autophagy inhibitor and agonists. We found that EP could inhibit PAECs proliferation. After EP treatment, expression of p-PI3K/Akt was upregulated in vitro and in vivo. LC3-II and Beclin-1 were inhibited and their expression was lower after autophagy inhibitor was given, while after administration of autophagy agonists, their expression was higher than that in the EP alone group. Besides, EP attenuated PAH, and right ventricular hypertrophy and pulmonary vascular remodeling were also reversed. EP can reduce PAH and reverse vascular remodeling which is associated with inhibition of autophagy in PAECs based on PI3K-Akt signaling pathway. The results of this study can provide surgical opportunities for patients with severe PAH caused by congenital heart disease in clinical cardiovascular surgery.
Collapse
Affiliation(s)
- Xin Lv
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No. 107, West Wenhua Road, Jinan, 250012, Shandong, China
| | - Jianhua Li
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No. 107, West Wenhua Road, Jinan, 250012, Shandong, China
| | - Ruyuan Wei
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No. 107, West Wenhua Road, Jinan, 250012, Shandong, China
| | - Lingwei Meng
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No. 107, West Wenhua Road, Jinan, 250012, Shandong, China
| | - Xiangjin Kong
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No. 107, West Wenhua Road, Jinan, 250012, Shandong, China
| | - Kaiming Wei
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No. 107, West Wenhua Road, Jinan, 250012, Shandong, China
| | - Mengmeng Tang
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No. 107, West Wenhua Road, Jinan, 250012, Shandong, China
| | - Kai Liu
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No. 107, West Wenhua Road, Jinan, 250012, Shandong, China.
| | - Chuanzhen Liu
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No. 107, West Wenhua Road, Jinan, 250012, Shandong, China.
- Shandong University, No. 27, South Shanda Road, Jinan, 250100, Shandong, China.
- Pantheum Biotechnology Co, LtdShandong, 250012, Jinan, China.
| |
Collapse
|
3
|
Sha S, Jin N, Xie X, Zhou R, Ruan Y, Ouyang Y. Ethyl pyruvate alleviates NLRP3/Caspase-1/GSDMD-mediated neuronal pyroptosis in neonatal rats with hypoxic-ischemic brain damage. Int J Dev Neurosci 2024; 84:594-604. [PMID: 38940222 DOI: 10.1002/jdn.10357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 06/29/2024] Open
Abstract
Pyroptosis is an inflammation-associated programmed cell death, and neuroinflammation is strongly associated with severe neurological deficits in neonatal hypoxic-ischemic encephalopathy (HIE). Ethyl pyruvate (EP), a known anti-inflammatory agent, has shown promise in the treatment of hypoxic-ischemic brain damage (HIBD) rats; nevertheless, the therapeutic mechanism of EP and its capacity to suppress neuronal pyroptosis in HIBD rats remain unclear. In both the neonatal Rice-Vannucci rat model and the OGD/R model, this study examined alterations in the NLRP3/Caspase-1/GSDMD classical pyroptosis pathway in hippocampal neurons during HIE and the potential inhibitory impact of ethyl pyruvate on this pathway. We used HE staining, immunofluorescence double staining, transmission electron microscopy, and western blot to demonstrate that EP effectively inhibited hippocampal neuronal pyroptosis and attenuated the activation of the NLRP3/Caspase-1/GSDMD signaling pathway in HIBD rats, which resulted in a reduction of neuroinflammation and facilitated neural recovery. The results suggest that EP may be a promising neuroprotective agent for treating HIE.
Collapse
Affiliation(s)
- Sha Sha
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ni Jin
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xinyi Xie
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruiyu Zhou
- Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanghao Ruan
- Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Ying Ouyang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Deng RM, Zhou J. Targeting NF-κB in Hepatic Ischemia-Reperfusion Alleviation: from Signaling Networks to Therapeutic Targeting. Mol Neurobiol 2024; 61:3409-3426. [PMID: 37991700 DOI: 10.1007/s12035-023-03787-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
Hepatic ischemia-reperfusion injury (HIRI) is a major complication of liver trauma, resection, and transplantation that can lead to liver dysfunction and failure. Scholars have proposed a variety of liver protection methods aimed at reducing ischemia-reperfusion damage, but there is still a lack of effective treatment methods, which urgently needs to find new effective treatment methods for patients. Many studies have reported that signaling pathway plays a key role in HIRI pathological process and liver function recovery mechanism, among which nuclear transfer factor-κB (NF-κB) signaling pathway is one of the signal transduction closely related to disease. NF-κB pathway is closely related to HIRI pathologic process, and inhibition of this pathway can delay oxidative stress, inflammatory response, cell death, and mitochondrial dysfunction. In addition, NF-κB can also interact with PI3K/Akt, MAPK, and Nrf2 signaling pathways to participate in HIRI regulation. Based on the role of NF-κB pathway in HIRI, it may be a potential target pathway for HIRI. This review emphasizes the role of inhibiting the NF-κB signaling pathway in oxidative stress, inflammatory response, cell death, and mitochondrial dysfunction in HIRI, as well as the effects of related drugs or inhibitors targeting NF-κB on HIRI. The objective of this review is to elucidate the role and mechanism of NF-κB pathway in HIRI, emphasize the important role of NF-κB pathway in the prevention and treatment of HIRI, and provide a theoretical basis for the target NF-κB pathway as a therapy for HIRI.
Collapse
Affiliation(s)
- Rui-Ming Deng
- Department of Anesthesiology, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province, 341000, People's Republic of China
- The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16 Meiguan Avenue, Ganzhou, Jiangxi Province, 341000, People's Republic of China
| | - Juan Zhou
- The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16 Meiguan Avenue, Ganzhou, Jiangxi Province, 341000, People's Republic of China.
- Department of Thyroid and Breast Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province, 341000, People's Republic of China.
| |
Collapse
|
5
|
Pyne S, Paria K. Optimization of extraction process parameters of caffeic acid from microalgae by supercritical carbon dioxide green technology. BMC Chem 2022; 16:31. [PMID: 35562772 PMCID: PMC9107252 DOI: 10.1186/s13065-022-00824-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/27/2022] [Indexed: 11/12/2022] Open
Abstract
Purpose In this study, the optimization of extraction process parameters of caffeic acid content from Spirulina platensis is performed by supercritical green technology. Methods Especially, the optimization of supercritical carbon dioxide (SC-CO2) extraction parameters was carried out employing Box-Behnken design (BBD) and response surface methodology (RSM). Alongside, the three levels of extraction parameters i.e. extraction pressure, extraction time and temperature have been fixed. As a response, the caffeic acid content of the extracts was determined by HPLC. The statistical analysis (ANOVA) of developed mathematical models was used in the process. Results The extract exhibited the highest content of caffeic acid as 72.11 µg/g of dw at the optimized extraction conditions of 360.08 bar pressure for 57.13 min extraction time at 38.31 °C temperature. Simultaneously this extract exhibited the highest content of total phenolic content (76.87 µg GAE/g dw), reducing power (2278 µg BHT/g dw), FRAP value (4.19 mM FeSO4 equivalent/g dw) and IC50 for DPPH activity (89.28 µg/mL). Conclusion It has been also noted that supercritical fluid extract can significantly retard the growth of microorganisms in litchi beverage. Consequently, we can also predict that isolated SC-CO2 antioxidant containing fraction would have hopeful for foodstuff preservative.
Collapse
Affiliation(s)
- Smritikana Pyne
- Department of Food Technology and Biochemical Engineering, Jadavpur University, Kolkata, 700032, West Bengal, India.
| | - Kishalay Paria
- Oriental Institute of Science and Technology, Vidyasagar University, Midnapore, 721102, West Bengal, India.
| |
Collapse
|
6
|
Lu X, Ding Y, Liu H, Sun M, Chen C, Yang Y, Wang H. The Role of Hydrogen Sulfide Regulation of Autophagy in Liver Disorders. Int J Mol Sci 2022; 23:ijms23074035. [PMID: 35409395 PMCID: PMC8999478 DOI: 10.3390/ijms23074035] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 02/01/2023] Open
Abstract
Autophagy is a complex process of degradation of senescent or dysfunctional organelles in cells. Dysfunctional autophagy is associated with many diseases such as cancers, immune dysfunction, and aging. Hydrogen sulfide (H2S) is considered to be the third gas signal molecule after nitrous oxide and carbon monoxide. In recent years, H2S has been found to have a variety of important biological functions, and plays an important role in a variety of physiological and pathological processes. In this review, we review the recent role and mechanism of H2S in regulating autophagy in liver disorders, in order to provide a basis for further research in the future.
Collapse
Affiliation(s)
- Xueqin Lu
- Institute of Nursing and Health, School of Nursing and Health, Henan University, Jinming Avenue, Kaifeng 475004, China; (X.L.); (Y.D.); (C.C.)
| | - Yueming Ding
- Institute of Nursing and Health, School of Nursing and Health, Henan University, Jinming Avenue, Kaifeng 475004, China; (X.L.); (Y.D.); (C.C.)
| | - Huiyang Liu
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (H.L.); (M.S.); (Y.Y.)
| | - Mengyao Sun
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (H.L.); (M.S.); (Y.Y.)
| | - Chaoran Chen
- Institute of Nursing and Health, School of Nursing and Health, Henan University, Jinming Avenue, Kaifeng 475004, China; (X.L.); (Y.D.); (C.C.)
| | - Yihan Yang
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (H.L.); (M.S.); (Y.Y.)
| | - Honggang Wang
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (H.L.); (M.S.); (Y.Y.)
- Correspondence:
| |
Collapse
|
7
|
Ethyl pyruvate, a versatile protector in inflammation and autoimmunity. Inflamm Res 2022; 71:169-182. [PMID: 34999919 PMCID: PMC8742706 DOI: 10.1007/s00011-021-01529-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 12/18/2022] Open
Abstract
Ethyl pyruvate (EP) has potent influence on redox processes, cellular metabolism, and inflammation. It has been intensively studied in numerous animal models of systemic and organ-specific disorders whose pathogenesis involves a strong immune component. Here, basic chemical and biological properties of EP are discussed, with an emphasis on its redox and metabolic activity. Further, its influence on myeloid and T cells is considered, as well as on intracellular signaling beyond its effect on immune cells. Also, the effects of EP on animal models of chronic inflammatory and autoimmune disorders are presented. Finally, a possibility to apply EP as a treatment for such diseases in humans is discussed. Scientific papers cited in this review were identified using the PubMed search engine that relies on the MEDLINE database. The reference list covers the most important findings in the field in the past twenty years.
Collapse
|
8
|
Wang C, Du HB, Zhao ZA, Zhai JY, Zhang LM, Niu CY, Zhao ZG. Autophagy Is Involved in Stellate Ganglion Block Reversing Posthemorrhagic Shock Mesenteric Lymph-Mediated Vascular Hyporeactivity. Front Physiol 2021; 12:728191. [PMID: 34621184 PMCID: PMC8491623 DOI: 10.3389/fphys.2021.728191] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/23/2021] [Indexed: 12/05/2022] Open
Abstract
Objective: The aim of this study was to clarify the role of autophagy in stellate ganglion block (SGB) reversing posthemorrhagic shock mesenteric lymph (PHSML)-mediated vascular hyporeactivity. Methods: Hemorrhagic shock model in conscious rats was employed to observe the effects of SGB (0.2 ml of 0.25% ropivacaine hydrochloride hydrate) and autophagy inhibitor 3-methyladenine (3-MA; 30 mg/kg) on the vascular reactivity of second-order rat mesenteric arteries in vitro, while the effects of PHSML (1 ml/kg) and autophagy agonist rapamycin (Rapa, 10 mg/kg) on the beneficial effect of SGB were investigated. The cellular viability, contractility, and autophagy-related protein expressions in vascular smooth muscle cells (VSMCs) were detected following treatments of PHSML, PHSML obtained from the rats that underwent hemorrhagic shock plus SGB (PHSML-SGB), and PHSML plus 3-MA (5 mM), respectively. Results: Hemorrhagic shock significantly decreased the vascular reactivity to gradient norepinephrine (NE), which is reversed by the SGB treatment and 3-MA administration. On the contrary, PHSML intravenous infusion and Rapa administration inhibited the vascular contractile responses in rats that underwent hemorrhagic shock plus SGB treatment. PHSML treatment significantly inhibited the cellular viability and contractility in VSMCs, increased the expressions of LC3-II and Beclin 1, and decreased the expression of p62, along with opposite appearances in these indices following PHSML-SGB treatment. In addition, 3-MA counteracted the adverse roles of PHSML in these indices in VSMCs. Conclusion: SGB inhibits PHSML-mediated vascular hyporeactivity by reducing the excessive autophagy in VSMCs.
