Next Article in Journal
Purification, Biochemical and Kinetic Characterization of a Novel Alkaline sn-1,3-Regioselective Triacylglycerol Lipase from Penicilliumcrustosum Thom Strain P22 Isolated from Moroccan Olive Mill Wastewater
Next Article in Special Issue
Aberrant Transferrin and Ferritin Upregulation Elicits Iron Accumulation and Oxidative Inflammaging Causing Ferroptosis and Undermines Estradiol Biosynthesis in Aging Rat Ovaries by Upregulating NF-Κb-Activated Inducible Nitric Oxide Synthase: First Demonstration of an Intricate Mechanism
Previous Article in Journal
Characteristics of the Cytotoxicity of Taraxacum mongolicum and Taraxacum formosanum in Human Breast Cancer Cells
Previous Article in Special Issue
Assessment of DDAH1 and DDAH2 Contributions to Psychiatric Disorders via In Silico Methods
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Effects of iNOS in Hepatic Warm Ischaemia and Reperfusion Models in Mice and Rats: A Systematic Review and Meta-Analysis

1
Institute for Laboratory Animal Science and Experimental Surgery, RWTH Aachen University, 52074 Aachen, Germany
2
Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka 573-1010, Japan
3
Department of Intensive Care Medicine, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(19), 11916; https://doi.org/10.3390/ijms231911916
Submission received: 9 September 2022 / Revised: 26 September 2022 / Accepted: 26 September 2022 / Published: 7 October 2022
(This article belongs to the Special Issue Nitric Oxide Synthases: Function and Regulation)

Abstract

:
Warm ischaemia is usually induced by the Pringle manoeuver (PM) during hepatectomy. Currently, there is no widely accepted standard protocol to minimise ischaemia-related injury, so reducing ischaemia-reperfusion damage is an active area of research. This systematic review and meta-analysis focused on inducible nitric oxide synthase (iNOS) as an early inflammatory response to hepatic ischaemia reperfusion injury (HIRI) in mouse- and rat-liver models. A systematic search of studies was performed within three databases. Studies meeting the inclusion criteria were subjected to qualitative and quantitative synthesis of results. We performed a meta-analysis of studies grouped by different HIRI models and ischaemia times. Additionally, we investigated a possible correlation of endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) regulation with iNOS expression. Of 124 included studies, 49 were eligible for the meta-analysis, revealing that iNOS was upregulated in almost all HIRIs. We were able to show an increase of iNOS regardless of ischemia or reperfusion time. Additionally, we found no direct associations of eNOS or NO with iNOS. A sex gap of primarily male experimental animals used was observed, leading to a higher risk of outcomes not being translatable to humans of all sexes.

1. Introduction

Hepatectomy is a well-established therapeutic option for benign and malignant liver disease. [1,2,3]. A total of 21,443 patients underwent hepatectomy between 2012 and 2016 in the United States (American College of Surgeons National Surgical Quality Improvement Program’s Participant Use Files; male 10,476, female 10,967), 28,708 (male 15,489 and female 13,219) between 2007 and 2010 in France (the Medicalization des Systemes D’Information databases), 110,332 (male 55,781 and female 54,551) between 2010 and 2015 in Germany (Diagnosis-Related Groups; ICD-10 and German operations and procedure key codes) and 20,575 in 2011 in Japan (the National Clinical Database of Japan) [4,5,6,7].
Hepatectomy is a complicated abdominal surgical technique associated with a prolonged surgical time and an increased risk of perioperative complications, such as bleeding. To minimize bleeding, the Pringle manoeuver (PM), a clamping technique for the portal triad, is used intermittently or continuously and enables nonselective inflow occlusion by controlling blood loss during hepatectomy [8,9,10]. However, these techniques also simultaneously induce warm ischaemia. A recent survey in Europe showed that 71% of surgeons apply PM on indication, whereas 19% use PM routinely. Only 10% of surgeons never use PM, although it is the most frequently used technique in hepatectomy [11]. Despite the huge importance of this technique, only three randomised human trials have been conducted to compare hepatectomy with and without intermittent PM [12,13,14]. These trials reported conflicting results, which has led to even more discussion on the value of PM.
Hepatic ischaemia-reperfusion injury (HIRI) can occur during interventions, causing direct hepatic ischaemia in liver surgery or transplantation. HIRI is a serious clinical condition leading to cellular injury and organ dysfunction, mainly through production of reactive oxygen species and inflammatory cytokines [15]. The implications of warm HIRI are apoptosis of hepatocytes and sinusoidal endothelial cells, necrosis or a combination of both [16]. During hepatic ischaemia-reperfusion (HIR), there are interactions among liver cells, Kupffer cells, neutrophils, hepatic sinusoidal endothelial cells, and fat-storing cells. Platelets and alexin are also involved [17]. These activated cells release a large quantity of proinflammatory cytokines and lipid inflammatory factors, which can lead to further inflammatory reactions and cell apoptosis. Preventing HIRI is, therefore, important for regeneration after liver operation.
The function of nitric oxide (NO) in HIRI is inconclusive and complex. Whether or not NO has cytoprotective or cytotoxic effects is controversial because it has various roles throughout the body. It is recognised that NO maintains a continuous balance between vasodilatation and vasoconstriction due to continuous production [18]. In this context, NO and its derivatives are known to have important roles in the pathophysiology of the liver [19]. NO has been proven to reduce HIRI through various mechanisms [20,21]; and it is an unstable nitrogen-centred radical produced in a redox reaction between L-arginine and oxygen molecules by NO synthase (NOS) catalysation. There are three types of NOS: endothelial nitric oxide synthase (eNOS), neuronal nitric oxide synthase (nNOS) and inducible nitric oxide synthase (iNOS). In many cell types, including liver endothelial cells and hepatocytes, eNOS is constitutively expressed [21,22]. In contrast, iNOS is expressed under pathological circumstances, such as endotoxemia, haemorrhagic shock, hepatitis and liver regeneration, but is upregulated in various nucleated cell types, including hepatocytes, neutrophils, T-lymphocytes, endothelial, biliary and Kupffer cells, under inflammatory conditions associated with HIRI [23,24,25,26,27]. In general, it is assumed that NO derived from eNOS in liver sinusoidal endothelial cells is cell-protective, whereas iNOS-derived NO contributes to pathological processes by acting as a proinflammatory mediator [4]. NO is produced mainly by eNOS catalysis and upregulation of iNOS expressions during acute warm HIR [28]. An excess level of NO produced by iNOS has been implicated as a factor in liver injury [29]. iNOS is induced to produce large amounts of NO by lipopolysaccharides and proinflammatory cytokines, such as interleukin-1 (IL-1) and tumour necrosis factor (TNF), which have a role in many inflammatory and immune reactions [30]. During early warm HIRI, NO produced by eNOS activation is protective, but NO induced by iNOS may have either a protective or deleterious effect [31]. Therefore, strategies that target deleterious NO effects comprise an active area of discussion as potential new therapies, which have been experimentally investigated in many disease models. Animal-based research may give insights into the mechanisms of iNOS in the HIRI model, but the knowledge gained from most single studies is limited. It remains unclear how NO induced by iNOS is regulated during warm HIRI. Furthermore, the relationships between iNOS and eNOS in HIRI could not be clarified in detail. Therefore, a systematic review addressing this specific problem may provide more detailed insight into this topic and provide information about various experimental results.

2. Results

2.1. Results of the Search

The literature search resulted in 793 records. After removing 365 duplicates, 428 records remained, and their titles and abstracts were screened. We excluded 279 records that did not meet the pre-specified inclusion criteria and screened the full texts of the remaining 149 references. Twenty-five records were excluded after full-text assessment, and the reasons for exclusion are shown in Figure 1. In total, we included 124 records in our synthesis, from which 49 records were also used in the meta-analysis. The search process is described fully in the flow diagram in Figure 1.

2.2. General Characteristics

The characteristics of the 124 included studies are presented in Table 1, Table 2, Table 3, Table 4 and Table 5. The studies were classified according to the models of the study.

2.3. Details for Rat Models

Eighty-nine studies containing rat models were identified. Fifty-six (63%) studies with 70% HIRI were reported (Table 1). The ischaemic time taken varied from 5 to 120 min, and most of the studies used 45 (6 studies), 60 (25 studies) or 90 min (9 studies). In these studies, the reperfusion time varied between 0.5 and 168 h. For other studies with different or unknown ischaemic times, the number of results by subgroup was too small (n < 3) for a meta-analysis.
Table 1. Overview of rat models with 70% HIRI.
Table 1. Overview of rat models with 70% HIRI.
ReferenceYearStrainReperfusion Time (h)Gender (%)BW (g)iNOS
Detection Method
NO
Detection
eNOS
Detection
Survival Rate
30 min ischemia time
Abd-Elbaset [32]2017Wistar0.5♂ 100%300–350IHC-+-
Fouad [33]2011SD72♂ 100%190–210IHC+--
Liu [34]2000Fischer4♂ 100%275–300RNA+--
Liu [35]1998Fischer4♂ 100%245–290SP-+-
Rhee [36]2002SD1/6♂ 100%100–150indirect+-+
Wang [37]2017SD1♂ 100%200–250Protein---
Wang [38]2003Wistar6/1/3/5/7/336♂ 100%240–260RNA/WB-+-
45 min ischemia time
El-Emam [39]2020SD24♂ 100%250–280ELISA---
Hur [40]1999SD0.5/1/3/5/12/24♂ 100%350–400RNA---
Koti [21]2005SD2♂ 100%250–300WB++-
Mostafa-Hedeab [41]2019WistarN/A♂ 100%140–250RNA-+-
Serracino-Iglott [42]2003Wistar1♂ 100%250–300WB/IHC-+-
Yang [43]2011SD2♂ 100%250–300IHC++-
60 min ischemia time
Curek [44]2010Wistar1♂ 100%350–450IHC---
Eum [45]2004SD5♂ 100%270–300RNA-+-
Eum [46]2004SD5/24♂ 100%260–300RNA---
Fernández [47]2009SD0.33♂ 100%180–200RNA---
Ferrigno [48]2020Wistar1♂ 100%N/AWB---
Ferrigno [49]2020Wistar1/2♂ 100%N/AWB-+-
Hataji [50]2010SD24♂ 100%N/AIHC++-
Hsu [51]2002SD2♂/♀ 50%200–275IHC/activity---
Kang [52]2011SD1/5♂ 100%270–300RNA/WB---
Kim [53]2012SD5♂ 100%150–170WB/RNA---
Kim [54]2010SD5♂ 100%270–300RNA/WB---
Kim [55]2004SD5♂ 100%260–320RNA-+-
Kurabayashi [56]2005SD5♂ 100%230–290IHC---
Lee [57]2008SD5♂ 100%270–300RNA-+-
Man [58]2005SD0.33/1/1.5/6/24♂ 100%220–280RNA---
Park [59]2007SD5♂ 100%270–300RNA-+-
Ramalho [60]2014Wistar6♂ 100%150–200WB++-
Ren [61]2019Wistar1/3/6/12♂ 100%220–280RNA---
Sonin [62]1999SD6♂ 100%250–330RNA---
Tao [63]2014Wistar6♂ 100%180–220ELISA--+
Trocha [64]2014Wistar4♂ 100%N/AELISA---
Unal [65]2017Wistar1♂ 100%350–450IHC+--
Yang [66]2007SD1/3/5♂ 100%230–250mRNA++-
Yun [67]2012SD5♂ 100%270–300RNA/WB---
Yun [68]2010SD1/2/4/6/8/12/24♂ 100%270–300RNA/WB---
90 min ischemia time
Bektas [69]2016Wistar2♂ 100%250–300IHC-+-
Grezzana-Filho [70]2020Wistar24♂ 100%250–310WB-+-
Kim [71]2004SD6♂ 100%260–300RNA-+-
Kuncewitch [72]2013SD24♂ 100%250–275WB--+
Lin [73]2004Wistar1.5♂ 100%300–350IHC++-
Longo [74]2016Wistar2♂ 100%200–250WB-+-
Takamatsu [75]2006SD3/6/12/24♂ 100%230–300RNA+--
Yao [76]2009SD1/3/6/24/168♂ 100%220–240Protein+-+
Yun [77]2012SD3/24♂ 100%270–300WB---
Studies not included in meta-analysis
20 min ischemia time
Wang [78]1998Fischar0.5♂ 100%240–320N/A+--
35 min ischemia time
Kireev [79]2012Wistar36♂ 100%N/ARNA-+-
Kireev [80]2013fa/fa Zucker36♂ 100%496RNA-+-
40 min ischemia time
Duan [81]2017SD2♂ 100%190–210WB++-
Trocha [82]2010Wistar1♂ 100%240–303ELISA-+-
100 min ischemia time
Hara [83]2005N/A12N/AN/AWB---
120 min ischemia time
Ishizaki [84]2008SD1/3/6/9/12/24♂ 100%240–270RNA/WB+--
Unknown ischemia time
Hsieh [85]2015SD3.5/24♂ 100%250–300RNA---
ELISA, Enzyme-linked immunosorbent assay; IHC, immune histochemistry; N/A, not available; RNA, ribonuclein acid; SP, spectrophotometric analysis; SD, Spraque– Dawley; WB, Western blot; + reported; - not reported.
Figure 2 shows the distribution of warm ischaemia times compared with their reperfusion times for each study of 70% HIRI in rat models.
As shown in Table 2, 22 (24.7%) of the included studies reported a HIRI of 100%. The ischaemic time taken varied from 5 to 120 min, and most of the studies involved 30 (6 studies), 45 (7 studies) or 60 (4 studies) min. In these studies, the reperfusion time varied between 0 and 144 h. For other studies with different ischaemic times, the number of results by subgroup was too small (n < 3) for a meta-analysis.
Table 2. Overview of rat models with 100% HIRI.
Table 2. Overview of rat models with 100% HIRI.
ReferenceYearStrainReperfusion Time (h)GenderBW (g)iNOS
Detection Method
NO
Detection
eNOS
Detection
Survival Rate
30 min ischemia time
Acquaviva [86]2009Wistar3♂ 100%200–220WB-+-
Chen [87]2014SD2♂ 100%250–300-+--
Lanteri [88]2007Wistar0.5/3♂ 100%200–220WB-+-
Morisuee [89]2003Wistar3/4/6♂ 100%350–450IHC+-+
Nii [90]2014Wistar2♂ 100%250–300RNA---
Uchiami [91]2002Wistar0.5/1/2♂ 100%280–320RNA---
45 min ischemia time
Atef [92]2017Wistar1♂ 100%200–250RNA++-
El-Shintany [93]2015albino2♂ 100%180–200IHC+--
Ibrahim [94]2020albino24♂ 100%200–300ELISA-+-
Ibrahim [95]2014albino1♂ 100%200–230IHC+--
Sankary [96]1999SD0.25/0.5/1/2/3/144♂ 100%200–250---+
Sehitoglu [97]2019Wistar24♂ 100%250–300IHC---
Yaylak [98]2008Wistar0.75♂ 100%150–220IHC---
60 min ischemia time
Miyake [28]2013Wistar5/3♂ 100%250–300WB/RNA+-+
Rodríguez-Reynoso [99]2001SD1/2♂ 100%250–300RNA+-+
Sankary [96]1999SD0.25/0.5/1/2/3/144♂ 100%200–250---+
Yang [100]2007SD5♂ 100%230–250mRNA++-
Studies not included in meta-analysis
5/15 min ischemia time
Miyake [28]2013Wistar5,3♂ 100%250–300WB/RNA++-
20 min ischemia time
Harada [101]2003SD2/6/24♂ 100%225–250RNA---
Suetsugu [102]2005SD72♂ 100%240–255WB---
40 min ischemia time
Abdel-Gaber [103]2015Albino1♂ 100%200–230IHC++-
90 min ischemia time
Koeppel [26]2007SD3/6/12/24♂ 100%280–350RNA-+-
Rodriguez-Reynos [104]2018SD1/5/168♂ 100%250–300RNA+-+
Xue [105]2010SD24/48/72/168♂ 100%200–250RNA+++
ELISA, Enzyme-linked immunosorbent assay; IHC, immune histochemistry; N/A, not available; RNA, ribonuclein acid; SD, Spraque–Dawley; WB, Western blot; + reported; - not reported.
Figure 3 shows the distribution of warm ischaemia times compared with their reperfusion times for each study of 100% HIRI in rat models.
Rat HIRI models with additional procedures, such as ischaemic preconditioning, cirrhotic liver, atrial hepatectomy or splenic–caval shunt, were not included in the meta-analysis because of significant methodological heterogeneity with regard to the experimental model. These references are presented in Table 3, divided by the presence or absence of hepatectomy and ordered by the size of the ischaemia area. In total, seven studies performed partial hepatectomies.
Table 3. Overview of other rat models (not included in the meta-analysis).
Table 3. Overview of other rat models (not included in the meta-analysis).
ModelReferenceYearStrainReperfusion Time (h)GenderBW (g)iNOS Detection MethodNO DetectioneNOS DetectionSurvival Rate
30% HIRI
60 min ischemia time
Wang [106]2012SD6♂ 100%250–300WB---
40% HIRI
45 min ischemia time
Björnsson [107]2015SD4♂ 100%313–444RNA/IHC+--
40% HIRI
60 min ischemia time
Björnsson [108]2014SD1/4♂ 100%258–444RNA+--
32% HIRI + 68% PH
30 min ischemia time
Liang [109]2009SD3/8/48/168♀ 100%200–220IHC--+
50%HIRI + 50%PH
45 min ischemia time
Iwasaki [110]2019Wistar1/3/24/168♂ 100%200–300RNA/WB++-
70% HIRI + 30%PH
45 min ischemia time
Shen [111]2007SD3/12/24/168♂ 100%220–300SP+++
70% HIRI + 30%PH
45 min ischemia time
Shen [112]2007SD3/12/24♂ 100%250–300SP+++
70% HIRI + 30%PH
60 min ischemia time
El-Gohary [113]2017Wistar1/5♂ 100%200–250WB-+-
70% HIRI + 30%PH
60 min ischemia time
Zhang [114]2015SD6/168♂ 100%250–300WB+--
70% HIRI + 70%PH
30 min ischemia time
Duval [115]2010SD0/0.5/1/3/6/9/12/15/18/21/24/30/48/72♂ 100%200–225RNA---
100% HIRI + 70%PH
15 min ischemia time
Kawai [116]2010Wistar1♂ 100%250–300RNA/IHC++-
120, 150 min ischemia time + SCSSankary [96]1999SD≤144♂ 100%200–250IHC---
IHC, immune histochemistry; PH, Partial hepatectomy; RNA, ribonuclein acid; SCS, splenic-caval shunt; SD, Spraque–Dawley; SP, Spectrophotometric analysis; WB, Western blot; + reported; - not reported.

