1
|
Rajendran P. Unveiling the power of flavonoids: A dynamic exploration of their impact on cancer through matrix metalloproteinases regulation. Biomedicine (Taipei) 2024; 14:12-28. [PMID: 38939095 PMCID: PMC11204124 DOI: 10.37796/2211-8039.1447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/15/2023] [Accepted: 12/22/2023] [Indexed: 06/29/2024] Open
Abstract
Cancer stands as a significant contributor to global mortality rates, primarily driven by its progression and widespread dissemination. Despite notable strides in cancer therapy, the efficacy of current treatment strategies is compromised due to their inherent toxicity and the emergence of chemoresistance. Consequently, there is a critical need to evaluate alternative therapeutic approaches, with natural compounds emerging as promising candidates, showcasing demonstrated anticancer capabilities in various research models. This review manuscript presents a comprehensive examination of the regulatory mechanisms governing the expression of matrix metalloproteinases (MMPs) and delves into the potential therapeutic role of flavonoids as agents exhibiting specific anticancer activity against MMPs. The primary aim of this study is to elucidate the diverse functions associated with MMP production in cancer and to investigate the potential of flavonoids in modulating MMP expression to inhibit metastasis.
Collapse
Affiliation(s)
- Peramaiyan Rajendran
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, 31982, Saudi Arabia
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, Tamil Nadu, India
| |
Collapse
|
2
|
Wang JW, Li YJ, Wu HH, Hsu HH, Chang MY, Wang RY, Tian YC. The essential role of the ERK activation in large T antigen of BK polyomavirus regulated cell migration. Virus Res 2023; 336:199220. [PMID: 37689160 PMCID: PMC10507160 DOI: 10.1016/j.virusres.2023.199220] [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: 08/11/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
Recent studies have suggested that BK polyomavirus (BKPyV) may be associated with the development of urothelial carcinoma. In Merkel cell carcinoma, TAg and tAg are the major viral proteins of Merkel cell polyomavirus with oncogenic potential. In this study, we aimed to distinguish the role of TAg and tAg in cell migration. Our result demonstrated that ERK was phosphorylated in human renal tubular cells expressing its TAg and tAg after BKPyV infection. Treatment with the ERK inhibitor U0126 suppressed BKPyV gene expression and reduced BKPyV replication. Both TAg and tAg induced cell migration via ERK-dependent signaling. Furthermore, the expression of TAg and tAg had a significant regulatory effect on focal adhesion molecules in renal proximal tubular cells, which strongly suggests that alterations in the focal adhesion complexes are critically involved in TAg and tAg-induced cell migration. Gelatin zymography profiling revealed that TAg regulates the expression and activity of MMP-2 and MMP-9, but not tAg. Interestingly, TAg regulates the expression and activity of MMP-9 through ERK signaling, whereas MMP-2 is regulated through an ERK-independent pathway. Unbalanced ERK pathway activity is frequently observed in many cancers, while MMP proteins are usually overexpressed in aggressive tumors. These findings support the view that BKPyV is an oncogenic virus.
Collapse
Affiliation(s)
- Jiun-Wen Wang
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 333, Taiwan; Kidney Research Center and Department of Nephrology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Yi-Jung Li
- Kidney Research Center and Department of Nephrology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; Department of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Hsin-Hsu Wu
- Kidney Research Center and Department of Nephrology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan 333, Taiwan
| | - Hsiang-Hao Hsu
- Kidney Research Center and Department of Nephrology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Ming-Yang Chang
- Kidney Research Center and Department of Nephrology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Robert Yl Wang
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 333, Taiwan; Kidney Research Center and Department of Nephrology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan.
| | - Ya-Chung Tian
- Kidney Research Center and Department of Nephrology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; Department of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan 333, Taiwan.
| |
Collapse
|
3
|
Gao D, Yuan S, Ji X, Su Y, Qi Z. The neuroprotective role of prolonged normobaric oxygenation applied during ischemia and in the early stage of reperfusion in cerebral ischemic rats. Brain Res 2023; 1816:148464. [PMID: 37328087 DOI: 10.1016/j.brainres.2023.148464] [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: 04/10/2023] [Revised: 05/19/2023] [Accepted: 06/12/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Recanalization is the main treatment option for ischemic stroke. However, prognosis remains poor for about half of patients after recanalization, possibly due to the "no-reflow" phenomenon at the early phase of recanalization. Normobaric oxygenation (NBO) during ischemia can reportedly maintain the partial pressure of oxygen and exert a protective effect in ischemic brain tissue. OBJECTIVES AND METHODS This study investigated whether prolonged NBO treatment during ischemia and the early phase of reperfusion (i/rNBO) has neuroprotective effects and to elucidate the underlying mechanisms in rats with middle cerebral artery occlusion plus reperfusion. RESULTS NBO treatment significantly elevated the level of O2 in the atmosphere and arterial blood without altering the level of CO2. The infarcted cerebral volume was significantly reduced by application of i/rNBO as compared to iNBO (applied during ischemia) or rNBO (applied at the early phase of reperfusion), indicating better protective effects of i/rNBO. i/rNBO more effectively suppressed s-nitrosylation of MMP-2 (amplifying inflammation) as compared to iNBO and rNBO, dramatically downregulated the cleavage of poly(ADP-ribose)polymerase-1 (PARP-1, acting as the substrate of MMP-2), and suppressed neuronal apoptosis, as determined by the TUNEL assay and staining for NeuN. These results demonstrated that application of i/rNBO in the early stage of reperfusion significantly alleviated neuronal apoptosis via suppression of the MMP-2/PARP-1 pathway. CONCLUSIONS The mechanism underlying the neuroprotective role of i/rNBO involved prolonged NBO treatment for cerebral ischemia, suggesting that i/rNBO may allow expansion of the time window for NBO application in stroke patients following vascular recanalization.
