1
|
Chung HY, Lee GS, Nam SH, Lee JH, Han JP, Song S, Kim GD, Jung C, Hyeon DY, Hwang D, Choi BO, Yeom SC. Morc2a variants cause hydroxyl radical-mediated neuropathy and are rescued by restoring GHKL ATPase. Brain 2024; 147:2114-2127. [PMID: 38227798 DOI: 10.1093/brain/awae017] [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: 07/31/2023] [Revised: 12/21/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024] Open
Abstract
Mutations in the Microrchidia CW-type zinc finger 2 (MORC2) GHKL ATPase module cause a broad range of neuropathies, such as Charcot-Marie-Tooth disease type 2Z; however, the aetiology and therapeutic strategy are not fully understood. Previously, we reported that the Morc2a p.S87L mouse model exhibited neuropathy and muscular dysfunction through DNA damage accumulation. In the present study, we analysed the gene expression of Morc2a p.S87L mice and designated the primary causing factor. We investigated the pathological pathway using Morc2a p.S87L mouse embryonic fibroblasts and human fibroblasts harbouring MORC2 p.R252W. We subsequently assessed the therapeutic effect of gene therapy administered to Morc2a p.S87L mice. This study revealed that Morc2a p.S87L causes a protein synthesis defect, resulting in the loss of function of Morc2a and high cellular apoptosis induced by high hydroxyl radical levels. We considered the Morc2a GHKL ATPase domain as a therapeutic target because it simultaneously complements hydroxyl radical scavenging and ATPase activity. We used the adeno-associated virus (AAV)-PHP.eB serotype, which has a high CNS transduction efficiency, to express Morc2a or Morc2a GHKL ATPase domain protein in vivo. Notably, AAV gene therapy ameliorated neuropathy and muscular dysfunction with a single treatment. Loss-of-function characteristics due to protein synthesis defects in Morc2a p.S87L were also noted in human MORC2 p.S87L or p.R252W variants, indicating the correlation between mouse and human pathogenesis. In summary, CMT2Z is known as an incurable genetic disorder, but the present study demonstrated its mechanisms and treatments based on established animal models. This study demonstrates that the Morc2a p.S87L variant causes hydroxyl radical-mediated neuropathy, which can be rescued through AAV-based gene therapy.
Collapse
Affiliation(s)
- Hye Yoon Chung
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Geon Seong Lee
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Soo Hyun Nam
- Samsung Medical Center, Cell & Gene Therapy Institute, Seoul 06351, Korea
| | - Jeong Hyeon Lee
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Jeong Pil Han
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Sumin Song
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Gap-Don Kim
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Choonkyun Jung
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
| | - Do Young Hyeon
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Daehee Hwang
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Bioinformatics Institute, Bio-MAX, Seoul National University, Seoul 08826, Republic of Korea
| | - Byung-Ok Choi
- Samsung Medical Center, Cell & Gene Therapy Institute, Seoul 06351, Korea
- Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences & Technology, Seoul 06351, Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Su Cheong Yeom
- Graduate School of International Agricultural Technology and Institute of Green-Bio Science and Technology, Seoul National University, Kangwon 25354, Korea
- Department of Agricultural Biotechnology, WCU Biomodulation Major, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
2
|
Li Z, Xu L, Li JY, Lei L, Liang PZ, Wu Q, Yang F, Ren TB, Yin X, Yuan L, Zhang XB. Superoxide Anion-Mediated Afterglow Mechanism-Based Water-Soluble Zwitterion Dye Achieving Renal-Failure Mice Detection. J Am Chem Soc 2023; 145:26736-26746. [PMID: 38015824 DOI: 10.1021/jacs.3c08579] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Afterglow materials-based biological imaging has promising application prospects, due to negligible background. However, currently available afterglow materials mainly include inorganic materials as well as some organic nanoparticles, which are difficult to translate to the clinic, resulting from non-negligible metabolic toxicity and even leakage risk of inorganic heavy metals. Although building small organic molecules could solve such obstacles, organic small molecules with afterglow ability are extremely scarce, especially with a sufficient renal metabolic capacity. To address these issues, herein, we designed water-soluble zwitterion Cy5-NF with renal metabolic capacity and afterglow luminescence, which relied on an intramolecular cascade reaction between superoxide anion (O2•-, instead of 1O2) and Cy5-NF to release afterglow luminescence. Of note, compared with different reference contrast agents, zwitterion Cy5-NF not only had excellent afterglow properties but also had a rapid renal metabolism rate (half-life period, t1/2, around 10 min) and good biocompatibility. Unlike prior afterglow nanosystems possessing a large size, for the first time, zwitterion Cy5-NF has achieved the construction of water-soluble renal metabolic afterglow contrast agents, which showed higher sensitivity and signal-to-background ratio in afterglow imaging than fluorescence imaging for the kidney. Moreover, zwitterion Cy5-NF had a longer kidney retention time in renal-failure mice (t1/2 more than 15 min). More importantly, zwitterion Cy5-NF can be metabolized very quickly even in severe renal-failure mice (t1/2 around 25-30 min), which greatly improved biosecurity. Therefore, we are optimistic that the O2•--mediated afterglow mechanism-based water-soluble zwitterion Cy5-NF is very promising for clinical application, especially rapid detection of kidney failure.
Collapse
Affiliation(s)
- Zhe Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Li Xu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Jin-Yu Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Lingling Lei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Ping-Zhao Liang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Qian Wu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Feiyu Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Tian-Bing Ren
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Xia Yin
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Lin Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Xiao-Bing Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
3
|
Zha D, Yang Y, Huang X, Wang Z, Lin H, Yang L, Xu L, Wu Y, Huang H, Wang Y, Xin Z, Wu X, Xiao YF, Li TS, Deng KY, Xin HB, Qian Y. Nicaraven protects against endotoxemia-induced inflammation and organ injury through modulation of AMPK/Sirt1 signaling in macrophages. Eur J Pharmacol 2023; 946:175666. [PMID: 36944380 DOI: 10.1016/j.ejphar.2023.175666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 03/23/2023]
Abstract
Endotoxemia is a disease characterized by systemic inflammatory responses and organ injury caused by lipopolysaccharide (LPS) infection, with high mortality. Nicaraven (AVS), a potent hydroxyl radical scavenger, has been proven to regulate the inflammatory response in tumors. To investigate the protective effects and mechanisms of AVS in endotoxemia, mice were injected intraperitoneally with LPS to induce endotoxemia. AVS treatment significantly decreased the levels of pro-inflammatory cytokines in the serum, reduced neutrophil infiltration, attenuated multiple organ injury, and increased the survival rate in LPS-challenged mice. In the LPS-induced inflammatory model of macrophages, AVS inhibited macrophage activation, suppressed nitric oxide (NO) production, and inhibited the expression and secretion of pro-inflammatory cytokines. Mechanistically, AVS treatment up-regulated silence information regulator transcript-1 (Sirt1) expression in a time- and dose-dependent manner. AVS treatment activated the AMP-dependent protein kinase (AMPK)/Sirt1 signaling pathway and suppressed the activation of nuclear factor kappa B (NF-κB) in macrophages exposed to LPS. However, the anti-inflammatory effects of AVS could be reversed by the AMPK, the Sirt1 inhibitor, or the histone deacetylase inhibitor. We confirmed that the AMPK inhibitor inhibited AVS-mediated AMPK/Sirt1 activation and NF-κB p65 acetylation. These results suggested that AVS alleviated endotoxemia by activating the AMPK/Sirt1 signaling pathway in macrophages.