2.4. Details for Mouse Models

Thirty-nine studies in mouse models were included. As shown in Table 4, 37 (95%) studies with 70% HIRI were reported. The ischaemic time varied from 30 to 90 min. Ischaemic times of 45 (6 studies), 60 (19 studies) and 90 (7 studies) min were included in the meta-analysis.
Table 4. Overview of mouse models with 70% HIRI.
Table 4. Overview of mouse models with 70% HIRI.
ReferenceYearStrainReper-fusion Time (h)GenderAge (Weeks)iNOS
Detection Method
NO
Detec.
eNOS
Detec.
Survival Rate
45 min ischemia time
Bae [117]2014C57Bl/61/6/24♂ 100%8–10RNA---
Datta [118]2014C57Bl/6 (eNOS −/− and WT)2♂ 100%8–12WB-+-
Hines [119]2001C57Bl/6 (iNOS −/− and WT)1/3/6♂ 100%N/ARNA---
Hines [120]2002C57Bl/6 (iNOS −/−, eNOS −/− and WT)1/3♂ 100%N/ARNA-+-
Kawachi [121]2000C57Bl/6 (NOS −/−, eNOS −/− and WT)5♂ 100%N/ARNA-+-
Lee [122]2001C57Bl/6 (iNOS −/−, eNOS −/− and WT)6♂/♀ 50%N/ARNA---
60 min ischemia time
Chen [123]2017C57Bl/6 (NLRC5 −/− and WT)N/A♂ 100%8–10ELISA---
Gao [124]2016Cav-1tm1Mls/J (Cav-1 −/−and WT)1/6/12♂ 100%8–12WB/ICH---
Guo [125]2011BALB/c2/4/24♂ 100%N/ASP++-
Guo [126]2011BALB/c2/4/12♂ 100%N/AWB/SP-+-
Jeyabalan [127]2008C57Bl/63/6♂ 100%8–12RNA++-
Kim [128]2015C57Bl/66♂ 100%6–8RNA---
Klune [129]2012C57BL/6 (IRF2 −/−, IRF2 +/− and WT)6♂ 100%8–12RNA---
Lee [122]2001C57Bl/6 (iNOS −/−, eNOS −/− and WT)6♂/♀ 50%N/ARNA---
Luedde [130]2005Ikk2 −/−, Nemo −/−6/24♂ 100%8–10IHC---
Moon [131]2008C57Bl/62♂ 100%N/AWB---
Mukhopadhyay [132]2011C57Bl/62/6/24♂ 100%N/ARNA---
Qiao [133]2020C57Bl/6 (iNOS −/− and WT)6/24♂ 100%8–10WB---
Qiao [134]2019C57Bl/6 (iNOS −/− and WT)6/24♂ 100%~8RNA/WB---
Sanches [135]2014Swiss6♂ 100%N/AWB++-
Shaker [136]2016C57Bl/63/12♂ 100%8–10RNA---
Shi [137]2012C57Bl/62/6/168♂ 100%8–9WB--+
Tsung [138]2006C57Bl/6 (IRF−1 −/− and WT)1/3/6♂ 100%8–12RNA---
Tsurui [139]2005C57Bl/62/6♂ 100%8–12RNA-+-
Zhao [140]2017BALB/c (H−2d)0/6♂ 100%6–8RNA---
90 min ischemia time
Ajamieh [141]2015foz/foz and WT24♂ 100%8RNA-+-
Duarte [142]2012C57Bl/6 (TIMP −/− and WT)6/48/168♂ 100%N/ARNA--+
Freitas [143]2010C57Bl/66♂ 100%8–12RNA---
Hamada [144]2009C57Bl/6 (iNOS −/− and WT) and C57Bl/6 (MMP−9 −/− and WT)3/6/24♂ 100%8–10WB+--
Lee [145]2016C57BL/66♂ 100%8WB---
Okaya [146]2004C57Bl/6 (PPARa −/− and WT)8♂ 100%8–12WB++-
Zhou [147]2020C57Bl/66♂ 100%8ELISA/RNA---
Studies not included in meta-analysis
30 min ischemia time
Tao [148]2016C57Bl/66♂ 100%12RNA/WB---
Lee [122]2001C57Bl/6 (iNOS −/−, eNOS −/− and WT)6♂/♀N/ARNA---
40 min ischemia time
Patouraux [149]2014C57Bl/6 (Opn −/− and WT)4♂ 100%10–12RNA---
75 min ischemia time
Kobayashi [150]2008BALB/c24N/A6–8WB/IHC---
Lee [122]2001C57Bl/6 (iNOS −/−, eNOS −/− and WT)6♂/♀ 50%N/ARNA---
ELISA, Enzyme-linked immunosorbent assay; IHC, immune histochemistry; N/A, not available; RNA, ribonuclein acid; SP, Spectrophotometric analysis; WB, Western blot; + reported; - not reported.
Figure 4 shows the distribution of warm ischaemia times compared with their reperfusion times for each study of 70% HIRI in mouse models.
Two studies, one with a 100% HIRI model and the other with an unclear ischaemic area, were not included in the meta-analysis (Table 5).
Table 5. Other mouse models not included in the meta-analysis.
Table 5. Other mouse models not included in the meta-analysis.
ModelReferenceYearStrainGenderAge (Weeks)Reper-fusion Time (h)iNOS Detection MethodNO Detec.eNOS Detec.Survival Rate
100% HIRI
3, 20 min ischemia time
Jessup [151]2005Nude and C57bl/6 (IL-10 −/−, IL-6 −/− and WT)♂ 100%84RNA+-+
70% HIRI + 30% PHGodwin [152]2014C57bl/6♂ 100%N/A24RNA---
N/A, not available; PH, Partial hepatectomy; RNA, ribonuclein acid; + reported; - not reported.
Different analysis methods, such as RNA and immunohistochemistry (IHC), were used to measure iNOS We present an overview of the different methods for iNOS detection in Table 6. Additionally, we state the number of valuable results for the meta-analysis for each of these individual methods.
For the meta-analysis, the method of RNA detection was used most frequently, followed by Western blot and IHC. Among all methods, RNA detection could be evaluated for meta-analysis most frequently (13.4%). Enzyme-linked immunosorbent assay (ELISA) and spectrometric detection methods were less likely to be quantified and displayed in the current studies.

2.5. Risk of Bias

One hundred and fifteen studies were judged for risk of bias, as shown in Figure 5 and Figure 6. We found that 22.2% of all studies had a low risk by using a random sequence generation, although none of them gave insight into the method used. The risk of bias was equally distributed (40.8% to 43.6%) between low and high risk in baseline characteristics, with a small proportion (16.1%) with unclear risk. Most of the studies (92.0%) gave a statement about animals being housed randomly and equally. For incomplete outcome data, the majority of studies (61.3%) were at high risk, and even 96.4% had no reporting bias. A high risk for other sources of bias was found in 24.3% of the studies because of private funding or analysis errors, with the rest being positively low.
We could not find any statement for allocation concealment, blinding of participants and personal and random outcome assessment in any of the included studies, and these questions were judged as unclear. Two studies did, however, refer to a blinding of outcome assessment with low risk. Apart from histological examinations, none of the other studies referred to any blinded outcome assessment for their data assessment, which meant that the risk of bias was unknown.

2.6. Effects of Intervention

For rats and mice, we performed independent meta-analyses for different warm ischaemia models separated into 70% and 100% warm ischaemia on the liver and various warm ischaemia time points. Graphs were grouped by reperfusion time points to show the course.

2.6.1. Analysis of Rat Models with 70% HIRI and 30 min Warm Ischaemia Time

For the studies of rats with 70% HIRI models, three meta-analyses were performed for 30, 60 and 90 min warm ischaemia times.
Four studies reported iNOS values of 70% HIRI and 30 min warm ischaemia time for 60 animals (Figure 7). Considering the reported event rates across studies, we found that three studies had an increased iNOS level when undergoing HIRI. Ischaemia-reperfusion injury caused an increase in iNOS in three studies whereas two studies showed a decrease at a warm ischaemia time of 30 min when compared with the iNOS levels in the sham operation (SHAM) groups for all studies (SMD of 0.51, 95% CI = −3.20–4.21, I2 = 93%).

2.6.2. Analysis of Rat Models with 70% HIRI and 60 min Warm Ischaemia Time

Eleven studies reported iNOS values of 70% HIRI and 60 min warm ischaemia time for 159 animals (Figure 8). Considering the reported event rates across studies, we found that 10 studies had an increased iNOS level when undergoing HIRI. Ischaemia-reperfusion injury results caused a large increase in iNOS at a warm ischaemia time of 60 min when compared with the iNOS levels in the SHAM groups for all studies (SMD of 4.81, 95% CI = 3.23–6.397, I2 = 81%).

2.6.3. Analysis of Rat Models with 70% HIRI and 90 min Warm Ischaemia Time

Five studies reported iNOS values of 70% HIRI and 90 min warm ischaemia time for 83 animals (Figure 9). Considering the reported event rates across studies, we noted that all studies had an increased iNOS level when undergoing HIRI. Ischaemia-reperfusion injury was caused in a large increase in iNOS at a warm ischaemia time of 90 min when compared with the iNOS levels in the SHAM groups for all studies (SMD of 4.06, 95% CI = 2.29–5.82, I2 = 74%).

2.6.4. Analysis of Rat Models with 100% HIRI and 45 min Warm Ischaemia Time

For rats with 100% HIRI, we analysed a 45 min warm ischaemia time in subgroups with different reperfusion time points. The 100% HIRI 30 min and 60 min groups could not be analysed because fewer than three studies showed valid values.
Five studies reported iNOS values of 100% HIRI and 45 min warm ischaemia time for 68 animals (Figure 10). Considering the reported event rates across studies, we identified that four studies showed an increased iNOS level in the animals that were undergoing HIRI. Ischaemia–reperfusion injury caused a large increase in iNOS at a warm ischaemia time of 45 min when compared with the iNOS levels in the SHAM groups for all studies (SMD of 4.53, 95% CI = 1.89–7.16, I2 = 82%).

2.6.5. Analysis of Mouse Models with 70% HIRI and 60 min Warm Ischaemia Time

Mice with 70% HIRI and 45, 60 and 90 min warm ischaemia times were analysed in subgroups with different reperfusion time points. A meta-analysis could only be performed in the 60 min group because of an insufficient number of studies (n ≥ 3 required by our protocol).
Three studies reported iNOS values of 70% HIRI and 60 min warm ischaemia time for 38 animals (Figure 11). Considering the reported event rates across studies, we noted that all studies showed an increased iNOS level in the animals undergoing HIRI. Ischaemia–reperfusion injury caused a large increase in iNOS at a warm ischaemia time of 45 min when compared with the iNOS levels in the SHAM groups for all studies (SMD of 4.06, 95% CI = 2.58–5.55, I2 = 24%). Heterogeneity was moderate because of the small numbers of animals and differences in the detection methods.
Our meta-analysis showed iNOS to be elevated in HIRI versus SHAM-operated animals regardless of species, model or ischaemic time. Our meta-analysis using a random effect showed that iNOS was positively associated with ischaemia damage. The same effects were found for a rat model of 100% HIRI and 45 min ischaemia time. The results of a mouse model of 70% HIRI and 60 min ischaemia time were consistent with the previous results and exhibited low heterogeneity. Overall, the heterogeneity was moderate.

2.6.6. Analysis Comparing NO Values between SHAM and HIRI Groups in Rats

In addition to iNOS, we also investigated NO as a free radical scavenger associated with the iNOS cascade. For the meta-analysis of nitrate/nitrite values, 20 studies were included. Because of missing values, only rat studies of different models were analysed. The studies were grouped by warm ischaemia time for comparison.
Twenty-three studies reported NO values for various HIRI models with different warm ischaemia and reperfusion times for 362 animals (Figure 12). Considering the reported event rates across studies, we estimated that seven studies had decreased NO levels after HIRI. In contrast, 16 studies reported an increase in NO values (SMD of 1.25, 95% CI = −0.00–2.50, I2 = 100%). Heterogeneity was high because of the methodological and clinical diversity.
The subgroups within this analysis used different reperfusion time points, which might have affected the results of NO expression. Therefore, we grouped the above results by reperfusion time to determine if there was a trend regardless of the HIRI damage influenced by warm ischaemia time. These meta-analysis results were consistent with the results above.

2.6.7. Analysis of eNOS Values Compared between the SHAM and HIRI Groups in Rats

In addition to iNOS and NO, we performed a meta-analysis to determine if eNOS is affected by HIRI when compared with SHAM-operated animals. Only results for rat studies could be analysed at three different warm ischaemia time points (45, 60 and 90 min).
Eleven studies reported eNOS values for various HIRI models when compared with the SHAM groups with different warm ischaemia and reperfusion times for 156 animals (Figure 13). Considering the reported event rates across studies, there was no significant effect for eNOS for 45, 60 or 90 min warm ischaemia time (SMD −0.78, 95% CI = −2.18–2.90, I2 = 90%). The results may have been affected by the low animal numbers, different analysis methodologies (Western blot vs. RNA) and an unknown reperfusion time in the study of Mostafa–Hedeab [41]. Therefore, because of the methodological and clinical diversity, heterogeneity for subgroups was high.

3. Discussion

3.1. Summary of Main Results

In this present study, we analysed the reporting in 124 animal studies focusing on the relationship between HIRI and iNOS. To the best of our knowledge, this is the first systematic review on the role of iNOS in HIRI. In our quality assessment, studies were divided into subgroups based on outcomes. The literature was subsequently reviewed with a focus on an overview of experimental models, sex distribution, weight or age, warm ischaemia and reperfusion time, iNOS detection method, NOS-related parameters and survival rates for the HIRI and treatment groups with drug treatment.