Collapse
Affiliation(s)
- Daiquan Gao
- Department of Neurology, Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Shuhua Yuan
- Department of Neurology, Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xunming Ji
- Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yingying Su
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.
| | - Zhifeng Qi
- Department of Neurology, Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Wendt TS, Gonzales RJ. Ozanimod differentially preserves human cerebrovascular endothelial barrier proteins and attenuates matrix metalloproteinase-9 activity following in vitro acute ischemic injury. Am J Physiol Cell Physiol 2023; 325:C951-C971. [PMID: 37642239 DOI: 10.1152/ajpcell.00342.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023]
Abstract
Endothelial integrity is critical in mitigating a vicious cascade of secondary injuries following acute ischemic stroke (AIS). Matrix metalloproteinase-9 (MMP-9), a contributor to endothelial integrity loss, is elevated during stroke and is associated with worsened stroke outcome. We investigated the FDA-approved selective sphingosine-1-phosphate receptor 1 (S1PR1) ligand, ozanimod, on the regulation/activity of MMP-9 as well as endothelial barrier components [platelet endothelial cell adhesion molecule 1 (PECAM-1), claudin-5, and zonula occludens 1 (ZO-1)] in human brain microvascular endothelial cells (HBMECs) following hypoxia plus glucose deprivation (HGD). We previously reported that S1PR1 activation improves HBMEC integrity; however, mechanisms underlying S1PR1 involvement in endothelial cell barrier integrity have not been clearly elucidated. We hypothesized that ozanimod would attenuate an HGD-induced increase in MMP-9 activity that would concomitantly attenuate the loss of integral barrier components. Male HBMECs were treated with ozanimod or vehicle and exposed to 3 h of normoxia (21% O2) or HGD (1% O2). Immunoblotting, zymography, qRT-PCR, and immunocytochemical labeling techniques assessed processes related to MMP-9 and barrier markers. We observed that HGD acutely increased MMP-9 activity and reduced claudin-5 and PECAM-1 levels, and ozanimod attenuated these responses. In situ analysis, via PROSPER, suggested that attenuation of MMP-9 activity may be a primary factor in maintaining these integral barrier proteins. We also observed that HGD increased intracellular mechanisms associated with augmented MMP-9 activation; however, ozanimod had no effect on these select factors. Thus, we conclude that ozanimod has the potential to attenuate HGD-mediated decreases in HBMEC integrity in part by decreasing MMP-9 activity as well as preserving barrier properties.NEW & NOTEWORTHY We have identified a potential novel mechanism by which ozanimod, a selective sphingosine-1-phosphate receptor 1 (S1PR1) agonist, attenuates hypoxia plus glucose deprivation (HGD)-induced matrix metalloproteinase-9 (MMP-9) activity and disruptions in integral human brain endothelial cell barrier proteins. Our results suggest that ischemic-like injury elicits increased MMP-9 activity and alterations of barrier integrity proteins in human brain microvascular endothelial cells (HBMECs) and that ozanimod via S1PR1 attenuates these HGD-induced responses, adding to its therapeutic potential in cerebrovascular protection during the acute phase of ischemic stroke.
Collapse
Affiliation(s)
- Trevor S Wendt
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona, United States
| | - Rayna J Gonzales
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona, United States
| |
Collapse
|
5
|
Betsou A, Lambropoulou M, Georgakopoulou AE, Kostomitsopoulos N, Konstandi O, Anagnostopoulos K, Tsalikidis C, Simopoulos CE, Valsami G, Tsaroucha AK. The hepatoprotective effect of silibinin after hepatic ischemia/reperfusion in a rat model is confirmed by immunohistochemistry and qRT-PCR. J Pharm Pharmacol 2021; 73:1274-1284. [PMID: 33847359 DOI: 10.1093/jpp/rgab062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 03/19/2021] [Indexed: 01/18/2023]
Abstract
OBJECTIVES We investigated the positive effect of silibinin after IV administration as silibinin-hydroxypropyl-β-cyclodextrin lyophilized product, by measuring gene expression and liver tissue protein levels of tumor necrosis factor-α, interleukin-6, monocyte chemoattractant protein-1, matrix metalloproteinases matrix metalloproteinases and tissue inhibitor of matrix metalloproteinases-2. METHODS 63 Wistar rats of age 13.24±4.40 weeks underwent ischemia/reperfusion (I/R) injury of the liver. The animals were randomized into three groups: Sham (S; n = 7); Control (C; n-28); silibinin (Si; n-28). The C and Si groups underwent 45 min ischemia. Si received silibinin-hydroxypropyl-β-cyclodextrin intravenously immediately before reperfusion at a dose of 5 mg/kg. Both groups were further divided into 4 subgroups, based on euthanasia time (i.e., 60, 120, 180 and 240 min). KEY FINDINGS qRT-PCR results confirmed the statistically significant reduction of the expression of the pro-inflammatory factors at 240 min after I/R injury (tumor necrosis factor-α: P < 0.05; MCR1: P < 0.05) and matrix metalloproteinases (matrix metalloproteinases 2: P < 0.05; matrix metalloproteinases 3: P < 0.05) and the increase of tissue inhibitor of matrix metalloproteinases-2 in liver tissue in the Si group. Moreover, results of immunohistochemistry levels confirmed that at 240 min pro-inflammatory factors (tumor necrosis factor-α: P < 0.05; MCR1: P < 0.05) and matrix metalloproteinases ( matrix metalloproteinases 2: P < 0.05; matrix metalloproteinases 3: P < 0.05) had a statistically significantly lower expression in the Si group while tissue inhibitor of matrix metalloproteinases-2 had a higher expression. CONCLUSIONS Silibinin may have a beneficial effect on the protection of the liver.