Collapse
Affiliation(s)
- Chen Wang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
| | - Hui-Bo Du
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
| | - Zhen-Ao Zhao
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China.,Pathophysiology Experimental Teaching Center of Basic Medical College, Hebei North University, Zhangjiakou, China.,Key Laboratory of Critical Disease Mechanism and Intervention in Hebei, Hebei Medical University & Hebei North University, Shijiazhuang & Zhangjiakou, China
| | - Jia-Yi Zhai
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
| | - Li-Min Zhang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China.,Pathophysiology Experimental Teaching Center of Basic Medical College, Hebei North University, Zhangjiakou, China.,Key Laboratory of Critical Disease Mechanism and Intervention in Hebei, Hebei Medical University & Hebei North University, Shijiazhuang & Zhangjiakou, China
| | - Chun-Yu Niu
- Key Laboratory of Critical Disease Mechanism and Intervention in Hebei, Hebei Medical University & Hebei North University, Shijiazhuang & Zhangjiakou, China.,Basic Medical College, Hebei Medical University, Shijiazhuang, China
| | - Zi-Gang Zhao
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China.,Pathophysiology Experimental Teaching Center of Basic Medical College, Hebei North University, Zhangjiakou, China.,Key Laboratory of Critical Disease Mechanism and Intervention in Hebei, Hebei Medical University & Hebei North University, Shijiazhuang & Zhangjiakou, China
| |
Collapse
|
9
|
Lu C, Wang C, Xiao H, Chen M, Yang Z, Liang Z, Wang H, Liu Y, Yang Y, Wang Q. Ethyl pyruvate: A newly discovered compound against ischemia-reperfusion injury in multiple organs. Pharmacol Res 2021; 171:105757. [PMID: 34302979 DOI: 10.1016/j.phrs.2021.105757] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/21/2021] [Accepted: 07/02/2021] [Indexed: 12/23/2022]
Abstract
Ischemia-reperfusion injury (IRI) is a process whereby an initial ischemia injury and subsequent recovery of blood flow, which leads to the propagation of an innate immune response and the changes of structural and functional of multiple organs. Therefore, IRI is considered to be a great challenge in clinical treatment such as organ transplantation or coronary angioplasty. In recent years, ethyl pyruvate (EP), a derivative of pyruvate, has received great attention because of its stability and low toxicity. Previous studies have proved that EP has various pharmacological activities, including anti-inflammation, anti-oxidative stress, anti-apoptosis, and anti-fibrosis. Compelling evidence has indicated EP plays a beneficial role in a variety of acute injury models, such as brain IRI, myocardial IRI, renal IRI, and hepatic IRI. Moreover, EP can not only effectively inhibit multiple IRI-induced pathological processes, but also improve the structural and functional lesion of tissues and organs. In this study, we review the recent progress in the research on EP and discuss their implications for a better understanding of multiple organ IRI, and the prospects of targeting the EP for therapeutic intervention.
Collapse
Affiliation(s)
- Chenxi Lu
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Guangming Road, Shenmu, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Changyu Wang
- Department of Cardiology, Xi'an No.3 Hospital, School of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China
| | - Haoxiang Xiao
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Guangming Road, Shenmu, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Mengfan Chen
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Guangming Road, Shenmu, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Zhi Yang
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Guangming Road, Shenmu, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an, China
| | - Zhenxing Liang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou, China
| | - Haiying Wang
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Guangming Road, Shenmu, China
| | - Yonglin Liu
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Guangming Road, Shenmu, China
| | - Yang Yang
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Guangming Road, Shenmu, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an, China.
| | - Qiang Wang
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Guangming Road, Shenmu, China.
| |
Collapse
|
10
|
Yan B, Luo J, Kaltenmeier C, Du Q, Stolz DB, Loughran P, Yan Y, Cui X, Geller DA. Interferon Regulatory Factor-1 (IRF1) activates autophagy to promote liver ischemia/reperfusion injury by inhibiting β-catenin in mice. PLoS One 2020; 15:e0239119. [PMID: 33137133 PMCID: PMC7605671 DOI: 10.1371/journal.pone.0239119] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 07/27/2020] [Indexed: 01/08/2023] Open
Abstract
Autophagy is an important factor in liver ischemia-reperfusion injury. In the current study we investigate the function of interferon regulatory factor-1 (IRF1) in regulating autophagy to promote hepatic ischemia reperfusion injury (IR). The high expression of IRF1 during hepatic IR exhibited increased liver damage and was associated with activation of autophagy shown by Western blot markers, as well as immunofluorescent staining for autophagosomes. These effects were diminished by IRF1 deficiency in IRF1 knock out (KO) mice. Moreover, the autophagy inhibitor 3-MA decreased IR-induced liver necrosis and markedly abrogated the rise in liver injury tests (AST/ALT). β-catenin expression decreased during liver IR and was increased in the IRF1 KO mice. Immunoprecipitation assay showed the binding between IRF1 and β-catenin. Overexpression of IRF1 induced autophagy and also inhibited the expression of β-catenin. β-catenin inhibitor increased autophagy while β-catenin agonist suppressed autophagy in primary mouse hepatocytes. These results indicate that IRF1 induced autophagy aggravates hepatic IR injury in part by inhibiting β-catenin and suggests that targeting IRF1 may be an effective strategy in reducing hepatic IR injury.
Collapse
Affiliation(s)
- Bing Yan
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Jing Luo
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Christof Kaltenmeier
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Qiang Du
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Donna B. Stolz
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA, United States of America
| | - Patricia Loughran
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA, United States of America
| | - Yihe Yan
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Xiao Cui
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - David A. Geller
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
- * E-mail:
| |
Collapse
|
11
|
Salidroside Ameliorates Mitochondria-Dependent Neuronal Apoptosis after Spinal Cord Ischemia-Reperfusion Injury Partially through Inhibiting Oxidative Stress and Promoting Mitophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3549704. [PMID: 32774670 PMCID: PMC7396093 DOI: 10.1155/2020/3549704] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/30/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022]
Abstract
Ischemia-reperfusion injury is the second most common injury of the spinal cord and has the risk of neurological dysfunction and paralysis, which can seriously affect patient quality of life. Salidroside (Sal) is an active ingredient extracted from Herba Cistanche with a variety of biological attributes such as antioxidant, antiapoptotic, and neuroprotective activities. Moreover, Sal has shown a protective effect in ischemia-reperfusion injury of the liver, heart, and brain, but its effect in ischemia-reperfusion injury of the spinal cord has not been elucidated. Here, we demonstrated for the first time that Sal pretreatment can significantly improve functional recovery in mice after spinal cord ischemia-reperfusion injury and significantly inhibit the apoptosis of neurons both in vivo and in vitro. Neurons have a high metabolic rate, and consequently, mitochondria, as the main energy-supplying suborganelles, become the main injury site of spinal cord ischemia-reperfusion injury. Mitochondrial pathway-dependent neuronal apoptosis is increasingly confirmed by researchers; therefore, Sal's effect on mitochondria naturally attracted our attention. By means of a range of experiments both in vivo and in vitro, we found that Sal can reduce reactive oxygen species production through antioxidant stress to reduce mitochondrial permeability and mitochondrial damage, and it can also enhance the PINK1-Parkin signaling pathway and promote mitophagy to eliminate damaged mitochondria. In conclusion, our results show that Sal is beneficial to the protection of spinal cord neurons after ischemia-reperfusion injury, mainly by reducing apoptosis associated with the mitochondrial-dependent pathway, among which Sal's antioxidant and autophagy-promoting properties play an important role.
Collapse
|
12
|
El-Emam SZ, Soubh AA, Al-Mokaddem AK, Abo El-Ella DM. Geraniol activates Nrf-2/HO-1 signaling pathway mediating protection against oxidative stress-induced apoptosis in hepatic ischemia-reperfusion injury. Naunyn Schmiedebergs Arch Pharmacol 2020; 393:1849-1858. [PMID: 32417955 DOI: 10.1007/s00210-020-01887-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/28/2020] [Indexed: 12/20/2022]
Abstract
Geraniol (GOH) is a natural essential oil that possesses antioxidant, anti-inflammatory, and antiapoptotic properties by various signaling pathways. Liver ischemia-reperfusion injury (IRI) is a serious event that triggers liver dysfunction or even failure. Nuclear factor erythroid 2-related factor 2 (Nrf2), a transcriptional factor, maintains cellular defense mechanism through antioxidant and anti-inflammatory properties. To detect GOH effect against liver IRI through the activation of the Nrf2/HO-1 antioxidant pathway, five groups of rats were randomized to normal, sham, IR, GOH, and GOH/IR. Blood samples and liver tissues were collected to measure various biochemical parameters related to liver function, and oxidative stress as well as inflammatory and apoptotic indicators besides liver tissue histopathology was evaluated by light microscopy. GOH induces activation of Nrf2 along with the upregulation of HO-1 expression. Also, the antioxidant activity of GOH was shown by the elevation of total antioxidant capacity and GSH levels, together with normalizing malondialdehyde. Regarding the anti-inflammatory effect of GOH, it suppresses the levels of TNF-α, iNOS, and COX-2. Additionally, the antiapoptotic effect of GOH, Bax, and caspase-3, 9 were reduced in liver tissue. GOH is a promising hepatoprotective agent in liver IRI through the activation of Nrf2/HO-1 antioxidant pathway.
Collapse
Affiliation(s)
- Soad Z El-Emam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, 6th of October City, Giza, 12566, Egypt
| | - Ayman A Soubh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, ACU University, 4th Industrial Zone, Banks Complex, 6th of October City, Giza, 12451, Egypt
| | - Asmaa K Al-Mokaddem
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Dina M Abo El-Ella
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, 6th of October City, Giza, 12566, Egypt.
| |
Collapse
|
13
|
Guo W, Fang H, Cao S, Chen S, Li J, Shi J, Tang H, Zhang Y, Wen P, Zhang J, Wang Z, Shi X, Pang C, Yang H, Hu B, Zhang S. Six-Transmembrane Epithelial Antigen of the Prostate 3 Deficiency in Hepatocytes Protects the Liver Against Ischemia-Reperfusion Injury by Suppressing Transforming Growth Factor-β-Activated Kinase 1. Hepatology 2020; 71:1037-1054. [PMID: 31393024 PMCID: PMC7155030 DOI: 10.1002/hep.30882] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/24/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Hepatic ischemia-reperfusion (I/R) injury remains a major challenge affecting the morbidity and mortality of liver transplantation. Effective strategies to improve liver function after hepatic I/R injury are limited. Six-transmembrane epithelial antigen of the prostate 3 (Steap3), a key regulator of iron uptake, was reported to be involved in immunity and apoptotic processes in various cell types. However, the role of Steap3 in hepatic I/R-induced liver damage remains largely unclear. APPROACH AND RESULTS In the present study, we found that Steap3 expression was significantly up-regulated in liver tissue from mice subjected to hepatic I/R surgery and primary hepatocytes challenged with hypoxia/reoxygenation insult. Subsequently, global Steap3 knockout (Steap3-KO) mice, hepatocyte-specific Steap3 transgenic (Steap3-HTG) mice, and their corresponding controls were subjected to partial hepatic warm I/R injury. Hepatic histology, the inflammatory response, and apoptosis were monitored to assess liver damage. The molecular mechanisms of Steap3 function were explored in vivo and in vitro. The results demonstrated that, compared with control mice, Steap3-KO mice exhibited alleviated liver damage after hepatic I/R injury, as shown by smaller necrotic areas, lower serum transaminase levels, decreased apoptosis rates, and reduced inflammatory cell infiltration, whereas Steap3-HTG mice had the opposite phenotype. Further molecular experiments showed that Steap3 deficiency could inhibit transforming growth factor-β-activated kinase 1 (TAK1) activation and downstream c-Jun N-terminal kinase (JNK) and p38 signaling during hepatic I/R injury. CONCLUSIONS Steap3 is a mediator of hepatic I/R injury that functions by regulating inflammatory responses as well as apoptosis through TAK1-dependent activation of the JNK/p38 pathways. Targeting hepatocytes, Steap3 may be a promising approach to protect the liver against I/R injury.