3.2. Rat and Mouse Models within This Systematic Review

This systematic review included rat and mouse models of HIRI with various ranges of ischaemia. Among those HIRI models, an ischaemic area of >60% of liver tissue is clinically relevant. The model of 100% HIRI with portal decompression is ideal for warm ischaemia after PM for hepatic resection and LTx because it best mimics HIRI clinically [153]. The hepatic tolerance to warm HIRI caused by PM is thought to be associated with the duration of liver ischaemia [154]. However, the lack of abundant collateral circulation to the liver in rats and mice limits the ability to assess prolonged ischaemic damage in this 100% HIRI model [153]. The 60–70% HIRI models mimic the conditions for hepatic resection (e.g., posterior segment resection) with occlusion of blood flow in the right lobe. In this systematic review, the 100% HIRI model was found in 23 studies using rat models, and only one study using this technique in a mouse model. Eleven studies could be used in the meta-analysis.
The 70% HIRI model has been widely used in experimental studies of hepatic ischaemia-reperfusion [16]. This technique causes ischaemia by occluding the hepatic artery, portal vein and bile ducts in the left and median lobes. Blood flow through the right and caudal liver lobes allows assessment of prolonged ischaemic damage. In this systematic review, we identified 61 studies that used a 70% HIRI model in rats and 36 studies in mice, of which 40 studies could be used in the meta-analysis.
On the other hand, the 30% HIRI model demonstrated that the blood supply to the right lobe of the liver was interrupted by occlusions at the level of the hepatic artery and portal vein [155]. This model has haemodynamic circulation, which enables the assessment of prolonged ischaemic insults, but it is difficult to apply clinically.

3.3. Warm Ischaemia and Reperfusion Models

The standard PM in humans is performed as an ischaemic procedure of the whole liver for 15 min, followed by unclamping for 5 min, and these cycles are repeated until the liver resection is complete [156]. Therefore, a 15 or 20 min ischaemia time at 100% HIRI [28,101,102], 20 min at 70% HIRI [84] or 15 min at 100% HIRI + 70% partial hepatectomy (PH) [114], if PH is added, is in accordance with human clinical practice. However, these studies were not subjected to meta-analysis. The reason for the small number of studies is that the response to ischaemia differs between humans and rodents; thus, the same conditions are not given. So, far, only six studies have used an ischaemia time <20 min. Among all studies (93), the ischaemia time ranged between 30 and 60 min, and 24 studies opted for a higher reperfusion time than 24 h. The ischaemia time is unknown for one study.
Regarding the reperfusion time, the three groups that were included in the meta-analysis had similar sampling points. In the 70% HIRI rat model, iNOS expression was observed from 1 to 24 (1, 2, 4, 5, 6 and 24) h and from 2 to 24 (2, 3 and 24) h after initiation of reperfusion in response to 60 and 90 min of ischaemia, respectively. The 100% HIRI rat model showed iNOS expression from 0.75 to 24 (0.75, 1, 2 and 24) h after initiation of reperfusion in response to 45 min of ischaemia, respectively. In the 100% HIRI mouse model, the expression of INOS was observed from 0.7 to 24 h after the start of reperfusion in response to 45 min of ischaemia. In rat hepatocytes, iNOS mRNA expression at 3 h after inflammatory cytokine stimulation in primary cultured cells, followed by iNOS protein and NO increase at 8 h afterwards, has been reported [157]. Divided into an early phase (0–4 h after HIRI), a middle phase (5–8 h) and a late phase (later), the similarity in the focus on iNOS concentrations can be seen by analysing iNOS mRNA in the early phase, iNOS protein and NO measurements in the middle phase and the associated responses in the late phase. However, the commonality of reperfusion times is impossible to find exactly because of the different conditions of the models and the different focus of the authors.
In this systematic review, we sought to determine if a certain ischaemia time is associated with a reperfusion time so that we could determine if a mouse or rat model is best suited to investigate the iNOS pathways. Unfortunately, we showed that the warm ischaemia time and reperfusion time varied between mouse and rat models of the same HIRI. A standardisation for comparing the outcomes is needed not only with regard to animal models but also to clinical situations and possible translation of results.

3.4. Overall Completeness and Applicability of Evidence

3.4.1. Sex Distribution

Regarding the general characteristics, various experiments have been conducted without standardisation. We also observed a sex bias in our analysis. In our selected studies, mostly male mice and rats (97.4%) were used [158]. Female rats have a 4-day sexual cycle, which would affect the experiment because oestrogen has a hepatoprotective effect [159]. Sex dimorphism in hepatic ischaemia-reperfusion in the murine liver ischaemia-reperfusion model has been demonstrated [160]. It was found that 100% of the female mice survived indefinitely whereas all male mice died within 5 days. The protective effect in females appeared to be due to ovarian oestrogen, as ovariectomy of females or administration of a selective oestrogen antagonist to females resulted in an enhanced liver injury and greater mortality rate. It was shown that oestrogen protects against ischaemic-reperfusion injury of the liver, which was associated with increased levels of serum NO and decreased levels of TNFα [161]. In another animal study, the mechanism of the effects of oestrogen included its antioxidative nature [162,163,164]. These findings might explain why the studies in this systematic review opted to use males. In humans, different mechanisms have been hypothesised, including the effect of sex hormones on oxidative, metabolic and immunological pathways, as well as differential gene transcription in response to acute and chronic liver injury [165]. According to the European Liver Transplant Registry data, 46,334 patients underwent liver transplant in Europe between December 2002 and December 2012, with 32,656 (70.5%) males and 13,678 (29.5%) females [166]. In 2011–2012, the proportion of male donors was 56.1% and female donors was 43.9% [166]. Thus, ischaemia-reperfusion injury can occur in both males and females, but most studies in rodents have been exclusively conducted in males. Additionally, there are sex differences in the liver metabolism of human cytochrome P450 isoforms in rodents [167,168]. It is important to study novel therapies at the basic science level in both sexes [169]. Therefore, it is quite possible that studies using only males may not translate into certain clinical situations.

3.4.2. iNOS Generation and Function

Transcriptional regulation is the main control of iNOS expression. In general, proinflammatory cytokines, such as TNF-alpha, IL-1, interferon-gamma and lipopolysaccharides, first bind to cell surface receptors and activate kinases, leading to the phosphorylation of various intracellular proteins. Specific transcription factors, including nuclear factor κB and transcription factors, such as nuclear factor 1 transducer and activator of transcription 1a, are activated. The activated factor translocates into the nucleus, binds to the promoter region of the iNOS gene and induces expression of iNOS [170].
Overexpression or deregulation of iNOS causes excessive NO levels, which can lead to toxic effects and are associated with a number of human diseases, such as septic shock, cardiac dysfunction, pain, diabetes and cancer [171]. Large amounts of NO play an important role in the inflammatory response and the innate immune system by helping to protect against incoming pathogens [172]. We speculate that decreased iNOS in humans would reduce inflammatory responses and defences against the innate immune system, but this has not been reported.
In this systematic review, iNOS was found to have been measured by a variety of analytical methods in mRNA and protein (Western blot, IHC, ELISA and spectrometry). The heterogeneity of iNOS assays made comparisons difficult. However, mRNA assays are widely used (13.4%) and provide quantitative comparability, but they cannot prove that iNOS is transcribed instead of metabolised. Therefore, we require a more uniform measurement method with standardised scaling focusing on the presence of the effective protein.

3.4.3. iNOS in HIRI versus SHAM

High NO concentrations due to the promotion of iNOS may lead to harmful products, such as peroxynitrite (ONOO−), and other reactive oxygen species (ROS) are formed [173]. ROSs cause not only direct damage to cells, but also activate a cascade of molecular mediators, leading to acute inflammatory changes with microvascular alterations, increased apoptosis and increased necrosis of liver cells [174]. In this study, we found that HIRI increased iNOS in rats and mice without transgenic backgrounds. In iNOS knockout mice, mRNA of iNOS was not expressed at reperfusion [121].

3.4.4. Increased iNOS May Induce Hepatic Damage Via Significant Production of ROS

Within our meta-analysis, we demonstrated that iNOS was elevated when HIRI was compared with SHAM regardless of species and warm ischaemia time. This is consistent with the common knowledge that iNOS is upregulated after surgery with such high impact on the liver.

3.5. Agreements and Disagreements with Other Studies or Reviews

3.5.1. Nitric Oxide in HIRI Models versus SHAM

In human clinical practice, it has been reported that NO was produced in the liver during HIRI and that iNOS is involved in the production of NO. Plasma nitrate and nitrite were determined by the Griess reaction [175]. In addition to iNOS, a meta-analysis was performed to determine if NO is affected by HIRI relative to animals undergoing SHAM surgery. The present analysis did not show any effect on NO in HIRI, or any relationship with iNOS, which may be because of the lack of standardisation of conditions (e.g., reperfusion time and ischaemia time) and NO detection (method and time) in animal experiments.

3.5.2. eNOS in HIRI Models versus SHAM

The hepatoprotective effect of eNOS against HIRI is related to increasing NO levels, and inhibition of eNOS overexpression also protects against HIRI. Multiple eNOS/NO signalling pathways (e.g., Akt-eNOS/NO, AMPK-eNOS/NO, HIF-1α-eNOS/NO) participate in the anti-HIRI process [31].
The effect of NO on HIRI has been reported as potentially harmful, beneficial or both [176]. The NO from eNOS is considered to have protective effects and to maintain homeostasis [177,178]. On the other hand, high NO production from iNOS has potentially toxic effects because it may react with superoxide anions to form toxic peroxynitrite [179,180]. In a study of HIRI using genetically engineered mice, iNOS knockout mice showed decrease [122], increase [119] or no change [121] in toxicity caused by NO. In addition, eNOS mRNA was strongly expressed at 1 h post-perfusion in iNOS-knockout mice [120]. From these reports, the role of iNOS and eNOS in HIRI, as well as their interaction with each other, remains unclear. This systematic review excluded genetically modified mice. Therefore, we were not able to further investigate the role of eNOS in HIRI.
The meta-analysis of eNOS of animals undergoing HIRI versus SHAM operation did not show any effect, and thus did not reveal any relationship with iNOS whatsoever.

3.5.3. iNOS and Survival

The indications for reducing iNOS are ischaemia-reperfusion injury, sepsis and pain [181]. Attempts to treat ischaemia-reperfusion injury and septic shock with drugs intended to reduce iNOS in clinical practice have been largely unsuccessful and none have been used in clinical practice [182]. One animal study found that the iNOS inhibitor 2-aminoethyl-isothiourea [78] may not have benefits for HIRI universally. ONO-1714 has been used to reduce HIRI [75]. The non-specific NOS inhibitors, NG-nitro-L-arginine (L-NNA) [183] and L-NAME [34], have been shown to increase HIRI. On the other hand, L-NAME has shown a hepatoprotective effect by inhibiting iNOS mRNA on HIRI after portal triad clamping and reperfusion for nonselective inflow occlusion and partial hepatectomy of cholestatic livers induced by bile duct ligation in rats [110]. L-NAME may be protective or harmful, depending on the model and timing of the experiment, which might be explained by L-NAME being a nonselective NOS inhibitor and by competitive inhibition. L-NAME is a potent inhibitor not only of iNOS but also of eNOS. The use of nonselective NOS inhibitors in clinical practice may increase side effects because eNOS is also inhibited. On the other hand, selective inhibition of iNOS is expected to have fewer side effects, but it is currently still under investigation.
Surgical approaches to ischaemia-reperfusion injury, aimed at reducing iNOS, also have not been used in clinical practice. Evidence of the effect of intermittent hepatic ischaemia (IPC) on NO from eNOS and iNOS is limited. IPC is considered to protect short-term ischaemia against subsequent long-term ischaemic injury. Koti et al. and Guo et al. reported that eNOS was upregulated in rats receiving liver IPC + HIRI compared to rats receiving HIRI [21,125]. The increased eNOS expression was associated with significantly higher plasma NOx levels in the IPC + HIRI group than in the HIRI group. Expression of iNOS was not found in all experimental groups, including both liver IPC + HIRI and HIRI groups [21,125]. On the other hand, Longo et al. reported that the expression of eNOS and iNOS was not increased in the livers of rats receiving intrahepatic IPC + HIRI relative to the levels in rats receiving HIRI [74]. In remote IPC, increased NO due to eNOS expression in the liver [81] and iNOS in skeletal muscle is thought to act against HIRI protection.
A study of increased iNOS mRNA by administration of metron factor-1 (MF-1) reported increased survival. In HIRI, MF-1 improved hepatic microcirculation by increasing NO expression, and NO-induced damage was not obvious [105]. In these studies, focusing on survival experiments, there was no evidence of a link between increased survival and changes in iNOS alone. Apart from NOS-related and general (e.g., liver function enzymes, inflammatory cytokines, pathology) analyses, there were no analyses in the literature focused on other mechanisms unaffected by iNOS. In discussion, the overall improvement in the condition was considered to have contributed to the increased survival rate.
Another strategy to improve the survival rate is to avoid HIRI in the first place. New surgical tools are available to dissect liver tissue without the need for a Pringle manoeuver, thus preventing ischemia-reperfusion injury. Such instruments as the Habib sealer or ultrasonic scalpels can coagulate the tissue while cutting [184,185]. Studies in patients showed fewer postoperative complications and a reduction of blood loss as well as reduced liver manipulation and significant reduced warm ischemia time [186,187,188].
Considering the fact that many included studies investigated reducing iNOS expression in HIRI models, it is interesting that there is a lack of studies linking the iNOS reduction to a possible prolonged survival.

3.6. Potential Biases in the Review Process

Our systematic review had several limitations. The aim of this systematic review was to provide a wide overview into current approaches and the state of research on the effects of warm ischaemia and reperfusion injury in mouse- and rat-liver models on iNOS and other related parameters. Our systematic search was therefore designed to be broad and include as many related studies as possible. Consequently, we retrieved a variety of differing approaches and were able to qualitatively assess them. Nonetheless, this very inclusive approach might have also precluded some results that might have been obtained if we had used a search protocol that more specifically focused on some of the subsets we identified. Although we are confident in presenting a somewhat comprehensive summary of the review question, it is possible that due to the broadness of our search protocol some eligible studies were not covered. Although we have searched three independent databases it is possible that the search in Web of Science is limited due to licensed access. Additionally, this database has no equivalent to MeSH-terms and we might have missed some studies. One limitation was our exclusion of non-English literature. Another limitation is the fact that we performed the meta-analysis exclusively for HIRI versus SHAM-operated animals. We were therefore not able to implement all included studies as some did not use a SHAM model in their experiments.
Unfortunately, we were not able to include many studies due to incomplete data, such as animal number. We tried to reach out to the authors at least twice but received very little response. We are also aware of the possibility that negative results have not been published. As we included only studies with available data for iNOS, this might have limited the outcome. Additionally, we were only able to implement results if a table or graph provided them. In the case of immunohistochemistry, we were barely able to carry out counting because only example pictures have been reported. Additionally, many publishers limit their word count, so the authors may have had to adjust the length of their reports and exclude some relevant elements. Another limitation is that we could not adjust for the reporting quality of the studies as it related to word count limits of the publishing journals because of some practical obstacles (changes in publication policies over the years, difficulties in contacting every single publisher, and the fact that several journals have been sold or the publisher has changed over the decades); however, we have included the available online supplements in our assessment. Nonetheless, interpretation of these findings must be performed carefully. We cannot rule out the possibility that certain confounders that were not accounted for in the analysis contributed to the observed significant result. Due to the high risk of bias in incomplete outcome data, as well as an unclear randomised assessment of data, we cannot rule out a compromised scientific interpretation of study results.

4. Materials and Methods

This systematic review and meta-analysis were conducted according to the Cochrane standard and was reported according to the principles of the Preferred Reported Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline. The PRISMA checklist can be found in the Supplementary Materials. The protocol is available at PROSPERO (registration number CRD 42020191298).