Collapse
Affiliation(s)
- Afrodite Betsou
- Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Maria Lambropoulou
- Laboratory of Histology-Embryology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | | | | | - Ourania Konstandi
- Faculty of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Christos Tsalikidis
- Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- 2nd Department of Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Constantinos E Simopoulos
- Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- 2nd Department of Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Georgia Valsami
- School of Health Sciences, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexandra K Tsaroucha
- Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- 2nd Department of Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- Laboratory of Experimental Surgery and Surgical Research, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- Laboratory of Bioethics, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
6
|
Wang Q, Sundar IK, Blum JL, Ratner JR, Lucas JH, Chuang TD, Wang Y, Liu J, Rehan VK, Zelikoff JT, Rahman I. Prenatal Exposure to Electronic-Cigarette Aerosols Leads to Sex-Dependent Pulmonary Extracellular-Matrix Remodeling and Myogenesis in Offspring Mice. Am J Respir Cell Mol Biol 2021; 63:794-805. [PMID: 32853043 DOI: 10.1165/rcmb.2020-0036oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Electronic-cigarette (e-cig) vaping is a serious concern, as many pregnant women who vape consider it safe. However, little is known about the harmful effects of prenatal e-cig exposure on adult offspring, especially on extracellular-matrix (ECM) deposition and myogenesis in the lungs of offspring. We evaluated the biochemical and molecular implications of maternal exposure during pregnancy to e-cig aerosols on the adult offspring of both sexes, with a particular focus on pulmonary ECM remodeling and myogenesis. Pregnant CD-1 mice were exposed to e-cig aerosols with or without nicotine, throughout gestation, and lungs were collected from adult male and female offspring. Compared with the air-exposed control group, female mice exposed to e-cig aerosols, with or without nicotine, demonstrated increased lung protein abundance of LEF-1 (lymphoid enhancer-binding factor 1), fibronectin, and E-cadherin, whereas altered E-cadherin and PPARγ (peroxisome proliferator-activated receptor γ) levels were observed only in males exposed to e-cig aerosols with nicotine. Moreover, lipogenic and myogenic mRNAs were dysregulated in adult offspring in a sex-dependent manner. PAI-1 (plasminogen activator inhibitor-1), one of the ECM regulators, was significantly increased in females exposed prenatally to e-cig aerosols with nicotine and in males exposed to e-cig aerosols compared with control animals exposed to air. MMP9 (matrix metalloproteinase 9), a downstream target of PAI-1, was downregulated in both sexes exposed to e-cig aerosols with nicotine. No differences in lung histology were observed among any of the treatment groups. Overall, adult mice exposed prenatally to e-cig aerosols could be predisposed to developing pulmonary disease later in life. Thus, these findings suggest that vaping during pregnancy is unsafe and increases the propensity for later-life interstitial lung diseases.
Collapse
Affiliation(s)
- Qixin Wang
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester Medical Center, University of Rochester, Rochester, New York
| | - Isaac K Sundar
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester Medical Center, University of Rochester, Rochester, New York
| | - Jason L Blum
- Department of Environmental Medicine, School of Medicine, New York University, New York, New York; and
| | - Jill R Ratner
- Department of Environmental Medicine, School of Medicine, New York University, New York, New York; and
| | - Joseph H Lucas
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester Medical Center, University of Rochester, Rochester, New York
| | - Tsai-Der Chuang
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation, Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine, University of California Los Angeles, Torrance, California
| | - Ying Wang
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation, Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine, University of California Los Angeles, Torrance, California
| | - Jie Liu
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation, Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine, University of California Los Angeles, Torrance, California
| | - Virender K Rehan
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation, Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine, University of California Los Angeles, Torrance, California
| | - Judith T Zelikoff
- Department of Environmental Medicine, School of Medicine, New York University, New York, New York; and
| | - Irfan Rahman
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester Medical Center, University of Rochester, Rochester, New York
| |
Collapse
|
7
|
Wang Q, Sundar IK, Li D, Lucas JH, Muthumalage T, McDonough SR, Rahman I. E-cigarette-induced pulmonary inflammation and dysregulated repair are mediated by nAChR α7 receptor: role of nAChR α7 in SARS-CoV-2 Covid-19 ACE2 receptor regulation. Respir Res 2020; 21:154. [PMID: 32552811 PMCID: PMC7301079 DOI: 10.1186/s12931-020-01396-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022] Open
Abstract
Electronic cigarette (e-cig) vaping is increasing rapidly in the United States, as e-cigs are considered less harmful than combustible cigarettes. However, limited research has been conducted to understand the possible mechanisms that mediate toxicity and pulmonary health effects of e-cigs. We hypothesized that sub-chronic e-cig exposure induces inflammatory response and dysregulated repair/extracellular matrix (ECM) remodeling, which occur through the α7 nicotinic acetylcholine receptor (nAChRα7). Adult wild-type (WT), nAChRα7 knockout (KO), and lung epithelial cell-specific KO (nAChRα7 CreCC10) mice were exposed to e-cig aerosol containing propylene glycol (PG) with or without nicotine. Bronchoalveolar lavage fluids (BALF) and lung tissues were collected to determine e-cig induced inflammatory response and ECM remodeling, respectively. Sub-chronic e-cig exposure with nicotine increased inflammatory cellular influx of macrophages and T-lymphocytes including increased pro-inflammatory cytokines in BALF and increased SARS-Cov-2 Covid-19 ACE2 receptor, whereas nAChRα7 KO mice show reduced inflammatory responses associated with decreased ACE2 receptor. Interestingly, matrix metalloproteinases (MMPs), such as MMP2, MMP8 and MMP9, were altered both at the protein and mRNA transcript levels in female and male KO mice, but WT mice exposed to PG alone showed a sex-dependent phenotype. Moreover, MMP12 was increased significantly in male mice exposed to PG with or without nicotine in a nAChRα7-dependent manner. Additionally, sub-chronic e-cig exposure with or without nicotine altered the abundance of ECM proteins, such as collagen and fibronectin, significantly in a sex-dependent manner, but without the direct role of nAChRα7 gene. Overall, sub-chronic e-cig exposure with or without nicotine affected lung inflammation and repair responses/ECM remodeling, which were mediated by nAChRα7 in a sex-dependent manner.
