Open Access

Hepatic recruitment of CD11b+Ly6C+ inflammatory monocytes promotes hepatic ischemia/reperfusion injury

  • Authors:
    • Peng Song
    • Junbin Zhang
    • Yunwei Zhang
    • Zhiping Shu
    • Peng Xu
    • Long He
    • Chao Yang
    • Jinxiang Zhang
    • Hui Wang
    • Yiqing Li
    • Qin Li
  • View Affiliations

  • Published online on: December 8, 2017     https://doi.org/10.3892/ijmm.2017.3315
  • Pages: 935-945
  • Copyright: © Song et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Monocytes infiltrate damaged liver tissue during noninfectious liver injury and often have dual roles, perpetuating inflammation and promoting resolution of inflammation and fibrosis. However, how monocyte subsets distribute and are differentially recruited in the liver remain unclear. In the current study, the subpopulations of infiltrating monocytes were examined following liver ischemia/reperfusion (I/R) injury in mice using flow cytometry. CD11b+Ly6C high (Ly6Chi) cells (inflammatory monocytes) and CD11b+Ly6C low cells (reparative monocytes) were recruited into the liver following I/R injury. Treatment with clodronate‑loaded liposomes, which transiently deplete systemic macrophages, alleviated hepatic damage. Mice genetically deficient in C‑C motif chemokine ligand 2 (CCL2), or its receptor C‑C chemokine receptor 2 (CCR2), exhibited diminished hepatic damage compared with wild‑type mice following I/R, by controlling intrahepatic inflammatory Ly6Chi monocyte accumulation. In addition, the CCR2 specific inhibitor RS504393 alleviated hepatic I/R injury. The results suggest that the CCR2/CCL2 axis has an important role in monocyte infiltration and may represent a novel target for the treatment of liver I/R injury.

Introduction

Liver ischemia/reperfusion (I/R) injury is an important cause of liver damage occurring during liver surgery or transplantation, significantly influences the prognosis of liver function (1). A variety of immune cells, including monocytes, Kupffer cells, CD4+ lymphocytes, neutrophils, hepatocytes, and cytokines are involved in liver I/R injury (24). Hepatic macrophages have an important role in maintaining homeostasis of the liver and in the pathogenesis of liver injury. I/R can cause the activation of Kupffer cells to release transforming growth factor-α, and other pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). An increase in the inflammatory state results in the activation of the immune system as a major intermediary during the process of I/R injury (5), which can drive injury resolution or lead to chronic inflammation (6,7).

Intrahepatic monocyte infiltration of the damaged liver tissue represents an important constitution of the innate immune response (8). When the liver is under pathological stress, such as steatohepatitis and viral hepatitis, Kupffer cells can differentiate from infiltrated bone marrow-derived mononuclear cells (9). In addition, it has been reported that Kupffer cells can be constantly replenished by blood monocytes, even in steady state conditions (10). Two major monocyte subsets in mice have been identified that closely resemble human monocytes and vary in migratory and differentiation properties (11). In humans, classical CD14+CD16+ monocytes express C-C chemokine receptor 2 (CCR2), CD64, and selectin L, whereas non-classical CD14+CD16 monocytes lack CCR2 (12). Their counterparts in mice are CCR2+Ly6C high (Ly6Chi) and CCR2Ly6C low (Ly6Clo) monocytes, respectively (12,13). Ly6Chi monocytes are considered to be precursors to macrophages and dendritic cells during inflammatory conditions, whereas Ly6Clo monocytes represent steady state precursor cells for tissue macrophages (14-16). The differential recruitment of these monocyte subsets appears to be crucially controlled by chemokines released from injured tissue (17-19). As such, enhanced hepatic expression of the ligands for CCR2, including monocyte chemoattractant protein-1 (MCP-1)/C-C motif chemokine ligand 2 (CCL2), CCR6 and macrophage inflammatory protein-3α/CCL20, has been reported in patients with liver cirrhosis (17,20).

CCR2 is the specific receptor for MCP-1/CCL2, but is also the receptor for CCL7, CCL8, CCL11, CCL12 and CCL13. Recently, CCL2 has been reported to have a critical role in the pathogenesis of various liver diseases (21-23). CCL2 interacts with its receptor, CCR2, and induces mononuclear cells in the blood to cross the endothelial barrier towards sites of inflammation during an inflammatory response (24). Upregulated expression of CCL2 has been reported in various liver diseases (21,25). Although the CCR2/CCL2 interaction has been reported to be associated with various liver diseases (26), the underlying mechanism of this association has not been clearly elucidated. Specifically, the role of CCL2/CCR2 in the immune response of the liver during I/R injury remains unknown. The current study aimed to identify the infiltrating monocyte subsets in liver I/R injury and determine how the CCL2/CCR2 axis contributes to the hepatic monocyte recruitment during I/R injury.