4.1. Search Strategy

A systematic literature search was performed encompassing PubMed (1946–April 2022); [title/abstract], [MeSH]), Web of Science (1990–April 2022) and Embase databases (1947–April 2022; [ti.ab], [exp/mj]). This literature search was performed to identify all potentially relevant articles on triggered liver regeneration after warm ischaemia in mouse and rat models with a focus on iNOS. The applied search terms are shown in Figure 14. The search terms were limited to Title/Abstract in PubMed and adjusted accordingly for the other two databases. These results were imported to Endnote Version X8 (Clarivate Analytics, NY, USA). Endnote was used to identify duplicates and to generate an Excel file for independent screening and comparison thereafter. Since Web of Science has licensed access, the available collections for this systematic review were:
  • Web of Science Core Collection Indexes;
  • Science Citation Index Expanded (SCI-Expanded) (1900-);
  • Social Sciences Citation Index (SSCI) (1900-);
  • Arts & Humanities Citation Index (A&HCI) (1975-);
  • Conference Proceedings Citation Index—Science (CPCI-S) (1990-);
  • Conference Proceedings Citation Index—Social Sciences & Humanities (CPCI-SSH) (1990-);
  • Book Citation Index—Science (BKCI-S) (2005-);
  • Book Citation Index—Social Sciences & Humanities (BKCI-SSH) (2005-);
  • Current Chemical Reactions (CCR-Expanded) (1985-);
  • Index Chemicus (IC) (1993-);
  • BIOSIS Previews (1926-): Journals, patents and conference proceedings in biomedicine;
  • MEDLINE (1950-);
  • Russian Science Citation Index (2005-);
  • SciELO Citation Index (1997-);
  • Journal Citation Reports.

4.2. Searching Other Resources

We identified other potentially eligible studies by searching the reference lists of systematic reviews and the 124 included studies.

4.3. Criteria for Considering Studies for This Review

The study selection was based on the following inclusion and exclusion criteria.

4.3.1. Types of Studies

We included research publications in international peer-reviewed literature (including inter-library loan requests, open access and closed access) available as full-text publications in English. Conference abstracts, review articles, case studies, editorials, withdrawn articles and those in languages other than English were excluded.

4.3.2. Types of Species

Experimental animal studies using living mice or rats were included in this systematic review. All other species and iNOS or eNOS knockout rats and mice were excluded. A search for other species revealed that mice and rats were the only rodents used in combination with our search terms. A further search listed only twelve studies in which pigs and dogs were used as large animal models combined with iNOS and HIRI.

4.3.3. Types of Intervention

Experimental animal studies that performed laparotomy and warm HIRI with and without partial hepatectomy under anaesthesia were included if warm ischaemia was applied to the whole or parts of the liver graft. Studies were also included if they performed partial hepatectomy and if techniques, such as PM, were used during which the graft underwent warm ischaemia. Liver transplantation was excluded because of the effect on the organ and body yielding various immune system responses. Additionally, grafts for transplantation that received cold ischaemia during storage combined with warm ischaemia afterwards were excluded. Therefore, the liver transplantation data will not be comparable to data from animals that underwent simple warm ischaemia following reperfusion. Additionally, partial hepatectomy without warm ischaemia and reperfusion and partial hepatectomy >70% resection were excluded, as were ex vivo studies and those involving treatment for warm ischaemia and reperfusion with drugs or ischaemic preconditioning.

4.3.4. Types of Comparison

We included a comparison of animals that underwent sham operations versus animals that underwent warm ischaemia and reperfusion, as well as control animals versus animals with various treatments for meta-analysis. Studies without SHAM or HIRI controls were excluded.
Types of outcome measures included were all studies with the parameter of iNOS measured in liver, serum or plasma. If no detection of iNOS was made the study had to be excluded.

4.4. Outcomes

The following outcomes were assessed by meta-analysis:
  • We compared iNOS values of SHAM to HIRI groups using subgroups of different reperfusion times:
  • Rat 70% HIRI at 30 min warm ischaemia time, data grouped by reperfusion time;
  • Rat 70% HIRI at 60 min warm ischaemia time, data grouped by reperfusion time;
  • Rat 100% HIRI at 45 min warm ischaemia time, data grouped by reperfusion time;
  • Mouse 70% HIRI at 60 min warm ischaemia time, data grouped by reperfusion time.
  • Effects of parameters related to iNOS:
  • NO parameters of SHAM groups versus HIRI groups in rat 70% HIRI data grouped by warm ischaemia times;
  • eNOS parameters of SHAM groups versus HIRI groups.

4.5. Differences between Protocol and Review

We specified data handling within the meta-analysis by adding subgroups to distinguish between different reperfusion times for Figure 7, Figure 8, Figure 9, Figure 10, Figure 11, Figure 12 and Figure 13. Furthermore, we did not investigate into survival rates. The protocol states that a formal screening was performed while the preliminary searches were yet not finished. We want to clarify that this was a peer-review screening and the results of this screening were implemented into the systematic review if the studies met our final inclusion criteria.

4.6. Data Collection and Analysis

Two authors (RN, MS) independently screened the results of the search strategies for eligibility for this review by reading the titles and abstracts using EndNote Software (EndNote X8, Clarivate Analytics, London, UK). We coded the abstracts as either ‘include‘ or ‘exclude‘. In the case of disagreement, or if it was unclear whether we should include the abstract or not, we obtained the full-text publication for further discussion. Both review authors assessed the full-text articles of the selected studies. Disagreements concerning the suitability of an article were discussed by the study team. A third reviewer was not needed as all disagreements were resolved. If applicable, online supplementary information was also retrieved for further analysis.
We documented the study-selection process in a flow chart, as recommended in the PRISMA statement [189], and show the total numbers of retrieved references and the numbers of included and excluded studies.

4.7. Handling of Missing Data

We contacted authors for missing data at least twice to confirm study characteristics and obtain details for the risk of bias assessment. If studies only showed their results in graphs, we used a digital ruler to measure the data from the graph to the best of our ability if possible.
The following data were extracted from each of the included studies:
  • Species/strain and liver model;
  • Warm ischaemia and reperfusion time for the HIRI and SHAM groups;
  • Sex distribution of mice and rats for each study;
  • Weight or age of experimental animals for each study;
  • iNOS detection method;
  • NO parameter and eNOS parameters assessed parallel to iNOS for the SHAM and HIRI groups.

4.8. Risk of Bias and Quality Assessment

Two authors (R.N., M.S.) independently evaluated the methodological quality of the included studies using the risk of bias tool for animals according to the Systematic Review Center for Laboratory Animal Experimentation (SYRCLEs) risk of bias. Two different authors (C.K., S.B.) performed the risk of bias assessment for the studies by Ishizaki [84] and Iwasaki [110] to avoid any conflicts of interest.
We adapted the tool based on the needs of this systematic review and meta-analysis as recommended by SYRCLEs [190]. The following aspects were covered in our study’s risk of bias tool: (a) random sequence generation, (b) baseline characteristics, (c) allocation concealment, (d) random housing, (e) blinding of caregivers and investigators, (f) random outcome assessment, (g) blinding of outcome assessors, (h) incomplete outcome data, (i) selective reporting and (j) other bias.

4.9. Data Analysis and Statistics

Data were compared and plotted using Review Manager Software (Version 5.3, Nordic Cochrane Center, Copenhagen, Denmark). We performed the meta-analysis only when enough data (n ≥ 3) were available. In tests for overall effect, p < 0.05 was accepted as indicative of statistical significance. A random-effects model for the meta-analysis was chosen to compensate for methodological experimental heterogeneity such as parameter assessment, as well as various treatments. For group size, the mean values were used. Continuous data are presented as the standardised mean difference (SMD) with 95% confidence intervals (CI) as studies used different scales to measure the same outcome. Heterogeneity was tested using I2 [191].

5. Conclusions

Our systematic review and meta-analysis showed that iNOS was clearly increased in HIRI in both mice and rats regardless of the warm ischaemia time or reperfusion time (end point). We did not identify any association between iNOS upregulation and an effect on NO or eNOS levels.
We found a lack of comparability of different detection methods for iNOS leading to a decreased number of studies including into meta-analysis. Additionally, the number of animals per group was often unclear. We recommend that studies need to be more precise and publish complete results for later evaluation.
We were not able to find a direct link between iNOS reduction and prolonged survival within the literature. However, further investigation into the causality of a possible association of iNOS with survival is needed. Furthermore, the specific circumstances that determine if iNOS-generated NO will have a positive effect on survival also require more study.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms231911916/s1, PRISMA checklist.

Author Contributions

Conceptualization, R.N., M.S. and R.H.T.; software, data curation, visualization, M.S.; methodology, validation, formal analysis, resources, writing—original draft preparation, R.N. and M.S.; investigation, R.N., M.S., C.K. and C.B.; writing—review and editing, C.B. and R.H.T.; supervision, R.H.T.; project administration, R.H.T. and M.S.; funding acquisition, R.H.T. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the German Research Foundation (Deutsche Forschungs Gemeinschaft—DFG) FOR-2591 to R.H.T. (TO 542/5-2).

Institutional Review Board Statement

The paper is exempt from ethical committee approval as no additional animal experiments were performed for this systematic review.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data of this study are openly available in ‘Zenodo’ at https://doi.org/10.5281/zenodo.5082151 (accessed on 8 July 2021).

Acknowledgments

We thank the scientists who conducted the primary research for providing us with information on their eligibility for this systematic review.

Conflicts of Interest

Two included studies [33,34] in this systematic review are related to the authors’ groups. These studies were reviewed by two additional independent authors (C.B. and C.K.). to confirm their eligibility for this review. The authors declare no other conflicts of interest.