Collapse
Affiliation(s)
- Qixin Wang
- Department of Environmental Medicine, University of Rochester Medical Center, Box 850, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Isaac K Sundar
- Department of Environmental Medicine, University of Rochester Medical Center, Box 850, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Dongmei Li
- Department of Clinical and Translational Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Joseph H Lucas
- Department of Environmental Medicine, University of Rochester Medical Center, Box 850, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Thivanka Muthumalage
- Department of Environmental Medicine, University of Rochester Medical Center, Box 850, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Samantha R McDonough
- Department of Environmental Medicine, University of Rochester Medical Center, Box 850, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Box 850, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| |
Collapse
|
8
|
Wang Q, Sundar I, Li D, Lucas J, Muthumalage T, McDonough S, Rahman I. E-cigarette-Induced Pulmonary Inflammation and Dysregulated Repair are Mediated by nAChR α7 Receptor: Role of nAChR α7 in ACE2 Covid-19 receptor regulation. RESEARCH SQUARE 2020:rs.2.23829. [PMID: 32702718 PMCID: PMC7336696 DOI: 10.21203/rs.2.23829/v2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Electronic cigarette (e-cig) vaping is increasing rapidly in the United States, as e-cigs are considered less harmful than combustible cigarettes. However, limited research has been conducted to understand the possible mechanism that mediate, toxicity and pulmonary health effects of e-cigs. We hypothesized that sub-chronic e-cig exposure induces inflammatory response and dysregulated repair/extracellular matrix (ECM) remodeling, which occur through the α7 nicotinic acetylcholine receptor (nAChR α7). Adult wild-type (WT), nAChRα7 knockout (KO), and lung epithelial cell-specific KO (nAChRα7 CreCC10) mice were exposed to e-cig aerosol containing propylene glycol (PG) with or without nicotine. Bronchoalveolar lavage fluids (BALF) and lungs tissues were collected to determine e-cig induced inflammatory response and ECM remodeling, respectively. Sub-chronic e-cig exposure with nicotine increased the inflammatory cellular influx of macrophages and T-lymphocytes including increased pro-inflammatory cytokines in BALF and increased ACE2 Covid-19 receptor, whereas nAChR α7 KO mice show reduced inflammatory responses associated with decreased ACE2 receptor. Interestingly, matrix metalloproteinases (MMPs), such as MMP2, MMP8, and MMP9 were altered both at the protein and mRNA transcript levels in female and male, but WT mice exposed to PG alone showed a sex-dependent phenotype. Moreover, MMP12 was increased significantly in male mice exposed to PG with or without nicotine in a nAChR α7-dependent manner. Additionally, sub-chronic e-cig exposure with or without nicotine altered the abundance of ECM proteins, such as collagen and fibronectin significantly in a sex-dependent manner, but without the direct role of nAChR α7 gene. Overall, sub-chronic e-cig exposure with or without nicotine affected lung inflammation and repair responses/ECM remodeling, which were mediated by nAChR α7 in a sex-dependent manner.
Collapse
|
9
|
Dusabimana T, Kim SR, Kim HJ, Park SW, Kim H. Nobiletin ameliorates hepatic ischemia and reperfusion injury through the activation of SIRT-1/FOXO3a-mediated autophagy and mitochondrial biogenesis. Exp Mol Med 2019; 51:1-16. [PMID: 31028246 PMCID: PMC6486618 DOI: 10.1038/s12276-019-0245-z] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 01/23/2023] Open
Abstract
Hepatic ischemia and reperfusion injury are characterized by impaired autophagy, mitochondrial dysfunction, and subsequent compromise of cellular homeostasis following hepatic surgery or transplantation. Nobiletin, a natural flavonoid, is a beneficial antioxidant that possesses anti-inflammatory and anti-cancer activities. We investigated the effect of nobiletin on hepatic IR injury and described the underlying mechanisms. C57BL/6 mice were subjected to 60 min of partial hepatic ischemia, treated with nobiletin (5 mg/kg) or vehicle at the start of reperfusion, and killed at 5 h of reperfusion. Hepatic ischemia and reperfusion increased hepatocellular oxidative damage, inflammation, and cell death, but these changes were alleviated upon nobiletin treatment. Nobiletin increased the expression of proteins that control autophagy, mitochondrial dynamics, and biogenesis. Specifically, the SIRT-1/FOXO3a and PGC-1α pathways were activated by nobiletin. IR-induced AKT activation was associated with FOXO3a phosphorylation, which resulted in a significant reduction in the nuclear FOXO3a levels and potentially attenuated autophagy-regulatory gene expression. Nobiletin increased FOXO3a expression and its nuclear translocation via the inhibition of AKT. Specific inhibition of SIRT-1 abolished the protective effect of nobiletin, causing decreased FOXO3a expression, followed by autophagy induction and decreased PGC-1α expression and mitochondrial dynamics. Taken together, our data indicate that SIRT-1 directly mediates the protective effect of nobiletin against hepatic ischemia and reperfusion injury. The activation of autophagy and mitochondrial function through the SIRT-1/FOXO3a and PGC-1α pathways indicate that nobiletin could have therapeutic potential for treating hepatic ischemia and reperfusion injury. Nobiletin, an antioxidant found in citrus peel, may protect the liver from reperfusion injury, damage following blood flow interruption. When blood flow is restricted and then restored, as in transplant, surgery, or shock, cells are injured, largely due to damage to the cellular powerhouses, the mitochondria. Nobiletin is known to have many benefits, including anti-cancer and anti-inflammatory activities, but its mechanism of action is not well understood. Sang Won Park and Hwajin Kim, at the Gyeongsang National University School of Medicine, in Jinju, South Korea, and co-workers, investigated how nobiletin might protect the liver against interruption of blood flow. They found that nobiletin triggered cells to dismantle damaged mitochondria and produce new, functioning mitochondria, greatly reducing liver damage. These results illuminate how nobiletin works and may lead to better treatments for liver reperfusion injury.