Collapse
Affiliation(s)
- Duoduo Zha
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China
| | - Yaqin Yang
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China
| | - Xiang Huang
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China
| | - Ziwei Wang
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China
| | - Hongru Lin
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China
| | - Lingyi Yang
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China
| | - Luyan Xu
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China
| | - Yijia Wu
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China
| | - Houda Huang
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China
| | - Yihan Wang
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China
| | - Zhaochen Xin
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China
| | - Xuehan Wu
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China
| | - Yun-Fei Xiao
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Ke-Yu Deng
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China
| | - Hong-Bo Xin
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China.
| | - Yisong Qian
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
4
|
Nicaraven-loaded electrospun wound dressings promote diabetic wound healing via proangiogenic and immunomodulatory functions: a preclinical investigation. Drug Deliv Transl Res 2023; 13:222-236. [PMID: 35648292 DOI: 10.1007/s13346-022-01176-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2022] [Indexed: 12/13/2022]
Abstract
The current study developed a biopolymer-based wound dressing by electrospinning of Nicaraven-loaded collagen solution. Firstly, collagen was dissolved in acetic acid, and then Nicaraven was added to the polymeric solution at three different concentrations of 2 w/w%, 4 w/w%, and 6 w/w%. The resulting solution was then electrospun. Various experiments were performed to characterize the produced wound dressings. In vitro studies showed that Nicaraven-loaded scaffolds were not toxic against L929 fibroblast cells and protected them against oxidative stress. Wound healing potential of different formulations of Nicaraven-loaded collagen wound dressings was studied in a rat model of the excisional diabetic wound. The study showed that the collagen/4% Nicaraven and collagen/6% Nicaraven wound dressings exhibited a significantly higher percentage of wound closure, the thickness of the epithelium, and collagen deposition compared with collagen/2% Nicaraven, collagen-only, and sterile gauze groups. Gene expression study showed that the developed wound dressings reduced the tissue expression levels of glutathione peroxidase, NFKβ, and matrix metalloproteinase 9 (MMP9) genes. In addition, in the wounds treated with collagen/4% Nicaraven and collagen/6% Nicaraven scaffolds, wound healing was associated with a higher tissue expression level of b-FGF, VEGF, and collagen type I genes. Overall, wound healing activity of collagen/4% Nicaraven and collagen/6% Nicaraven wound dressings was not significantly different. This study implies that collagen wound dressings incorporated with 4% and 6% Nicaraven can be considered a potential candidate to treat diabetic wounds in the clinic.
Collapse
|
5
|
The Effect of Antioxidant Added to Preservation Solution on the Protection of Kidneys before Transplantation. Int J Mol Sci 2022; 23:ijms23063141. [PMID: 35328560 PMCID: PMC8954097 DOI: 10.3390/ijms23063141] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/07/2022] [Accepted: 03/13/2022] [Indexed: 02/04/2023] Open
Abstract
Ischemia–reperfusion injury is a key clinical problem of transplantology. Current achievements in optimizing organ rinse solutions and storage techniques have significantly influenced the degree of graft damage and its survival after transplantation. In recent years, intensive research has been carried out to maintain the viability of tissues and organs outside the integral environment of the body. Innovative solutions for improving the biochemical functions of the stored organ have been developed. The article discusses directions for modifying preservation solutions with antioxidants. Clinical and experimental studies aimed at optimizing these fluids, as well as perfusion and organ preservation techniques, are presented.
Collapse
|
6
|
Eryilmaz S, Turkyilmaz Z, Karabulut R, Gulburun MA, Poyraz A, Gulbahar O, Arslan B, Sonmez K. The effects of hydrogen-rich saline solution on intestinal anastomosis performed after intestinal ischemia reperfusion injury. J Pediatr Surg 2020; 55:1574-1578. [PMID: 31466816 DOI: 10.1016/j.jpedsurg.2019.07.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/02/2019] [Accepted: 07/19/2019] [Indexed: 01/31/2023]
Abstract
AIM We investigated the effects of hydrogen-rich saline solution (HRSS) on intestinal anastomosis performed after intestinal ischemia reperfusion injury (IRI). MATERIALS AND METHODS Thirty Wistar albino female rats were randomly divided into five groups. Only laparotomy was performed in the Sham group. In the other four groups, an intestinal IRI was performed for 45 min by clamping the superior mesenteric artery. After intestinal IRI, anastomosis was performed by cutting the intestine from the proximal 15 cm of the ileocecal valve at the first and 24th hours. HRSS was given intraperitoneally 5 ml/kg before reperfusion and for four more days in the HRSS1 and HRSS24groups, while no treatment was given to the I/R1 and I/R24 groups. After 5 days, all groups underwent relaparotomy. The anastomotic bursting pressures were measured in all groups, except the Sham group. The tumor necrosis factor-α (TNF-α), interleukin 6 (IL-6), myeloperoxidase (MPO) and malondialdehyde (MDA) levels were measured in the tissues taken from the anastomosis line. The tissue sections were evaluated histopathologically and the apoptosis index was determined by applying the TUNEL method. The results were analyzed one-way analysis of variance (ANOVA) and Pearson's chi-squared test. RESULTS Although the MPO, MDA, IL-6 and TNF-α tissue values were not statistically significant among the groups, the degree of tissue damage and apoptosis levels were lower and the anastomotic bursting pressures values were higher in the HRSS1 and HRSS24 groups compared to the I/R1 and I/R24 groups. CONCLUSION HRSS is effective in reducing the intestinal damage caused by an IRI: HRSS has the potential to reduce the detrimental effects of intestinal anastomosis performed after an intestinal IRI.
Collapse
Affiliation(s)
- Sibel Eryilmaz
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Zafer Turkyilmaz
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ramazan Karabulut
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey.
| | - Merve Altin Gulburun
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Aylar Poyraz
- Department of Pediatric Pathology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ozlem Gulbahar
- Department of Pediatric Biochemistry, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Burak Arslan
- Department of Pediatric Biochemistry, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Kaan Sonmez
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
7
|
Zhang X, Moriwaki T, Kawabata T, Goto S, Liu KX, Guo CY, Li TS. Nicaraven Attenuates Postoperative Systemic Inflammatory Responses-Induced Tumor Metastasis. Ann Surg Oncol 2019; 27:1068-1074. [PMID: 31873930 PMCID: PMC7060163 DOI: 10.1245/s10434-019-08076-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Inflammation has been demonstrated to promote cancer metastasis. Due to the well-known systemic inflammatory responses (SIR) after major surgery, it is critical to investigate and attenuate SIR-induced tumor metastasis of cancer patients suffering surgical procedures. METHODS C57BL/6 mice were intravenously injected with Lewis lung cancer cells at 6, 24, and 72 h after the induction of intestinal ischemia/reperfusion (I/R) injury. We found that the number of tumor nodules significantly increased in lungs of mice injected with cancer cells at 6 h but not at 24 and 72 h after I/R injury. The administration of nicaraven 30 min before and 24 h after I/R injury effectively attenuated the enhanced tumor metastasis to lungs. Protein array showed the increase of various cytokines in plasma of mice at 6 h after I/R injury, but many of them were attenuated by the administration of nicaraven. Immunostaining indicated the increase of Ly6g-, CD206-, and CD11c-positive inflammatory cells in the lungs, but it was also attenuated by nicaraven administration. CONCLUSIONS Postoperative SIR-induced tumor metastasis have been clearly evidenced in our experimental model, and the administration of nicaraven may ameliorate the SIR-induced tumor metastasis by suppressing inflammatory responses.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Takahito Moriwaki
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Tsuyoshi Kawabata
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Shinji Goto
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Ke-Xiang Liu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin, 130041, China
| | - Chang-Ying Guo
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, No. 519 Beijing East Road, Nanchang, 330006, China
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.
| |
Collapse
|
8
|
Zhang X, Li TS. ASO Author Reflections: How Should a Surgeon Care and Manage Cancer Metastasis? Ann Surg Oncol 2019; 27:1075-1076. [PMID: 31832918 DOI: 10.1245/s10434-019-08131-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Indexed: 11/18/2022]
Affiliation(s)
- Xu Zhang
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan.