Collapse
Affiliation(s)
- Wen‐Zhi Guo
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Hong‐Bo Fang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Sheng‐Li Cao
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - San‐Yang Chen
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Jie Li
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Ji‐Hua Shi
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Hong‐Wei Tang
- Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Yi Zhang
- Department of SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina
| | - Pei‐Hao Wen
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Jia‐Kai Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Zhi‐Hui Wang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Xiao‐Yi Shi
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Chun Pang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Han Yang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Bo‐Wen Hu
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Shui‐Jun Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| |
Collapse
|
14
|
Effects of Ethyl Pyruvate on Bile Duct Ligation-Induced Liver Fibrosis by Regulating Nrf2 Pathway and Proinflammatory Cytokines in Rats. Gastroenterol Res Pract 2019; 2019:2969802. [PMID: 31933629 PMCID: PMC6942817 DOI: 10.1155/2019/2969802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 05/02/2019] [Accepted: 05/19/2019] [Indexed: 12/14/2022] Open
Abstract
Aim The aim of this paper is to investigate the effects of ethyl pyruvate (EP) on experimental liver fibrosis induced by bile duct ligation (BDL) and explore the underlying molecular mechanisms. Material and Method Rats were randomly divided into three groups: the sham group, the BDL group, and the BDL+EP group. Liver fibrosis was induced by common bile duct ligation and was evaluated by serum biochemical parameter levels, Masson's trichrome staining, α-SMA expression, and collagen I deposition. The levels of Nrf2 signaling pathway-related antioxidant genes (Nrf2, SOD2, NQO1, and GSH-Px) in liver tissues were also measured. Meanwhile, the mRNA expression levels of HMGB1, IL-1β, TNF-α, and HSP27 were analyzed. In BDL-induced liver fibrosis rats, the successfully established model was confirmed by the significant increase of serum ALT and AST levels, the high liver fibrosis score, α-SMA expression, and collagen deposition. Results Compared with the BDL group, EP administration could diminish fibrosis level and substantially increase the expression of Nrf2 signaling pathway-related antioxidant genes. Furthermore, EP significantly suppressed the mRNA expression levels of HMGB1, IL-1β, TNF-α, and HSP27. Conclusions The results suggested that EP administration could effectively inhibit the liver fibrosis induced by BDL in rat, which may be associated with the enhanced activity of Nrf2 to mediate antioxidant enzyme system and downregulate the inflammatory genes.
Collapse
|
15
|
Protective Role of mTOR in Liver Ischemia/Reperfusion Injury: Involvement of Inflammation and Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7861290. [PMID: 31827701 PMCID: PMC6885218 DOI: 10.1155/2019/7861290] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/24/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023]
Abstract
Liver ischemia/reperfusion (IR) injury is a common phenomenon after liver resection and transplantation, which often results in liver graft dysfunction such as delayed graft function and primary nonfunction. The mammalian target of rapamycin (mTOR) is an evolutionarily highly conserved serine/threonine protein kinase, which coordinates cell growth and metabolism through sensing environmental inputs under physiological or pathological conditions, involved in the pathophysiological process of IR injury. In this review, we mainly present current evidence of the beneficial role of mTOR in modulating inflammation and autophagy under liver IR to provide some evidence for the potential therapies for liver IR injury.
Collapse
|
16
|
Yin J, Yin Z, Wang B, Zhu C, Sun C, Liu X, Gong G. Angiopoietin-1 Protects Spinal Cord Ischemia and Reperfusion Injury by Inhibiting Autophagy in Rats. Neurochem Res 2019; 44:2746-2754. [PMID: 31630316 DOI: 10.1007/s11064-019-02893-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 10/10/2019] [Accepted: 10/17/2019] [Indexed: 10/25/2022]
Abstract
Spinal cord ischemia and reperfusion (SCIR) injury can induce autophagy, which is involved in the survival of neurons. However, whether autophagy plays a neuroprotective or a detrimental role in SCIR injury remains controversial. Angiopoietin-1 (Ang-1), an endothelial growth factor, has been shown to have neuroprotective effects. The present study aimed to explore the neuroprotective mechanisms of Ang-1 in neuronal cells in a rat model of SCIR injury in vivo. Ang-1 protein and rapamycin were injected intrathecally. Basso Beattie Bresnahan (BBB) scoring and hematoxylin and eosin staining were used to assess the degree of SCIR injury. Proteins that reflected the level of autophagy expression, such as Beclin-1 and LC3, were evaluated by western blotting. The results indicated that SCIR injury resulted in loss in lower limb motor function. Ang-1 protein inhibited the expression of Beclin-1 and LC3, which improved the BBB score and alleviated spinal cord injury. In contrast, rapamycin, an autophagy activator, caused the opposite effect. This study provides evidence that Ang-1 plays a neuroprotective role by inhibiting of autophagy expression in SCIR injury. Overall, findings could be useful for the treatment of SCIR injury.
Collapse
Affiliation(s)
- Jian Yin
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, People's Republic of China
| | - Zhaoyang Yin
- Department of Orthopedics, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First People's Hospital of Lianyungang, Lianyungang, 222000, People's Republic of China
| | - Bin Wang
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, People's Republic of China
| | - Chao Zhu
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, People's Republic of China
| | - Chao Sun
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, People's Republic of China
| | - Xinhui Liu
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, People's Republic of China.
| | - Ge Gong
- Department of Geriatrics, Jinling Hospital, Medical School of Nanjing University, 211002, Nanjing, People's Republic of China.
| |
Collapse
|
17
|
Dar WA, Sullivan E, Bynon JS, Eltzschig H, Ju C. Ischaemia reperfusion injury in liver transplantation: Cellular and molecular mechanisms. Liver Int 2019; 39:788-801. [PMID: 30843314 PMCID: PMC6483869 DOI: 10.1111/liv.14091] [Citation(s) in RCA: 259] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 02/13/2019] [Accepted: 02/22/2019] [Indexed: 12/11/2022]
Abstract
Liver disease causing end organ failure is a growing cause of mortality. In most cases, the only therapy is liver transplantation. However, liver transplantation is a complex undertaking and its success is dependent on a number of factors. In particular, liver transplantation is subject to the risks of ischaemia-reperfusion injury (IRI). Liver IRI has significant effects on the function of a liver after transplantation. The cellular and molecular mechanisms governing IRI in liver transplantation are numerous. They involve multiple cells types such as liver sinusoidal endothelial cells, hepatocytes, Kupffer cells, neutrophils and platelets acting via an interconnected network of molecular pathways such as activation of toll-like receptor signalling, alterations in micro-RNA expression, production of ROS, regulation of autophagy and activation of hypoxia-inducible factors. Interestingly, the cellular and molecular events in liver IRI can be correlated with clinical risk factors for IRI in liver transplantation such as donor organ steatosis, ischaemic times, donor age, and donor and recipient coagulopathy. Thus, understanding the relationship of the clinical risk factors for liver IRI to the cellular and molecular mechanisms that govern it is critical to higher levels of success after liver transplantation. This in turn will help in the discovery of therapeutics for IRI in liver transplantation - a process that will lead to improved outcomes for patients suffering from end-stage liver disease.
Collapse
Affiliation(s)
- Wasim A. Dar
- Department of Surgery, McGovern Medical School at UT Health, Houston, TX
| | - Elise Sullivan
- Department of Anesthesia, McGovern Medical School at UT Health, Houston, TX
| | - John S. Bynon
- Department of Surgery, McGovern Medical School at UT Health, Houston, TX
| | - Holger Eltzschig
- Department of Anesthesia, McGovern Medical School at UT Health, Houston, TX
| | - Cynthia Ju
- Department of Anesthesia, McGovern Medical School at UT Health, Houston, TX
| |
Collapse
|
18
|
Wang J, Wu D, Wang H. Hydrogen sulfide plays an important protective role by influencing autophagy in diseases. Physiol Res 2019; 68:335-345. [PMID: 30904008 DOI: 10.33549/physiolres.933996] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Autophagy can regulate cell growth, proliferation, and stability of cell environment. Its dysfunction can be involved in a variety of diseases. Hydrogen sulfide (H(2)S) is an important signaling molecule that regulates many physiological and pathological processes. Recent studies indicate that H(2)S plays an important protective role in many diseases through influencing autophagy, but its mechanism is not fully understood. This article reviewed the progress about the effect of H(2)S on autophagy in diseases in recent years in order to provide theoretical basis for the further research on the interaction of H(2)S and autophagy and the mechanisms involved.
Collapse
Affiliation(s)
- J Wang
- School of Basic Medical Science, Henan University, Kaifeng, Henan, China.
| | | | | |
Collapse
|
19
|
Yu Q, Wu L, Liu T, Li S, Feng J, Mao Y, Fan X, Guo C, Wu J. Protective effects of levo-tetrahydropalmatine on hepatic ischemia/reperfusion injury are mediated by inhibition of the ERK/NF-κB pathway. Int Immunopharmacol 2019; 70:435-445. [PMID: 30856394 DOI: 10.1016/j.intimp.2019.02.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/13/2019] [Accepted: 02/13/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Hepatic ischemia/reperfusion (IR) injury is a common medical phenomenon that occurs during a number of clinical conditions, such as liver transplantation, severe injuries, and shock. In our study, we determined the protective functions of levo-tetrahydropalmatine (L-THP) on hepatic IR injury in mice by inhibiting the ERK/NF-κB signaling pathway. METHOD BALB/c mice were randomly divided into six groups as follows: normal control (NC); sham; L-THP (40 mg/kg); IR; L-THP (20 mg/kg) + IR; and L-THP (40 mg/kg) + IR. Liver tissues and sera were collected at three time points after reperfusion (2, 8, and 24 h). The liver enzyme, inflammatory factor, and other protein levels in the serum and liver tissues were detected. RESULTS L-THP pretreatment alleviated hepatocyte injury caused by IR and reduced the production of proinflammatory cytokines, such as IL-6 and TNF-α. Furthermore, L-THP could inhibit the ERK/NF-κB signaling pathway to attenuate hepatocyte apoptosis and autophagy. And the protective effect of L-THP is positively correlated with its dose. CONCLUSION L-THP protects the liver from IR injury by inhibiting the release of inflammatory factors and alleviating liver cell apoptosis and autophagy. The protective functions of L-THP may be partly based on the downregulation of the ERK/NF-κB pathway.