Collapse
Affiliation(s)
- Theodomir Dusabimana
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, 52727, Republic of Korea.,Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju, 52727, Republic of Korea
| | - So Ra Kim
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, 52727, Republic of Korea
| | - Hye Jung Kim
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, 52727, Republic of Korea.,Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju, 52727, Republic of Korea
| | - Sang Won Park
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, 52727, Republic of Korea. .,Department of Convergence Medical Sciences, Institute of Health Sciences, Gyeongsang National University Graduate School, Jinju, 52727, Republic of Korea.
| | - Hwajin Kim
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju, 52727, Republic of Korea.
| |
Collapse
|
10
|
Kato H, Duarte S, Miller MG, Busuttil RW, Coito AJ. Overproduction of Tenascin-C Driven by Lipid Accumulation in the Liver Aggravates Hepatic Ischemia/Reperfusion Injury in Steatotic Mice. Liver Transpl 2019; 25:288-301. [PMID: 30358115 PMCID: PMC6355355 DOI: 10.1002/lt.25365] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 10/17/2018] [Indexed: 01/08/2023]
Abstract
The purpose of this study was to assess the significance of tenascin-C (Tnc) expression in steatotic liver ischemia/reperfusion injury (IRI). The critical shortage in donor organs has led to the use of steatotic livers in transplantation regardless of their elevated susceptibility to hepatic IRI. Tnc is an endogenous danger signal extracellular matrix molecule involved in various aspects of immunity and tissue injury. In the current study, mice were fed with a steatosis-inducing diet and developed approximately 50% hepatic steatosis, predominantly macrovesicular, before being subjected to hepatic IRI. We report here that lipid accumulation in hepatocytes inflated the production of Tnc in steatotic livers and in isolated hepatic stellate cells. Moreover, we show that the inability of Tnc-/- deficient steatotic mice to express Tnc significantly protected these mice from liver IRI. Compared with fatty controls, Tnc-/- steatotic mice showed significantly reduced serum transaminase levels and enhanced liver histological preservation at both 6 and 24 hours after hepatic IRI. The lack of Tnc expression resulted in impaired lymphocyte antigen 6 complex, locus (Ly6G) neutrophil and macrophage antigen-1 (Mac-1) leukocyte recruitment as well as in decreased expression of proinflammatory mediators (interleukin 1β, tumor necrosis factor α, and chemokine [C-X-C motif] ligand 2) after liver reperfusion. Myeloperoxidase (MPO) is the most abundant cytotoxic enzyme secreted by neutrophils and a key mediator of neutrophil-induced oxidative tissue injuries. Using an in vitro model of steatosis, we also show that Tnc markedly potentiated the effect of steatotic hepatocytes on neutrophil-derived MPO activity. In conclusion, our data support the view that inhibition of Tnc is a promising therapeutic approach to lessen inflammation in steatotic livers and to maximize their successful use in organ transplantation.
Collapse
Affiliation(s)
- Hiroyuki Kato
- The Dumont‐UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles CA
| | - Sergio Duarte
- The Dumont‐UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles CA
| | - Mary G. Miller
- The Dumont‐UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles CA
| | - Ronald W. Busuttil
- The Dumont‐UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles CA
| | - Ana J. Coito
- The Dumont‐UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles CA
| |
Collapse
|
11
|
Nakamura K, Kageyama S, Kupiec-Weglinski JW. The Evolving Role of Neutrophils in Liver Transplant Ischemia-Reperfusion Injury. CURRENT TRANSPLANTATION REPORTS 2019; 6:78-89. [PMID: 31602356 PMCID: PMC6786799 DOI: 10.1007/s40472-019-0230-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Purpose of Review Hepatic ischemia-reperfusion injury (IRI), an inevitable event during liver transplantation, represents a major risk factor for the primary graft dysfunction as well as the development of acute and chronic rejection. Neutrophils, along macrophages, are pivotal in the innate immune-driven liver IRI, whereas the effective neutrophil-targeting therapies remain to be established. In this review, we summarize progress in our appreciation of the neutrophil biology and discuss neutrophil-based therapeutic perspectives. Recent Findings New technological advances enable to accurately track neutrophil movements and help to understand molecular mechanisms in neutrophil function, such as selective recruitment to IR-stressed tissue, formation of neutrophil extracellular traps, or reverse migration into circulation. In addition to pro-inflammatory and tissue-destructive functions, immune regulatory and tissue-repairing phenotype associated with distinct neutrophil subsets have been identified. Summary Newly recognized and therapeutically attractive neutrophil characteristics warrant comprehensive preclinical and clinical attention to target IRI in transplant recipients.
Collapse
Affiliation(s)
- Kojiro Nakamura
- The Dumont-UCLA Transplant Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| | - Shoichi Kageyama
- The Dumont-UCLA Transplant Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| | - Jerzy W Kupiec-Weglinski
- The Dumont-UCLA Transplant Center, Department of Surgery, Division of Liver and Pancreas Transplantation, David Geffen School of Medicine at University of California, Los Angeles, CA 90095
| |
Collapse
|
12
|
Hardy E, Hardy-Sosa A, Fernandez-Patron C. MMP-2: is too low as bad as too high in the cardiovascular system? Am J Physiol Heart Circ Physiol 2018; 315:H1332-H1340. [DOI: 10.1152/ajpheart.00198.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Matrix metalloproteinase (MMP)-2 cleaves a broad spectrum of substrates, including extracellular matrix components (responsible for normal tissue remodeling) and cytokines (modulators of the inflammatory response to physiological insults such as tissue damage). MMP-2 expression is elevated in many cardiovascular pathologies (e.g., myocardial infarction, hypertensive heart disease) where tissue remodeling and inflammatory responses are perturbed. Thus, it has generally been assumed that blockade of MMP-2 activity will yield therapeutic effects. Here, we provide a counterargument to this dogma based on 1) preclinical studies on Mmp2-null ( Mmp2−/−) mice and 2) clinical studies on patients with inactivating MMP2 gene mutations. Furthermore, we put forward the hypothesis that, when MMP-2 activity falls below baseline, the bioavailability of proinflammatory cytokines normally cleaved and inactivated by MMP-2 increases, leading to the production of cytokines and cardiac secretion of phospholipase A2activity into the circulation, which stimulate systemic inflammation that perturbs lipid metabolism in target organs. Finally, we suggest that insufficient understanding of the consequences of MMP-2 deficiency remains a major factor in the failure of MMP-2 inhibitor-based therapeutic approaches. This paucity of knowledge precludes our ability to effectively intervene in cardiovascular and noncardiovascular pathologies at the level of MMP-2.