| |
Collapse
|
9
|
Nicaraven reduces cancer metastasis to irradiated lungs by decreasing CCL8 and macrophage recruitment. Cancer Lett 2018; 418:204-210. [DOI: 10.1016/j.canlet.2018.01.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/07/2018] [Accepted: 01/09/2018] [Indexed: 11/24/2022]
|
10
|
Lukiswanto BS, Yuniarti WM, Motulo YY. Effects of hyperbaric therapy on liver morphofunctional of rabbits ( Oryctolagus cuniculus) after hind limb ischemia-reperfusion injury. Vet World 2017; 10:1337-1342. [PMID: 29263596 PMCID: PMC5732340 DOI: 10.14202/vetworld.2017.1337-1342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 10/18/2017] [Indexed: 12/11/2022] Open
Abstract
AIM The objective of this research was to study and to prove the effectiveness of hyperbaric oxygen therapy (HBOT) starting time on liver morphofunctional changes after ischemia-reperfusion in the hind limb of rabbits. MATERIALS AND METHODS This research used a complete randomized design with 4 groups and 6 repetitions on each. After 6 h artery femoral is ligation, reperfusion was performed for 100 min (G1), HBOT for 90 min after 10 min reperfusion (G2), 10 min reperfusion (G3), and HBOT 90 min after 60 min reperfusion (G4). Then, all of the rabbits were sacrificed. The liver and blood were taken for histopathological changes examination as well as for measuring the level of aspartate aminotransferase (AST) and alanine aminotransferase (ALT). The statistical test using Kruskal-Wallis and Mann-Whitney showed that the score of degeneration, necrosis, and portal inflammation in groups without HBOT (G1 and G3) were not significantly different, as well as in group with HBOT (G2 and G4) (p>0.05). However, the scores of histopathological changes in G1 and G3 were significantly different from those in G2 and G4 (p<0.05). The levels of AST and ALT in the groups without hyperbaric therapy (G1 and G3) were not significantly different from those in the groups treated with hyperbaric therapy (G2 and G4) (p>0.05). RESULT Hind limb ischemia injury reperfusion can trigger damage for liver morphology, but not lead to liver dysfunction. Reperfusion can trigger increased activity of neutrophils, while neutrophil infiltration in the organ will lead to dysfunction. HBOT can inhibit the activity of neutrophils and the dysfunction of organs caused by ischemic reperfusion. CONCLUSION HBOT for 90 min, both 10 and 60 min after the reperfusion, can protect hepatocytes from damage.
Collapse
Affiliation(s)
- Bambang Sektiari Lukiswanto
- Department of Veterinary Clinic, Faculty of Veterinary Medicine, Universitas Airlangga, Jl. Mulyorejo, Kampus C, Surabaya 60115, Indonesia
| | - Wiwik Misaco Yuniarti
- Department of Veterinary Clinic, Faculty of Veterinary Medicine, Universitas Airlangga, Jl. Mulyorejo, Kampus C, Surabaya 60115, Indonesia
| | - Y. Yosis Motulo
- Department of Surgery, Division of Thoracic, Cardiac, and Vascular Surgery, Dr. Soetomo General Hospital, Surabaya 60115, Indonesia
| |
Collapse
|
11
|
Inan M, Bakar E, Cerkezkayabekir A, Sanal F, Ulucam E, Subaşı C, Karaöz E. Mesenchymal stem cells increase antioxidant capacity in intestinal ischemia/reperfusion damage. J Pediatr Surg 2017; 52:1196-1206. [PMID: 28118930 DOI: 10.1016/j.jpedsurg.2016.12.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 12/03/2016] [Accepted: 12/23/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) may have beneficial effects in reversing intestinal damage resulting from circulatory disorders. The hypothesis of this study is that MSCs increase antioxidant capacity of small bowel tissue following intestinal ischemia reperfusion (I/R) damage. METHODS A total of 100 rats were used for the control group and three experimental groups, as follows: the sham control, local MSC, and systemic MSC groups. Each group consisted of 10 animals on days 1, 4, and 7 of the experiment. Ischemia was established by clamping the superior mesenteric artery (SMA) for 45min; following this, reperfusion was carried out for 1, 4, and 7days in all groups. In the local and systemic groups, MSCs were administered intravenously and locally just after the ischemia, and they were investigated after 1, 4, and 7days. The superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (Gpx) activities, as well as malondialdehyde (MDA) and total protein levels, were measured. Histopathological analysis was performed using light and electron microscopy. The indicators of proliferation from the effects of anti- and pro-inflammatory cytokines were evaluated using immunohistochemistry. RESULTS MDA was increased (P<0.05) in the sham control group and decreased (P<0.05) in the MSC groups. SOD, CAT, and Gpx were decreased in the local MSC group (P<0.05). The highest level of amelioration was observed on day 7 in the local MSC group via light and electron microscopy. It was found that the MSCs arrived at the damaged intestinal wall in the MSC groups immediately after injection. Pro-inflammatory cytokines interleukin-1β (IL1β), transforming growth factor-β1 (TGFβ1), tumor necrosis factor-α (TNFα), IL6, MIP2, and MPO decreased (P<0.05), while anti-inflammatory cytokines EP3 and IL1ra increased (p<0.05) in the local and systemic MSC groups. In addition, proliferation indicators, such as PCNA and KI67, increased (P<0.05) in the local and systemic MSC groups. CONCLUSIONS Parallel to our hypothesis, MSC increases the antioxidant capacity of small bowel tissue after intestinal I/R damage. The MSCs migrated to the reperfused small intestine by homing and reduced oxidative stress via the effects of SOD, CAT, and Gpx, as well as reducing the MDA level; thus, they could increase antioxidant capacity of intestine and have a therapeutic effect on the damaged tissue. We think that this effect was achieved via scavenging of oxygen radicals, suppression of pro-inflammatory cytokines, and increasing the expression of anti-inflammatory cytokines.
Collapse
Affiliation(s)
- M Inan
- Department of Pediatric Surgery, Trakya University Faculty of Medicine, Edirne, Turkey.
| | - E Bakar
- Department of Pharmaceutical Technology, Trakya University Faculty of Pharmacy, Edirne, Turkey
| | - A Cerkezkayabekir
- Division of Molecular Biology, Department of Biology, Trakya University Faculty of Science, Edirne, Turkey
| | - F Sanal
- Division of Molecular Biology, Department of Biology, Trakya University Faculty of Science, Edirne, Turkey
| | - E Ulucam
- Department of Anatomy, Trakya University Faculty of Medicine, Edirne, Turkey
| | - C Subaşı
- Department of Histology and Embryology, Faculty of Medicine, İstinye University, İstanbul, Turkey
| | - E Karaöz
- Department of Histology and Embryology, Faculty of Medicine, İstinye University, İstanbul, Turkey
| |
Collapse
|
12
|
Yan C, Luo L, Urata Y, Goto S, Guo CY, Li TS. Nicaraven, a Potential Radioprotective Agent, has Very Limited Effects on the Survival of Cancer Cells and the Growth of Established Tumors. Radiat Res 2017; 187:339. [DOI: 10.1667/rr4614.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Chen Yan
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Lan Luo
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Yoshishige Urata
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Shinji Goto
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Chang-Ying Guo
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| |
Collapse
|
13
|
Fahrner R, Dondorf F, Ardelt M, Settmacher U, Rauchfuss F. Role of NK, NKT cells and macrophages in liver transplantation. World J Gastroenterol 2016; 22:6135-6144. [PMID: 27468206 PMCID: PMC4945975 DOI: 10.3748/wjg.v22.i27.6135] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/25/2016] [Accepted: 06/15/2016] [Indexed: 02/07/2023] Open
Abstract
Liver transplantation has become the treatment of choice for acute or chronic liver disease. Because the liver acts as an innate immunity-dominant organ, there are immunological differences between the liver and other organs. The specific features of hepatic natural killer (NK), NKT and Kupffer cells and their role in the mechanism of liver transplant rejection, tolerance and hepatic ischemia-reperfusion injury are discussed in this review.