Collapse
Affiliation(s)
- Qiang Yu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, China; Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Tong Liu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Sainan Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yuqing Mao
- Department of Gerontology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China.
| | - Xiaoming Fan
- Department of Gastroenterology, Jinshan Hospital of Fudan University, Jinshan, Shanghai 201508, China
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, China; Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, China.
| |
Collapse
|
20
|
Zhang W, Yang C, Guo W, Guo X, Bian J, Zhou Q, Chen M, Zhou J, Chen Z, Wang P, Lv X, Xiao Z, Liu C. [rotective effect of bone marrow mesenchymal stem cells-derived exosomes against testicular ischemia-reperfusion injury in rats]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 38:910-916. [PMID: 30187884 DOI: 10.3969/j.issn.1673-4254.2018.08.02] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE To investigate the protective effect of bone marrow mesenchymal stem cells (BMSCs)-derived exosomesagainst testicular ischemia-reperfusion injury (IRI) in rats. METHODS Rat BMSCs were isolated, cultured and identified in theprimary culture. The exosomes were extracted from the BMSCs and characterized using nanoparticle tracking analysis, transmission electron microscopy, and Western blotting. Twenty-four healthy male SD rats were randomly divided into shamoperation group, testicular IRI with saline treatment group and IRI with exosome treatment group. The contralateral testes ofthe rats were collected for pathological observation, aseessment of superoxide dismutase (SOD) and malondialdehyde (MDA), and detection of HMGB1, caspases-3 and cleaved caspase-3 expressions using Western blotting. RESULTS We successfullyobtained exosomes from rat BMSCs. Testicular IRI significantly impaired testicular spermatogenesis, which was markedlyimproved by treatment with the exosomes (P < 0.05). Testicular IRI also caused significant increase in the protein expression ofHMGB1, caspase-3 and cleaved caspase-3 in the testicular tissue, and treatment with the exosomes obviously amelioratedthese changes (P < 0.05). CONCLUSIONS BMSCs-derived exosomes protects against testicular IRI due to the anti-oxidant, antiinflammatory and anti-apoptosis activities of the exosomes.
Collapse
Affiliation(s)
- Wansong Zhang
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Cheng Yang
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Wenbin Guo
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Xiaobin Guo
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Jun Bian
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Qizhao Zhou
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Mingkun Chen
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Junhao Zhou
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Zijian Chen
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Peng Wang
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Xianyuan Lv
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Zhuoyu Xiao
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Cundong Liu
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| |
Collapse
|
21
|
Chakhtoura M, Chain RW, Sato PY, Qiu CC, Lee MH, Meissler JJ, Eisenstein TK, Koch WJ, Caricchio R, Gallucci S. Ethyl Pyruvate Modulates Murine Dendritic Cell Activation and Survival Through Their Immunometabolism. Front Immunol 2019; 10:30. [PMID: 30761126 PMCID: PMC6362406 DOI: 10.3389/fimmu.2019.00030] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/08/2019] [Indexed: 12/16/2022] Open
Abstract
Attenuating the innate immunity activation could ameliorate inflammation and disease in settings such as transplant rejection or autoimmunity. Recently, a pivotal role for metabolic re-programming in TLR-induced dendritic cell (DC) activation has emerged. Ethyl pyruvate (EP), a pyruvate derivative, possesses anti-inflammatory properties in vitro and in animal models of disease. However, its effects on DCs remain elusive. We found that EP attenuated LPS-induced activation of murine GM-CSF bone marrow-derived dendritic cells (DCs) in vitro, reducing pro-inflammatory cytokine and IL-10 production, costimulatory molecule and MHC expression, the type I Interferon (IFN-I) response, the LPS-induced cell death, and the ability of DCs to stimulate allogeneic T cells. DC activation induced by TLR7 and TLR9 ligands was also suppressed by EP in vitro. Finally, EP decreased TLR-induced activation stimulated in vivo in conventional DCs and inflammatory monocytes. Investigating EP mechanisms, we found that EP decreased glycolysis and mitochondrial respiration, upon and in absence of TLR stimulation, by reducing ERK, AKT, and nitric oxide (NO) activation. These results indicate that EP inhibits most of the DC biological responses to TLR triggering, altering the metabolic reprogramming necessary for DC activation.
Collapse
Affiliation(s)
- Marita Chakhtoura
- Laboratory of Dendritic Cell Biology, Department of Microbiology-Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Robert W Chain
- Laboratory of Dendritic Cell Biology, Department of Microbiology-Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Priscila Y Sato
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Connie C Qiu
- Laboratory of Dendritic Cell Biology, Department of Microbiology-Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Michael H Lee
- Laboratory of Dendritic Cell Biology, Department of Microbiology-Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Joseph J Meissler
- Department of Microbiology-Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Toby K Eisenstein
- Department of Microbiology-Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Walter J Koch
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Roberto Caricchio
- Division of Rheumatology, Department of Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Stefania Gallucci
- Laboratory of Dendritic Cell Biology, Department of Microbiology-Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
22
|
Abstract
High-mobility group box 1 (HMGB1) is one of the most abundant proteins in eukaryotes and the best characterized damage-associated molecular pattern (DAMP). The biological activities of HMGB1 depend on its subcellular location, context and post-translational modifications. Inside the nucleus, HMGB1 is engaged in many DNA events such as DNA repair, transcription regulation and genome stability; in the cytoplasm, its main function is to regulate the autophagic flux while in the extracellular environment, it possesses more complicated functions and it is involved in a large variety of different processes such as inflammation, migration, invasion, proliferation, differentiation and tissue regeneration. Due to this pleiotropy, the role of HMGB1 has been vastly investigated in various pathological diseases and a large number of studies have explored its function in cardiovascular pathologies. However, in this contest, the precise mechanism of action of HMGB1 and its therapeutic potential are still very controversial since is debated whether HMGB1 is involved in tissue damage or plays a role in tissue repair and regeneration. The main focus of this review is to provide an overview of the effects of HMGB1 in different ischemic heart diseases and to discuss its functions in these pathological conditions.
Collapse
|
23
|
Deng J, Feng J, Liu T, Lu X, Wang W, Liu N, Lv Y, Liu Q, Guo C, Zhou Y. Beraprost sodium preconditioning prevents inflammation, apoptosis, and autophagy during hepatic ischemia-reperfusion injury in mice via the P38 and JNK pathways. Drug Des Devel Ther 2018; 12:4067-4082. [PMID: 30568428 PMCID: PMC6276616 DOI: 10.2147/dddt.s182292] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE The goal of this study was to determine the effects of beraprost sodium (BPS) preconditioning on hepatic ischemia-reperfusion (IR) injury and its underlying mechanisms of action. MATERIALS AND METHODS Mice were randomly divided into sham, IR, IR+BPS (50 µg/kg), and IR+BPS (100 µg/kg) groups. Saline or BPS was given to the mice by daily gavage for 1 week before the hepatic IR model was established. Liver tissues and orbital blood were collected at 2, 8, and 24 hours after reperfusion for the determination of liver enzymes, inflammatory mediators, apoptosis- and autophagy-related proteins, key proteins in P38 and c-Jun N-terminal kinase (JNK) cascades, and evaluation of liver histopathology. RESULTS BPS preconditioning effectively reduced serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, improved pathological damage, ameliorated production of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), and affected expressions of Bax, Bcl-2, Caspase-3, Caspase-8, and Caspase-9, microtubule-associated protein 1 light chain 3 (LC3), Beclin-1, and P62. The protective effects of BPS preconditioning were associated with reduced P38 and JNK phosphorylation. CONCLUSION BPS preconditioning ameliorated hepatic IR injury by suppressing inflammation, apoptosis, and autophagy, partially via inhibiting activation of the P38 and JNK cascades.
Collapse
Affiliation(s)
- Jingfan Deng
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, People’s Republic of China, ;
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, People’s Republic of China, ;
| | - Tong Liu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, People’s Republic of China, ;
| | - Xiya Lu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, People’s Republic of China, ;
| | - Wenwen Wang
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, People’s Republic of China, ;
| | - Ning Liu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, People’s Republic of China
| | - Yang Lv
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, People’s Republic of China, ;
| | - Qing Liu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, People’s Republic of China, ;
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, People’s Republic of China, ; ,Correspondence: Chuanyong Guo; Yingqun Zhou, Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang Road, Jing’an, Shanghai 200072, People’s Republic of China, Tel +86 21 6630 2535; +86 21 3605 0414, Fax +86 21 6630 3983, Email ;
| | - Yingqun Zhou
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, People’s Republic of China, ; ,Correspondence: Chuanyong Guo; Yingqun Zhou, Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Number 301, Middle Yanchang Road, Jing’an, Shanghai 200072, People’s Republic of China, Tel +86 21 6630 2535; +86 21 3605 0414, Fax +86 21 6630 3983, Email ;
| |
Collapse
|
24
|
Zhao J, Cheng J, Li C, Xu M, Ma C, Qin L, Yi K, Liao N. Ethyl Pyruvate Attenuates CaCl 2-Induced Tubular Epithelial Cell Injury by Inhibiting Autophagy and Inflammatory Responses. Kidney Blood Press Res 2018; 43:1585-1595. [DOI: 10.1159/000494445] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 10/12/2018] [Indexed: 11/19/2022] Open
|
25
|
Li X, Feng D, Chen G. An Update On Medical Treatment for Intracerebral Hemorrhage. Transl Stroke Res 2018; 9:10.1007/s12975-018-0664-5. [PMID: 30206812 DOI: 10.1007/s12975-018-0664-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Dongxia Feng
- Department of Neurosurgery, Baylor Scott & White Medical Center, Texas A&M University College of Medicine, Temple, 76508, USA
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China.
| |
Collapse
|
26
|
Palanissami G, Paul SFD. RAGE and Its Ligands: Molecular Interplay Between Glycation, Inflammation, and Hallmarks of Cancer—a Review. Discov Oncol 2018; 9:295-325. [DOI: 10.1007/s12672-018-0342-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022] Open
|
27
|
Mennander AA. Time to celebrate! J Thorac Cardiovasc Surg 2018; 155:1659-1660. [DOI: 10.1016/j.jtcvs.2017.11.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 11/13/2017] [Indexed: 10/18/2022]
|
28
|
Wagner N, Dieteren S, Franz N, Köhler K, Mörs K, Nicin L, Schmidt J, Perl M, Marzi I, Relja B. Ethyl pyruvate ameliorates hepatic injury following blunt chest trauma and hemorrhagic shock by reducing local inflammation, NF-kappaB activation and HMGB1 release. PLoS One 2018; 13:e0192171. [PMID: 29420582 PMCID: PMC5805235 DOI: 10.1371/journal.pone.0192171] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 01/17/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The treatment of patients with multiple trauma including blunt chest/thoracic trauma (TxT) and hemorrhagic shock (H) is still challenging. Numerous studies show detrimental consequences of TxT and HS resulting in strong inflammatory changes, organ injury and mortality. Additionally, the reperfusion (R) phase plays a key role in triggering inflammation and worsening outcome. Ethyl pyruvate (EP), a stable lipophilic ester, has anti-inflammatory properties. Here, the influence of EP on the inflammatory reaction and liver injury in a double hit model of TxT and H/R in rats was explored. METHODS Female Lewis rats were subjected to TxT followed by hemorrhage/H (60 min, 35±3 mm Hg) and resuscitation/R (TxT+H/R). Reperfusion was performed by either Ringer`s lactated solution (RL) alone or RL supplemented with EP (50 mg/kg). Sham animals underwent all surgical procedures without TxT+H/R. After 2h, blood and liver tissue were collected for analyses, and survival was assessed after 24h. RESULTS Resuscitation with EP significantly improved haemoglobin levels and base excess recovery compared with controls after TxT+H/R, respectively (p<0.05). TxT+H/R-induced significant increase in alanine aminotransferase levels and liver injury were attenuated by EP compared with controls (p<0.05). Local inflammation as shown by increased gene expression of IL-6 and ICAM-1, enhanced ICAM-1 and HMGB1 protein expression and infiltration of the liver with neutrophils were also significantly attenuated by EP compared with controls after TxT+H/R (p<0.05). EP significantly reduced TxT+H/R-induced p65 activation in liver tissue. Survival rates improved by EP from 50% to 70% after TxT+H/R. CONCLUSIONS These data support the concept that the pronounced local pro-inflammatory response in the liver after blunt chest trauma and hemorrhagic shock is associated with NF-κB. In particular, the beneficial anti-inflammatory effects of ethyl pyruvate seem to be regulated by the HMGB1/NF-κB axis in the liver, thereby, restraining inflammatory responses and liver injury after double hit trauma in the rat.