Collapse
Affiliation(s)
- Eugenio Hardy
- Biotechnology Laboratory, Study Center for Research and Biological Evaluations, Institute of Pharmacy and Foods, University of Havana, Havana, Cuba
| | | | - Carlos Fernandez-Patron
- Department of Biochemistry, Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
13
|
Zeng G, Ding W, Li Y, Sun M, Deng L. Morroniside protects against cerebral ischemia/reperfusion injury by inhibiting neuron apoptosis and MMP2/9 expression. Exp Ther Med 2018; 16:2229-2234. [PMID: 30186462 PMCID: PMC6122141 DOI: 10.3892/etm.2018.6457] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/09/2018] [Indexed: 12/22/2022] Open
Abstract
The aim of the present study was to investigate the effect of morroniside against matrix metalloproteinase (MMP)2/9 and focal cerebral ischemia/reperfusion (I/R) injury in rats. A rat model of focal cerebral I/R injury rats was established and rats were administered with 30, 90 or 270 mg/kg/day morroniside for 7 days. The expression of MMP2/9 and neuronal apoptosis were assessed. In addition, the expression of active caspase-3, B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X protein (Bax) were measured. The results revealed that MMP2 and MMP9 expression was upregulated and the percentage of apoptotic neurons was increased in rats with focal cerebral I/R injury compared with the control. However, treatment with morroniside significantly inhibited I/R-induced MMP2/9 expression and neuron apoptosis compared with the untreated I/R injury group. Morroniside administration also decreased the expression of active caspase-3 and increased the Bcl-2/Bax ratio compared with untreated rats with focal cerebral I/R injury. The inhibitory effect of morroniside on MMP2/9 expression and neuron apoptosis was dose dependent. In summary, the results of the present study suggest that morroniside is able to protect against cerebral I/R injury in the brain and may have potential as a therapeutic treatment for patients who have suffered a stroke.
Collapse
Affiliation(s)
- Guoyong Zeng
- Department of Neurology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi 341000, P.R. China
| | - Weijiang Ding
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yin Li
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Meiying Sun
- Department of Neurology, The People's Hospital of Fuzhou, Fuzhou, Jiangxi 344000, P.R. China
| | - Liying Deng
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
14
|
Duarte S, Matian P, Ma S, Busuttil RW, Coito AJ. Adeno-Associated Virus-Mediated Gene Transfer of Tissue Inhibitor of Metalloproteinases-1 Impairs Neutrophil Extracellular Trap Formation and Ameliorates Hepatic Ischemia and Reperfusion Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1820-1832. [PMID: 29870740 DOI: 10.1016/j.ajpath.2018.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/19/2018] [Accepted: 05/03/2018] [Indexed: 01/01/2023]
Abstract
Matrix metalloproteinase-9 (MMP-9) is abundantly expressed by infiltrating leukocytes and contributes to the pathogenesis of hepatic ischemia and reperfusion injury (IRI). On the other hand, its physiological inhibitor, the tissue inhibitor of metalloproteinases-1 (TIMP-1), is available in insufficient levels to hamper MMP-9 activity during hepatic IRI. In this study, we generated recombinant adeno-associated virus type 8 vectors (rAAV8) encoding mouse TIMP-1 driven by a liver-specific thyroxine-binding globulin promoter as a strategy to increase the levels of TIMP-1 during liver IRI. Biodistribution analysis confirmed selective overexpression of TIMP-1 in livers of rAAV8-TIMP-1 vector treated C57BL/6 mice. rAAV8-TIMP-1-treated mice showed reduced MMP-9 activity, diminished leukocyte trafficking and activation, lowered transaminase levels, and improved histology after liver IRI. Moreover, the rAAV8-TIMP-1 vector therapy enhanced significantly the 7-day survival rate of TIMP-1-/- mice subjected to hepatic IRI. Neutrophils are the first cells recruited to inflamed tissues and, once activated, they release nuclear DNA-forming web-like structures, known as neutrophil extracellular traps. It was found that TIMP-1 has the ability to reduce formation of neutrophil extracellular traps and, consequently, limit the impact of neutrophil extracellular trap-mediated cytotoxicity in hepatic IRI. This is the first report demonstrating that TIMP-1 overexpression is hepatoprotective in ischemia and reperfusion injury. Hence, TIMP-1 may represent a promising molecule for drug development to treat liver IRI.