References

  1. Mirnezami, R.; Mirnezami, A.H.; Chandrakumaran, K.; Abu Hilal, M.; Pearce, N.W.; Primrose, J.N.; Sutcliffe, R.P. Short- and long-term outcomes after laparoscopic and open hepatic resection: Systematic review and meta-analysis. HPB 2011, 13, 295–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Shelat, V.G.; Cipriani, F.; Basseres, T.; Armstrong, T.H.; Takhar, A.S.; Pearce, N.W.; AbuHilal, M. Pure laparoscopic liver resection for large malignant tumors: Does size matter? Ann. Surg. Oncol. 2015, 22, 1288–1293. [Google Scholar] [CrossRef] [PubMed]
  3. Shelat, V.G.; Serin, K.; Samim, M.; Besselink, M.G.; Al Saati, H.; Gioia, P.D.; Pearce, N.W.; Abu Hilal, M. Outcomes of repeat laparoscopic liver resection compared to the primary resection. World J. Surg. 2014, 38, 3175–3180. [Google Scholar] [CrossRef] [PubMed]
  4. Chacon, E.; Eman, P.; Dugan, A.; Davenport, D.; Marti, F.; Ancheta, A.; Gupta, M.; Shah, M.; Gedaly, R. Effect of operative duration on infectious complications and mortality following hepatectomy. HPB 2019, 21, 1727–1733. [Google Scholar] [CrossRef]
  5. Farges, O.; Goutte, N.; Bendersky, N.; Falissard, B. Incidence and risks of liver resection: An all-inclusive French nationwide study. Ann. Surg. 2012, 256, 697–704. [Google Scholar] [CrossRef]
  6. Filmann, N.; Walter, D.; Schadde, E.; Bruns, C.; Keck, T.; Lang, H.; Oldhafer, K.; Schlitt, H.J.; Schön, M.R.; Herrmann, E.; et al. Mortality after liver surgery in Germany. Br. J. Surg. 2019, 106, 1523–1529. [Google Scholar] [CrossRef]
  7. Kenjo, A.; Miyata, H.; Gotoh, M.; Kitagawa, Y.; Shimada, M.; Baba, H.; Tomita, N.; Kimura, W.; Sugihara, K.; Mori, M. Risk stratification of 7732 hepatectomy cases in 2011 from the National Clinical Database for Japan. J. Am. Coll. Surg. 2014, 218, 412–422. [Google Scholar] [CrossRef]
  8. Pringle, J.H.V. Notes on the Arrest of Hepatic Hemorrhage Due to Trauma. Ann. Surg. 1908, 48, 541–549. [Google Scholar] [CrossRef]
  9. Belghiti, J.; Noun, R.; Malafosse, R.; Jagot, P.; Sauvanet, A.; Pierangeli, F.; Marty, J.; Farges, O. Continuous versus intermittent portal triad clamping for liver resection: A controlled study. Ann. Surg. 1999, 229, 369–375. [Google Scholar] [CrossRef]
  10. Ishizaki, Y.; Yoshimoto, J.; Miwa, K.; Sugo, H.; Kawasaki, S. Safety of prolonged intermittent pringle maneuver during hepatic resection. Arch. Surg. 2006, 141, 649–653. [Google Scholar] [CrossRef]
  11. van der Bilt, J.D.; Livestro, D.P.; Borren, A.; van Hillegersberg, R.; Borel Rinkes, I.H. European survey on the application of vascular clamping in liver surgery. Dig. Surg. 2007, 24, 423–435. [Google Scholar] [CrossRef]
  12. Lee, K.F.; Cheung, Y.S.; Wong, J.; Chong, C.C.; Wong, J.S.; Lai, P.B. Randomized clinical trial of open hepatectomy with or without intermittent Pringle manoeuvre. Br. J. Surg. 2012, 99, 1203–1209. [Google Scholar] [CrossRef]
  13. Capussotti, L.; Muratore, A.; Ferrero, A.; Massucco, P.; Ribero, D.; Polastri, R. Randomized clinical trial of liver resection with and without hepatic pedicle clamping. Br. J. Surg. 2006, 93, 685–689. [Google Scholar] [CrossRef]
  14. Man, K.; Fan, S.T.; Ng, I.O.; Lo, C.M.; Liu, C.L.; Wong, J. Prospective evaluation of Pringle maneuver in hepatectomy for liver tumors by a randomized study. Ann. Surg. 1997, 226, 704–711. [Google Scholar] [CrossRef]
  15. Zaki, H.F.; Abdelsalam, R.M. Vinpocetine protects liver against ischemia-reperfusion injury. Can. J. Physiol. Pharm. 2013, 91, 1064–1070. [Google Scholar] [CrossRef]
  16. Massip-Salcedo, M.; Roselló-Catafau, J.; Prieto, J.; Avíla, M.A.; Peralta, C. The response of the hepatocyte to ischemia. Liver Int. 2007, 27, 6–16. [Google Scholar] [CrossRef] [Green Version]
  17. Dudzinski, D.M.; Igarashi, J.; Greif, D.; Michel, T. The regulation and pharmacology of endothelial nitric oxide synthase. Annu. Rev. Pharm. Toxicol. 2006, 46, 235–276. [Google Scholar] [CrossRef]
  18. Iwakiri, Y.; Kim, M.Y. Nitric oxide in liver diseases. Trends Pharm. Sci. 2015, 36, 524–536. [Google Scholar] [CrossRef] [Green Version]
  19. Canbakan, B.; Akin, H.; Tahan, G.; Tarcin, O.; Eren, F.; Atug, O.; Tahan, V.; Imeryuz, N.; Yapicier, O.; Avsar, E.; et al. The effects of pegylated interferon alpha 2b on bile-duct ligation induced liver fibrosis in rats. Ann. Hepatol. 2009, 8, 234–240. [Google Scholar] [CrossRef]
  20. Erwin, P.A.; Lin, A.J.; Golan, D.E.; Michel, T. Receptor-regulated dynamic S-nitrosylation of endothelial nitric-oxide synthase in vascular endothelial cells. J. Biol. Chem. 2005, 280, 19888–19894. [Google Scholar] [CrossRef]
  21. Koti, R.S.; Tsui, J.; Lobos, E.; Yang, W.; Seifalian, A.M.; Davidson, B.R. Nitric oxide synthase distribution and expression with ischemic preconditioning of the rat liver. FASEB J. 2005, 19, 1155–1157. [Google Scholar] [CrossRef]
  22. McNaughton, L.; Puttagunta, L.; Martinez-Cuesta, M.A.; Kneteman, N.; Mayers, I.; Moqbel, R.; Hamid, Q.; Radomski, M.W. Distribution of nitric oxide synthase in normal and cirrhotic human liver. Proc. Natl. Acad. Sci. USA 2002, 99, 17161–17166. [Google Scholar] [CrossRef] [Green Version]
  23. Amersi, F.; Shen, X.D.; Moore, C.; Melinek, J.; Busuttil, R.W.; Kupiec-Weglinski, J.W.; Coito, A.J. Fibronectin-alpha 4 beta 1 integrin-mediated blockade protects genetically fat Zucker rat livers from ischemia/reperfusion injury. Am. J. Pathol. 2003, 162, 1229–1239. [Google Scholar] [CrossRef]
  24. Cescon, M.; Grazi, G.L.; Grassi, A.; Ravaioli, M.; Vetrone, G.; Ercolani, G.; Varotti, G.; D’Errico, A.; Ballardini, G.; Pinna, A.D. Effect of ischemic preconditioning in whole liver transplantation from deceased donors. A pilot study. Liver Transpl. 2006, 12, 628–635. [Google Scholar] [CrossRef]
  25. Kimura, H.; Katsuramaki, T.; Isobe, M.; Nagayama, M.; Meguro, M.; Kukita, K.; Nui, A.; Hirata, K. Role of inducible nitric oxide synthase in pig liver transplantation. J. Surg. Res. 2003, 111, 28–37. [Google Scholar] [CrossRef]
  26. Koeppel, T.A.; Mihaljevic, N.; Kraenzlin, B.; Loehr, M.; Jesenofsky, R.; Post, S.; Palma, P. Enhanced iNOS gene expression in the steatotic rat liver after normothermic ischemia. Eur. Surg. Res. 2007, 39, 303–311. [Google Scholar] [CrossRef]
  27. Meguro, M.; Katsuramaki, T.; Nagayama, M.; Kimura, H.; Isobe, M.; Kimura, Y.; Matsuno, T.; Nui, A.; Hirata, K. A novel inhibitor of inducible nitric oxide synthase (ONO-1714) prevents critical warm ischemia-reperfusion injury in the pig liver. Transplantation 2002, 73, 1439–1446. [Google Scholar] [CrossRef] [Green Version]
  28. Miyake, T.; Yokoyama, Y.; Kokuryo, T.; Mizutani, T.; Imamura, A.; Nagino, M. Endothelial nitric oxide synthase plays a main role in producing nitric oxide in the superacute phase of hepatic ischemia prior to the upregulation of inducible nitric oxide synthase. J. Surg. Res. 2013, 183, 742–751. [Google Scholar] [CrossRef]
  29. Colasanti, M.; Suzuki, H. The dual personality of NO. Trends Pharm. Sci. 2000, 21, 249–252. [Google Scholar] [CrossRef]
  30. Jiang, W.W.; Kong, L.B.; Li, G.Q.; Wang, X.H. Expression of iNOS in early injury in a rat model of small-for-size liver transplantation. Hepatobiliary Pancreat. Dis. Int. 2009, 8, 146–151. [Google Scholar]
  31. Zhang, Y.Q.; Ding, N.; Zeng, Y.F.; Xiang, Y.Y.; Yang, M.W.; Hong, F.F.; Yang, S.L. New progress in roles of nitric oxide during hepatic ischemia reperfusion injury. World J. Gastroenterol. 2017, 23, 2505–2510. [Google Scholar] [CrossRef] [PubMed]
  32. Abd-Elbaset, M.; Arafa, E.S.A.; El Sherbiny, G.A.; Abdel-Bakky, M.S.; Elgendy, A.N.A.M. Thymoquinone mitigate ischemia-reperfusion-induced liver injury in rats: A pivotal role of nitric oxide signaling pathway. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2017, 390, 69–76. [Google Scholar] [CrossRef] [PubMed]
  33. Fouad, A.A.; Jresat, I. Therapeutic potential of cannabidiol against ischemia/reperfusion liver injury in rats. Eur. J. Pharmacol. 2011, 670, 216–223. [Google Scholar] [CrossRef] [PubMed]
  34. Liu, P.; Xu, B.; Spokas, E.; Lai, P.S.; Wong, P.Y. Role of endogenous nitric oxide in TNF-alpha and IL-1beta generation in hepatic ischemia-repefusion. Shock 2000, 13, 217–223. [Google Scholar] [CrossRef] [PubMed]
  35. Liu, P.; Yin, K.; Nagele, R.; Wong, P.Y. Inhibition of nitric oxide synthase attenuates peroxynitrite generation, but augments neutrophil accumulation in hepatic ischemia-reperfusion in rats. J. Pharm. Exp. 1998, 284, 1139–1146. [Google Scholar]
  36. Rhee, J.E.; Jung, S.E.; Shin, S.D.; Suh, G.J.; Noh, D.Y.; Youn, Y.K.; Oh, S.K.; Choe, K.J. The effects of antioxidants and nitric oxide modulators on hepatic ischemic-reperfusion injury in rats. J. Korean Med. Sci. 2002, 17, 502–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Wang, J.; Yan, B.J. A polysaccharide (PNPA) from Pleurotus nebrodensis ameliorates hepatic ischemic/reperfusion (I/R) injury in rats. Int. J. Biol. Macromol. 2017, 105, 447–451. [Google Scholar] [CrossRef]
  38. Wang, L.M.; Tian, X.F.; Song, Q.Y.; Gao, Z.M.; Luo, F.W.; Yang, C.M. Expression and role of inducible nitric oxide synthase in ischemia-reperfusion liver in rats. Hepatobiliary Pancreat. Dis. Int. 2003, 2, 252–258. [Google Scholar]
  39. El-Emam, S.Z.; Soubh, A.A.; Al-Mokaddem, A.K.; Abo El-Ella, D.M. Geraniol activates Nrf-2/HO-1 signaling pathway mediating protection against oxidative stress-induced apoptosis in hepatic ischemia-reperfusion injury. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2020, 393, 1849–1858. [Google Scholar] [CrossRef]
  40. Hur, G.M.; Ryu, Y.S.; Yun, H.Y.; Jeon, B.H.; Kim, Y.M.; Seok, J.H.; Lee, J.H. Hepatic ischemia/reperfusion in rats induces iNOS gene transcription by activation of NF-κB. Biochem. Biophys. Res. Commun. 1999, 261, 917–922. [Google Scholar] [CrossRef]
  41. Mostafa-Hedeab, G.; Hanyelhady; El-Nahass, E.S.; Sabry, D. Tadalafil mitigate experimental liver ischemia-reperfusion injury in rats via anti-oxidant, anti-inflammatory and anti-apoptotic action. Int. J. Pharm. Res. 2019, 11, 47–54. [Google Scholar]
  42. Serracino-Inglott, F.; Virlos, I.T.; Habib, N.A.; Williamson, R.C.; Mathie, R.T. Differential nitric oxide synthase expression during hepatic ischemia-reperfusion. Am. J. Surg. 2003, 185, 589–595. [Google Scholar] [CrossRef]
  43. Yang, L.Q.; Tao, K.M.; Liu, Y.T.; Cheung, C.W.; Irwin, M.G.; Wong, G.T.; Lv, H.; Song, J.G.; Wu, F.X.; Yu, W.F. Remifentanil preconditioning reduces hepatic ischemia-reperfusion injury in rats via inducible nitric oxide synthase expression. Anesthesiology 2011, 114, 1036–1047. [Google Scholar] [CrossRef]
  44. Curek, G.D.; Cort, A.; Yucel, G.; Demir, N.; Ozturk, S.; Elpek, G.O.; Savas, B.; Aslan, M. Effect of astaxanthin on hepatocellular injury following ischemia/reperfusion. Toxicology 2010, 267, 147–153. [Google Scholar] [CrossRef]
  45. Eum, H.A.; Lee, S.M. Effect of Trolox on altered vasoregulatory gene expression in hepatic ischemia/reperfusion. Arch. Pharm. Res. 2004, 27, 225–231. [Google Scholar] [CrossRef]
  46. Eum, H.A.; Lee, S.M. Effects of Trolox on the activity and gene expression of cytochrome P450 in hepatic ischemia/reperfusion. Br. J. Pharm. 2004, 142, 35–42. [Google Scholar] [CrossRef] [Green Version]
  47. Fernandez, V.; Tapia, G.; Varela, P.; Cornejo, P.; Videla, L.A. Upregulation of liver inducible nitric oxide synthase following thyroid hormone preconditioning: Suppression by N-acetylcysteine. Biol. Res. 2009, 42, 487–495. [Google Scholar] [CrossRef]
  48. Ferrigno, A.P.; Pasqua, L.G.D.G.; Berardo, C.; Richelmi, P.; Cadamuro, M.; Fabris, L.; Perlini, S.; Adorini, L.; Vairetti, M. Obeticholic acid reduces biliary and hepatic matrix metalloproteinases activity in rat hepatic ischemia/reperfusion injury. PLoS ONE 2020, 15, e0238543. [Google Scholar] [CrossRef]
  49. Ferrigno, A.; Di Pasqua, L.G.; Palladini, G.; Berardo, C.; Verta, R.; Richelmi, P.; Perlini, S.; Collotta, D.; Collino, M.; Vairetti, M. Transient expression of reck under hepatic ischemia/reperfusion conditions is associated with mapk signaling pathways. Biomolecules 2020, 10, 747. [Google Scholar] [CrossRef]
  50. Hataji, K.; Watanabe, T.; Oowada, S.; Nagaya, M.; Kamibayashi, M.; Murakami, E.; Kawakami, H.; Ishiuchi, A.; Kumai, T.; Nakano, H.; et al. Effects of a calcium-channel blocker (CV159) on hepatic ischemia/reperfusion injury in rats: Evaluation with selective NO/pO2 electrodes and an electron paramagnetic resonance spin-trapping method. Biol. Pharm. Bull. 2010, 33, 77–83. [Google Scholar] [CrossRef] [Green Version]
  51. Hsu, C.M.; Wang, J.S.; Liu, C.H.; Chen, L.W. Kupffer cells protect liver from ischemia-reperfusion injury by an inducible nitric oxide synthase-dependent mechanism. Shock 2002, 17, 280–285. [Google Scholar] [CrossRef] [PubMed]
  52. Kang, J.W.; Koh, E.J.; Lee, S.M. Melatonin protects liver against ischemia and reperfusion injury through inhibition of toll-like receptor signaling pathway. J. Pineal. Res. 2011, 50, 403–411. [Google Scholar] [CrossRef] [PubMed]
  53. Kim, S.J.; Lee, S.M. Effect of baicalin on toll-like receptor 4-mediated ischemia/reperfusion inflammatory responses in alcoholic fatty liver condition. Toxicol. Appl. Pharmacol. 2012, 258, 43–50. [Google Scholar] [CrossRef] [PubMed]
  54. Kim, S.J.; Moon, Y.J.; Lee, S.M. Protective effects of baicalin against ischemia/reperfusion injury in rat liver. J. Nat. Prod. 2010, 73, 2003–2008. [Google Scholar] [CrossRef]
  55. Kim, Y.H.; Lee, S.M. Role of Kupffer cells in the vasoregulatory gene expression during hepatic ischemia/reperfusion. Arch. Pharmacal. Res. 2004, 27, 111–117. [Google Scholar] [CrossRef]
  56. Kurabayashi, M.; Takeyoshi, I.; Yoshinari, D.; Koibuchi, Y.; Ohki, T.; Matsumoto, K.; Morishita, Y. NO donor ameliorates ischemia-reperfusion injury of the rat liver with iNOS attenuation. J. Investig. Surg. 2005, 18, 193–200. [Google Scholar] [CrossRef]