Collapse
Affiliation(s)
- Nils Wagner
- Department of Trauma Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany
| | - Scott Dieteren
- Department of Trauma Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany
| | - Niklas Franz
- Department of Trauma Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany
| | - Kernt Köhler
- Institute of Veterinary Pathology, Justus Liebig University Giessen, Giessen, Germany
| | - Katharina Mörs
- Department of Trauma Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany
| | - Luka Nicin
- Department of Trauma Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany
| | - Julia Schmidt
- Department of Trauma Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany
| | - Mario Perl
- BG-Trauma Center Murnau, Murnau, Germany
| | - Ingo Marzi
- Department of Trauma Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany
| | - Borna Relja
- Department of Trauma Surgery, University Hospital Frankfurt, Goethe-University, Frankfurt, Germany
- * E-mail:
| |
Collapse
|
29
|
Zhang M, Hu X, Li S, Lu C, Li J, Zong Y, Qi W, Yang H. Hepatoprotective effects of ethyl pyruvate against CCl4-induced hepatic fibrosis via inhibition of TLR4/NF-κB signaling and up-regulation of MMPs/TIMPs ratio. Clin Res Hepatol Gastroenterol 2018; 42:72-81. [PMID: 28601590 DOI: 10.1016/j.clinre.2017.04.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 04/01/2017] [Accepted: 04/12/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIM Liver fibrosis is a worldwide clinical issue. It has been well established that liver fibrosis is characterized of excessive extracellular matrix (ECM) deposition in chronically damaged livers. Accumulating evidences have suggested that ethyl pyruvate (EP) is a potential useful agent for preventing from hepatic injury. The aim of this study was to evaluate the protective effects of the EP against liver fibrosis induced by carbon tetrachloride (CCl4) in rats. METHOD Rats were randomly divided into control group, CCl4 group and CCl4+EP group, which with and without EP administration. Liver fibrosis was evaluated by serum biochemical parameters levels, Masson's trichromic staining and immunohistochemistry. Q-RTPCR was used to indicate genes expression. ELISA was used to detect proteins level. RESULTS This study demonstrates that Toll-like receptors 4 (TLR4)/nuclear factor kappa B (NF-κB) signal is an important regulator of liver fibrosis while TLR4/NF-κB mRNA and protein levels reduced during HSCs activation. In addition, down-regulated high-mobility group box 1 (HMGB1) expression reduced NF-κB transcription and phosphorylation, which inhibited HSCs activation by blocking the TLR4 signal. Moreover, EP contributed to an increase in the ratio of matrix metalloproteinase (MMPs) to tissue inhibitor of matrix metalloproteinase (TIMPs), which might facilitate the degradation of the ECM. In CCl4-induced liver fibrosis rats, additional EP injection resulted in decreased ECM deposition and improved liver function. CONCLUSION In conclusion, the present findings indicated that EP might be an effective agent for anti-fibrotic therapy.
Collapse
Affiliation(s)
- Mingxiao Zhang
- Department of Morphology Lab, Chengdu Medical College, Sichuan Chengdu, China; Luoyang Vocational and Technical College, Luoyang, Henan, China.
| | - Xiaosong Hu
- Department of Morphology Lab, Chengdu Medical College, Sichuan Chengdu, China.
| | - Shuai Li
- Department of Morphology Lab, Chengdu Medical College, Sichuan Chengdu, China
| | - Chen Lu
- Department of Morphology Lab, Chengdu Medical College, Sichuan Chengdu, China
| | - Juan Li
- Department of Morphology Lab, Chengdu Medical College, Sichuan Chengdu, China
| | - Yonghua Zong
- Department of Morphology Lab, Chengdu Medical College, Sichuan Chengdu, China
| | - Wenqian Qi
- Department of Morphology Lab, Chengdu Medical College, Sichuan Chengdu, China
| | - Huijun Yang
- Department of Morphology Lab, Chengdu Medical College, Sichuan Chengdu, China.
| |
Collapse
|
30
|
Kayiran O, Cuzdan SS, Uysal A, Kocer U. Ethyl pyruvate improves skin flap survival after ischaemia reperfusion injury. Indian J Med Res 2018; 146:369-374. [PMID: 29355144 PMCID: PMC5793472 DOI: 10.4103/ijmr.ijmr_1428_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND & OBJECTIVES Reperfusion after ischaemia is detrimental to the tissues. The oxidative stress created and cytokines released are mostly responsible in this process. In this study, ethyl pyruvate, a known agent for its anti-inflammatory and antioxidant properties, was used to investigate the effects on ischaemia/reperfusion injury on skin island flaps in rats. METHODS Sixty rats were randomly distributed in three groups (non-ischaemic, ischaemic and medication groups). Ethyl pyruvate was administered in the medication group with a dose of 50 mg/kg. After 24 h and one week, the animals were sacrificed, and the flaps were analyzed macroscopically, histopathologically, biochemically (total nitrite, malondialdehyde and myeloperoxidase). RESULTS Biochemical markers indicating oxidative stress, were found elevated in ischaemic group, whereas medication with ethyl pyruvate significantly reduced these values. There was a significant reduction (P<0.05) in the levels of these markers between ischaemic and medication groups. Ethyl pyruvate improved all the parameters significantly. INTERPRETATION & CONCLUSION Ethyl pyruvate showed strong scavenger activity against reactive oxygen species. It could be a potential candidate to improve the flap viability in reconstructive microsurgery, especially in free tissue transfers. However, more studies are warranted in experimental models to confirm these findings.
Collapse
Affiliation(s)
- Oguz Kayiran
- Department Plastic & Reconstructive Surgery, Izmir University, Izmir, Turkey
| | | | - Afsin Uysal
- Plastic & Reconstructive Surgery Clinic, TOBB ETU Hospital, Ankara, Turkey
| | - Ugur Kocer
- Plastic & Reconstructive Surgery Clinic, Ankara Training & Research Hospital, Ankara, Turkey
| |
Collapse
|
31
|
Chen C, Chen W, Li Y, Dong Y, Teng X, Nong Z, Pan X, Lv L, Gao Y, Wu G. Hyperbaric oxygen protects against myocardial reperfusion injury via the inhibition of inflammation and the modulation of autophagy. Oncotarget 2017; 8:111522-111534. [PMID: 29340072 PMCID: PMC5762340 DOI: 10.18632/oncotarget.22869] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/27/2017] [Indexed: 01/01/2023] Open
Abstract
Our previous study demonstrated that hyperbaric oxygen (HBO) preconditioning protected against myocardial ischemia reperfusion injury (MIRI) and improved myocardial infarction. However, HBO’s effect on MIRI-induced inflammation and autophagy remains unclear. In this study, we investigate the potential impact and underlying mechanism of HBO preconditioning on an MIRI-induced inflammatory response and autophagy using a ligation of the left anterior descending (LAD) coronary artery rat model. Our results showed that HBO restored myocardial enzyme levels and decreased the apoptosis of cardiomyocytes, which were induced by MIRI. Moreover, HBO significantly suppressed MIRI-induced inflammatory cytokines. This effect was associated with the inhibition of the TLR4-nuclear factor kappa-B (NF-κB) pathway. Interestingly, lower expression levels of microtubule-associated protein 1 light chain 3B (LC3B) and Beclin-1 were observed in the HBO-treatment group. Furthermore, we observed that HBO reduced excessive autophagy by activating the mammalian target of the rapamycin (mTOR) pathway, as evidenced by higher expression levels of threonine protein kinase (Akt) and phosphorylated-mTOR. In conclusion, HBO protected cardiomocytes during MIRI by attenuating inflammation and autophagy. Our results provide a new mechanistic insight into the cardioprotective role of HBO against MIRI.
Collapse
Affiliation(s)
- Chunxia Chen
- Department of Hyperbaric Oxygen, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Wan Chen
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Yaoxuan Li
- Department of Neurology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Yanling Dong
- Department of Neurology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Xiaoming Teng
- Department of Neurology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Zhihuan Nong
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, Guangxi 530022, P. R. China
| | - Xiaorong Pan
- Department of Hyperbaric Oxygen, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Liwen Lv
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Ying Gao
- Department of Biology and Tennessee Center for Botanical Medicine Research, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| | - Guangwei Wu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| |
Collapse
|
32
|
Wu L, Wang C, Li J, Li S, Feng J, Liu T, Xu S, Wang W, Lu X, Chen K, Xia Y, Fan X, Guo C. Hepatoprotective effect of quercetin via TRAF6/JNK pathway in acute hepatitis. Biomed Pharmacother 2017; 96:1137-1146. [PMID: 29174851 DOI: 10.1016/j.biopha.2017.11.109] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/12/2017] [Accepted: 11/20/2017] [Indexed: 01/08/2023] Open
Abstract
Quercetin, as a member of the flavonoids family, has many beneficial properties. The aim of our study was to evaluate the protective effect of quercetin in ConA-induced hepatitis in mice, and to clarify its mechanism of action. Hepatitis was induced by using ConA (25 mg/kg), and quercetin was administered intragastrically at the dose of 100 mg/kg or 200 mg/kg for 5 days before ConA injection. The serum levels of liver enzymes, inflammatory cytokines and other marker proteins were determined at 2 h, 8 h and 24 h after ConA injection. Following ConA injection, serum levels of liver enzymes and inflammatory cytokines were significantly increased. Quercetin ameliorated liver damage and histopathological changes, and suppressed the release of inflammatory cytokines. The expression of Bax, Bcl-2, Beclin-1, LC3, P62 and caspase 9 were markedly affected by quercetin pretreatment. The expression of TRAF6 and p-JNK were decreased in the quercetin groups. Quercetin attenuated apoptosis and autophagy in ConA-induced autoimmune hepatitis by inhibiting TRAF6/JNK pathway.
Collapse
Affiliation(s)
- Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Chengfen Wang
- Putuo District People's Hospital, Tongji University School of Medicine, Shanghai 200060, China.
| | - Jingjing Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Sainan Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Tong Liu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Shizan Xu
- Department of Gastroenterology, Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China.
| | - Wenwen Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Xiya Lu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Kan Chen
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Yujing Xia
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| | - Xiaoming Fan
- Department of Gastroenterology, Jinshan Hospital of Fudan University, Jinshan, Shanghai 201508, China.