Collapse
Affiliation(s)
- Sergio Duarte
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Patrick Matian
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Stacy Ma
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Ronald W Busuttil
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Ana J Coito
- The Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
15
|
Ito T, Kuriyama N, Kato H, Matsuda A, Mizuno S, Usui M, Sakurai H, Isaji S. Sinusoidal protection by sphingosine-1-phosphate receptor 1 agonist in liver ischemia-reperfusion injury. J Surg Res 2018; 222:139-152. [PMID: 29273365 DOI: 10.1016/j.jss.2017.09.048] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 09/20/2017] [Accepted: 09/29/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Functional and structural damages in sinusoidal endothelial cells (SECs) have a crucial role during hepatic ischemia-reperfusion injury (IRI). In regulating endothelial function, sphingosine-1-phosphate receptor 1 (S1PR1), which is a G protein-coupled receptor, has an important role. The present study aimed to clarify whether SEW2871, a selective S1PR1 agonist, can attenuate hepatic damage caused by hepatic IRI, focusing on SEC functions. METHODS In vivo, using a 60-min partial-warm IRI model, mice were treated with SEW2871 or without it (with vehicle). In vitro, isolated SECs pretreated with SEW2871 or without it (with vehicle) were incubated with hydrogen peroxide. RESULTS Compared with the IRI + vehicle group, SEW2871 administration significantly improved serum transaminase levels and liver damage, attenuated infiltration of Ly-6G and mouse macrophage antigen-1-positive cells, suppressed the expression of vascular cell adhesion molecule-1 and proinflammatory cytokines in the liver, and enhanced the expressions of endothelial nitric oxide synthase (eNOS) and vascular endothelial (VE) cadherin in the liver (eNOS/β-actin [median]: 0.24 versus 0.53, P = 0.008; VE-cadherin/β-actin [median]: 0.21 versus 0.94, P = 0.008). In vitro, compared with the vehicle group, pretreatment of SECs with SEW2871 significantly increased the expressions of eNOS and VE-cadherin (eNOS/β-actin [median]: 0.22 versus 0.29, P = 0.008; VE-cadherin/β-actin [median]: 0.38 versus 0.67, P = 0.008). As results of investigation of prosurvival signals, SEW2871 significantly increased Akt phosphorylation in SECs and decreased lactate dehydrogenase levels in supernatants of SECs. CONCLUSIONS These results indicate that S1PR1 agonist induces attenuation of hepatic IRI, which might be provided by preventing SEC damage. S1PR1 may be a therapeutic target for the prevention of early sinusoidal injury after hepatic IRI.
Collapse
Affiliation(s)
- Takahiro Ito
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Naohisa Kuriyama
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Mie, Japan.
| | - Hiroyuki Kato
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | | | - Shugo Mizuno
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Masanobu Usui
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Hiroyuki Sakurai
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Shuji Isaji
- Department of Hepatobiliary Pancreatic and Transplant Surgery, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| |
Collapse
|
16
|
Neutrophils: a cornerstone of liver ischemia and reperfusion injury. J Transl Med 2018; 98:51-62. [PMID: 28920945 DOI: 10.1038/labinvest.2017.90] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 07/05/2017] [Accepted: 07/09/2017] [Indexed: 12/12/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) is the main cause of morbidity and mortality due to graft rejection after liver transplantation. During IRI, an intense inflammatory process occurs in the liver. This hepatic inflammation is initiated by the ischemic period but occurs mainly during the reperfusion phase, and is characterized by a large neutrophil recruitment to the liver. Production of cytokines, chemokines, and danger signals results in activation of resident hepatocytes, leukocytes, and Kupffer cells. The role of neutrophils as the main amplifiers of liver injury in IRI has been recognized in many publications. Several studies have shown that elimination of excessive neutrophils or inhibition of their function leads to reduction of liver injury and inflammation. However, the mechanisms involved in neutrophil recruitment during liver IRI are not well known. In addition, the molecules necessary for this type of migration are poorly defined, as the liver presents an atypical sinusoidal vasculature in which the classical leukocyte migration paradigm only partially applies. This review summarizes recent advances in neutrophil-mediated liver damage, and its application to liver IRI. Basic mechanisms of activation of neutrophils and their unique mechanisms of recruitment into the liver vasculature are discussed. In particular, the role of danger signals, adhesion molecules, chemokines, glycosaminoglycans (GAGs), and metalloproteinases is explored. The precise definition of the molecular events that govern the recruitment of neutrophils and their movement into inflamed tissue may offer new therapeutic alternatives for hepatic injury by IRI and other inflammatory diseases of the liver.
Collapse
|
17
|
Roderfeld M. Matrix metalloproteinase functions in hepatic injury and fibrosis. Matrix Biol 2017; 68-69:452-462. [PMID: 29221811 DOI: 10.1016/j.matbio.2017.11.011] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/29/2017] [Accepted: 11/29/2017] [Indexed: 01/18/2023]
Abstract
Liver fibrosis is the most common final outcome for chronic liver diseases. The complex pathogenesis includes hepatic parenchymal damage as a result of a persistent noxe, activation and recruitment of immune cells, activation of hepatic stellate cells, and the synthesis of fibrotic extracellular matrix (ECM) components leading to scar formation. Clinical studies and animal models demonstrated that fibrosis can be reversible. In this regard matrix metalloproteinases (MMPs) have been focused as therapeutic targets due to their ability to modulate tissue turnover during fibrogenesis as well as regeneration and, of special interest, due to their influence on cellular behavior like proliferation, gene expression, and apoptosis that, in turn, impact fibrosis and regeneration. The current review aims to summarize and update the knowledge about expression pattern and the central roles of MMPs in hepatic fibrosis.
Collapse
Affiliation(s)
- Martin Roderfeld
- Department of Gastroenterology, Justus-Liebig-University Giessen, Gaffkystr. 11c, D-35392 Giessen, Germany.
| |
Collapse
|
18
|
Naim A, Pan Q, Baig MS. Matrix Metalloproteinases (MMPs) in Liver Diseases. J Clin Exp Hepatol 2017; 7:367-372. [PMID: 29234202 PMCID: PMC5715451 DOI: 10.1016/j.jceh.2017.09.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/24/2017] [Indexed: 02/07/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are proteinases capable of degrading components of the extracellular matrix and numerous nonmatrix proteins. MMPs along with tissue inhibitors of MMPs, have been implicated in the pathogenesis of liver diseases. Although, the precise mechanism-of-actions of MMPs in various liver related disorders is largely unknown, however, data from diverse experimental models indicate that these proteinases influence cellular activities including proliferation and survival, gene expression, as well as multiple aspects of inflammation. Hence, MMP's are likely key players in the outcomes related to liver disease.