Materials and methods

Animals

Male C57BL/6 mice [wild-type (WT); weight, 22–25 g] were purchased from the Center for Animal Experiment of Wuhan University (Wuhan, China). Male CCL2−/− or CCR2−/− C57BL/6 mice were bred at the sterile Animal Management Centre of Tongji Medical College (Wuhan, China) and used between the ages of 8–12 weeks. The original experiments used 10 mice/group [including hematoxylin and eosin (H&E) staining, serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), mRNA/protein levels of TNF-α and IL-6 in liver tissue; recruitment of monocytes into I/R injured liver lobes; effect of macrophage depletion as well as CCR2 or CCL2 knockout on liver I/R injury]. For the additional experiments performed (quantification of intrahepatic CD4+ T cells and CD8+ T cells; immunofluorescence staining of hepatic infiltrated monocyte subsets; effect of CCR2 inhibition on hepatic I/R injury), 4-6 animals were used per group. All animal studies were performed in accordance with the National Institutes of Health Guidelines for Care and Use of Laboratory Animals and were approved by the Scientific Affairs Committee on Animal Research and Ethics of Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (Wuhan, China).

Liver I/R model

A non-lethal model of segmental (70%) hepatic warm ischemia was performed as described previously (27). Briefly, mice were anesthetized with sodium pentobarbital [60 mg/kg; intraperitoneally (i.p.)], which is a traditional anesthetic used in animal research of liver, lung or cardiac I/R injury (2830). A midline laparotomy was performed and ligaments surrounding each lobe were dissected carefully. The portal vein, the hepatic artery, and the bile duct supplying the median and the left lateral lobes of the liver were clamped with an artery clip. The caudal and right lobes retained portal and arterial inflow and venous outflow, preventing intestinal venous congestion. Following surgery, the abdominal cavity was properly closed and then the mice were placed in an incubator maintained at 32°C. Liver lobes were inspected for ischemia by visualizing the pale blanching of the ischemic lobes intermittently. After 60 min, the clamps were removed and liver reperfusion was initiated. For sham controls, the animals underwent anesthesia, laparotomy, and exposure of the portal triad without hepatic I/R. Mice were sacrificed after 6 h of reperfusion. Blood and tissues of liver lobes from sham and I/R injury mice were collected and stored at −80°C for future analysis.

Serum transaminase

Cardiac puncture blood was collected following hepatic I/R or sham into tubes (without anticoagulant), allowed to clot and serum was separated by centrifugation at 200 x g at 4°C for 10 min. For CCR2 inhibitor pretreatment, mice were injected with RS-504393 (25 mg/kg; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) or dimethyl sulfoxide (DMSO) 1 h prior to I/R injury. Liver injury was estimated by measuring the increased activities of serum ALT and AST, which were measured using a Synchron CX7 analyzer (Beckman Coulter, Inc., Brea, CA, USA) in the Clinical Biochemical Laboratory of Wuhan Union Hospital (Wuhan, China).

Histopathology

At predetermined time points (6 h after reperfusion), mice were sacrificed humanely by CO2 suffocation. The liver lobes were collected from I/R mice and sham mice and fixed in 4% formalin at room temperature (25°C) for 4 h. Fixed tissues were embedded in paraffin, cut into 5 µm thick sections and were placed onto glass slides. Slides were then stained with hematoxylin and eosin (H&E) using a conventional protocol. The necrotic area was assessed by 2 independent pathologists, and more than 10 random fields of view were quantified using the Image-Pro Plus v 6.0 software (Media Cybernetics, Silver Spring, MD, USA).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from the lobes of the ischemic and sham liver using TRIzol. RT was performed using the RT kit (Thermo Fisher Scientific, Inc., Waltham, MA, USA) at 65°C for 5 min. qPCR was performed using the SYBR-Green PCR mix (Thermo Fisher Scientific, Inc.) in a LightCycler 480 system (Roche Molecular Diagnostics, Pleasanton, CA, USA). The PCR program was 95°C, 2 min, ×1; 95°C, 10 sec, 60°C, 1 min, ×40. β-actin was used as an internal reference. Many previous I/R studies (31,32) also used this gene for data normalization, indicating I/R has no effect on the expression of β-actin. Primers were synthesized by Sangon Biotech Co., Ltd. (Shanghai, China) and the sequences were as follows: TNF-α, 5′-ACT GAA CTT CGG GGT GATCG-3′ (forward) and 5′-GGC TAC AGG CTT GTC ACTCG-3′ (reverse); IL-6, 5′-AGT TGC CTT CTT GGG ACTGA-3′ (forward) and 5′-TCC ACG ATT TCC CAG AGAAC-3′ (reverse); β-actin, 5′-GTG ACG TTG ACA TCC GTA AAGA-3′ (forward) and 5′-GCC GGA CTC ATC GTA CTCC-3′ (reverse). Data were analyzed using the 2−ΔΔCq method (33).

Serum/liver cytokines

The serum expression levels of TNF-α and IL-6 was measured using commercial ELISA kits [TNF-α (MTA00B); IL-6 (M6000B); R&D Systems, Inc., Minneapolis, MN, USA] following standardized techniques (34). All samples and standards were performed in duplicate. ELISA was also performed on tissues of liver lobes from sham and I/R mice.