Collapse
|
14
|
Shimada S, Wakayama K, Fukai M, Shimamura T, Ishikawa T, Fukumori D, Shibata M, Yamashita K, Kimura T, Todo S, Ohsawa I, Taketomi A. Hydrogen Gas Ameliorates Hepatic Reperfusion Injury After Prolonged Cold Preservation in Isolated Perfused Rat Liver. Artif Organs 2016; 40:1128-1136. [PMID: 27140066 DOI: 10.1111/aor.12710] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 12/30/2015] [Accepted: 01/07/2016] [Indexed: 12/12/2022]
Abstract
Hydrogen gas reduces ischemia and reperfusion injury (IRI) in the liver and other organs. However, the precise mechanism remains elusive. We investigated whether hydrogen gas ameliorated hepatic I/R injury after cold preservation. Rat liver was subjected to 48-h cold storage in University of Wisconsin solution. The graft was reperfused with oxygenated buffer with or without hydrogen at 37° for 90 min on an isolated perfusion apparatus, comprising the H2 (+) and H2 (-) groups, respectively. In the control group (CT), grafts were reperfused immediately without preservation. Graft function, injury, and circulatory status were assessed throughout the perfusion. Tissue samples at the end of perfusion were collected to determine histopathology, oxidative stress, and apoptosis. In the H2 (-) group, IRI was indicated by a higher aspartate aminotransferase (AST), alanine aminotransferase (ALT) leakage, portal resistance, 8-hydroxy-2-deoxyguanosine-positive cell rate, apoptotic index, and endothelial endothelin-1 expression, together with reduced bile production, oxygen consumption, and GSH/GSSG ratio (vs. CT). In the H2 (+) group, these harmful changes were significantly suppressed [vs. H2 (-)]. Hydrogen gas reduced hepatic reperfusion injury after prolonged cold preservation via the maintenance of portal flow, by protecting mitochondrial function during the early phase of reperfusion, and via the suppression of oxidative stress and inflammatory cascades thereafter.
Collapse
Affiliation(s)
| | | | - Moto Fukai
- Transplant Surgery, Hokkaido University Graduate School of Medicine
| | - Tsuyoshi Shimamura
- Central Clinical Facilities, Division of Organ Transplantation, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | | | - Daisuke Fukumori
- Department of Surgical Gastroenterology and Transplantation, University of Copenhagen, Copenhagen, Denmark
| | - Maki Shibata
- Department of Biological Process of Aging, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo
| | | | - Taichi Kimura
- Laboratory of Cancer Research, Department of Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido
| | - Satoru Todo
- St. Maria Hospital Laboratory, Kurume, Fukuoka, Japan
| | - Ikuroh Ohsawa
- Department of Biological Process of Aging, Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo
| | | |
Collapse
|
15
|
Rocha-Santos V, Figueira ERR, Rocha-Filho JA, Coelho AMM, Pinheiro RS, Bacchella T, Machado MCC, D'Albuquerque LAC. Pentoxifylline enhances the protective effects of hypertonic saline solution on liver ischemia reperfusion injury through inhibition of oxidative stress. Hepatobiliary Pancreat Dis Int 2015; 14:194-200. [PMID: 25865693 DOI: 10.1016/s1499-3872(15)60348-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Liver ischemia reperfusion (IR) injury triggers a systemic inflammatory response and is the main cause of organ dysfunction and adverse postoperative outcomes after liver surgery. Pentoxifylline (PTX) and hypertonic saline solution (HTS) have been identified to have beneficial effects against IR injury. This study aimed to investigate if the addition of PTX to HTS is superior to HTS alone for the prevention of liver IR injury. METHODS Male Wistar rats were allocated into three groups. Control rats underwent 60 minutes of partial liver ischemia, HTS rats were treated with 0.4 mL/kg of intravenous 7.5% NaCl 15 minutes before reperfusion, and HPTX group were treated with 7.5% NaCl plus 25 mg/kg of PTX 15 minutes before reperfusion. Samples were collected after reperfusion for determination of ALT, AST, TNF-alpha, IL-6, IL-10, mitochondrial respiration, lipid peroxidation, pulmonary permeability and myeloperoxidase. RESULTS HPTX significantly decreased TNF-alpha 30 minutes after reperfusion. HPTX and HTS significantly decreased ALT, AST, IL-6, mitochondrial dysfunction and pulmonary myeloperoxidase 4 hours after reperfusion. Compared with HTS only, HPTX significantly decreased hepatic oxidative stress 4 hours after reperfusion and pulmonary permeability 4 and 12 hours after reperfusion. CONCLUSION This study showed that PTX added the beneficial effects of HTS on liver IR injury through decreases of hepatic oxidative stress and pulmonary permeability.
Collapse
Affiliation(s)
- Vinicius Rocha-Santos
- Department of Gastroenterology, Laboratory of Medical Investigations LIM37 Discipline of Liver and Gastrointestinal Transplantation and Discipline of Anesthesiology, Hospital das Clinicas, University of Sao Paulo School of Medicine, Sao Paulo, SP, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Shimada S, Fukai M, Wakayama K, Ishikawa T, Kobayashi N, Kimura T, Yamashita K, Kamiyama T, Shimamura T, Taketomi A, Todo S. Hydrogen sulfide augments survival signals in warm ischemia and reperfusion of the mouse liver. Surg Today 2014; 45:892-903. [PMID: 25362520 DOI: 10.1007/s00595-014-1064-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 06/19/2014] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND PURPOSE Hydrogen sulfide (H2S) ameliorates hepatic ischemia and reperfusion injury (IRI), but the precise mechanism remains elusive. We investigated whether sodium hydrogen sulfide (NaHS), a soluble derivative of H2S, would ameliorate hepatic IRI, and if so, via what mechanism. METHODS Mice were subjected to partial warm ischemia for 75 min followed by reperfusion. Either NaHS or saline was administered intravenously 10 min before reperfusion. The liver and serum were collected 3, 6, and 24 h after reperfusion. RESULTS In the NaHS(-) group, severe IRI was apparent by the ALT leakage, tissue injury score, apoptosis, lipid peroxidation, and inflammation (higher plasma TNF-α, IL-6, IL-1β, IFN-γ, IL-23, IL-17, and CD40L), whereas IRI was significantly ameliorated in the NaHS(+) group. These effects could be explained by the augmented nuclear translocation of Nrf2, and the resulting up-regulation of HO-1 and thioredoxin-1. Phosphorylation of the PDK-1/Akt/mTOR/p70S6k axis, which is known to mediate pro-survival and anti-apoptotic signals, was significantly augmented in the NaHS(+) group, with a higher rate of PCNA-positive cells thereafter. CONCLUSION NaHS ameliorated hepatic IRI by direct and indirect anti-oxidant activities by augmenting pro-survival, anti-apoptotic, and anti-inflammatory signals via mechanisms involving Nrf-2, and by accelerating hepatic regeneration via mechanisms involving Akt-p70S6k.