Collapse
Key Words
- Col, collagen
- ECM, extra cellular matrix
- GBD, global burden of disease
- HCC, hepato-cellular carcinoma
- IRI, ischemia and reperfusion injury
- MMP, matrix metalloproteases
- NAFLD, non-alcoholic fatty liver disease
- NFkB, nuclear factor kappa-B
- TIMPs, tissue inhibitors of MMPs
- TNF, tumor necrosis factor
- cirrhosis
- extracellular matrix (ECM)
- hepatocellular carcinoma
- liver fibrosis
- matrix metalloproteinases
Collapse
Affiliation(s)
- Adnan Naim
- Centre for Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Mirza S. Baig
- Centre for Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India,Address for correspondence: Mirza S. Baig, Centre for Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore 453552, MP, India.Mirza S. Baig, Centre for Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI)IndoreMP453552India
| |
Collapse
|
19
|
Matrix metalloproteinases as regulators of inflammatory processes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2036-2042. [DOI: 10.1016/j.bbamcr.2017.05.010] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/06/2017] [Accepted: 05/09/2017] [Indexed: 12/26/2022]
|
20
|
Zhao Z, Tang Z, Zhang W, Liu J, Li B. Magnesium isoglycyrrhizinate protects against renal‑ischemia‑reperfusion injury in a rat model via anti‑inflammation, anti‑oxidation and anti‑apoptosis. Mol Med Rep 2017; 16:3627-3633. [PMID: 28714024 DOI: 10.3892/mmr.2017.6993] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 03/14/2017] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate whether magnesium isoglycyrrhizinate protects against renal ischemia‑reperfusion injury (RIRI), and to verify the underlying mechanisms. An RIRI rat model was induced by removing the right kidney, and exposing and clamping the left kidney. RIRI model rats were administered 30 mg/kg magnesium isoglycyrrhizinate for 3 days. Blood urea nitrogen (BUN) and serum creatinine levels in the blood of RIRI model rat were examined, compared with sham‑operated controls. Magnesium isoglycyrrhizinate suppressed the activities of tumor necrosis factor‑α, interleukin (IL)‑1β, IL‑6, superoxide dismutase, glutathione peroxidase, inducible nitric oxide synthase (iNOS) and caspase‑3 in RIRI model rats. Renal iNOS, matrix metalloproteinase (MMP)‑2, phosphorylated‑signal transducers and activators of transcription 3 (STAT3) and intercellular adhesion molecule‑1 (ICAM‑1) protein expression levels were suppressed by magnesium isoglycyrrhizinate treatment in RIRI model rats. These findings suggested that magnesium isoglycyrrhizinate protects RIRI via anti‑inflammatory, ‑oxidative and ‑apoptotic mechanisms in an RIRI rat model. These results implicate magnesium isoglycyrrhizinate pretreatment as a potential approach to protect against RIRI via suppression of the iNOS, ICAM‑1, MMP‑2 and STAT3 signaling pathways.
Collapse
Affiliation(s)
- Zhigang Zhao
- Department of Emergency, Wuhan General Hospital of Guangzhou Military Command, Wuhan, Hubei 430070, P.R. China
| | - Zhongzhi Tang
- Department of Emergency, Wuhan General Hospital of Guangzhou Military Command, Wuhan, Hubei 430070, P.R. China
| | - Wenkai Zhang
- Department of Emergency, Wuhan General Hospital of Guangzhou Military Command, Wuhan, Hubei 430070, P.R. China
| | - Jie Liu
- Department of Emergency, Wuhan General Hospital of Guangzhou Military Command, Wuhan, Hubei 430070, P.R. China
| | - Bo Li
- Department of Emergency, Wuhan General Hospital of Guangzhou Military Command, Wuhan, Hubei 430070, P.R. China
| |
Collapse
|
21
|
Suyavaran A, Thirunavukkarasu C. Preconditioning methods in the management of hepatic ischemia reperfusion- induced injury: Update on molecular and future perspectives. Hepatol Res 2017; 47:31-48. [PMID: 26990696 DOI: 10.1111/hepr.12706] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/12/2016] [Accepted: 03/11/2016] [Indexed: 12/14/2022]
Abstract
Hepatic IR (ischemia reperfusion) injury is a commonly encountered obstacle in the post-operative management of hepatic surgery. Hepatic IR occurs during 'Pringle maneuver' for reduction of blood loss or during a brief period of cold storage followed by reperfusion of liver grafts. The stress induced during hepatic IR, triggers a spectrum of cellular responses leading to the varying degrees of hepatic complications which in turn affect the post operative care. Different preconditioning methods either activate or subdue different sets of molecular signals, resulting in varied levels of protection against hepatic IR injury. Yet, there is a serious lacuna in the knowledge regarding the choice of preconditioning methods and the resulting molecular changes in order to assess the efficiency and choice of these methods correctly. This review provides an update on the various preconditioning approaches such as surgical/ischemic, antioxidant, pharmaceutical and genetic preconditioning strategies published during last six years (2009-2015). Further, we discuss the attenuation or inhibition of specific inflammatory, apoptotic and necrotic markers in the various experimental models of liver IR subjected to different preconditioning strategies. While enlisting the controversies in the ischemic preconditioning strategy, we bring out the uncertainties in the existing molecular targets and their reliability in the attenuation of hepatic IR injury. Future research studies would include the novel preconditioning strategies employ i) the targeted gene silencing of key molecular targets inducing IR, ii) hyper expression of beneficial molecular signals against IR via gene transfer techniques. The above studies would see the combination of these latest techniques with the established preconditioning strategies for better post-operative hepatic management.
Collapse
Affiliation(s)
- Arumugam Suyavaran
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | | |
Collapse
|