Isolation of hepatic non-parenchymal cells (NPCs)

Hepatic NPCs were obtained from the liver using a collagenase digestion method, as described previously (35). Briefly, the animals were anesthetized and the liver was perfused in situ by way of the portal vein with PBS containing 0.05% collagenase. The liver was removed, placed in PBS and incubated at 37°C for 15 min. The incubated liver was then torn using cell scrapers. The cell suspension were shaken on a shaking table at 37°C for 20 min and then filtered through a 70-µm nylon mesh. NPCs were isolated by gradient centrifugation at 400 x g for 16 min with acceleration and braking at 0 using OptiPrep™ (Axis-Shield Diagnostics Ltd., Dundee, UK) according to manufacturer's instructions. NPCs were then washed by high-speed centrifugation (1,500 rpm for 5 min).

Depletion of Kupffer cells

Kupffer cells were depleted by using clodronate in a liposomal formulation (36,37). Briefly, a dose of 0.2 ml/20 g animal body weight of liposome-encapsulated clodronate (FormuMax Scientific, Inc., Sunnyvale, CA, USA) was i.p. injected 48 h before I/R. Macrophage depletion was successfully achieved (~90%), as examined by flow cytometry (anti-F4/80-PerCP-Cy5.5, clone BM8 (cat. no. 123128); from BioLegend, Inc., San Diego, CA, USA) in the liver 48 h after a single injection.

Flow cytometric analysis

Red blood cells in the isolated NPCs were lysed using Hybri-Max red blood cell lysis buffer (Sigma-Aldrich; Merck KGaA). Cell numbers were determined by a sequential gating scheme. The cells were then incubated with fluorescent-labeled anti-mouse antibodies in PBS containing 1% bovine serum albumin for 30 min at 4°C. Antibodies used in this experiment were as follows: anti-CD45-Pacific Blue (clone 30-F11; cat. no. 103126), anti-Ly6CAPC-Cy7 (clone HK1.4; cat. no. 128026), anti-CD11b-PE-Cy7 (clone M1/70; cat. no. 101216), and anti-CD4-PerCP-Cy5.5 (clone GK1.5; cat. no. 100434) from BioLegend; anti-CD8-APC (clone 53-6.7; cat. no. 561093) from BD Biosciences (San Diego, CA, USA); anti-CD3-PE-Cy7 (clone 17A2; cat. no. 100220) from BioLegend, Inc. For intracellular cytokine staining, NPCs were isolated following sham or I/R injury of the 3 groups of mice (n=4–6 mice/group) and cultured with GolgiStop (cat. no. 554724; BD Biosciences) for 6 h. The cells were then stained with CD11b, Ly6C and anti-TNF-α-PE (IC410P) from R&D Systems. The corresponding isotype IgGs were used when necessary as controls. Following staining, cells were washed, fixed in 1% para-formaldehyde in 1X PBS, and resuspended at ~5×106/ml for flow cytometry. Cells were analyzed on a CyAn ADP analyzer (Beckman Coulter, Inc.). FlowJo version 7.6 software (Tree Star, Inc., Ashland, OR, USA) was used to analyze the data (38).

Data analysis

The results were expressed as the mean ± standard error. Student's t-test was used to compare the difference between two groups. For multigroup comparison, one-way analysis of variance was used followed by post hoc Mann Whitney U-test. All of the data analysis was performed using SPSS 15.0 software (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Hepatic pathology in I/R injured liver

Compared with sham animals with normal liver histology, mice undergoing I/R injury exhibited periportal necrosis after 6 h, as determined by H&E staining (Fig. 1A). I/R-induced liver injury was also indicated by elevated serum levels of ALT and AST (Fig. 1B). The levels of inflammatory cytokines TNF-α and IL-6 were significantly higher in liver tissues of I/R mice than in sham animals at the mRNA (Fig. 1C) and protein (Fig. 1D) levels. For the positive control, mice were treated with CCl4 (0.3 ml/kg) and elevated TNF-α and IL-6 in liver tissue were observed (data not shown).

Ly6 high (Ly6Chi) and low (Ly6Clo)monocytes are increased in I/R-injured livers

To investigate the cells that secrete the cytokines demonstrated, the population of intrahepatic leukocytes present in the damaged liver was characterized. The flow cytometric plots of CD11b and Ly6C expression in isolated hepatic NPCs from one sham mouse and one I/R mouse, representative of the 10 mice in each group, are presented in Fig. 2. The population of CD11b+Ly6Chi cells (inflammatory monocytes) increased following I/R injury, from 2.1 to 6.9% of CD45+ immune cells. CD11b+Ly6Clo cells (reparative monocytes) also increased from 9.21 to 20.6% of CD45+ immune cells following hepatic I/R injury (Fig. 2). Statistical analysis was then calculated based on data from all animals of each group and there was significant difference in CD11b+Ly6Chi/low populations between the sham group and I/R group. These results suggest that the increased infiltration of monocytes into liver may have an important role in hepatic pathology in I/R-injured liver.