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| |
Collapse
|
33
|
Soh S, Jun JH, Song JW, Shin EJ, Kwak YL, Shim JK. Ethyl pyruvate attenuates myocardial ischemia-reperfusion injury exacerbated by hyperglycemia via retained inhibitory effect on HMGB1. Int J Cardiol 2017; 252:156-162. [PMID: 29169909 DOI: 10.1016/j.ijcard.2017.11.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/19/2017] [Accepted: 11/13/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Hyperglycemia (HG) exacerbates myocardial ischemia/reperfusion (I/R) injury and renders protective strategies ineffective by amplified inflammatory response via enhanced high-mobility group box-1 (HMGB1) release. This study investigated the role of ethyl pyruvate (EP) against myocardial I/R injury under a clinically relevant HG condition. METHODS Sprague-Dawley rats (n=76) were randomly assigned to 6 groups: normoglycemia (NG)-Sham, NG-I/R-control (C, saline), NG-I/R-EP treatment (50mg/kg) upon reperfusion, HG-Sham, HG-I/R-C, and HG-I/R-EP treatment upon reperfusion. HG was induced by 1.2g/kg dextrose. I/R was induced by ligation of the left anterior descending artery for 30min followed by 4h of reperfusion. RESULTS HG resulted in exacerbation of myocardial infarct size by 19% with amplified activation of HMGB1-receptors of advanced glycation end products/toll like receptors-NF-κB pathway compared to NG following I/R, which all could be attenuated by EP. EP treatment was associated with diminished tumor necrosis factor-α, interleukin-1β, and interleukin-6 expressions. It also served to normalize the increase in pro-apoptotic Bax and the decrease in anti-apoptotic Bcl-2 protein levels. These effects were associated with decreased myocardial apoptosis and infarct size (by 30% and 36% in the NG and HG groups, respectively) regardless of the glycemic condition. CONCLUSION HG exacerbated myocardial I/R injury through amplified inflammatory response via increased HMGB1 level. EP treatment upon reperfusion conveyed significant myocardial protection against the I/R injury under both NG and HG conditions. Common to both glycemic conditions, associated mechanisms involved attenuated increase in HMGB1 level and suppression of its down-stream pathways.
Collapse
Affiliation(s)
- Sarah Soh
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea; Severance Cardiovascular Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - Ji Hae Jun
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - Jong Wook Song
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea; Severance Cardiovascular Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea; Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - Eun-Jung Shin
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - Young-Lan Kwak
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea; Severance Cardiovascular Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea; Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - Jae-Kwang Shim
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea; Severance Cardiovascular Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea; Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea.
| |
Collapse
|
34
|
Zenerino C, Nuzzo AM, Giuffrida D, Biolcati M, Zicari A, Todros T, Rolfo A. The HMGB1/RAGE Pro-Inflammatory Axis in the Human Placenta: Modulating Effect of Low Molecular Weight Heparin. Molecules 2017; 22:molecules22111997. [PMID: 29149067 PMCID: PMC6150179 DOI: 10.3390/molecules22111997] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/10/2017] [Accepted: 11/14/2017] [Indexed: 01/11/2023] Open
Abstract
We evaluated whether physiological and pre-eclamptic (PE) placentae, characterized by exacerbated inflammation, presented alterations in pro-inflammatory High Mobility Group Box 1 (HMGB1) and its Receptor of Advanced Glycation End products (RAGE) expression. Moreover, we investigated, in physiological placental tissue, the ability of Low Molecular Weight Heparin (LMWH) to modify HMGB1 structural conformation thus inhibiting RAGE binding and HMGB1/RAGE axis inflammatory activity. HMGB1, RAGE, IL-6 and TNFα (HMGB1/RAGE targets) mRNA expression were assessed by Real Time PCR. HMGB1, RAGE protein levels were assessed by western blot assay. Physiological term placental explants were treated by 0.5 U LMWH for 24 or 48 h. HMGB1 and RAGE expression and association were evaluated in LMWH explants by RAGE immunoprecipitation followed by HMGB1 immunoblot. HMGB1 spatial localization was evaluated by immuofluorescent staining (IF). HMGB1 expression was increased in PE relative to physiological placentae while RAGE was unvaried. 24 h LMWH treatment significantly up-regulated HMGB1 expression but inhibited HMGB1/RAGE complex formation in physiological explants. RAGE expression decreased in treated relative to untreated explants at 48 h. IF showed HMGB1 localization in both cytoplasm and nucleus of mesenchymal and endothelial cells but not in the trophoblast. IL-6 and TNFα gene expression were significantly increased at 24 h relative to controls, while they were significantly down-regulated in 48 h vs. 24 h LMWH explants. Our data depicted a new molecular mechanism through which LMWH exerts its anti-inflammatory effect on PE placentae, underlying the importance of HMGB1/RAGE axis in PE inflammatory response.
Collapse
Affiliation(s)
- Cristian Zenerino
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy.
| | - Anna Maria Nuzzo
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy.
| | - Domenica Giuffrida
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy.
| | - Marilisa Biolcati
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy.
| | - Alessandra Zicari
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy.
| | - Tullia Todros
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy.
| | - Alessandro Rolfo
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy.
| |
Collapse
|
35
|
Jun JH, Song JW, Shin EJ, Kwak YL, Choi N, Shim JK. Ethyl pyruvate is renoprotective against ischemia-reperfusion injury under hyperglycemia. J Thorac Cardiovasc Surg 2017; 155:1650-1658. [PMID: 29195627 DOI: 10.1016/j.jtcvs.2017.10.069] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 10/03/2017] [Accepted: 10/23/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hyperglycemia (HG) is common in cardiovascular surgeries due to diabetes, inflammation, and the neuroendocrine stress response. HG aggravates renal ischemia-reperfusion (I/R) injury through an increased inflammatory response, and blunts the protective effect of various measures. Ethyl pyruvate (EP) provides anti-inflammatory effects against I/R injury via inhibition of high-mobility group box 1 protein (HMGB1) release. This study aimed to determine the renoprotective effect of EP against I/R injury under HG. METHODS Sprague-Dawley rats were randomly assigned at random to 8 groups: normoglycemia (NG)-sham, NG-I/R-control, NG-EP-I/R (pretreatment), NG-I/R-EP (posttreatment), HG-sham, HG-I/R-control, HG-EP-I/R, and HG-I/R-EP. Renal I/R was induced by 45 minutes of ischemia (clamping of renal arteries), followed by 24 hours of reperfusion. EP (50 mg/kg) was administered intraperitoneally at 1 h before ischemia (pretreatment) or on reperfusion (posttreatment). RESULTS I/R injury under HG significantly aggravated the degree of renal tubular apoptosis and damage compared with the NG groups, which could be attenuated by both pretreatment and posttreatment of EP. I/R-induced increases in HMGB1 and Toll-like receptors (TLRs), activation of NF-kB, and resultant alterations in interleukin-1β, tumor necrosis factor-α, proapoptotic Bax, and antiapoptotic Bcl-2 were all favorably modulated by EP treatment in both the NG and HG groups compared with their corresponding control groups. CONCLUSIONS Despite aggravation of renal I/R injury by HG through amplified inflammation, EP administration showed similar suppression of the HMGB1-TLR-NF-kB pathway in the HG and NG groups. EP retained anti-inflammatory, antiapoptotic, and renoprotective effects in the HG groups, whether administered before ischemia or on reperfusion.
Collapse
Affiliation(s)
- Ji Hae Jun
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Wook Song
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun-Jung Shin
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Lan Kwak
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Nakcheol Choi
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Kwang Shim
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Wu L, Zhang Q, Dai W, Li S, Feng J, Li J, Liu T, Xu S, Wang W, Lu X, Yu Q, Chen K, Xia Y, Lu J, Zhou Y, Fan X, Guo C. Quercetin Pretreatment Attenuates Hepatic Ischemia Reperfusion-Induced Apoptosis and Autophagy by Inhibiting ERK/NF- κB Pathway. Gastroenterol Res Pract 2017; 2017:9724217. [PMID: 29123547 PMCID: PMC5662816 DOI: 10.1155/2017/9724217] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/20/2017] [Accepted: 06/12/2017] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Hepatic ischemia reperfusion (IR) injury is a common phenomenon in transplantation or trauma. The aim of the present study was to determine the protective effect of quercetin (QE) on hepatic IR injury via the ERK/NF-κB pathway. METHODS Mice were randomized into the sham, IR, QE100 + IR, and QE200 + IR groups. Quercetin was administered intragastrically daily at two doses (100 mg/kg and 200 mg/kg) for 5 days prior to IR injury. The expression levels of liver enzymes, inflammatory cytokines, and other marker proteins were determined at 2, 8, and 24 hours after IR. And they were compared among these groups. RESULTS Compared with the IR group, the treatment of QE reduced the release of cytokines, leading to inhibition of apoptosis and autophagy via downregulation of the ERK/NF-κB pathway in this model of hepatic IR injury. CONCLUSION Apoptosis and autophagy caused by hepatic IR injury were inhibited by QE following a reduction in the release of inflammatory cytokines, and the relationship between the two may be associated with inactivation of the ERK/NF-κB pathway.
Collapse
Affiliation(s)
- Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Qinghui Zhang
- Department of Clinical Laboratory, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215300, China
| | - Weiqi Dai
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Sainan Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jingjing Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Tong Liu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Shizan Xu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China
| | - Wenwen Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiya Lu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Qiang Yu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China
| | - Kan Chen
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yujing Xia
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jie Lu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yingqun Zhou
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiaoming Fan
- Department of Gastroenterology, Jinshan Hospital of Fudan University, Jinshan, Shanghai 201508, China
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
37
|
Jung SM, Lee J, Baek SY, Lee J, Jang SG, Hong SM, Park JS, Cho ML, Park SH, Kwok SK. Ethyl pyruvate ameliorates inflammatory arthritis in mice. Int Immunopharmacol 2017; 52:333-341. [PMID: 28987932 DOI: 10.1016/j.intimp.2017.09.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Ethyl pyruvate (EP) is the ethyl ester of pyruvate and has antioxidative and anti-inflammatory effects. This study aimed to evaluate the therapeutic effect of EP in inflammatory arthritis and to identify the underlying mechanisms. METHODS Mice with collagen-induced arthritis (CIA) were treated with the vehicle control or EP at 20mg/kg, and clinical and histological analyses were performed on the animals. The differentiation of murine CD4+ T cells into T helper 17 (Th17) cells in the presence of EP was investigated in vitro. The effects of EP on osteoclastogenesis were determined by staining for tartrate-resistant acid phosphatase, and measuring the mRNA levels of osteoclastogenesis-related genes. The expression of high-mobility group box 1 (HMGB1) was evaluated after EP therapy using immunohistochemical staining and Western blotting. RESULTS EP significantly improved the clinical and histological features of arthritis in CIA mice. EP suppressed the differentiation of CD4+ T cells into Th17 cells, and inhibited the expression of RORγt. The generation of osteoclasts and osteoclastogenic markers from murine and human monocytes was significantly reduced in the presence of EP. The expression of HMGB1 in the synovium was significantly lower in CIA mice treated with EP, compared to control CIA mice. During osteoclastogenesis, HMGB1 release from monocytes was inhibited in the presence of EP. CONCLUSIONS EP attenuated synovial inflammation and bone destruction in the experimental arthritis model through suppression of IL-17 and HMGB-1. The data suggests that EP could be a novel therapeutic agent for the treatment of inflammatory arthritis, such as rheumatoid arthritis.
Collapse
Affiliation(s)
- Seung Min Jung
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jaeseon Lee
- Rheumatism Research Center, Catholic Institutes of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Ye Baek
- Rheumatism Research Center, Catholic Institutes of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Juhyun Lee
- Rheumatism Research Center, Catholic Institutes of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Se Gwang Jang
- Rheumatism Research Center, Catholic Institutes of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Min Hong
- Rheumatism Research Center, Catholic Institutes of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin-Sil Park
- Rheumatism Research Center, Catholic Institutes of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Institutes of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Rheumatism Research Center, Catholic Institutes of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Rheumatism Research Center, Catholic Institutes of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
38
|
Wang R, Shen Z, Yang L, Yin M, Zheng W, Wu B, Liu T, Song H. Protective effects of heme oxygenase-1-transduced bone marrow-derived mesenchymal stem cells on reduced‑size liver transplantation: Role of autophagy regulated by the ERK/mTOR signaling pathway. Int J Mol Med 2017; 40:1537-1548. [PMID: 28901391 PMCID: PMC5627878 DOI: 10.3892/ijmm.2017.3121] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 08/21/2017] [Indexed: 12/17/2022] Open
Abstract
Autophagy is a critical lysosomal pathway that degrades cytoplasmic components to maintain cell homeostasis and provide substrates for energy metabolism. A study revealed that heme oxygenase-1 (HO-1)-transduced bone marrow-derived mesenchymal stem cells (BM-MSCs) could protect 50% reduced-size liver transplantation (RSLT) in a rat model. However, the mechanisms remain mostly unknown. The aim of the present study was to explore the effects and related mechanism of autophagy on the protection conferred by HO-1-transduced BM-MSCs (HO-1/BM-MSCs) on 50% RSLT in a rat model. The authors established an acute rejection model following 50% RSLT in rats, with recipients divided into three groups receiving treatment with BM-MSCs, HO-1/BM-MSCs or normal saline (NS) injected through the dorsal penile vein. Transplanted liver tissues at 0, 1, 3, 5, 7, 10 and 14 days following transplantation were acquired for further analysis. The results indicated that the expression of autophagy-related proteins LC3 and Beclin-1 increased, the levels of ERK and p-ERK increased, and the levels of mammalian target of rapamycin (mTOR) and p-mTOR decreased in the HO-1/BM-MSCs. These observations indicated that autophagy is involved in the protective effects of HO-1/BM-MSCs on liver grafts following RSLT, possibly via upregulation of autophagy-related proteins through the ERK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Raorao Wang
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Zhongyang Shen
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Liu Yang
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Mingli Yin
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Weiping Zheng
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Bin Wu
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Tao Liu
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Hongli Song
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| |
Collapse
|
39
|
Li W, Lou J, Wei L, Bai H, Zhang Y, He Y. Ethyl pyruvate protects PC12 cells from oxygen-glucose deprivation: A potential role in ischemic cerebrovascular disease. Biomed Pharmacother 2017; 92:168-174. [DOI: 10.1016/j.biopha.2017.05.067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 04/26/2017] [Accepted: 05/12/2017] [Indexed: 02/02/2023] Open
|
40
|
Feng J, Zhang Q, Mo W, Wu L, Li S, Li J, Liu T, Xu S, Fan X, Guo C. Salidroside pretreatment attenuates apoptosis and autophagy during hepatic ischemia-reperfusion injury by inhibiting the mitogen-activated protein kinase pathway in mice. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:1989-2006. [PMID: 28721018 PMCID: PMC5501634 DOI: 10.2147/dddt.s136792] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ischemia–reperfusion injury (IRI) contributes to liver damage in many clinical situations, such as liver resection and liver transplantation. In the present study, we investigated the effects of the antioxidant, anti-inflammatory, and anticancer agent salidroside (Sal) on hepatic IRI in mice. The mice were randomly divided into six groups: normal control, Sham, Sal (20 mg/kg), IRI, IRI + Sal (10 mg/kg), and IRI + Sal (20 mg/kg). We measured liver enzymes, proinflammatory cytokines, TNF-α and interleukin-6, and apoptosis- and autophagy-related marker proteins at 2, 8, and 24 hours after reperfusion. Components of mitogen-activated protein kinase (MAPK) signaling, including P-38, jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK), were also measured using an MAPK activator anisomycin to deduce their roles in hepatic IRI. Our results show that Sal safely protects hepatocytes from IRI by reducing levels of liver enzymes in the serum. These findings were confirmed by histopathology. We concluded that Sal protects hepatocytes from IRI partly by inhibiting the activation of MAPK signaling, including the phosphorylation of P38, JNK, and ERK. This ameliorates inflammatory reactions, apoptosis, and autophagy in the mouse liver.
Collapse
Affiliation(s)
- Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai
| | - Qinghui Zhang
- Department of Clinical Laboratory, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, JiangSu
| | - Wenhui Mo
- Department of Gastroenterology, Minhang Hospital, Fudan University, Shanghai
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai
| | - Sainan Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai
| | - Jingjing Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai
| | - Tong Liu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai
| | - Shizan Xu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai
| | - Xiaoming Fan
- Department of Gastroenterology, Jinshan Hospital of Fudan University, Jinshan, Shanghai, China
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai
| |
Collapse
|
41
|
Abstract
Although acute liver failure (ALF) is a rare disease, it continues to have high mortality and morbidity rates due to its many causes. High mobility group box 1 (HMGB1), originally reported as a ubiquitous non-histone chromosomal protein, is a multi-functional protein with varying functions depending on its location, such as in the nucleus, cytoplasm and extracellular space. The role of extracellular HMGB1 as an inflammatory mediator has been well studied, and the elevation of serum HMGB1 has been reported in several diseases that are closely associated with ALF. Areas covered: In this review, we focus on the relationship between causes of acute liver failure, such as viral infection, drug-induced liver injury, ischemia/reperfusion injury, and acute-on-chronic liver failure, and the role of HMGB1. Furthermore, we also consolidate and summarize the current reports of HMGB1-targeting therapies in hepatic injury models. Expert commentary: HMGB1 could be a novel therapeutic candidate for ALF, and the clinical testing of HMGB1-targeting therapies for ALF patients is expected.
Collapse
Affiliation(s)
- Tetsu Yamamoto
- a Department of Digestive and General Surgery , Shimane University Faculty of Medicine , Izumo , Japan
| | - Yoshitsugu Tajima
- a Department of Digestive and General Surgery , Shimane University Faculty of Medicine , Izumo , Japan
| |
Collapse
|
42
|
Hydrogen Sulfide Inhibits Autophagic Neuronal Cell Death by Reducing Oxidative Stress in Spinal Cord Ischemia Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:8640284. [PMID: 28685010 PMCID: PMC5480044 DOI: 10.1155/2017/8640284] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 04/01/2017] [Accepted: 04/23/2017] [Indexed: 11/25/2022]
Abstract
Autophagy is upregulated in spinal cord ischemia reperfusion (SCIR) injury; however, its expression mechanism is largely unknown; moreover, whether autophagy plays a neuroprotective or neurodegenerative role in SCIR injury remains controversial. To explore these issues, we created an SCIR injury rat model via aortic arch occlusion. Compared with normal controls, autophagic cell death was upregulated in neurons after SCIR injury. We found that autophagy promoted neuronal cell death during SCIR, shown by a significant number of terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling- (TUNEL-) positive cells colabeled with the autophagy marker microtubule-associated protein 1 light chain 3, while the autophagy inhibitor 3-methyladenine reduced the number of TUNEL-positive cells and restored neurological and motor function. Additionally, we showed that oxidative stress was the main trigger of autophagic neuronal cell death after SCIR injury and N-acetylcysteine inhibited autophagic cell death and restored neurological and motor function in SCIR injury. Finally, we found that hydrogen sulfide (H2S) inhibited autophagic cell death significantly by reducing oxidative stress in SCIR injury via the AKT-the mammalian target of rapamycin (mTOR) pathway. These findings reveal that oxidative stress induces autophagic cell death and that H2S plays a neuroprotective role by reducing oxidative stress in SCIR.
Collapse
|
43
|
Pinto S, Cunha C, Barbosa M, Vaz AR, Brites D. Exosomes from NSC-34 Cells Transfected with hSOD1-G93A Are Enriched in miR-124 and Drive Alterations in Microglia Phenotype. Front Neurosci 2017; 11:273. [PMID: 28567000 PMCID: PMC5434170 DOI: 10.3389/fnins.2017.00273] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/28/2017] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal adult-onset neurodegenerative disorder affecting motor neurons (MNs). Evidences indicate that ALS is a non-cell autonomous disease in which glial cells participate in both disease onset and progression. Exosomal transfer of mutant copper-zinc superoxide dismutase 1 (mSOD1) from cell-to-cell was suggested to contribute to disease dissemination. Data from our group and others showed that exosomes from activated cells contain inflammatory-related microRNAs (inflamma-miRNAs) that recapitulate the donor cell. While glia-derived exosomes and their effects in neurons have been addressed by several studies, only a few investigated the influence of motor neuron (MN)-derived exosomes in other cell function, the aim of the present study. We assessed a set of inflamma-miRs in NSC-34 MN-like cells transfected with mutant SOD1(G93A) and extended the study into their derived exosomes (mSOD1 exosomes). Then, the effects produced by mSOD1 exosomes in the activation and polarization of the recipient N9 microglial cells were investigated. Exosomes in coculture with N9 microglia and NSC-34 cells [either transfected with either wild-type (wt) human SOD1 or mutant SOD1(G93A)] showed to be transferred into N9 cells. Increased miR-124 expression was found in mSOD1 NSC-34 cells and in their derived exosomes. Incubation of mSOD1 exosomes with N9 cells determined a sustained 50% reduction in the cell phagocytic ability. It also caused a persistent NF-kB activation and an acute generation of NO, MMP-2, and MMP-9 activation, as well as upregulation of IL-1β, TNF-α, MHC-II, and iNOS gene expression, suggestive of induced M1 polarization. Marked elevation of IL-10, Arginase 1, TREM2, RAGE, and TLR4 mRNA levels, together with increased miR-124, miR-146a, and miR-155, at 24 h incubation, suggest the switch to mixed M1 and M2 subpopulations in the exosome-treated N9 microglial cells. Exosomes from mSOD1 NSC-34 MNs also enhanced the number of senescent-like positive N9 cells. Data suggest that miR-124 is translocated from the mSOD1 MNs to exosomes, which determine early and late phenotypic alterations in the recipient N9-microglial cells. In conclusion, modulation of the inflammatory-associated miR-124, in mSOD1 NSC-34 MNs, with potential benefits in the cargo of their exosomes may reveal a promising therapeutic strategy in halting microglia activation and associated effects in MN degeneration.
Collapse
Affiliation(s)
- Sara Pinto
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Carolina Cunha
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Marta Barbosa
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Ana R Vaz
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Dora Brites
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| |
Collapse
|
44
|
Chen K, Li JJ, Li SN, Feng J, Liu T, Wang F, Dai WQ, Xia YJ, Lu J, Zhou YQ, Guo CY. 15-Deoxy-Δ 12,14-prostaglandin J 2 alleviates hepatic ischemia-reperfusion injury in mice via inducing antioxidant response and inhibiting apoptosis and autophagy. Acta Pharmacol Sin 2017; 38:672-687. [PMID: 28216619 PMCID: PMC5457695 DOI: 10.1038/aps.2016.108] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 08/25/2016] [Indexed: 12/11/2022]
Abstract
Hepatic ischemia-reperfusion (I/R) injury is a common clinical impairment that occurs in many circumstances and leads to poor prognosis. Both apoptosis and autophagy have been shown to contribute to cell death in hepatic I/R injury. 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) is one of the best-studied anti-inflammatory prostaglandins, which has been verified to exert anti-inflammatory and cell-protective functions in various types of cells and animal models. In this study we explored the effects of 15d-PGJ2 on both apoptosis and autophagy in mouse hepatic I/R injury and its possible mechanisms. A model of segmental (70%) hepatic warm ischemia was established in Balb/c mice, and the pathological changes in serum and liver tissues were detected at 6, 12, and 24 h post-surgery, while 15d-PGJ2 (2.5, 7.5, 15 μg, iv) was administered 30 min prior the surgery. Pretreatment with 15d-PGJ2 (7.5, 15 μg) significantly ameliorated I/R-induced hepatic injury evidenced by dose-dependent reduction of serum ALT and AST levels as well as alleviated tissue damages. 15d-PGJ2 pretreatment significantly decreased the serum TNF-α and IL-1β levels and the hepatic expression of F4/80, a major biomarker of macrophages. 15d-PGJ2 pretreatment upregulated the Bcl-2/Bax ratio, thus reducing the number of apoptotic cells in the livers. 15d-PGJ2 pretreatment considerably suppressed the expression of Beclin-1 and LC3, thus decreasing the number of autophagosomes in the livers. Furthermore, 15d-PGJ2 pretreatment activated Nrf2 and inhibited a ROS/HIF1α/BNIP3 pathway in the livers. Pretreatment with the PPARγ receptor blocker GW9662 (2 μg, ip) partly reversed the protective effects of 15d-PGJ2 on hepatic I/R injury. In conclusion, our results confirm the protective effect of 15d-PGJ2 on hepatic I/R injury, an effect that may rely on a reduction in the activation of Kupffer cells and on activation of the Nrf2 pathway, which lead to inhibition of ROS generation, apoptosis, and autophagy.