  57. Lee, C.H.; Kim, S.H.; Lee, S.M. Effect of pyrrolidine dithiocarbamate on hepatic vascular stress gene expression during ischemia and reperfusion. Eur. J. Pharm. 2008, 595, 100–107. [Google Scholar] [CrossRef]
  58. Man, K.; Ng, K.T.; Lee, T.K.; Lo, C.M.; Sun, C.K.; Li, X.L.; Zhao, Y.; Ho, J.W.; Fan, S.T. FTY720 attenuates hepatic ischemia-reperfusion injury in normal and cirrhotic livers. Am. J. Transpl. 2005, 5, 40–49. [Google Scholar] [CrossRef]
  59. Park, S.W.; Choi, S.M.; Lee, S.M. Effect of melatonin on altered expression of vasoregulatory genes during hepatic ischemia/reperfusion. Arch. Pharmacal. Res. 2007, 30, 1619–1624. [Google Scholar] [CrossRef]
  60. Ramalho, L.N.Z.; Pasta, A.A.C.; Terra, V.A.; Augusto, M.J.; Sanches, S.C.; Souza-Neto, F.P.; Cecchini, R.; Gulin, F.; Ramalho, F.S. Rosmarinic acid attenuates hepatic ischemia and reperfusion injury in rats. Food Chem. Toxicol. 2014, 74, 270–278. [Google Scholar] [CrossRef]
  61. Ren, Y.; Wang, L.H.; Deng, F.S.; Li, J.S.; Jiang, L. Protective effect and mechanism of alpha-lipoic acid on partial hepatic ischemia-reperfusion injury in adult male rats. Physiol. Res. 2019, 68, 739–745. [Google Scholar] [CrossRef]
  62. Sonin, N.V.; Garcia-Pagan, J.C.; Nakanishi, K.; Zhang, J.X.; Clemens, M.G. Patterns of vasoregulatory gene expression in the liver response to ischemia/reperfusion and endotoxemia. Shock 1999, 11, 175–179. [Google Scholar] [CrossRef]
  63. Tao, X.; Wan, X.; Xu, Y.; Xu, L.; Qi, Y.; Yin, L.; Han, X.; Lin, Y.; Peng, J. Dioscin attenuates hepatic ischemia-reperfusion injury in rats through inhibition of oxidative-nitrative stress, inflammation and apoptosis. Transplantation 2014, 98, 604–611. [Google Scholar] [CrossRef]
  64. Trocha, M.; Merwid-Lad, A.; Chlebda-Sieragowska, E.; Szuba, A.; Piesniewska, M.; Fereniec-Golebiewska, L.; Kwiatkowska, J.; Szelag, A.; Sozanski, T. Age-related changes in ADMA-DDAH-NO pathway in rat liver subjected to partial ischemia followed by global reperfusion. Exp. Gerontol. 2014, 50, 45–51. [Google Scholar] [CrossRef]
  65. Unal, B.; Ozcan, F.; Tuzcu, H.; Kirac, E.; Elpek, G.O.; Aslan, M. Inhibition of neutral sphingomyelinase decreases elevated levels of nitrative and oxidative stress markers in liver ischemia-reperfusion injury. Redox. Rep. 2017, 22, 147–159. [Google Scholar] [CrossRef] [Green Version]
  66. Yang, S.L.; Lou, Y.J. Sodium nitroprusside decreased leukotriene C4 generation by inhibiting leukotriene C4 synthase expression and activity in hepatic ischemia-reperfusion injured rats. Biochem. Pharmacol. 2007, 73, 724–735. [Google Scholar] [CrossRef]
  67. Yun, N.; Kang, J.W.; Lee, S.M. Protective effects of chlorogenic acid against ischemia/reperfusion injury in rat liver: Molecular evidence of its antioxidant and anti-inflammatory properties. J. Nutr. Biochem. 2012, 23, 1249–1255. [Google Scholar] [CrossRef]
  68. Yun, N.; Eum, H.A.; Lee, S.M. Protective role of heme oxygenase-1 against liver damage caused by hepatic ischemia and reperfusion in rats. Antioxid. Redox. Signal 2010, 13, 1503–1512. [Google Scholar] [CrossRef]
  69. Bektas, S.; Karakaya, K.; Can, M.; Bahadir, B.; Guven, B.; Erdogan, N.; Ozdamar, S.O. The effects of tadalafil and pentoxifylline on apoptosis and nitric oxide synthase in liver ischemia/reperfusion injury. Kaohsiung J. Med. Sci. 2016, 32, 339–347. [Google Scholar] [CrossRef] [Green Version]
  70. Grezzana-Filho, T.J.M.L.; Santos, J.L.L.; Gabiatti, G.; Boffi, C.; Santos, E.B.; Cerski, C.T.S.; Chedid, M.F.; Corso, C.O. Induction of selective liver hypothermia prevents significant ischemia/reperfusion injury in wistar rats after 24 hours. Acta Cir. Bras. 2020, 35, 1–12. [Google Scholar] [CrossRef]
  71. Kim, S.H.; Lee, S.M. Expression of hepatic vascular stress genes following ischemia/reperfusion and subsequent endotoxemia. Arch. Pharm. Res. 2004, 27, 769–775. [Google Scholar] [CrossRef]
  72. Kuncewitch, M.; Yang, W.L.; Molmenti, E.; Nicastro, J.; Coppa, G.F.; Wang, P. Wnt agonist attenuates liver injury and improves survival after hepatic ischemia/reperfusion. Shock 2013, 39, 3–10. [Google Scholar] [CrossRef] [Green Version]
  73. Lin, H.I.; Wang, D.; Leu, F.J.; Chen, C.F.; Chen, H.I. Ischemia and reperfusion of liver induces eNOS and iNOS expression: Effects of a NO donor and NOS inhibitor. Chin. J. Physiol. 2004, 47, 121–127. [Google Scholar]
  74. Longo, L.; Sinigaglia-Fratta, L.X.; Weber, G.R.; Janz-Moreira, A.; Kretzmann, N.A.; Grezzana-Filho Tde, J.; Possa-Marroni, N.; Corso, C.O.; Schmidt-Cerski, C.T.; Reverbel-da-Silveira, T.; et al. Hypothermia is better than ischemic preconditioning for preventing early hepatic ischemia/reperfusion in rats. Ann. Hepatol. 2016, 15, 110–120. [Google Scholar] [CrossRef]
  75. Takamatsu, Y.; Shimada, K.; Yamaguchi, K.; Kuroki, S.; Chijiiwa, K.; Tanaka, M. Inhibition of inducible nitric oxide synthase prevents hepatic, but not pulmonary, injury following ischemia-reperfusion of rat liver. Dig. Dis. Sci. 2006, 51, 571–579. [Google Scholar] [CrossRef]
  76. Yao, X.M.; Chen, H.; Li, Y. Protective effect of bicyclol on liver injury induced by hepatic warm ischemia/reperfusion in rats. Hepatol. Res. 2009, 39, 833–842. [Google Scholar] [CrossRef]
  77. Yun, N.; Kim, S.H.; Lee, S.M. Differential consequences of protein kinase C activation during early and late hepatic ischemic preconditioning. J. Physiol. Sci. 2012, 62, 199–209. [Google Scholar] [CrossRef]
  78. Wang, Y.; Lawson, J.A.; Jaeschke, H. Differential effect of 2-aminoethyl-isothiourea, an inhibitor of the inducible nitric oxide synthase, on microvascular blood flow and organ injury in models of hepatic ischemia-reperfusion and endotoxemia. Shock 1998, 10, 20–25. [Google Scholar] [CrossRef]
  79. Kireev, R.A.; Cuesta, S.; Ibarrola, C.; Bela, T.; Gonzalez, E.M.; Vara, E.; Tresguerres, J.A.F. Age-related differences in hepatic ischemia/reperfusion: Gene activation, liver injury, and protective effect of melatonin. J. Surg. Res. 2012, 178, 922–934. [Google Scholar] [CrossRef]
  80. Kireev, R.; Bitoun, S.; Cuesta, S.; Tejerina, A.; Ibarrola, C.; Moreno, E.; Vara, E.; Tresguerres, J.A. Melatonin treatment protects liver of Zucker rats after ischemia/reperfusion by diminishing oxidative stress and apoptosis. Eur. J. Pharm. 2013, 701, 185–193. [Google Scholar] [CrossRef]
  81. Duan, Y.F.; An, Y.; Zhu, F.; Jiang, Y. Remote ischemic preconditioning protects liver ischemia-reperfusion injury by regulating eNOS-NO pathway and liver microRNA expressions in fatty liver rats. Hepatobiliary Pancreat Dis. Int. 2017, 16, 387–394. [Google Scholar] [CrossRef]
  82. Trocha, M.; Merwid-Lad, A.; Szuba, A.; Chlebda, E.; Piesniewska, M.; Sozanski, T.; Szelag, A. Effect of simvastatin on nitric oxide synthases (eNOS, iNOS) and arginine and its derivatives (ADMA, SDMA) in ischemia/reperfusion injury in rat liver. Pharm. Rep. 2010, 62, 343–351. [Google Scholar] [CrossRef]
  83. Hara, Y.; Teramoto, K.; Kumashiro, Y.; Sato, E.; Nakamura, N.; Takatsu, S.; Kawamura, T.; Arii, S. Beneficial effect of tetrahydrobiopterin on the survival of rats exposed to hepatic ischemia-reperfusion injury. Transpl. Proc. 2005, 37, 442–444. [Google Scholar] [CrossRef] [PubMed]
  84. Ishizaki, M.; Kaibori, M.; Uchida, Y.; Tanaka, H.; Ozaki, T.; Saito, T.; Matsui, K.; Kamiyama, Y.; Nishizawa, M.; Okumura, T. Protective effect of FR183998, a Na+/H+ exchanger inhibitor, and its inhibition of inducible nitric oxide synthase induction in hepatic ischemia-reperfusion injury in rats. Am. J. Transplant. 2008, 8, 496. [Google Scholar]
  85. Hsieh, C.C.; Hsu, S.M.; Hwang, L.S.; Chiu, J.H.; Lu, W.C.; Wu, Y.L.; Hsieh, S.C. Protective effects of the extract from longan flower against hepatic ischemia/reperfusion injury in rats. J. Funct. Foods 2015, 15, 570–579. [Google Scholar] [CrossRef]
  86. Acquaviva, R.; Lanteri, R.; Li Destri, G.; Caltabiano, R.; Vanella, L.; Lanzafame, S.; Di Cataldo, A.; Li Volti, G.; Di Giacomo, C. Beneficial effects of rutin and L-arginine coadministration in a rat model of liver ischemia-reperfusion injury. Am. J. Physiol. Gastrointest. Liver Physiol. 2009, 296, G664–G670. [Google Scholar] [CrossRef] [PubMed]
  87. Chen, T.H.; Liao, F.T.; Yang, Y.C.; Wang, J.J. Inhibition of inducible nitric oxide synthesis ameliorates liver ischemia and reperfusion injury induced transient increase in arterial stiffness. Transpl. Proc. 2014, 46, 1112–1116. [Google Scholar] [CrossRef] [PubMed]
  88. Lanteri, R.; Acquaviva, R.; Di Giacomo, C.; Sorrenti, V.; Li Destri, G.; Santangelo, M.; Vanella, L.; Di Cataldo, A. Rutin in rat liver ischemia/reperfusion injury: Effect on DDAH/NOS pathway. Microsurgery 2007, 27, 245–251. [Google Scholar] [CrossRef]
  89. Morisue, A.; Wakabayashi, G.; Shimazu, M.; Tanabe, M.; Mukai, M.; Matsumoto, K.; Kawachi, S.; Yoshida, M.; Yamamoto, S.; Kitajima, M. The role of nitric oxide after a short period of liver ischemia-reperfusion. J. Surg. Res. 2003, 109, 101–109. [Google Scholar] [CrossRef]
  90. Nii, A.; Utsunomiya, T.; Shimada, M.; Ikegami, T.; Ishibashi, H.; Imura, S.; Morine, Y.; Ikemoto, T.; Sasaki, H.; Kawashima, A. Hydrolyzed whey peptide-based diet ameliorates hepatic ischemia-reperfusion injury in the rat nonalcoholic fatty liver. Surg. Today 2014, 44, 2354–2360. [Google Scholar] [CrossRef]
  91. Uchinami, H.; Yamamoto, Y.; Kume, M.; Yonezawa, K.; Ishikawa, Y.; Taura, K.; Nakajima, A.; Hata, K.; Yamaoka, Y. Effect of heat shock preconditioning on NF-kappaB/I-kappaB pathway during I/R injury of the rat liver. Am. J. Physiol. Gastrointest. Liver Physiol. 2002, 282, G962–G971. [Google Scholar] [CrossRef]
  92. Atef, Y.; El-Fayoumi, H.M.; Abdel-Mottaleb, Y.; Mahmoud, M.F. Effect of cardamonin on hepatic ischemia reperfusion induced in rats: Role of nitric oxide. Eur. J. Pharmacol. 2017, 815, 446–453. [Google Scholar] [CrossRef]
  93. El-Shitany, N.A.; El-Desoky, K. Cromoglycate, not ketotifen, ameliorated the injured effect of warm ischemia/reperfusion in rat liver: Role of mast cell degranulation, oxidative stress, proinflammatory cytokine, and inducible nitric oxide synthase. Drug Des. Dev. Ther. 2015, 9, 5237–5246. [Google Scholar] [CrossRef] [Green Version]
  94. Ibrahim, S.G.; El-Emam, S.Z.; Mohamed, E.A.; Abd Ellah, M.F. Dimethyl fumarate and curcumin attenuate hepatic ischemia/reperfusion injury via Nrf2/HO-1 activation and anti-inflammatory properties. Int. Immunopharmacol. 2020, 80, 106131. [Google Scholar] [CrossRef]
  95. Ibrahim, M.A.; Abdel-Gaber, S.A.; Amin, E.F.; Ibrahim, S.A.; Mohammed, R.K.; Abdelrahman, A.M. Molecular mechanisms contributing to the protective effect of levosimendan in liver ischemia-reperfusion injury. Eur. J. Pharm. 2014, 741, 64–73. [Google Scholar] [CrossRef]
  96. Sankary, H.N.; Yin, D.P.; Chong, A.S.F.; Ma, L.L.; Blinder, L.; Shen, J.K.; Foster, P.; Liu, L.P.; Li, C.; Williams, J.W. The portosystemic shunt protects liver against ischemic reperfusion injury. Transplantation 1999, 68, 958–963. [Google Scholar] [CrossRef]
  97. Sehitoglu, M.H.; Karaboga, I.; Kiraz, A.; Kiraz, H.A. The hepatoprotective effect of Aloe vera on ischemia-reperfusion injury in rats. N. Clin. Istanb. 2019, 6, 203–209. [Google Scholar]
  98. Yaylak, F.; Canbaz, H.; Caglikulekci, M.; Dirlik, M.; Tamer, L.; Ogetman, Z.; Polat, Y.; Kanik, A.; Aydin, S. Liver tissue inducible nitric oxide synthase (iNOS) expression and lipid peroxidation in experimental hepatic ischemia reperfusion injury stimulated with lipopolysaccharide: The role of aminoguanidine. J. Surg. Res. 2008, 148, 214–223. [Google Scholar] [CrossRef]
  99. Rodríguez-Reynoso, S.; Leal, C.; Portilla, E.; Olivares, N.; Muñiz, J. Effect of exogenous melatonin on hepatic energetic status during ischemia/reperfusion: Possible role of tumor necrosis factor-α and nitric oxide. J. Surg. Res. 2001, 100, 141–149. [Google Scholar] [CrossRef]
  100. Yang, S.L.; Chen, L.J.; Kong, Y.; Xu, D.; Lou, Y.J. Sodium nitroprusside regulates mRNA expressions of LTC4 synthesis enzymes in hepatic ischemia/reperfusion injury rats via NF-kappaB signaling pathway. Pharmacology 2007, 80, 11–20. [Google Scholar] [CrossRef]
  101. Harada, N.; Iimuro, Y.; Nitta, T.; Yoshida, M.; Uchinami, H.; Nishio, T.; Hatano, E.; Yamamoto, N.; Yamamoto, Y.; Yamaoka, Y. Inactivation of the small GTPase Rac1 protects the liver from ischemia/reperfusion injury in the rat. Surgery 2003, 134, 480–491. [Google Scholar] [CrossRef] [Green Version]
  102. Suetsugu, H.; Iimuro, Y.; Uehara, T.; Nishio, T.; Harada, N.; Yoshida, M.; Hatano, E.; Son, G.; Fujimoto, J.; Yamaoka, Y. Nuclear factor kappa B inactivation in the rat liver ameliorates short term total warm ischaemia/reperfusion injury. Gut 2005, 54, 835–842. [Google Scholar] [CrossRef] [Green Version]
  103. Abdel-Gaber, S.A.; Ibrahim, M.A.; Amin, E.F.; Ibrahim, S.A.; Mohammed, R.K.; Abdelrahman, A.M. Effect of selective versus non-selective cyclooxygenase inhibitors on ischemia-reperfusion-induced hepatic injury in rats. Life Sci. 2015, 134, 42–48. [Google Scholar] [CrossRef]
  104. Rodriguez-Reynos, S.; Leal-Cortes, C.; Portilla-de Buen, E.; Lopez-De la Torre, S.P. Ischemic Preconditioning Preserves Liver Energy Charge and Function on Hepatic Ischemia/Reperfusion Injury in Rats. Arch. Med. Res. 2018, 49, 373–380. [Google Scholar] [CrossRef]
  105. Xue, F.; Zhang, J.J.; Xu, L.M.; Zhang, C.; Xia, Q. Protective effects of HGF-MSP chimer (metron factor-1) on liver ischemia-reperfusion injury in rat model. J. Dig. Dis. 2010, 11, 299–305. [Google Scholar] [CrossRef]
  106. Wang, Y.; Wong, G.T.; Man, K.; Irwin, M.G. Pretreatment with intrathecal or intravenous morphine attenuates hepatic ischaemia-reperfusion injury in normal and cirrhotic rat liver. Br. J. Anaesth. 2012, 109, 529–539. [Google Scholar] [CrossRef] [Green Version]
  107. Björnsson, B.; Bojmar, L.; Olsson, H.; Sundqvist, T.; Sandström, P. Nitrite, a novel method to decrease ischemia/reperfusion injury in the rat liver. World J. Gastroenterol. 2015, 21, 1775–1783. [Google Scholar] [CrossRef]