Collapse
Affiliation(s)
- Shingo Shimada
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, N-15, W-7, Kita-ku, Sapporo, 060-8638, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Kawakatsu M, Urata Y, Imai R, Goto S, Ono Y, Nishida N, Li TS. Nicaraven attenuates radiation-induced injury in hematopoietic stem/progenitor cells in mice. PLoS One 2013; 8:e60023. [PMID: 23555869 PMCID: PMC3612087 DOI: 10.1371/journal.pone.0060023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 02/20/2013] [Indexed: 01/01/2023] Open
Abstract
Nicaraven, a chemically synthesized hydroxyl radical-specific scavenger, has been demonstrated to protect against ischemia-reperfusion injury in various organs. We investigated whether nicaraven can attenuate radiation-induced injury in hematopoietic stem/progenitor cells, which is the conmen complication of radiotherapy and one of the major causes of death in sub-acute phase after accidental exposure to high dose radiation. C57BL/6 mice were exposed to 1 Gy γ-ray radiation daily for 5 days in succession (a total of 5 Gy), and given nicaraven or a placebo after each exposure. The mice were sacrificed 2 days after the last radiation treatment, and the protective effects and relevant mechanisms of nicaraven in hematopoietic stem/progenitor cells with radiation-induced damage were investigated by ex vivo examination. We found that post-radiation administration of nicaraven significantly increased the number, improved the colony-forming capacity, and decreased the DNA damage of hematopoietic stem/progenitor cells. The urinary levels of 8-oxo-2′-deoxyguanosine, a marker of DNA oxidation, were significantly lower in mice that were given nicaraven compared with those that received a placebo treatment, although the levels of intracellular and mitochondrial reactive oxygen species in the bone marrow cells did not differ significantly between the two groups. Interestingly, compared with the placebo treatment, the administration of nicaraven significantly decreased the levels of the inflammatory cytokines IL-6 and TNF-α in the plasma of mice. Our data suggest that nicaraven effectively diminished the effects of radiation-induced injury in hematopoietic stem/progenitor cells, which is likely associated with the anti-oxidative and anti-inflammatory properties of this compound.
Collapse
Affiliation(s)
- Miho Kawakatsu
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yoshishige Urata
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Ryo Imai
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shinji Goto
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yusuke Ono
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Noriyuki Nishida
- Department of Molecular Microbiology and Immunology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
18
|
Czubkowski P, Socha P, Pawlowska J. Current status of oxidative stress in pediatric liver transplantation. Pediatr Transplant 2010; 14:169-77. [PMID: 20113425 DOI: 10.1111/j.1399-3046.2009.01256.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Generation of free radicals in children after liver transplantation is multifactorial from ischemia-reperfusion injury, immunosuppression and post-transplant complications. Thus, this group is at higher risk of oxidative imbalance with molecular and clinical consequences. We discuss pathogenesis and ways of action against oxidative stress in liver transplant recipients.
Collapse
Affiliation(s)
- Piotr Czubkowski
- Department of Gastroenterology, Hepatology and Immunology, The Children's Memorial Health Institute, Warsaw, Poland.
| | | | | |
Collapse
|
19
|
Synergistic induction of heme oxygenase-1 by nicaraven after subarachnoid hemorrhage to prevent delayed cerebral vasospasm. Eur J Pharmacol 2009; 620:16-20. [PMID: 19686725 DOI: 10.1016/j.ejphar.2009.08.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Revised: 07/24/2009] [Accepted: 08/04/2009] [Indexed: 11/21/2022]
Abstract
Cerebral vasospasm remains a major cause of morbidity and mortality in patients with subarachnoid hemorrhage. Heme oxygenase-1 (HO-1) is an oxidative stress-inducible enzyme with multiple protective functions against vascular and neurological diseases, including delayed cerebral vasospasm. In the present study, intravenous administration (i.v.) of nicaraven (1 mg/kg/min, for 2 days after subarachnoid hemorrhage) ameliorated delayed cerebral vasospasm in rat subarachnoid hemorrhage models, marked synergistic induction of HO-1 protein (> 2.5-fold than 'subarachnoid hemorrhage with saline i.v.'), and elicited a rapid increase of cGMP accumulation in the basilar arteries. In the sham-operated rats, nicaraven could not induce HO-1 expression. Antisense HO-1 oligodeoxynucleotides abrogated this HO-1 induction and the antivasospastic effect of nicaraven. In vitro study using Hela cells, nicaraven enhanced the human HO-1 promoter (-4.5 kbp) activity, which was pre-activated with the blood component oxyhemoglobin to mimic the ability of subarachnoid hemorrhage. These results suggest that this enhanced HO-1 expression through a combination of pathological state and pharmacological agent could be an effective strategy to improve the prognosis of heme- and oxidative stress-induced diseases, such as delayed cerebral vasospasm.
Collapse
|
20
|
Wu TJ, Khoo NH, Zhou F, Day BJ, Parks DA. Decreased hepatic ischemia-reperfusion injury by manganese–porphyrin complexes. Free Radic Res 2009; 41:127-34. [PMID: 17364938 DOI: 10.1080/10715760600801298] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Reactive oxygen and nitrogen species have been implicated in ischemia-reperfusion (I/R) injury. Metalloporphyrins (MP) are stable catalytic antioxidants that can scavenge superoxide, hydrogen peroxide, peroxynitrite and lipid peroxyl radicals. Studies were conducted with three manganese-porphyrin (MnP) complexes with varying superoxide dimutase (SOD) and catalase catalytic activity to determine if the MnP attenuates I/R injury in isolated perfused mouse livers. The release of the hepatocellular enzymes alanine aminotransferase (ALT), aspartate aminotransferase (AST) and lactate dehydrogenase (LDH) was maximal at 1 min reperfusion, decreased rapidly and increased gradually by 90 min. Manganese tetrakis-(N-ethyl-2 pyridyl) porphyrin (MnTE-2-PyP) decreased ALT, AST, LDH at 1-90 min reperfusion, while manganese tetrakis-(N-methyl-2 pyridyl) porphyrin (MnTM-2-PyP) and manganese tetrakis-(ethoxycarbonyl) porphyrin (MnTECP) decreased ALT and LDH from 5 to 90 min reperfusion. The release of thiobarbituric acid-reacting substances (TBARS) was diminished by MnTE-2-PyP and MnTM-2-PyP at 90 min. The extent of protein nitration (nitrotyrosine, NT) was decreased in all three MnPs treated livers. These results demonstrate that MnP complexes can attenuate hepatic I/R injury and may have therapeutic implications in disease states involving oxidants.
Collapse
Affiliation(s)
- Tzong-Jin Wu
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
21
|
Gedik E, Girgin S, Obay BD, Ozturk H, Ozturk H, Buyukbayram H. Iloprost, a prostacyclin (PGI2) analogue, reduces liver injury in hepatic ischemiareperfusion in rats. Acta Cir Bras 2009; 24:226-32. [DOI: 10.1590/s0102-86502009000300012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Accepted: 03/18/2009] [Indexed: 11/22/2022] Open
Abstract
PURPOSE: To evaluate the effects of iloprost a prostacyclin analogue on the hepatic IR injury in rats. METHODS: Forty male Sprague-Dawley rats (250-300 g) were divided into four groups each containing 10 rats;(1)- controls: data from unmanipulated animals; (2) sham group: rats subjected to the surgical procedure, except for liver I/R, and given saline; (3) I/R group: rats that underwent liver ischemia for 45 min followed by reperfusion for 45 min; (4) IR/ Iloprost group: rats pretreated with iloprost (10 µg kg-1, i.v). Liver tissues were taken to determine SOD, CAT, GSH, and MDA levels and for biochemical and histological evaluation. RESULTS: The plasma ALT and AST levels were increased in group 3 than in group 4. MDA values and the liver injury score decreased, while the SOD, CAT, and GSH values increased in group 4 compared to group 3. In group 3, hepatocytes were swollen with marked vacuolization. In group 4, there were regular sinusoidal structures with normal morphology without any signs of congestion. CONCLUSION: We demonstrated hepatoprotective effects of iloprost against severe ischemia and reperfusion injury in rat liver.