Collapse
Affiliation(s)
- Kan Chen
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jing-jing Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Sai-nan Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Tong Liu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Fan Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Wei-qi Dai
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yu-jing Xia
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jie Lu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ying-qun Zhou
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Chuan-yong Guo
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
45
|
Liu T, Zhang Q, Mo W, Yu Q, Xu S, Li J, Li S, Feng J, Wu L, Lu X, Zhang R, Li L, Cheng K, Zhou Y, Zhou S, Kong R, Wang F, Dai W, Chen K, Xia Y, Lu J, Zhou Y, Zhao Y, Guo C. The protective effects of shikonin on hepatic ischemia/reperfusion injury are mediated by the activation of the PI3K/Akt pathway. Sci Rep 2017; 7:44785. [PMID: 28322249 PMCID: PMC5359611 DOI: 10.1038/srep44785] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/13/2017] [Indexed: 12/20/2022] Open
Abstract
Hepatic ischemia/reperfusion (I/R) injury, which can result in severe liver injury and dysfunction, occurs in a variety of conditions such as liver transplantation, shock, and trauma. Cell death in hepatic I/R injury has been linked to apoptosis and autophagy. Shikonin plays a significant protective role in ischemia/reperfusion injury. The purpose of the present study was to investigate the protective effect of shikonin on hepatic I/R injury and explore the underlying mechanism. Mice were subjected to segmental (70%) hepatic warm ischemia to induce hepatic I/R injury. Two doses of shikonin (7.5 and 12.5 mg/kg) were administered 2 h before surgery. Balb/c mice were randomly divided into four groups: normal control, I/R, and shikonin preconditioning at two doses (7.5 and 12.5 mg/kg). The serum and liver tissues were collected at three time points (3, 6, and 24 h). Shikonin significantly reduced serum AST and ALT levels and improved pathological features. Shikonin affected the expression of Bcl-2, Bax, caspase 3, caspase 9, Beclin-1, and LC3, and upregulated PI3K and p-Akt compared with the levels in the I/R group. Shikonin attenuated hepatic I/R injury by inhibiting apoptosis and autophagy through a mechanism involving the activation of PI3K/Akt signaling.
Collapse
Affiliation(s)
- Tong Liu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - QingHui Zhang
- Department of Clinical Laboratory, Kunshan First People’s Hospital Affiliated to Jiangsu University, 215300, Kunshan, JiangSu, China
| | - Wenhui Mo
- Department of Gastroenterology, Minhang Hospital, Shanghai Medical School of Fudan University, Shanghai, 201100, China
| | - Qiang Yu
- Department of Gastroenterology, Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China
| | - Shizan Xu
- Department of Gastroenterology, Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China
| | - Jingjing Li
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Sainan Li
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiya Lu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Rong Zhang
- Department of Gastroenterology, Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China
| | - Linqiang Li
- The School of Medicine of Soochow University, Suzhou 215006, China
| | - Keran Cheng
- The School of Medicine of Soochow University, Suzhou 215006, China
| | - Yuqing Zhou
- The School of Medicine of Soochow University, Suzhou 215006, China
| | - Shunfeng Zhou
- The School of Medicine of Soochow University, Suzhou 215006, China
| | - Rui Kong
- The School of Medicine of Soochow University, Suzhou 215006, China
| | - Fan Wang
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Weiqi Dai
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Kan Chen
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yujing Xia
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jie Lu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yingqun Zhou
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yan Zhao
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
46
|
Zhu Q, Wang H, Wang H, Luo Y, Yu Y, Du Q, Fei A, Pan S. Protective effects of ethyl pyruvate on lipopolysaccharide‑induced acute lung injury through inhibition of autophagy in neutrophils. Mol Med Rep 2017; 15:1272-1278. [PMID: 28098908 PMCID: PMC5367357 DOI: 10.3892/mmr.2017.6118] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 11/24/2016] [Indexed: 01/02/2023] Open
Abstract
Among a number of clinical factors, bacterial infection is one of the most common causes of acute lung injury (ALI), a serious complication that carries a high risk of mortality (~40%). During the process of ALI, intense local and systemic inflammation is elicited, which exacerbates the injury. Neutrophil infiltration into airspace is observed in early stage of ALI, and is required for the full development of ALI through an array of mechanisms, including the release of granule contents and the production of pro‑inflammatory cytokines, due to the overactivation of complement and cytokines. The present study noted that ethyl pyruvate alleviated ALI in lipopolysaccharide (LPS)‑induced ALI mice. Increased autophagy in neutrophils from ALI mice was observed, while ethyl pyruvate diminished autophagy in neutrophils and constrained granule release, and therefore myeloperoxidase (MPO) in bronchoalveolar lavage fluid and the production of proinflammatory cytokines. Using neutrophil cells, it was identified that autophagy was required for neutrophil activation and granule release, and that ethyl pyruvate caused neutrophil autophagy, leading to the restriction of granule release, and thus contributing to the mitigation of ALI. If autophagy was obviated through knockdown of key regulator of autophagy Atg5, the effects of ethyl pyruvate on granule release by neutrophils disappeared. Taken together, the results demonstrated that ethyl pyruvate alleviates ALI through inhibition of autophagy‑induced granule release by neutrophils, and this mechanism suggested a novel potential therapeutic target in autophagy regulation for ALI.
Collapse
Affiliation(s)
- Qingteng Zhu
- Department of Emergency Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Hui Wang
- Department of Respiratory Medicine, Putuo Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Hairong Wang
- Department of Emergency Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Yong Luo
- Department of Respiratory Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Yang Yu
- Department of Emergency Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Qirong Du
- Department of Emergency Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Aihua Fei
- Department of Emergency Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - Shuming Pan
- Department of Emergency Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
47
|
Yang R, Zhu S, Tonnessen TI. Ethyl pyruvate is a novel anti-inflammatory agent to treat multiple inflammatory organ injuries. JOURNAL OF INFLAMMATION-LONDON 2016; 13:37. [PMID: 27980458 PMCID: PMC5135784 DOI: 10.1186/s12950-016-0144-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/18/2016] [Indexed: 12/19/2022]
Abstract
Ethyl pyruvate (EP) is a simple derivative of pyruvic acid, which is an important endogenous metabolite that can scavenge reactive oxygen species (ROS). Treatment with EP is able to ameliorate systemic inflammation and multiple organ dysfunctions in multiple animal models, such as acute pancreatitis, alcoholic liver injury, acute respiratory distress syndrome (ARDS), acute viral myocarditis, acute kidney injury and sepsis. Recent studies have demonstrated that prolonged treatment with EP can ameliorate experimental ulcerative colitis and slow multiple tumor growth. It has become evident that EP has pharmacological anti-inflammatory effect to inhibit multiple early inflammatory cytokines and the late inflammatory cytokine HMGB1 release, and the anti-tumor activity is likely associated with its anti-inflammatory effect. EP has been tested in human volunteers and in a clinical trial of patients undergoing cardiac surgery in USA and shown to be safe at clinical relevant doses, even though EP fails to improve outcome of the heart surgery, EP is still a promising agent to treat patients with multiple inflammatory organ injuries and the other clinical trials are on the way. This review focuses on how EP is able to ameliorate multiple organ injuries and summarize recently published EP investigations. The targets of the anti-inflammatory agent EP ![]()
Collapse
Affiliation(s)
- Runkuan Yang
- Department of Intensive Care Medicine, Tampere University Hospital, University of Tampere, 10 Bio katu, Tampere, 33014 Finland ; Department of Critical Care Medicine, University of Pittsburgh Medical School, 3550 Terrace Street, Pittsburgh, PA 15261 USA ; Department of Emergencies and Critical Care, Rikshospital of Oslo University, PO Box 4950, Nydalen, Oslo 0424 Norway
| | - Shengtao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Beijing, 100050 China
| | - Tor Inge Tonnessen
- Department of Emergencies and Critical Care, Rikshospital of Oslo University, PO Box 4950, Nydalen, Oslo 0424 Norway ; Institute for Clinical Medicine, University of Oslo, Blindern, Oslo 0316 Norway
| |
Collapse
|
48
|
Zeng M, Wei X, Wu Z, Li W, Zheng Y, Li B, Meng X, Fu X, Fei Y. Simulated ischemia/reperfusion-induced p65-Beclin 1-dependent autophagic cell death in human umbilical vein endothelial cells. Sci Rep 2016; 6:37448. [PMID: 27857190 PMCID: PMC5114588 DOI: 10.1038/srep37448] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 10/31/2016] [Indexed: 12/15/2022] Open
Abstract
Myocardial ischemia/reperfusion (I/R) injury detrimentally alters the prognosis of patients undergoing revascularization after acute myocardial infarction. Our previous study demonstrated that NF-κB-induced autophagy plays a detrimental role in cardiac I/R injury using a rabbit myocardial I/R model. In this study, we sought to explore the specific mechanism of this autophagy-mediated cell damage in an in vitro simulated ischemia/reperfusion (sI/R) model using human umbilical vein endothelial cells. Our current study demonstrates that simulated I/R induces autophagy in a p65-Beclin 1-dependent manner, which can be suppressed with the inhibition of NF-κB. Furthermore, rapamycin which promotes autophagy, exacerbates sI/R-induced cell death. While 3-methyladenine rescues cell damage. Our data thus suggest that I/R promotes NF-κB p65 activity mediated Beclin 1-mediated autophagic flux, thereby exacerbating myocardial injury.
Collapse
Affiliation(s)
- Min Zeng
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Xin Wei
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Zhiyong Wu
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Wei Li
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Yin Zheng
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Bing Li
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Xuqing Meng
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Xiuhong Fu
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| | - Yi Fei
- Medical Center, Hainan General Hospital, Haikou, 570311, China
| |
Collapse
|
49
|
Effects of insulin combined with ethyl pyruvate on inflammatory response and oxidative stress in multiple-organ dysfunction syndrome rats with severe burns. Am J Emerg Med 2016; 34:2154-2158. [DOI: 10.1016/j.ajem.2016.08.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 08/01/2016] [Accepted: 08/06/2016] [Indexed: 01/08/2023] Open
|
50
|
Shen M, Lu J, Dai W, Wang F, Xu L, Chen K, He L, Cheng P, Zhang Y, Wang C, Wu D, Yang J, Zhu R, Zhang H, Zhou Y, Guo C. Corrigendum to "Ethyl Pyruvate Ameliorates Hepatic Ischemia-Reperfusion Injury by Inhibiting Intrinsic Pathway of Apoptosis and Autophagy". Mediators Inflamm 2016; 2016:5434275. [PMID: 27807393 PMCID: PMC5078780 DOI: 10.1155/2016/5434275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 05/26/2016] [Indexed: 01/21/2023] Open
Abstract
[This corrects the article DOI: 10.1155/2013/461536.].
Collapse
Affiliation(s)
- Miao Shen
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Jie Lu
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Weiqi Dai
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Fan Wang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Ling Xu
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Kan Chen
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Lei He
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Ping Cheng
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Yan Zhang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Chengfen Wang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Dong Wu
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Jing Yang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Rong Zhu
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Huawei Zhang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Yingqun Zhou
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Chuanyong Guo
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| |
Collapse
|