  108. Björnsson, B.; Winbladh, A.; Bojmar, L.; Sundqvist, T.; Gullstrand, P.; Sandström, P. Conventional, but not remote ischemic preconditioning, reduces iNOS transcription in liver ischemia/reperfusion. World J. Gastroenterol. 2014, 20, 9506–9512. [Google Scholar] [CrossRef] [PubMed]
  109. Liang, R.; Nickkholgh, A.; Hoffmann, K.; Kern, M.; Schneider, H.; Sobirey, M.; Zorn, M.; Buchler, M.W.; Schemmer, P. Melatonin protects from hepatic reperfusion injury through inhibition of IKK and JNK pathways and modification of cell proliferation. J. Pineal Res. 2009, 46, 8–14. [Google Scholar] [CrossRef] [PubMed]
  110. Iwasaki, J.; Afify, M.; Bleilevens, C.; Klinge, U.; Weiskirchen, R.; Steitz, J.; Vogt, M.; Yagi, S.; Nagai, K.; Uemoto, S.; et al. The Impact of a Nitric Oxide Synthase Inhibitor (L-NAME) on Ischemia(-)Reperfusion Injury of Cholestatic Livers by Pringle Maneuver and Liver Resection after Bile Duct Ligation in Rats. Int. J. Mol. Sci. 2019, 20, 2114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Shen, S.Q.; Zhang, Y.; Xiang, J.J.; Xiong, C.L. Protective effect of curcumin against liver warm ischemia/reperfusion injury in rat model is associated with regulation of heat shock protein and antioxidant enzymes. World J. Gastroenterol. 2007, 13, 1953–1961. [Google Scholar] [CrossRef] [Green Version]
  112. Shen, S.Q.; Zhang, Y.; Xiong, C.L. The protective effects of 17beta-estradiol on hepatic ischemia-reperfusion injury in rat model, associated with regulation of heat-shock protein expression. J. Surg. Res. 2007, 140, 67–76. [Google Scholar] [CrossRef]
  113. El-Gohary, O.A. Obestatin improves hepatic injury induced by ischemia/reperfusion in rats: Role of nitric oxide. Gen. Physiol. Biophys. 2017, 36, 109–115. [Google Scholar] [CrossRef]
  114. Zhang, T.; Ma, Y.; Xu, K.Q.; Huang, W.Q. Pretreatment of parecoxib attenuates hepatic ischemia/reperfusion injury in rats. BMC Anesth. 2015, 15, 165. [Google Scholar] [CrossRef] [Green Version]
  115. Duval, H.; Mbatchi, S.F.; Grandadam, S.; Legendre, C.; Loyer, P.; Ribault, C.; Piquet-Pellorce, C.; Guguen-Guillouzo, C.; Boudjema, K.; Corlu, A. Reperfusion stress induced during intermittent selective clamping accelerates rat liver regeneration through JNK pathway. J. Hepatol. 2010, 52, 560–569. [Google Scholar] [CrossRef]
  116. Kawai, K.; Yokoyama, Y.; Kokuryo, T.; Watanabe, K.; Kitagawa, T.; Nagino, M. Inchinkoto, an herbal medicine, exerts beneficial effects in the rat liver under stress with hepatic ischemia-reperfusion and subsequent hepatectomy. Ann. Surg. 2010, 251, 692–700. [Google Scholar] [CrossRef]
  117. Bae, U.J.; Yang, J.D.; Ka, S.O.; Koo, J.H.; Woo, S.J.; Lee, Y.R.; Yu, H.C.; Cho, B.H.; Zhao, H.Y.; Ryu, J.H.; et al. SPA0355 attenuates ischemia/reperfusion-induced liver injury in mice. Exp. Mol. Med. 2014, 46, e109. [Google Scholar] [CrossRef]
  118. Datta, G.; Luong, T.V.; Fuller, B.J.; Davidson, B.R. Endothelial nitric oxide synthase and heme oxygenase-1 act independently in liver ischemic preconditioning. J. Surg. Res. 2014, 186, 417–428. [Google Scholar] [CrossRef]
  119. Hines, I.N.; Harada, H.; Bharwani, S.; Pavlick, K.P.; Hoffman, J.M.; Grisham, M.B. Enhanced post-ischemic liver injury in iNOS-deficient mice: A cautionary note. Biochem. Biophys. Res. Commun. 2001, 284, 972–976. [Google Scholar] [CrossRef]
  120. Hines, I.N.; Kawachi, S.; Harada, H.; Pavlick, K.P.; Hoffman, J.M.; Bharwani, S.; Wolf, R.E.; Grisham, M.B. Role of nitric oxide in liver ischemia and reperfusion injury. Mol. Cell Biochem. 2002, 234, 229–237. [Google Scholar] [CrossRef]
  121. Kawachi, S.; Hines, I.N.; Laroux, F.S.; Hoffman, J.; Bharwani, S.; Gray, L.; Leffer, D.; Grisham, M.B. Nitric oxide synthase and postischemic liver injury. Biochem. Biophys. Res. Commun. 2000, 276, 851–854. [Google Scholar] [CrossRef]
  122. Lee, V.G.; Johnson, M.L.; Baust, J.; Laubach, V.E.; Watkins, S.C.; Billiar, T.R. The roles of iNOS in liver ischemia-reperfusion injury. Shock 2001, 16, 355–360. [Google Scholar] [CrossRef]
  123. Chen, Z.; Ding, T.; Ma, C.G. Dexmedetomidine (DEX) protects against hepatic ischemia/reperfusion (I/R) injury by suppressing inflammation and oxidative stress in NLRC5 deficient mice. Biochem. Biophys. Res. Commun. 2017, 493, 1143–1150. [Google Scholar] [CrossRef]
  124. Gao, L.; Chen, X.; Peng, T.; Yang, D.; Wang, Q.; Lv, Z.; Shen, J. Caveolin-1 protects against hepatic ischemia/reperfusion injury through ameliorating peroxynitrite-mediated cell death. Free Radic. Biol. Med. 2016, 95, 209–215. [Google Scholar] [CrossRef]
  125. Guo, J.Y.; Yang, T.; Sun, X.G.; Zhou, N.Y.; Li, F.S.; Long, D.; Lin, T.; Li, P.Y.; Feng, L. Ischemic postconditioning attenuates liver warm ischemia-reperfusion injury through Akt-eNOS-NO-HIF pathway. J. Biomed. Sci. 2011, 18, 79. [Google Scholar] [CrossRef] [Green Version]
  126. Guo, Y.; Yang, T.; Lu, J.; Li, S.; Wan, L.; Long, D.; Li, Q.; Feng, L.; Li, Y. Rb1 postconditioning attenuates liver warm ischemia-reperfusion injury through ROS-NO-HIF pathway. Life. Sci. 2011, 88, 598–605. [Google Scholar] [CrossRef]
  127. Jeyabalan, G.; Klune, J.R.; Nakao, A.; Martik, N.; Wu, G.; Tsung, A.; Geller, D.A. Arginase blockade protects against hepatic damage in warm ischemia-reperfusion. Nitric. Oxide 2008, 19, 29–35. [Google Scholar] [CrossRef]
  128. Kim, H.J.; Joe, Y.; Yu, J.K.; Chen, Y.; Jeong, S.O.; Mani, N.; Cho, G.J.; Pae, H.O.; Ryter, S.W.; Chung, H.T. Carbon monoxide protects against hepatic ischemia/reperfusion injury by modulating the miR-34a/SIRT1 pathway. Biochim. Biophys. Acta 2015, 1852, 1550–1559. [Google Scholar] [CrossRef] [Green Version]
  129. Klune, J.R.; Dhupar, R.; Kimura, S.; Ueki, S.; Cardinal, J.; Nakao, A.; Nace, G.; Evankovich, J.; Murase, N.; Tsung, A.; et al. Interferon regulatory factor-2 is protective against hepatic ischemia-reperfusion injury. Am. J. Physiol.-Gastrointest. Liver Physiol. 2012, 303, G666–G673. [Google Scholar] [CrossRef] [PubMed]
  130. Luedde, T.; Assmus, U.; Wüstefeld, T.; Meyer zu Vilsendorf, A.; Roskams, T.; Schmidt-Supprian, M.; Rajewsky, K.; Brenner, D.A.; Manns, M.P.; Pasparakis, M.; et al. Deletion of IKK2 in hepatocytes does not sensitize these cells to TNF-induced apoptosis but protects from ischemia/reperfusion injury. J. Clin. Investig. 2005, 115, 849–859. [Google Scholar] [CrossRef] [Green Version]
  131. Moon, K.H.; Hood, B.L.; Mukhopadhyay, P.; Rajesh, M.; Abdelmegeed, M.A.; Kwon, Y.I.; Conrads, T.P.; Veenstra, T.D.; Song, B.J.; Pacher, P. Oxidative inactivation of key mitochondrial proteins leads to dysfunction and injury in hepatic ischemia reperfusion. Gastroenterology 2008, 135, 1344–1357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Mukhopadhyay, P.; Rajesh, M.; Horvath, B.; Batkai, S.; Park, O.; Tanchian, G.; Gao, R.Y.; Patel, V.; Wink, D.A.; Liaudet, L.; et al. Cannabidiol protects against hepatic ischemia/reperfusion injury by attenuating inflammatory signaling and response, oxidative/nitrative stress, and cell death. Free Radic Biol. Med. 2011, 50, 1368–1381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Qiao, Y.; Xiao, F.; Li, W.; Yu, M.; Du, P.; Fang, Z.; Sun, J. Hepatocellular HO-1 mediated iNOS-induced hepatoprotection against liver ischemia reperfusion injury. Biochem. Biophys. Res. Commun. 2020, 521, 1095–1100. [Google Scholar] [CrossRef]
  134. Qiao, Y.; Zhang, X.; Zhao, G.; Liu, Z.; Yu, M.; Fang, Z.; Li, X. Hepatocellular iNOS protects liver from ischemia/reperfusion injury through HSF1-dependent activation of HSP70. Biochem. Biophys. Res. Commun. 2019, 512, 882–888. [Google Scholar] [CrossRef]
  135. Sanches, S.C.; Ramalho, L.N.; Mendes-Braz, M.; Terra, V.A.; Cecchini, R.; Augusto, M.J.; Ramalho, F.S. Riboflavin (vitamin B-2) reduces hepatocellular injury following liver ischaemia and reperfusion in mice. Food Chem. Toxicol. 2014, 67, 65–71. [Google Scholar] [CrossRef]
  136. Shaker, M.E.; Trawick, B.N.; Mehal, W.Z. The novel TLR9 antagonist COV08-0064 protects from ischemia/reperfusion injury in non-steatotic and steatotic mice livers. Biochem. Pharm. 2016, 112, 90–101. [Google Scholar] [CrossRef]
  137. Shi, Y.R.H.; Ramshesh, V.K.; Schwartz, J.; Liu, Q.; Krishnasamy, Y.; Zhang, X.; Lemasters, J.J.; Smith, C.D.; Zhong, Z. Sphingosine kinase-2 inhibition improves mitochondrial function and survival after hepatic ischemia-reperfusion. J. Hepatol. 2012, 56, 137–145. [Google Scholar] [CrossRef]
  138. Tsung, A.; Stang, M.T.; Ikeda, A.; Critchlow, N.D.; Izuishi, K.; Nakao, A.; Chan, M.H.; Jeyabalan, G.; Yim, J.H.; Geller, D.A. The transcription factor interferon regulatory factor-1 mediates liver damage during ischemia-reperfusion injury. Am. J. Physiol. Gastrointest. Liver Physiol. 2006, 290, G1261–G1268. [Google Scholar] [CrossRef] [Green Version]
  139. Tsurui, Y.; Sho, M.; Kuzumoto, Y.; Hamada, K.; Akashi, S.; Kashizuka, H.; Ikeda, N.; Nomi, T.; Mizuno, T.; Kanehiro, H.; et al. Dual role of vascular endothelial growth factor in hepatic ischemia-reperfusion injury. Transplantation 2005, 79, 1110–1115. [Google Scholar] [CrossRef]
  140. Zhao, G.Y.; Fu, C.; Wang, L.; Zhu, L.; Yan, Y.T.; Xiang, Y.; Zheng, F.; Gong, F.L.; Chen, S.; Chen, G. Down-regulation of nuclear HMGB1 reduces ischemia-induced HMGB1 translocation and release and protects against liver ischemia-reperfusion injury. Sci. Rep. 2017, 7, 1–11. [Google Scholar] [CrossRef] [Green Version]
  141. Ajamieh, H.; Farrell, G.C.; McCuskey, R.S.; Yu, J.; Chu, E.; Wong, H.J.; Lam, W.; Teoh, N.C. Acute atorvastatin is hepatoprotective against ischaemia-reperfusion injury in mice by modulating eNOS and microparticle formation. Liver Int. 2015, 35, 2174–2186. [Google Scholar] [CrossRef]
  142. Duarte, S.; Hamada, T.; Kuriyama, N.; Busuttil, R.W.; Coito, A.J. TIMP-1 deficiency leads to lethal partial hepatic ischemia and reperfusion injury. Hepatology 2012, 56, 1074–1085. [Google Scholar] [CrossRef] [Green Version]
  143. Freitas, M.C.; Uchida, Y.; Zhao, D.; Ke, B.; Busuttil, R.W.; Kupiec-Weglinski, J.W. Blockade of Janus kinase-2 signaling ameliorates mouse liver damage due to ischemia and reperfusion. Liver Transpl. 2010, 16, 600–610. [Google Scholar] [CrossRef] [Green Version]
  144. Hamada, T.; Duarte, S.; Tsuchihashi, S.; Busuttil, R.W.; Coito, A.J. Inducible nitric oxide synthase deficiency impairs matrix metalloproteinase-9 activity and disrupts leukocyte migration in hepatic ischemia/reperfusion injury. Am. J. Pathol. 2009, 174, 2265–2277. [Google Scholar] [CrossRef] [Green Version]
  145. Lee, H.M.; Jang, H.J.; Kim, S.S.; Kim, H.J.; Lee, S.Y.; Oh, M.Y.; Kwan, H.C.; Jang, D.S.; Eom, D.W. Protective Effect of Eupatilin Pretreatment Against Hepatic Ischemia-Reperfusion Injury in Mice. Transpl. Proc. 2016, 48, 1226–1233. [Google Scholar] [CrossRef]
  146. Okaya, T.; Lentsch, A.B. Peroxisome proliferator-activated receptor-α regulates postischemic liver injury. Am. J. Physiol.-Gastrointest. Liver Physiol. 2004, 286, G606–G612. [Google Scholar] [CrossRef] [Green Version]
  147. Zhou, H.M.; Sun, J.; Zhong, W.Z.; Pan, X.X.; Liu, C.M.; Cheng, F.; Wang, P.; Rao, Z.Q. Dexmedetomidine preconditioning alleviated murine liver ischemia and reperfusion injury by promoting macrophage M2 activation via PPAR gamma/STAT3 signaling. Int. Immunopharmacol. 2020, 82, 106363. [Google Scholar] [CrossRef]
  148. Tao, J.; Shen, X.H.; Ai, Y.H.; Han, X.J. Tea polyphenols protect against ischemia/reperfusion-induced liver injury in mice through anti-oxidative and anti-apoptotic properties. Exp. Ther. Med. 2016, 12, 3433–3439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Patouraux, S.; Rousseau, D.; Rubio, A.; Bonnafous, S.; Lavallard, V.J.; Lauron, J.; Saint-Paul, M.C.; Bailly-Maitre, B.; Tran, A.; Crenesse, D.; et al. Osteopontin deficiency aggravates hepatic injury induced by ischemia-reperfusion in mice. Cell Death Dis. 2014, 5, e1208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Kobayashi, H.P.; Watanabe, T.; Oowada, S.; Hirayama, A.; Nagase, S.; Kamibayashi, M.; Otsubo, T. Effect of CV159-Ca(2+)/calmodulin blockade on redox status hepatic ischemia-reperfusion injury in mice evaluated by a newly developed in vivo EPR imaging technique. J. Surg. Res. 2008, 147, 41–49. [Google Scholar] [CrossRef] [PubMed]
  151. Jessup, J.M.; Samara, R.; Battle, P.; Laguinge, L.M. Carcinoembryonic antigen promotes tumor cell survival in liver through an IL-10-dependent pathway. Clin. Exp. Metastasis 2005, 21, 709–717. [Google Scholar] [CrossRef]
  152. Godwin, A.; Yang, W.L.; Sharma, A.; Khader, A.; Wang, Z.; Zhang, F.; Nicastro, J.; Coppa, G.F.; Wang, P. Blocking cold-inducible RNA-binding protein (CIRP) protects liver from ischemia/reperfusion injury. Shock 2014, 43, 24–30. [Google Scholar] [CrossRef] [Green Version]
  153. Mendes-Braz, M.; Elias-Miró, M.; Jiménez-Castro, M.B.; Casillas-Ramírez, A.; Ramalho, F.S.; Peralta, C. The current state of knowledge of hepatic ischemia-reperfusion injury based on its study in experimental models. J. Biomed. Biotechnol. 2012, 2012, 298657. [Google Scholar] [CrossRef] [Green Version]
  154. Man, K.; Fan, S.T.; Ng, I.O.; Lo, C.M.; Liu, C.L.; Yu, W.C.; Wong, J. Tolerance of the liver to intermittent pringle maneuver in hepatectomy for liver tumors. Arch. Surg. 1999, 134, 533–539. [Google Scholar] [CrossRef] [Green Version]
  155. Peralta, C.; Bartrons, R.; Riera, L.; Manzano, A.; Xaus, C.; Gelpí, E.; Roselló-Catafau, J. Hepatic preconditioning preserves energy metabolism during sustained ischemia. Am. J. Physiol. Gastrointest. Liver Physiol. 2000, 279, G163–G171. [Google Scholar] [CrossRef] [Green Version]
  156. Liu, S.; Li, X.; Li, H.; Guo, L.; Zhang, B.; Gong, Z.; Zhang, J.; Ye, Q. Longer duration of the Pringle maneuver is associated with hepatocellular carcinoma recurrence following curative resection. J. Surg. Oncol. 2016, 114, 112–118. [Google Scholar] [CrossRef]