Collapse
|
22
|
Shaik IH, Mehvar R. Effects of cytochrome p450 inhibition by cimetidine on the warm hepatic ischemia-reperfusion injury in rats. J Surg Res 2008; 159:680-8. [PMID: 19500799 DOI: 10.1016/j.jss.2008.09.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Revised: 08/11/2008] [Accepted: 09/11/2008] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cimetidine is an H(2)-antagonist with cytochrome P450 (P450) inhibitory activity. Recent studies showed that cimetidine improves warm ischemia-reperfusion (IR) injury in isolated rat heart and rabbit lung and in primary cultures of rat proximal tubule epithelial cells by inhibiting P450-mediated reactive oxygen species generation. Here, we studied the effects of cimetidine on the warm IR injury in the liver. METHODS Three groups of rats were treated with a single i.p. dose (0.6 mmol/kg) of cimetidine or ranitidine (an H(2) antagonist without a significant P450 inhibitory activity) or with saline 1.5 h before surgery. Livers were then subjected to 1 h of in vivo ischemia, followed by 1 h of ex vivo reperfusion using a physiologic buffer in a recirculating manner. A fourth group of animals, receiving saline pretreatment underwent sham operation instead of ischemia. Perfusate and bile samples were collected during the reperfusion, and the liver tissue was collected at the end of reperfusion period for measurement of various biochemical markers. RESULTS Warm IR resulted in a significant increase in the perfusate concentrations of liver enzymes (3- to 4.5-fold) and hepatic concentrations of lipid hydroperoxides (2-fold). Whereas the glutathione concentrations in the liver tissue were not affected by IR injury, the injury caused a significant decrease ( approximately 40%) in the biliary glutathione excretion. Cimetidine treatment completely or partially reversed all the IR-mediated changes, while ranitidine was ineffective. The protective effects of cimetidine were associated with a 60% decline in the microsomal CYP2C11 activity. CONCLUSIONS Whereas cimetidine, an H(2) blocker with substantial P450 inhibitory activity, is protective in warm IR injury, ranitidine, a similar drug with no significant P450 inhibitory activity, is devoid of any protective effects. Therefore, P450 inhibition appears to be the underlying mechanism in the protective effects of cimetidine in this model of IR injury.
Collapse
Affiliation(s)
- Imam H Shaik
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, USA
| | | |
Collapse
|
23
|
Vardanian AJ, Busuttil RW, Kupiec-Weglinski JW. Molecular mediators of liver ischemia and reperfusion injury: a brief review. Mol Med 2008; 14:337-45. [PMID: 18292799 DOI: 10.2119/2007-00134.vardanian] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Accepted: 02/08/2008] [Indexed: 12/20/2022] Open
Abstract
Ischemia and reperfusion injury is a dynamic process that involves multiple organ systems in various clinical states including transplantation, trauma, and surgery. Research into this field has identified key molecular and signaling players that mediate, modulate, or augment cellular, tissue, and organ injury during this disease process. Further elucidation of the molecular mechanisms should provide the rationale to identify much-needed novel therapeutic options to prevent or ameliorate organ damage due to ischemia and reperfusion in clinics.
Collapse
Affiliation(s)
- Andrew J Vardanian
- The Dumont UCLA Transplantation Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, United States of America
| | | | | |
Collapse
|
24
|
Fukuda KI, Asoh S, Ishikawa M, Yamamoto Y, Ohsawa I, Ohta S. Inhalation of hydrogen gas suppresses hepatic injury caused by ischemia/reperfusion through reducing oxidative stress. Biochem Biophys Res Commun 2007; 361:670-4. [PMID: 17673169 DOI: 10.1016/j.bbrc.2007.07.088] [Citation(s) in RCA: 289] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2007] [Accepted: 07/14/2007] [Indexed: 12/13/2022]
Abstract
We have recently showed that molecular hydrogen has great potential for selectively reducing cytotoxic reactive oxygen species, such as hydroxyl radicals, and that inhalation of hydrogen gas decreases cerebral infarction volume by reducing oxidative stress [I. Ohsawa, M. Ishikawa, K. Takahashi, M. Watanabe, K. Nishimaki, K. Yamagata, K.-I. Katsura, Y. Katayama, S. Asoh, S. Ohta, Hydrogen acts as a therapeutic antioxidant by selectively reducing cytotoxic oxygen radicals, Nat. Med., 13 (2007) 688-694]. Here we show that the inhalation of hydrogen gas is applicable for hepatic injury caused by ischemia/reperfusion, using mice. The portal triad to the left lobe and the left middle lobe of the liver were completely occluded for 90min, followed by reperfusion for 180min. Inhalation of hydrogen gas (1-4%) during the last 190min suppressed hepatic cell death, and reduced levels of serum alanine aminotransferase and hepatic malondialdehyde. In contrast, helium gas showed no protective effect, suggesting that the protective effect by hydrogen gas is specific. Thus, we propose that inhalation of hydrogen gas is a widely applicable method to reduce oxidative stress.
Collapse
Affiliation(s)
- Kei-ichi Fukuda
- Department of Biochemistry and Cell Biology, Institute of Development and Aging Sciences, Graduate School of Medicine, Nippon Medical School, Kawasaki, Kanagawa 211-8533, Japan
| | | | | | | | | | | |
Collapse
|
25
|
Jassem W, Armeni T, Quiles JL, Bompadre S, Principato G, Battino M. Protection of mitochondria during cold storage of liver and following transplantation: comparison of the two solutions, University of Wisconsin and Eurocollins. J Bioenerg Biomembr 2006; 38:49-55. [PMID: 16721654 DOI: 10.1007/s10863-006-9005-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2005] [Accepted: 12/27/2005] [Indexed: 11/29/2022]
Abstract
Injury to allografts during ischaemia/reperfusion contribute to the development of graft failure following transplantation with significant morbidity and mortality to patients. The development of University of Wisconsin solution has significantly improved the quality of graft preservation and transplant outcome relative to formerly used solutions such as Eurocollins. The aim of this study was to further characterize mitochondrial structural and functional alterations occurring in rat livers following cold storage and transplantation. Mitochondrial impairment after prolonged storage in Eurocollins included decreased cyt. c+c1, cyt. b and cyt. a+a3 concentration and dramatic falls in the activities of the respiratory chain enzymes ubiquinol-cyt. c oxidoreductase and cytochrome oxidase. Under the same conditions the highest hydroperoxide but lowest vitamin E concentrations were also found. Although both the Eurocollins and University of Wisconsin preservation solutions have limitations in preventing oxidative injuries following cold storage and reperfusion, our data indicate that mitochondrial impairment was higher in Eurocollins- than in University of Wisconsin-stored livers. Further improvements are necessary in maintaining the stability of mitochondria in order to optimize preservations solutions used in transplantations.
Collapse
Affiliation(s)
- Wayel Jassem
- Institute of Biology and Genetics, Università Politecnica delle Marche, Ancona, Italy
| | | | | | | | | | | |
Collapse
|
26
|
Suzuki F, Hashikura Y, Ise H, Ishida A, Nakayama J, Takahashi M, Miyagawa SI, Ikeda U. MCI-186 (edaravone), a free radical scavenger, attenuates hepatic warm ischemia-reperfusion injury in rats. Transpl Int 2005; 18:844-53. [PMID: 15948865 DOI: 10.1111/j.1432-2277.2005.00094.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hepatic warm ischemia-reperfusion injury (IRI) during hepatectomy and liver transplantation is a major cause of liver dysfunction in which the pathologic role of free radicals is a major concern. To assess the effect of MCI-186 (edaravone) on hepatic IRI, male Wistar rats were subjected to partial hepatic ischemia for 60 min after pretreatment with vehicle (group C) or MCI-186 (group M), or after both MCI-186 pretreatment and additional administration of MCI-186 12 h after reperfusion (group MX). Groups M and MX showed significantly lower levels of serum alanine aminotransferase and hepatic lipid peroxidation than group C, and also significantly lower expression levels of mRNA for cytokines, chemokines and intercellular adhesion molecule-1. There were fewer tissue monocytes and neutrophils in groups M and MX than in group C. These effects were more marked in group MX than in group M. Our findings suggest that treatment with MCI-186 attenuates hepatic IRI in this rat in vivo model.
Collapse
Affiliation(s)
- Fumitaka Suzuki
- Department of Organ Regeneration, Shinshu University Graduate School of Medicine, Asahi, Matsumoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Fukai M, Hayashi T, Yokota R, Shimamura T, Suzuki T, Taniguchi M, Matsushita M, Furukawa H, Todo S. Lipid peroxidation during ischemia depends on ischemia time in warm ischemia and reperfusion of rat liver. Free Radic Biol Med 2005; 38:1372-81. [PMID: 15855055 DOI: 10.1016/j.freeradbiomed.2005.02.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2004] [Revised: 01/28/2005] [Accepted: 02/02/2005] [Indexed: 11/20/2022]
Abstract
Prolonged hepatic warm ischemia has been incriminated in oxidative stress after reperfusion. However, the magnitude of oxidative stress during ischemia has been controversial. The aims of the present study were to elucidate whether lipid peroxidation progressed during ischemia and to clarify whether oxidative stress during ischemia aggravated the oxidative damage after reperfusion. Rats were subjected to 30 to 120 min of 70% warm ischemia alone or followed by reperfusion for 60 min. Lipid peroxidation (LPO) was evaluated by amounts of phosphatidylcholine hydroperoxide (PC-OOH) and phosphatidylethanolamine hydroperoxide (PE-OOH) as primary LPO products. Total amounts of malondialdehyde and 4-hydroxy-2-nonenal (MDA + 4-HNE), degraded from hydroperoxides, were also determined. PC-OOH and PE-OOH significantly increased at 60 and 120 min ischemia with concomitant increase of oxidized glutathione. These hydroperoxides did not increase at 60 min reperfusion after 60 min ischemia, whereas they did increase at 60 min reperfusion after 120 min ischemia with deactivation of phospholipid hydroperoxide glutathione peroxidase and superoxide dismutase. The amount of MDA + 4-HNE exhibited similar changes, but the velocity of production dropped with ischemic time longer than 60 min. In conclusion, oxidative stress progressed during ischemia and triggered the oxidative injury after reperfusion. Secondary LPO products are less sensitive, especially during ischemia, which may cause possible underestimation and discrepancy.
Collapse
Affiliation(s)
- Moto Fukai
- First Department of Surgery, Hokkaido University School of Medicine, N15 W7, Kita-ku, Sapporo 060-8638, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Chang EJ, Lee SH, Mun KC, Suh SI, Bae JH, Kim SP, Choi HJ, Cho KB, Hwang JS. Effect of artificial cells on hepatic function after ischemia–reperfusion injury in liver. Transplant Proc 2004; 36:1959-61. [PMID: 15518711 DOI: 10.1016/j.transproceed.2004.08.092] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND The liver suffers from ischemia/reperfusion injury during transplantation. Reactive oxygen species generated by xanthine oxidase during reperfusion of the ischemic liver may be partially responsible for the hepatic injury. Oxygen free radicals are removed by antioxidant enzymes such as superoxide dismutase (SOD), catalase, and glutathione peroxidase. Using glutaraldehyde and lysine we constructed crosslinked hemoglobin, containing SOD and catalase, and assessed its ability to protect against ischemia/reperfusion injury during transplantation. METHODS In contrast to the sham-operated control groups, blood was exchanged using crosslinked hemoglobin (polyHb) a PolyHb-SOD-catalase (PSC) group. After ischemia/reperfusion injury, several parameters of hepatic damage and oxygen free radicals were measured as well as microscopic examination. RESULTS Alanine aminotransferase, aspartate aminotransferase, superoxide production, hydrogen peroxide, and malondialdehyde levels were higher among the PolyHb group than sham-operated controls. The PolyHb group revealed a few apoptotic bodies, some acute inflammatory infiltrates in the sinusoids, nuclear fragmentations, cell shrinkage, and chromatin clumping with formation of apoptotic bodies in the apoptotic cells under microscopic examination. Alanine aminotransferase, aspartate aminotransferase, superoxide production, and hydrogen peroxide levels were lower in the PSC than the PolyHb group. Hepatic structures were well preserved in the PSC group. CONCLUSIONS Reactive oxygen species contribute to hepatic dysfunction with morphologic changes. PSC is effective to reduce hepatic damage by lowering oxygen free radical-mediated injury after ischemia/reperfusion in the liver.
Collapse
Affiliation(s)
- E J Chang
- Institute for Medical Science and Chronic Disease Research Center, Keimyung University School of Medicine, Daegu, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Hayashi T, Uchida K, Takebe G, Takahashi K. Rapid formation of 4-hydroxy-2-nonenal, malondialdehyde, and phosphatidylcholine aldehyde from phospholipid hydroperoxide by hemoproteins. Free Radic Biol Med 2004; 36:1025-33. [PMID: 15059643 DOI: 10.1016/j.freeradbiomed.2004.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2003] [Revised: 01/12/2004] [Accepted: 01/16/2004] [Indexed: 01/18/2023]
Abstract
4-Hydroxy-2-nonenal (HNE) and malondialdehyde (MDA) are well-known toxic products of lipid peroxidation. Phosphatidylcholine aldehydes are also known as oxidation products of phosphatidylcholine. The mechanism of the formation of these compounds in vivo has been a long-standing question. We observed that the rapid reaction of hemoproteins (methemoglobin, metmyoglobin, and cytochrome c) with 1-palmitoyl-2-(13-hydroperoxy-cis-9, trans-11-octadecadienoyl) phosphatidylcholine (PLPC-OOH), having a hydroperoxylinoleoyl residue, generated HNE, MDA, and the phosphatidylcholine aldehyde 1-palmitoyl-2-(9-oxononanoyl) phosphatidylcholine. The efficiencies (mol% yield) of the formation of HNE and MDA from decomposed PLPC-OOH by methemoglobin, metmyoglobin, and cytochrome c after incubation for 10 min were 1.6, 1.0, and 1.0% for HNE and 1.2, 0.6, and 0.9% for MDA, respectively. When 1-palmitoyl-2-linoleoyl phosphatidylcholine was incubated with lipoxidase and methemoglobin, the formation of HNE and the phosphatidylcholine aldehyde 1-palmitoyl-2-(9-oxononanoyl) phosphatidylcholine was observed. When 1-palmitoyl-2-arachidonyl phosphatidylcholine was used instead of 1-palmitoyl-2-linoleoyl phosphatidylcholine, the phosphatidylcholine aldehyde 1-palmitoyl-2-oxovaleroyl phosphatidylcholine was obtained. These data suggest that HNE and phosphatidylcholine aldehydes might be rapidly formed from phosphatidylcholine by lipoxygenase and hemoproteins. Furthermore, hemichrome, converted from methemoglobin by deoxycholic acid and ursodeoxycholic acid, showed marked decomposition of HNE. These results suggest that hemoproteins are related to both the formation and the decomposition of HNE.
Collapse
Affiliation(s)
- Takaaki Hayashi
- Hokkaido Institute of Public Health, Kita 12, Nishi 6, Kita-ku, Sapporo 060-0819, Japan
| | | | | | | |
Collapse
|
30
|
Yoshinaga H, Watanabe M, Manome Y. Possible role of nicaraven in neuroprotective effect on hippocampal slice culture. Can J Physiol Pharmacol 2003; 81:683-9. [PMID: 12897815 DOI: 10.1139/y03-060] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nicaraven is an agent that is especially beneficial in vasospasm or brain damage caused by subarachnoid hemorrhage. It ameliorates neurological deficits of patients and protects the central nervous system from ischemia. We investigated the neuroprotective effect of nicaraven against oxygen-glucose deprivation (OGD) induced or N-methyl-D-aspartic acid (NMDA) induced hippocampal neuronal cell death in organotypic brain slice cultures. The effect of nicaraven on hippocampal neuronal injury was evaluated by inhibition of uptake of propidium iodide (PI) into dead cells. The results demonstrated that nicaraven protected neuronal cells from both OGD- and NMDA-induced cell death. While nicaraven has a strong hydroxyl radical scavenging effect, another radical scavenger, N-acetyl-L-cysteine (NAC), inhibited cell death only caused by OGD. In contrast, the poly(ADP-ribose) synthetase (PARS) inhibitors 3-aminobenzamide (3-AB) and theophylline protected cells from both OGD- and NMDA-induced cell death. Since nicaraven has an inhibitory effect in PARS, as well as a radical scavenging effect, these results suggest that inhibition of hippocampal cell death caused by NMDA may be attributable to PARS inhibition by nicaraven.
Collapse
Affiliation(s)
- Hisao Yoshinaga
- Department of Microbiology, Jikei University School of Medicine, 3-25-8 Nishishinbashi, Minato-ku, Tokyo, Japan 105-8461
| | | | | |
Collapse
|
31
|
Yoshida M, Horiuchi T, Uchinami M, Tabo T, Kimura N, Yokomachi J, Doi K, Nakamura T, Tamagawa K, Tanaka K. Intermittent hepatic ischemia-reperfusion minimizes liver metastasis in rats. J Surg Res 2003; 111:255-60. [PMID: 12850471 DOI: 10.1016/s0022-4804(03)00082-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Surgical stresses, including hepatic ischemia-reperfusion (I/R), promote cancer growth and metastasis. We have reported that continuous hepatic I/R increases liver damage and promoted liver metastasis from colon cancer, whereas intermittent I/R causes less liver damage. We therefore examined whether intermittent I/R could reduce liver metastasis in a rat model. MATERIALS AND METHODS Adult male Fischer rats was divided between three groups: group A (control), which received laparotomy for 120 min with no liver ischemia; group B (continuous I/R), which received 60 min of 70% partial liver ischemia followed by 60 min of reperfusion; and group C (intermittent I/R), which received 15 min of 70% ischemia and 15 min of reperfusion, repeated four times. Just before closing the abdomen, all animals were inoculated intrasplenically with rat colon adenocarcinoma cells (RCN-H4). Tumor nodules on the liver surface were counted 3 weeks later. In addition, expression of E-selectin mRNA in liver was examined at 1, 3, and 6 h after completing I/R by a reverse transcription-polymerase chain reaction. RESULTS Continuous I/R (B) greatly promoted liver metastasis in both ischemic and nonischemic liver lobes, whereas intermittent I/R (C) showed significantly fewer metastasis than group B in both lobes. Significantly less E-selectin mRNA was expressed in group C than in group B. CONCLUSIONS Intermittent I/R limits expression of E-selectin mRNA and liver metastasis. Intermittent hepatic I/R is less stressful than continuous I/R, minimizing liver metastasis by colon cancer cells through avoidance of E-selectin up-regulation.
Collapse
Affiliation(s)
- Makoto Yoshida
- Second Department of Surgery, Fukui Medical University, Fukui, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Mun KC, Bae JH, Suh SI, Kim YH, Lee SH, Kim SP, Kwon TK, Hwang JS. Effect of modified polyhemoglobin on the oxidative damage after ischemia-reperfusion in the liver. Transplant Proc 2003; 35:126-7. [PMID: 12591334 DOI: 10.1016/s0041-1345(02)03979-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- K C Mun
- Institute for Medical Science, Keimyung University School of Medicine, Taegu, Korea
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Reduction of ischemia/reperfusion injury in organ transplants by cytoprotective strategies. Curr Opin Organ Transplant 2001. [DOI: 10.1097/00075200-200103000-00013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
34
|
Yamamoto H, Watanabe T, Mizuno H, Endo K, Hosokawa T, Kazusaka A, Gooneratne R, Fujita S. In vivo evidence for accelerated generation of hydroxyl radicals in liver of Long-Evans Cinnamon (LEC) rats with acute hepatitis. Free Radic Biol Med 2001; 30:547-54. [PMID: 11182525 DOI: 10.1016/s0891-5849(00)00496-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The Long-Evans Cinnamon (LEC) rats accumulate excess copper (Cu) in the liver in a manner similar to patients with Wilson's disease (WD) and spontaneously develop acute hepatitis with severe jaundice. Although hydroxyl radicals (*OH) have been proposed to be a cause of hepatitis by the accumulation of Cu, it is not clear whether or not *OH can be produced in the liver of hepatitic LEC rats in vivo and also can be involved in the onset of hepatitis. In the present study, *OH production in plasma and liver of hepatitic LEC rats was quantified by trapping *OH with salicylic acid (SA) as 2, 3-dihydroxybenzoic acid (2, 3-DHBA). The ratios of 2, 3-DHBA/SA were significantly higher in plasma and liver of hepatitic LEC rats than those of Wistar rats and LEC rats showing no signs of hepatitis. Furthermore, the ratios of 2, 3-DHBA/SA in plasma and liver of hepatitic LEC rats were almost the same as those of Wistar rats treated orally with CuSO(4) (0.5 mmol/kg) 2 h before acetylsalicylic acid (ASA) injection. We also evaluated the protective effects of D-mannitol (a *OH scavenger) treatment against acute hepatitis in LEC rats. D-mannitol (500 mg/kg) was administered intraperitoneally to 10-week-old LEC rats for 3 weeks. D-mannitol treatment suppressed the increases in serum aspartate aminotransferase activity and total bilirubin concentration. In addition, D-mannitol treatment significantly reduced hepatic mitochondrial lipid peroxidation, which is thought to be important in the pathogenesis of Cu-induced hepatotoxicity. These observations suggest that accelerated generation of *OH catalyzed by free Cu in the liver may, at least in part, play a role in the pathogenesis of acute hepatitis in LEC rats.
Collapse
Affiliation(s)
- H Yamamoto
- Medicinal Safety Research Laboratories, Sankyo Co., Ltd., Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
In the liver, the progressive accumulation of connective tissue, a complex and dynamic process termed fibrosis, represents a very frequent event following a repeated or chronic insult of sufficient intensity to trigger a "wound healing"-like reaction. The fibrotic process recognises the involvement of various cells and different factors in bringing about an excessive fibrogenesis with disruption of intercellular contacts and interactions and of extracellular matrix composition. However, Kupffer cells, together with recruited mononuclear cells, and hepatic stellate cells are by far the key-players in liver fibrosis. Their cross-talk is triggered and favoured by a series of chemical mediators, with a prominent role played by the transforming growth factor beta. Both expression and synthesis of this inflammatory and pro-fibrogenic cytokine are mainly modulated through redox-sensitive reactions. Further, involvement of reactive oxygen species and lipid peroxidation products can be clearly demonstrated in other fundamental events of hepatic fibrogenesis, like activation and effects of stellate cells, expression of metalloproteinases and of their specific inhibitors. The important outcome of such findings as regards the pathogenesis of liver fibrosis derives from the observation of a consistent and marked oxidative stress condition in many if not all chronic disease processes affecting hepatic tissue. Hence, reactive oxidant species likely contribute to both onset and progression of fibrosis as induced by alcohol, viruses, iron or copper overload, cholestasis, hepatic blood congestion.
Collapse
Affiliation(s)
- G Poli
- Department of Clinical and Biological Sciences, S. Luigi Gonzaga Hospital, University of Turin, 10043 Orbassano, Turin, Italy
| |
Collapse
|