Treatment with clodronate-loaded liposomes alleviates liver I/R injury

To determine whether monocyte-derived subsets actively promote liver damage or represent an elemental reaction following I/R injury, WT mice were treated with clodronate-loaded liposomes to transiently deplete systemic macrophages 48 h before I/R injury (39). As presented in Fig. 3A, the population of CD11b+Ly6Chi cells in I/R model mice decreased from 7.1 to 0.18% following pretreatment with clodronate-loaded liposomes. CD11b+Ly6Clo cells also decreased from 20.4 to 2.9% in macrophage-depleted mice (Fig. 3A). Following macrophage depletion by clodronate-loaded liposomes, I/R-induced hepatic necrosis was also dramatically decreased (Fig. 3B). Serum ALT and AST levels were also reduced in mice pretreated with clodronate-loaded liposome compared to animals pretreated with PBS (Fig. 3C). These results suggest that the recruitment of monocyte subsets into the injured liver may contribute to acute hepatic damage.

CCR2/CCL2 axis is critical for the recruitment of Ly6C+ intrahepatic monocyte-derived cells

Ly6C+ monocytes express high levels of CCR2, CCR1 and CCR6 (39,40). To elucidate the role of CCR2-CCL2 axis in the recruitment of monocytes to damaged liver tissue, I/R-induced liver injury and intrahepatic recruitment of Ly6C+ monocytes were examined in WT, CCR2−/− and CCL2−/− mice. Compared with WT mice that exhibited significant hepatic necrosis, CCR2−/− and CCL2−/− mice exhibited lower levels of necrosis following I/R injury (Fig. 4A). Serum ALT and AST levels were also decreased compared with the WT group (Fig. 4B). The levels of inflammatory cytokines, TNF-α and IL-6, were also decreased in the CCR2−/− and CCL2−/− mice (Fig. 4C and D). In addition, leukocyte infiltration was significantly reduced following injury in CCR2 or CCL2 deficient mice (Fig. 5A). There was a significantly lower accumulation of intrahepatic CD11b+Ly6C+ monocytes in CCR2−/− and CCL2−/− mice undergoing I/R. The percentage of CD11b+Ly6Chi inflammatory monocytes and CD11b+Ly6Clo reparative monocytes were decreased in CCR2−/− and CCL2−/− mice compared with WT mice (Fig. 5A). However, there was no difference in the number of CD11b+Ly6Chi cells between CCR2−/− and CCL2−/− sham group and the operation group (Fig. 5). Flow cytometric quantification also demonstrated that CCR2 or CCL2 knockout decreased the accumulation of CD4+ T cells and CD8+ T cells in mice that underwent hepatic I/R injury (Fig. 5B). The decrease in CD11b+Ly6C+ cells was also revealed by immunofluorescence staining of infiltrated monocyte subsets in injured liver tissue (Fig. 6A). Intracellular cytokine staining demonstrated that CCR2−/− or CCL2−/− mice had significantly lower levels of TNF-α than WT mice following I/R injury, whereas there was no induction of TNF-α in the three groups under sham conditions (Fig. 6B).

CCR2 inhibitor alleviates hepatic I/R injury

CCR2−/− or CCL2−/− mice exhibited decreased hepatic I/R injury compared with WT mice. CCR2 has a crucial role in the CCR2/CCL2 axis and certain CCR2 inhibitors have been reported to provide renal and glycemic benefits (41). It was tested whether a CCR2 inhibitor could alleviate hepatic I/R injury. When mice were pretreated with the specific CCR2 inhibitor, RS504393 (25 mg/kg), serum ALT and AST levels were significantly decreased, compared with the control group and vehicle group (Fig. 7A). The CCR2 inhibitor group also exhibited a decrease in the area of necrosis (Fig. 7B). In addition, inflammatory CD11b+Ly6Chi and reparative CD11b+Ly6Clo cells were markedly decreased following RS504393 treatment (Fig. 7C). Similar to the magnitude of inhibition in CCR2−/− deficient mice, CCR2 inhibition nearly entirely suppressed CD11b+Ly6Chi cells infiltration into the hepatic tissue after I/R injury. No difference in CD11b+Ly6Chi and CD11b+Ly6Clo cell populations was observed between the untreated control group and DMSO control group. These results demonstrate that CCR2 inhibition efficiently alleviated hepatic I/R injury and, thus, may be an effective potential drug for hepatic I/R injury.

Discussion

In the present study, the role of infiltrating monocyte subsets into the liver following liver I/R injury was investigated and demonstrated an important functional role for inflammatory CD11b+Ly6C+ monocytes in causing liver I/R injury. The CCR2/CCL2 axis has a critical role in mediating monocyte infiltration into the liver following I/R injury.

Immune cells, and the release of associated cytokines, have been reported to be involved in the occurrence of liver I/R injury, however the exact immunocyte populations have yet to be determined (42). Inflammatory cells also have a major role in liver repair and are recruited immediately following injury (43). CD11b+Ly6C+ and CD11b+Ly6C monocytes are the two most well-defined subsets of monocytes and have been proposed to infiltrate tissues during inflammation (44,45). The infiltration of CD11b+Ly6C+ monocyte subset has been demonstrated to initiate liver injury in infected mice and produced pathogenic TNF (46). By contrast, the accumulation of Ly6C monocytes/macrophages in the liver coincided with a drop in the pool of Ly6C+ monocytes and provided hepatoprotective function by secreting the anti-inflammatory cytokine IL-10 (46). The findings of the current study provided evidence of an important role for the recruitment of CD11b+Ly6C+ monocytes to the site of hepatic damage following liver I/R in mice. CD11b+Ly6Clo reparative monocytes are predominantly recruited following acute liver I/R injury, with a lesser extent to CD11b+Ly6Chi inflammatory monocytes. CD11b+Ly6Chi monocytes have been demonstrated to inhibit Th1 differentiation, but enhance development of regulatory T cells and exhibit immunosuppressive characteristics (14). The subsequent release of the inflammatory cytokines, TNF-α and IL-6, in the liver resulted in hepatocyte necrosis and elevated serum ALT and AST levels, which suggest that hepatic injury may be in part due to monocyte infiltration after I/R injury. Therefore, hepatic infiltration of CD11b+Ly6Chi monocytes may be a self-regulatory mechanism to control inflammation in I/R injury.

The effects of macrophages in monocyte intrahepatic recruitment most likely result from their capacity to express numerous growth factors and cytokines (47). Macrophage depletion can be achieved with the systemic injection of liposomes containing clodronate (37,48). In the current study, ~90% macrophage depletion was successfully achieved in the liver 48 h after a single injection of liposomes containing clodronate. Notably, the transient depletion of macrophages alleviated I/R-induced hepatic damage, coinciding with a ecrease in CD11b+Ly6C+ monocytes intrahepatic infiltration. This finding strongly suggests that macrophages contribute to the damage observed in the liver following I/R injury.

Accumulation of Ly6Chi monocytes in the injured liver is critically dependent on the chemokine receptor CCR2 and its ligand, CCL2 (49). Increasing experimental evidence suggests that CCR2 regulates Ly6Chi monocyte entry into inflamed tissue, albeit indirectly, by promoting the egress of Ly6Chi monocytes from the bone marrow into blood circulation (50). CCR2 critically controls intrahepatic Gr1hi monocyte accumulation by mediating their egress from bone marrow (49). The findings of the current study demonstrated that CCR2−/− and CCL2−/− mice had almost no excess intrahepatic infiltration of Ly6C+ monocytes, indicating a similar mechanism applies during liver I/R injury. In addition, upon organ injury, CCR2, CCL2, CCR8 and CCL1 have been previously demonstrated to promote the accumulation of the inflammatory Ly6C+Gr1+ monocyte subset as precursors of tissue macrophages in the liver (8). Furthermore, novel anti-CCL2 directed agents specifically blocked the infiltration of pro-inflammatory monocytes into injured murine liver. In the present study, specific CCR2 inhibitor RS504393 was used to pretreat mice prior to I/R injury and produced a magnitude of inhibition similar to that in CCR2−/− deficient mice. CCR2 inhibitor nearly completely suppressed the infiltration of CD11b+Ly6Chi cells into the hepatic tissue following I/R injury. These results suggest that CCR2 inhibition efficiently alleviated the hepatic I/R injury and thus, may be an effective potential drug for hepatic I/R injury. There is a limitation of the current study. Although the internal reference gene β-actin expressed at a high level, there are changes in the expression upon I/R treatment. Therefore, using an additional reference gene would strengthen the reliability and reproducibility of our study.

In conclusion, the current study defined intrahepatic monocyte-derived subsets in experimental murine liver I/R injury and identified that Ly6C+ monocytes have an important role in acute I/R hepatic damage. These findings are likely to be relevant for the treatment of liver disease. CCR2 inhibition alleviated the inflamed hepatic environment, suggesting a potential novel approach for interventions targeting pro-inflammatory actions of Ly6Chi monocytes in the liver following I/R injury. However, translation from animal model into human clinical studies requires further investigation.

Acknowledgments

This study was supported by grants from the National Natural Science Foundation of China (nos. 81070355 and 81570570). We also acknowledge Dr Yin Liu (College of Life Science, Wuhan University, Wuhan, China) for providing technical assistance with flow cytometry.

Abbreviations:

I/R injury

ischemia/reperfusion injury

CCR2

C-C chemokine receptor 2

CCL2

C-C motif chemokine ligand 2

Ly6Chi

Ly6C high

Ly6Clo

Ly6C low

ALT

alanine aminotransferase

AST

aspartate aminotransferase

NPCs

non-parenchymal cells

WT

wild-type

TNF-α

tumor necrosis factor-α

IL-6

interleukin-6

References

1 

Peralta C, Jiménez-Castro MB and Gracia-Sancho J: Hepatic ischemia and reperfusion injury: Effects on the liver sinusoidal milieu. J Hepatol. 59:1094–1106. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Kupiec-Weglinski JW and Busuttil RW: Ischemia and reperfusion injury in liver transplantation. Transplant Proc. 37:1653–1656. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Seki E, De Minicis S, Osterreicher CH, Kluwe J, Osawa Y, Brenner DA and Schwabe RF: TLR4 enhances TGF-beta signaling and hepatic fibrosis. Nat Med. 13:1324–1332. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Jaeschke H: Mechanisms of Liver Injury. II. Mechanisms of neutrophil-induced liver cell injury during hepatic ischemia-reperfusion and other acute inflammatory conditions. Am J Physiol Gastrointest Liver Physiol. 290:G1083–G1088. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Abu-Amara M, Yang SY, Tapuria N, Fuller B, Davidson B and Seifalian A: Liver ischemia/reperfusion injury: Processes in inflammatory networks - a review. Liver Transpl. 16:1016–1032. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Ju C and Tacke F: Hepatic macrophages in homeostasis and liver diseases: From pathogenesis to novel therapeutic strategies. Cell Mol Immunol. 13:316–327. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Wang F, Yin J, Lu Z, Zhang G, Li J, Xing T, Zhuang S and Wang N: Limb ischemic preconditioning protects against contrast-induced nephropathy via renalase. EBioMedicine. 9:356–365. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Tacke F: Functional role of intrahepatic monocyte subsets for the progression of liver inflammation and liver fibrosis in vivo. Fibrogenesis Tissue Repair. 5(Suppl 1): S272012. View Article : Google Scholar : PubMed/NCBI

9 

Dai LJ, Li HY, Guan LX, Ritchie G and Zhou JX: The therapeutic potential of bone marrow-derived mesenchymal stem cells on hepatic cirrhosis. Stem Cell Res. 2:16–25. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Klein I, Cornejo JC, Polakos NK, John B, Wuensch SA, Topham DJ, Pierce RH and Crispe IN: Kupffer cell heterogeneity: Functional properties of bone marrow derived and sessile hepatic macrophages. Blood. 110:4077–4085. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Tacke F and Randolph GJ: Migratory fate and differentiation of blood monocyte subsets. Immunobiology. 211:609–618. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Tacke F, Alvarez D, Kaplan TJ, Jakubzick C, Spanbroek R, Llodra J, Garin A, Liu J, Mack M, van Rooijen N, et al: Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J Clin Invest. 117:185–194. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Combadière C, Potteaux S, Rodero M, Simon T, Pezard A, Esposito B, Merval R, Proudfoot A, Tedgui A and Mallat Z: Combined inhibition of CCL2, CX3CR1, and CCR5 abrogates Ly6C(hi) and Ly6C(lo) monocytosis and almost abolishes atherosclerosis in hypercholesterolemic mice. Circulation. 117:1649–1657. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Ma H, Wan S and Xia CQ: Immunosuppressive CD11b+Ly6Chi monocytes in pristane-induced lupus mouse model. J Leukoc Biol. 99:1121–1129. 2016. View Article : Google Scholar

15 

Neal LM and Knoll LJ: Toxoplasma gondii profilin promotes recruitment of Ly6Chi CCR2+ inflammatory monocytes that can confer resistance to bacterial infection. PLoS Pathog. 10:e10042032014. View Article : Google Scholar

16 

Helk E, Bernin H, Ernst T, Ittrich H, Jacobs T, Heeren J, Tacke F, Tannich E and Lotter H: TNFα-mediated liver destruction by Kupffer cells and Ly6Chi monocytes during Entamoeba histolytica infection. PLoS Pathog. 9:e10030962013. View Article : Google Scholar

17 

Marra F, DeFranco R, Grappone C, Milani S, Pastacaldi S, Pinzani M, Romanelli RG, Laffi G and Gentilini P: Increased expression of monocyte chemotactic protein-1 during active hepatic fibrogenesis: Correlation with monocyte infiltration. Am J Pathol. 152:423–430. 1998.PubMed/NCBI

18 

Denney L, Kok WL, Cole SL, Sanderson S, McMichael AJ and HO LP: Activation of invariant NKT cells in early phase of experimental autoimmune encephalomyelitis results in differentiation of Ly6Chi inflammatory monocyte to M2 macrophages and improved outcome. J Immunol. 189:551–557. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Gibbons MA, MacKinnon AC, Ramachandran P, Dhaliwal K, Duffin R, Phythian-Adams AT, van Rooijen N, Haslett C, Howie SE, Simpson AJ, et al: Ly6Chi monocytes direct alternatively activated profibrotic macrophage regulation of lung fibrosis. Am J Respir Crit Care Med. 184:569–581. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Shimizu Y, Murata H, Kashii Y, Hirano K, Kunitani H, Higuchi K and Watanabe A: CC-chemokine receptor 6 and its ligand macrophage inflammatory protein 3alpha might be involved in the amplification of local necroinflammatory response in the liver. Hepatology. 34:311–319. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Baeck C, Wehr A, Karlmark KR, Heymann F, Vucur M, Gassler N, Huss S, Klussmann S, Eulberg D, Luedde T, et al: Pharmacological inhibition of the chemokine CCL2 (MCP-1) diminishes liver macrophage infiltration and steatohepatitis in chronic hepatic injury. Gut. 61:416–426. 2012. View Article : Google Scholar

22 

Haukeland JW, Damås JK, Konopski Z, Løberg EM, Haaland T, Goverud I, Torjesen PA, Birkeland K, Bjøro K and Aukrust P: Systemic inflammation in nonalcoholic fatty liver disease is characterized by elevated levels of CCL2. Hepatol. 44:1167–1174. 2006. View Article : Google Scholar

23 

Klueh U, Czajkowski C, Ludzinska I, Qiao Y, Frailey J and Kreutzer DL: Impact of CCL2 and CCR2 chemokine/receptor deficiencies on macrophage recruitment and continuous glucose monitoring in vivo. Biosens Bioelectron. 86:262–269. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Yadav A, Saini V and Arora S: MCP-1: chemoattractant with a role beyond immunity: a review. Clin Chim Acta. 411:1570–1579. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Zhao L, Lim SY, Gordon-Weeks AN, Tapmeier TT, Im JH, Cao Y, Beech J, Allen D, Smart S and Muschel RJ: Recruitment of a myeloid cell subset (CD11b/Gr1 mid) via CCL2/CCR2 promotes the development of colorectal cancer liver metastasis. Hepatology. 57:829–839. 2013. View Article : Google Scholar

26 

Braunersreuther V, Viviani GL, Mach F and Montecucco F: Role of cytokines and chemokines in non-alcoholic fatty liver disease. World J Gastroenterol. 18:727–735. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Castellaneta A, Yoshida O, Kimura S, Yokota S, Geller DA, Murase N and Thomson AW: Plasmacytoid dendritic cell-derived IFN-α promotes murine liver ischemia/reperfusion injury by induction of hepatocyte IRF-1. Hepatology. 60:267–277. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Le Page S, Niro M, Fauconnier J, Cellier L, Tamareille S, Gharib A, Chevrollier A, Loufrani L, Grenier C, Kamel R, et al: Increase in Cardiac Ischemia-Reperfusion Injuries in Opa1+/− Mouse Model. PLoS One. 11:e01640662016. View Article : Google Scholar

29 

Wei Q and Dong Z: Mouse model of ischemic acute kidney injury: Technical notes and tricks. Am J Physiol Renal Physiol. 303:F1487–F1494. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Wilson GC, Freeman CM, Kuethe JW, Quillin RC III, Nojima H, Schuster R, Blanchard J, Edwards MJ, Caldwell CC and Lentsch AB: CXC chemokine receptor-4 signaling limits hepatocyte proliferation after hepatic ischemia-reperfusion in mice. Am J Physiol Gastrointest Liver Physiol. 308:G702–G709. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Andrassy M, Volz HC, Igwe JC, Funke B, Eichberger SN, Kaya Z, Buss S, Autschbach F, Pleger ST, Lukic IK, et al: High-mobility group box-1 in ischemia-reperfusion injury of the heart. Circulation. 117:3216–3226. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Inoue Y, Shirasuna K, Kimura H, Usui F, Kawashima A, Karasawa T, Tago K, Dezaki K, Nishimura S, Sagara J, et al: NLRP3 regulates neutrophil functions and contributes to hepatic ischemia-reperfusion injury independently of inflammasomes. J Immunol. 192:4342–4351. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar

34 

He L, Sun F, Wang Y, Zhu J, Fang J, Zhang S, Yu Q, Gong Q, Ren B, Xiang X, et al: HMGB1 exacerbates bronchiolitis obliterans syndrome via RAGE/NF-κB/hPSe signaling to enhance latent TGF-β release from ECM. Am J Transl Res. 8:1971–1984. 2016.

35 

Duret C, Gerbal-Chaloin S, Ramos J, Fabre JM, Jacquet E, Navarro F, Blanc P, Sa-Cunha A, Maurel P and Daujat-Chavanieu M: Isolation, characterization, and differentiation to hepatocyte-like cells of nonparenchymal epithelial cells from adult human liver. Stem Cells. 25:1779–1790. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Van Rooijen N and Hendrikx E: Liposomes for specific depletion of macrophages from organs and tissues. Methods Mol Biol. 605:189–203. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Van Rooijen N and Sanders A: Liposome mediated depletion of macrophages: Mechanism of action, preparation of liposomes and applications. J Immunol Methods. 174:83–93. 1994. View Article : Google Scholar : PubMed/NCBI

38 

Lv Q, Yang F, Chen K and Zhang Y: Autophagy protects podocytes from sublytic complement induced injury. Exp Cell Res. 341:132–138. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Rossi L, Serafini S, Antonelli A, Pierigé F, Carnevali A, Battistelli V, Malatesta M, Balestra E, Caliò R, Perno CF, et al: Macrophage depletion induced by clodronate-loaded erythrocytes. J Drug Target. 13:99–111. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Heymann F, Trautwein C and Tacke F: Monocytes and macrophages as cellular targets in liver fibrosis. Inflamm Allergy Drug Targets. 8:307–318. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Sullivan T, Miao Z, Dairaghi DJ, Krasinski A, Wang Y, Zhao BN, Baumgart T, Ertl LS, Pennell A, Seitz L, et al: CCR2 antagonist CCX140-B provides renal and glycemic benefits in diabetic transgenic human CCR2 knockin mice. Am J Physiol Renal Physiol. 305:F1288–F1297. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Wang F, Zhang G, Lu Z, Geurts AM, Usa K, Jacob HJ, Cowley AW, Wang N and Liang M: Antithrombin III/SerpinC1 insufficiency exacerbates renal ischemia/reperfusion injury. Kidney Int. 88:796–803. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Brempelis KJ and Crispe IN: Infiltrating monocytes in liver injury and repair. Clin Transl Immunology. 5:e1132016. View Article : Google Scholar : PubMed/NCBI

44 

Höchst B, Mikulec J, Baccega T, Metzger C, Welz M, Peusquens J, Tacke F, Knolle P, Kurts C, Diehl L, et al: Differential induction of Ly6G and Ly6C positive myeloid derived suppressor cells in chronic kidney and liver inflammation and fibrosis. PLoS One. 10:e01196622015. View Article : Google Scholar : PubMed/NCBI

45 

Cuervo H, Guerrero NA, Carbajosa S, Beschin A, De Baetselier P, Gironès N and Fresno M: Myeloid-derived suppressor cells infiltrate the heart in acute Trypanosoma cruzi infection. J Immunol. 187:2656–2665. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Morias Y, Abels C, Laoui D, Van Overmeire E, Guilliams M, Schouppe E, Tacke F, deVries CJ, De Baetselier P and Beschin A: Ly6C monocytes regulate parasite-induced liver inflammation by inducing the differentiation of pathogenic Ly6C+ monocytes into macrophages. PLoS Pathog. 11:e10048732015. View Article : Google Scholar

47 

Alisi A, Carpino G, Oliveira FL, Panera N, Nobili V and Gaudio E: The role of tissue macrophage-mediated inflammation on NAFLD pathogenesis and its clinical implications. Mediators Inflamm. 8162421:2017. View Article : Google Scholar : PubMed/NCBI

48 

Weisser SB, van Rooijen N and Sly LM: Depletion and reconstitution of macrophages in mice. J Vis Exp. Aug 1–2012.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

49 

Karlmark KR, Weiskirchen R, Zimmermann HW, Gassler N, Ginhoux F, Weber C, Merad M, Luedde T, Trautwein C and Tacke F: Hepatic recruitment of the inflammatory Gr1+ monocyte subset upon liver injury promotes hepatic fibrosis. Hepatology. 50:261–274. 2009. View Article : Google Scholar : PubMed/NCBI

50 

Engel DR, Maurer J, Tittel AP, Weisheit C, Cavlar T, Schumak B, Limmer A, van Rooijen N, Trautwein C, Tacke F, et al: CCR2 mediates homeostatic and inflammatory release of Gr1(high) monocytes from the bone marrow, but is dispensable for bladder infiltration in bacterial urinary tract infection. J Immunol. 181:5579–5586. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2018
Volume 41 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Song P, Zhang J, Zhang Y, Shu Z, Xu P, He L, Yang C, Zhang J, Wang H, Li Y, Li Y, et al: Hepatic recruitment of CD11b+Ly6C+ inflammatory monocytes promotes hepatic ischemia/reperfusion injury. Int J Mol Med 41: 935-945, 2018
APA
Song, P., Zhang, J., Zhang, Y., Shu, Z., Xu, P., He, L. ... Li, Q. (2018). Hepatic recruitment of CD11b+Ly6C+ inflammatory monocytes promotes hepatic ischemia/reperfusion injury. International Journal of Molecular Medicine, 41, 935-945. https://doi.org/10.3892/ijmm.2017.3315
MLA
Song, P., Zhang, J., Zhang, Y., Shu, Z., Xu, P., He, L., Yang, C., Zhang, J., Wang, H., Li, Y., Li, Q."Hepatic recruitment of CD11b+Ly6C+ inflammatory monocytes promotes hepatic ischemia/reperfusion injury". International Journal of Molecular Medicine 41.2 (2018): 935-945.
Chicago
Song, P., Zhang, J., Zhang, Y., Shu, Z., Xu, P., He, L., Yang, C., Zhang, J., Wang, H., Li, Y., Li, Q."Hepatic recruitment of CD11b+Ly6C+ inflammatory monocytes promotes hepatic ischemia/reperfusion injury". International Journal of Molecular Medicine 41, no. 2 (2018): 935-945. https://doi.org/10.3892/ijmm.2017.3315