  157. Nakatake, R.; Tsuda, T.; Matsuura, T.; Miki, H.; Hishikawa, H.; Matsushima, H.; Ishizaki, M.; Matsui, K.; Kaibori, M.; Nishizawa, M.; et al. Genipin Inhibits the Induction of Inducible Nitric Oxide Synthase Through the Inhibition of NF-κB Activation in Rat Hepatocytes. Drug. Metab. Lett. 2017, 10, 254–263. [Google Scholar] [CrossRef]
  158. Butcher, R.L.; Collins, W.E.; Fugo, N.W. Plasma concentration of LH, FSH, prolactin, progesterone and estradiol-17beta throughout the 4-day estrous cycle of the rat. Endocrinology 1974, 94, 1704–1708. [Google Scholar] [CrossRef]
  159. Yokoyama, Y.; Nagino, M.; Nimura, Y. Which gender is better positioned in the process of liver surgery? Male or female? Surg. Today 2007, 37, 823–830. [Google Scholar] [CrossRef]
  160. Harada, H.; Pavlick, K.P.; Hines, I.N.; Hoffman, J.M.; Bharwani, S.; Gray, L.; Wolf, R.E.; Grisham, M.B. Selected contribution: Effects of gender on reduced-size liver ischemia and reperfusion injury. J. Appl. Physiol. 2001, 91, 2816–2822. [Google Scholar] [CrossRef]
  161. Eckhoff, D.E.; Bilbao, G.; Frenette, L.; Thompson, J.A.; Contreras, J.L. 17-Beta-estradiol protects the liver against warm ischemia/reperfusion injury and is associated with increased serum nitric oxide and decreased tumor necrosis factor-alpha. Surgery 2002, 132, 302–309. [Google Scholar] [CrossRef] [PubMed]
  162. Vegeto, E.; Belcredito, S.; Etteri, S.; Ghisletti, S.; Brusadelli, A.; Meda, C.; Krust, A.; Dupont, S.; Ciana, P.; Chambon, P.; et al. Estrogen receptor-alpha mediates the brain antiinflammatory activity of estradiol. Proc. Natl. Acad. Sci. USA 2003, 100, 9614–9619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Vegeto, E.; Bonincontro, C.; Pollio, G.; Sala, A.; Viappiani, S.; Nardi, F.; Brusadelli, A.; Viviani, B.; Ciana, P.; Maggi, A. Estrogen prevents the lipopolysaccharide-induced inflammatory response in microglia. J. Neurosci. 2001, 21, 1809–1818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Arnal, J.F.; Clamens, S.; Pechet, C.; Negre-Salvayre, A.; Allera, C.; Girolami, J.P.; Salvayre, R.; Bayard, F. Ethinylestradiol does not enhance the expression of nitric oxide synthase in bovine endothelial cells but increases the release of bioactive nitric oxide by inhibiting superoxide anion production. Proc. Natl. Acad. Sci. USA 1996, 93, 4108–4113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Burra, P. Liver abnormalities and endocrine diseases. Best Pr. Res. Clin. Gastroenterol. 2013, 27, 553–563. [Google Scholar] [CrossRef] [PubMed]
  166. Germani, G.; Zeni, N.; Zanetto, A.; Adam, R.; Karam, V.; Belli, L.S.; O’Grady, J.; Mirza, D.; Klempnauer, J.; Cherqui, D.; et al. Influence of donor and recipient gender on liver transplantation outcomes in Europe. Liver Int. 2020, 40, 1961–1971. [Google Scholar] [CrossRef]
  167. Tang, J.; Cao, Y.; Rose, R.L.; Brimfield, A.A.; Dai, D.; Goldstein, J.A.; Hodgson, E. Metabolism of chlorpyrifos by human cytochrome P450 isoforms and human, mouse, and rat liver microsomes. Drug Metab. Dispos. 2001, 29, 1201–1204. [Google Scholar]
  168. Löfgren, S.; Hagbjörk, A.L.; Ekman, S.; Fransson-Steen, R.; Terelius, Y. Metabolism of human cytochrome P450 marker substrates in mouse: A strain and gender comparison. Xenobiotica 2004, 34, 811–834. [Google Scholar] [CrossRef]
  169. Yoon, D.Y.; Mansukhani, N.A.; Stubbs, V.C.; Helenowski, I.B.; Woodruff, T.K.; Kibbe, M.R. Sex bias exists in basic science and translational surgical research. Surgery 2014, 156, 508–516. [Google Scholar] [CrossRef]
  170. Kleinert, H.; Schwarz, P.M.; Förstermann, U. Regulation of the expression of inducible nitric oxide synthase. Biol. Chem. 2003, 384, 1343–1364. [Google Scholar] [CrossRef]
  171. Sharma, J.N.; Al-Omran, A.; Parvathy, S.S. Role of nitric oxide in inflammatory diseases. Inflammopharmacology 2007, 15, 252–259. [Google Scholar] [CrossRef]
  172. Kröncke, K.D.; Fehsel, K.; Kolb-Bachofen, V. Inducible nitric oxide synthase in human diseases. Clin. Exp. Immunol. 1998, 113, 147–156. [Google Scholar] [CrossRef]
  173. Kim, P.K.; Zamora, R.; Petrosko, P.; Billiar, T.R. The regulatory role of nitric oxide in apoptosis. Int. Immunopharmacol. 2001, 1, 1421–1441. [Google Scholar] [CrossRef]
  174. Datta, G.; Fuller, B.J.; Davidson, B.R. Molecular mechanisms of liver ischemia reperfusion injury: Insights from transgenic knockout models. World J. Gastroenterol. 2013, 19, 1683–1698. [Google Scholar] [CrossRef]
  175. Sugawara, Y.; Kubota, K.; Ogura, T.; Esumi, H.; Inoue, K.; Takayama, T.; Makuuchi, M. Increased nitric oxide production in the liver in the perioperative period of partial hepatectomy with Pringle’s maneuver. J. Hepatol. 1998, 28, 212–220. [Google Scholar] [CrossRef]
  176. Abu-Amara, M.; Yang, S.Y.; Seifalian, A.; Davidson, B.; Fuller, B. The nitric oxide pathway—Evidence and mechanisms for protection against liver ischaemia reperfusion injury. Liver Int. 2012, 32, 531–543. [Google Scholar] [CrossRef]
  177. Shah, V.; Chen, A.F.; Cao, S.; Hendrickson, H.; Weiler, D.; Smith, L.; Yao, J.; Katusic, Z.S. Gene transfer of recombinant endothelial nitric oxide synthase to liver in vivo and in vitro. Am. J Physiol. Gastrointest. Liver Physiol. 2000, 279, G1023–G1030. [Google Scholar] [CrossRef]
  178. Yamashita, T.; Kawashima, S.; Ohashi, Y.; Ozaki, M.; Rikitake, Y.; Inoue, N.; Hirata, K.; Akita, H.; Yokoyama, M. Mechanisms of reduced nitric oxide/cGMP-mediated vasorelaxation in transgenic mice overexpressing endothelial nitric oxide synthase. Hypertension 2000, 36, 97–102. [Google Scholar] [CrossRef]
  179. Behar-Cohen, F.F.; Heydolph, S.; Faure, V.; Droy-Lefaix, M.T.; Courtois, Y.; Goureau, O. Peroxynitrite cytotoxicity on bovine retinal pigmented epithelial cells in culture. Biochem. Biophys. Res. Commun. 1996, 226, 842–849. [Google Scholar] [CrossRef]
  180. Fan, C.; Zwacka, R.M.; Engelhardt, J.F. Therapeutic approaches for ischemia/reperfusion injury in the liver. J. Mol. Med. 1999, 77, 577–592. [Google Scholar] [CrossRef]
  181. Cinelli, M.A.; Do, H.T.; Miley, G.P.; Silverman, R.B. Inducible nitric oxide synthase: Regulation, structure, and inhibition. Med. Res. Rev. 2020, 40, 158–189. [Google Scholar] [CrossRef] [PubMed]
  182. Víteček, J.; Lojek, A.; Valacchi, G.; Kubala, L. Arginine-based inhibitors of nitric oxide synthase: Therapeutic potential and challenges. Mediat. Inflamm. 2012, 2012, 318087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Kobayashi, H.; Nonami, T.; Kurokawa, T.; Takeuchi, Y.; Harada, A.; Nakao, A.; Takagi, H. Role of endogenous nitric oxide in ischemia-reperfusion injury in rat liver. J. Surg. Res. 1995, 59, 772–779. [Google Scholar] [CrossRef] [PubMed]
  184. Jiao, L.R.; Ayav, A.; Navarra, G.; Sommerville, C.; Pai, M.; Damrah, O.; Khorsandi, S.; Habib, N.A. Laparoscopic liver resection assisted by the laparoscopic Habib Sealer. Surgery 2008, 144, 770–774. [Google Scholar] [CrossRef] [PubMed]
  185. Efanov, M.; Kazakov, I.; Alikhanov, R.; Vankovich, A.; Koroleva, A.; Kovalenko, D.; Salimgereeva, D.; Tsvirkun, V.; Khatkov, I. A randomized prospective study of the immediate outcomes of the use of a hydro-jet dissector and an ultrasonic surgical aspirator for laparoscopic liver resection. HPB 2021, 23, 1332–1338. [Google Scholar] [CrossRef] [PubMed]
  186. Rau, H.G.; Wichmann, M.W.; Schinkel, S.; Buttler, E.; Pickelmann, S.; Schauer, R.; Schildberg, F.W. Surgical techniques in hepatic resections: Ultrasonic aspirator versus Jet-Cutter. A prospective randomized clinical trial. Zent. Chir. 2001, 126, 586–590. [Google Scholar] [CrossRef] [PubMed]
  187. Kleinert, R.; Wahba, R.; Bangard, C.; Prenzel, K.; Hölscher, A.H.; Stippel, D. Radiomorphology of the Habib sealer-induced resection plane during long-time followup: A longitudinal single center experience after 64 radiofrequency-assisted liver resections. HPB Surg. 2010, 2010, 403097. [Google Scholar] [CrossRef]
  188. Yang, Y.; Peng, Y.; Chen, K.; Wei, Y.; Li, B.; Liu, F. Laparoscopic liver resection with “ultrasonic scalpel mimic CUSA” technique. Surg. Endosc. 2022, 1–8. [Google Scholar] [CrossRef]
  189. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS. Med. 2009, 6, e1000097. [Google Scholar] [CrossRef] [Green Version]
  190. Hooijmans, C.R.; Rovers, M.M.; de Vries, R.B.; Leenaars, M.; Ritskes-Hoitinga, M.; Langendam, M.W. SYRCLE’s risk of bias tool for animal studies. BMC Med. Res. Methodol. 2014, 14, 43. [Google Scholar] [CrossRef] [Green Version]
  191. Higgins, J.P.; Thompson, S.G.; Deeks, J.J.; Altman, D.G. Measuring inconsistency in meta-analyses. BMJ 2003, 327, 557–560. [Google Scholar] [CrossRef]
Figure 1. Flow chart of literature selection.
Figure 1. Flow chart of literature selection.
Ijms 23 11916 g001
Figure 2. Distribution of reperfusion times compared with their ischaemia times in 70% ischaemia models in rats. Studies with various reperfusion times are listed for each individual time point. Time points are listed regardless of the survival time of animals.
Figure 2. Distribution of reperfusion times compared with their ischaemia times in 70% ischaemia models in rats. Studies with various reperfusion times are listed for each individual time point. Time points are listed regardless of the survival time of animals.
Ijms 23 11916 g002
Figure 3. Distribution of reperfusion time compared with their ischaemia times in 100% ischaemia models in rats. Studies with various reperfusion times are listed for each individual time point. Time points are listed regardless of the survival time of animals.
Figure 3. Distribution of reperfusion time compared with their ischaemia times in 100% ischaemia models in rats. Studies with various reperfusion times are listed for each individual time point. Time points are listed regardless of the survival time of animals.
Ijms 23 11916 g003
Figure 4. Distribution of reperfusion times compared with their ischaemia times in 70% ischaemia models in mice. Studies with various reperfusion times are listed for each individual time point. Time points are listed regardless of the survival time of animals.
Figure 4. Distribution of reperfusion times compared with their ischaemia times in 70% ischaemia models in mice. Studies with various reperfusion times are listed for each individual time point. Time points are listed regardless of the survival time of animals.
Ijms 23 11916 g004
Figure 5. Risk of bias assessment for the 125 studies included.
Figure 5. Risk of bias assessment for the 125 studies included.
Ijms 23 11916 g005
Figure 6. Detailed risk of bias assessment for all 10 signalling questions.
Figure 6. Detailed risk of bias assessment for all 10 signalling questions.
Ijms 23 11916 g006
Figure 7. Forest plot of 70% HIRI in rats and 30 min warm ischaemia time grouped by reperfusion time between 0.5 and 72 h.
Figure 7. Forest plot of 70% HIRI in rats and 30 min warm ischaemia time grouped by reperfusion time between 0.5 and 72 h.
Ijms 23 11916 g007
Figure 8. Forest plot of 70% HIRI in rats and 60 min warm ischaemia time grouped by reperfusion time between 1 and 24 h.
Figure 8. Forest plot of 70% HIRI in rats and 60 min warm ischaemia time grouped by reperfusion time between 1 and 24 h.
Ijms 23 11916 g008
Figure 9. Forest plot of 70% HIRI in rats and 90 min warm ischaemia time grouped by reperfusion time between 2 and 24 h.
Figure 9. Forest plot of 70% HIRI in rats and 90 min warm ischaemia time grouped by reperfusion time between 2 and 24 h.
Ijms 23 11916 g009
Figure 10. Forest plot of 100% HIRI in rats and 45 min warm ischaemia time grouped by reperfusion time between 0.75 and 24 h.
Figure 10. Forest plot of 100% HIRI in rats and 45 min warm ischaemia time grouped by reperfusion time between 0.75 and 24 h.
Ijms 23 11916 g010
Figure 11. Forest plot of 70% HIRI in mice and 60 min warm ischaemia time grouped by reperfusion time between 1 and 6 h.
Figure 11. Forest plot of 70% HIRI in mice and 60 min warm ischaemia time grouped by reperfusion time between 1 and 6 h.
Ijms 23 11916 g011
Figure 12. Forest plot of NO detection in SHAM groups versus HIRI grouped by different warm ischaemia durations.
Figure 12. Forest plot of NO detection in SHAM groups versus HIRI grouped by different warm ischaemia durations.
Ijms 23 11916 g012
Figure 13. Forest plot of eNOS detection in SHAM versus HIRI grouped by different warm ischaemia durations.
Figure 13. Forest plot of eNOS detection in SHAM versus HIRI grouped by different warm ischaemia durations.
Ijms 23 11916 g013
Figure 14. Search terms and their combination used for PubMed and Embase.
Figure 14. Search terms and their combination used for PubMed and Embase.
Ijms 23 11916 g014
Table 6. Methods of iNOS detection and number of methods valuable for meta-analysis.
Table 6. Methods of iNOS detection and number of methods valuable for meta-analysis.
MethodN° of Used MethodN° Used for Meta-Analysis% of Used Methods
ELISA732.2
IHC2486.0
RNA611813.4
Spectrometry532.2
WB3786.0
Total1344029.8
ELISA, Enzyme-linked immunosorbent assay; IHC, immune histochemistry; RNA, ribonuclein acid; SP, Spectrophotometric analysis; WB, Western blot.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Nakatake, R.; Schulz, M.; Kalvelage, C.; Benstoem, C.; Tolba, R.H. Effects of iNOS in Hepatic Warm Ischaemia and Reperfusion Models in Mice and Rats: A Systematic Review and Meta-Analysis. Int. J. Mol. Sci. 2022, 23, 11916. https://doi.org/10.3390/ijms231911916

AMA Style

Nakatake R, Schulz M, Kalvelage C, Benstoem C, Tolba RH. Effects of iNOS in Hepatic Warm Ischaemia and Reperfusion Models in Mice and Rats: A Systematic Review and Meta-Analysis. International Journal of Molecular Sciences. 2022; 23(19):11916. https://doi.org/10.3390/ijms231911916

Chicago/Turabian Style

Nakatake, Richi, Mareike Schulz, Christina Kalvelage, Carina Benstoem, and René H. Tolba. 2022. "Effects of iNOS in Hepatic Warm Ischaemia and Reperfusion Models in Mice and Rats: A Systematic Review and Meta-Analysis" International Journal of Molecular Sciences 23, no. 19: 11916. https://doi.org/10.3390/ijms231911916

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop