scholarly journals ADAM Metalloproteinase Domain 17 Regulates Cholestasis-Associated Liver Injury and Sickness Behavior Development in Mice

2022 ◽  
Vol 12 ◽  
Author(s):  
Wagdi Almishri ◽  
Liam A. Swain ◽  
Charlotte D’Mello ◽  
Tyson S. Le ◽  
Stefan J. Urbanski ◽  
...  

Disintegrin and metalloproteinase domain-containing protein 17 (ADAM17) is a ubiquitously expressed membrane-bound enzyme that mediates shedding of a wide variety of important regulators in inflammation including cytokines and adhesion molecules. Hepatic expression of numerous cytokines and adhesion molecules are increased in cholestatic liver diseases including primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC), however, the pathophysiological role of ADAM17 in regulating these conditions remains unknown. Therefore, we evaluated the role of ADAM17 in a mouse model of cholestatic liver injury due to bile duct ligation (BDL). We found that BDL enhanced hepatic ADAM17 protein expression, paralleled by increased ADAM17 bioactivity. Moreover, inhibition of ADAM17 bioactivity with the specific inhibitor DPC 333 significantly improved both biochemical and histological evidence of liver damage in BDL mice. Patients with cholestatic liver disease commonly experience adverse behavioral symptoms, termed sickness behaviors. Similarly, BDL in mice induces reproducible sickness behavior development, driven by the upregulated expression of cytokines and adhesion molecules that are in turn regulated by ADAM17 activity. Indeed, inhibition of ADAM17 activity significantly ameliorated BDL-associated sickness behavior development. In translational studies, we evaluated changes in ADAM17 protein expression in liver biopsies obtained from patients with PBC and PSC, compared to normal control livers. PSC and PBC patients demonstrated increased hepatic ADAM17 expression in hepatocytes, cholangiocytes and in association with liver-infiltrating immune cells compared to normal controls. In summary, cholestatic liver injury in mice and humans is associated with increased hepatic ADAM17 expression. Furthermore, inhibition of ADAM17 activity improves both cholestatic liver injury and associated sickness behavior development, suggesting that ADAM17 inhibition may represent a novel therapeutic approach for treating patients with PBC/PSC.

2018 ◽  
Vol 314 (3) ◽  
pp. G319-G333 ◽  
Author(s):  
Nadine Gehrke ◽  
Michael Nagel ◽  
Beate K. Straub ◽  
Marcus A. Wörns ◽  
Marcus Schuchmann ◽  
...  

Cholestatic liver injury results from impaired bile flow or metabolism and promotes hepatic inflammation and fibrogenesis. Toxic bile acids that accumulate in cholestasis induce apoptosis and contribute to early cholestatic liver injury, which is amplified by accompanying inflammation. The aim of the current study was to evaluate the role of the antiapoptotic caspase 8-homolog cellular FLICE-inhibitory (cFLIP) protein during acute cholestatic liver injury. Transgenic mice exhibiting hepatocyte-specific deletion of cFLIP (cFLIP−/−) were used for in vivo and in vitro analysis of cholestatic liver injury using bile duct ligation (BDL) and the addition of bile acids ex vivo. Loss of cFLIP in hepatocytes promoted acute cholestatic liver injury early after BDL, which was characterized by a rapid release of proinflammatory and chemotactic cytokines (TNF, IL-6, IL-1β, CCL2, CXCL1, and CXCL2), an increased presence of CD68+ macrophages and an influx of neutrophils in the liver, and resulting apoptotic and necrotic hepatocyte cell death. Mechanistically, liver injury in cFLIP−/− mice was aggravated by reactive oxygen species, and sustained activation of the JNK signaling pathway. In parallel, cytoprotective NF-κB p65, A20, and the MAPK p38 were inhibited. Increased injury in cFLIP−/− mice was accompanied by activation of hepatic stellate cells and profibrogenic regulators. The antagonistic caspase 8-homolog cFLIP is a critical regulator of acute, cholestatic liver injury. NEW & NOTEWORTHY The current paper explores the role of a classical modulator of hepatocellular apoptosis in early, cholestatic liver injury. These include activation of NF-κB and MAPK signaling, production of inflammatory cytokines, and recruitment of neutrophils in response to cholestasis. Because these signaling pathways are currently exploited in clinical trials for the treatment of nonalcoholic steatohepatitis and cirrhosis, the current data will help in the development of novel pharmacological options in these indications.


2021 ◽  
Vol 12 ◽  
Author(s):  
Chuanrui Ma ◽  
Jiaqing Xiang ◽  
Guixiao Huang ◽  
Yaxi Zhao ◽  
Xinyu Wang ◽  
...  

Background and purpose: FXR is a promising target for the treatment of human cholestatic liver disease (CLD). SIRT1 is a deacetylase which promotes FXR activity through deacetylating FXR. Pterostilbene (PTE) is an activator of SIRT1. However, the role of PTE in cholestasis has so far not been investigated. We examined whether PTE treatment alleviate liver injury in DDC or ANIT-induced experimental cholestasis, and explored the underlying mechanisms.Experimental approach: Mice with DDC- or ANIT-induced cholestasis were treated with different dose of PTE. Primary hepatocytes and bone marrow derived macrophages were used in vitro to assess the molecular mechanism by which PTE may improve CLD. Identical doses of UDCA or PTE were administered to DDC- or ANIT-induced cholestasis mice.Key results: PTE intervention attenuated DDC or ANIT-induced cholestasis. PTE inhibited macrophage infiltration and activation in mouse liver through the SIRT1-p53 signaling pathway, and it improved hepatic bile metabolism through the SIRT1-FXR signaling pathway. Compare with UDCA, the same doses of PTE was more effective in improving cholestatic liver injury caused by DDC or ANIT.Conclusion and implications: SIRT1 activation in macrophages may be an effective CLD treatment avenue. Using CLD models, we thus identified PTE as a novel clinical candidate compound for the treatment of CLD.


2020 ◽  
Vol 3 (Supplement_1) ◽  
pp. 58-59
Author(s):  
A J Mathews ◽  
F Vicentini ◽  
L Swain ◽  
M Swain ◽  
K A Sharkey

Abstract Background Cholestatic liver disease is associated with immune-mediated inflammatory liver injury. This disorder is also associated with brain dysfunction and behavioural changes, notably fatigue, depression and social withdrawal. The mechanisms leading to these central nervous system abnormalities are unknown, however, they are associated with neuroinflammation. Microglia and astrocytes are two glial populations that play key roles in neuroinflammation. Activated glia display morphological changes, secrete cytokines, and mediate electrophysiological changes, altering the normal functioning of the brain. The aryl hydrocarbon receptor (AhR) is a transcription factor involved in the immune response. AhR is present on glia and its’ activation has been shown to reduce neuroinflammation. The role of the AhR in cholestatic liver disease has yet to be examined. Aims To study the function of the AhR in a model of cholestic liver disease. We will test the hypothesis that activation of AhR in the brain will reduce neuroinflammation and behavioral deficits observed in cholestatic mice. Methods Male C57Bl/6J mice had cholestasis induced by bile duct ligation (BDL); comparisons were made to sham-operated controls. Mice were tested for social interaction with a 4-week old juvenile in their home cage and the number of social interaction attempts quantified. Next, mice were euthanized, brains were removed and processed for immunohistochemistry. Brain sections were stained for markers of microglia (IBA-1) and astrocytes (GFAP). Microglia were counted and astrocyte activation was qualitatively assessed. PCR was used to quantify gene expression of AhR and its downstream gene targets (eg. CYP1A1) in mice that recived treatment with beta-napthoflavone (BNF), an AhR agonist, or in vehicle treated controls. Results BDL mice made significantly fewer attempts to interact with the juvenile as compared to controls (P<0.05). We also observed a significant increase in IBA-1 immunoreactive cell numbers in both the CA1 region of the hippocampus and the hypothalamic paraventricular nucleus (PVN, P<0.05). BDL mice also displayed marked increases in GFAP+ staining in the PVN, but not the CA1, in contrast to sham controls. Lastly, we found that BNF significantly upregulated CYP1A1 (P<0.05) in the liver and prefrontal cortex of mice. We are currently examining whether BNF can reduce neuroinflammation and improve decreased social interaction in cholestatic mice. Conclusions Cholestatic liver damage was associated with impaired social behavior. Further, glial activation, an indicator of neuroinflammation was increased in components of the limbic system associated with the response to stress, learning, and memory. Future experiments will address whether activation of the AhR will ameliorate neuroinflammation and behavioral changes observed in mice with cholestatic liver injury. Funding Agencies CCC, CIHR


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3678-3678
Author(s):  
Yan Zhang ◽  
Zhipeng Yao ◽  
Tao Li ◽  
Muhua Cao ◽  
Ruishuang Ma ◽  
...  

Abstract Introduction: The mechanisms involved in cholestatic liver injury remain elusive although emerging evidence indicate neutrophils, rather than bile acids (BAs), are the predominant source of damage. Neutrophil extracellular traps (NETs) are double-edge swords that serve to ensnare and kill microbial pathogens but also contribute to excessive inflammation and tissue damage. However, the role of NETs during bile duct ligation (BDL)-induced cholestatic liver injury is largely unknown. Moreover, whether neutrophils and BAs cooperatively participate in this process also needs to be investigated. Our objectives were to localize and determine the magnitude of NETs in the hepatic microvasculature, define the role of NETs on hematological and histological changes, and characterize the role of BAs in the induction of NETs in BDL mice. Methods: BDL was induced in C57BL/6 mice by ligation of the common bile duct. After 3 days, liver tissue was collected and NETs were detected by immunostaining and western blot. Serum myeloperoxidase (MPO)-DNA complexes were measured by ELISA. Cholestatic liver injury was characterized by hematological and histological assessment. NETs were dismantled by DNase I or depleted with peptidylarginine deiminase 4 (PAD4) inhibitor Cl-amidine. The apoptosis of hepatocytes was measured by TUNEL and the induction of chemokines on Kupffer cells was analyzed by quantitative polymerase chain reaction (qPCR). BAs and inflammatory cytokines were utilized collaboratively to induce NETs formation both in vitro and in vivo. Diphenylene iodonium (DPI) was used to inhibit reactive oxygen species (ROS) formation. Plasma was obtained from healthy controls and patients with entrahepatic cholestasis. Results: We found that NETs formed in the sinusoids of cholestatic liver lobes in vivo, which was associated with significantly increased serum MPO-DNA complexes (P<0.01) and tissue levels of citrullinated-histone 3 (P<0.05) in BDL mice compared with sham operated mice. The formation of NETs exacerbated BDL-induced liver injury as evidenced by markedly increased levels of serum alamine transaminase (ALT) and percentage of necrotic tissue area (both P<0.05 vs. sham group, Figure 1). Depleting of NETs with DNase I or Cl-amidine decreased NETs formation, protected hepatocytes and dramatically alleviated cholestatic liver injury, indicating the pathophysiological role of NETs in BDL mice (Figure 1). In addition, NETs promoted the apoptosis of hepatocytes and induced the release of chemokines from Kupffer cells in BDL mice compared to sham group. Both effects could be inhibited by DNase I or Cl-amidine. Moreover, BAs induced mouse neutrophils, primed with inflammatory cytokines, to release NETs in vitro in a dose-dependent manner and mediated by PAD4 and ROS. Intravenously injection of BAs and inflammatory cytokines to healthy mice lead to the formation of NETs and exacerbated liver injury, suggesting that depleting NETs can prevent organ damage. Furthermore, we found NETs markers were significantly increased in the serum of cholestatic patients, correlating positively with increased levels of ALT and BAs. Conclusions: Our results suggest that BAs promote NETs formation via PAD4 and ROS. Development of NETs subsequently initiates inflammatory responses and exacerbates organ damage during BDL-induced cholestatic liver injury, and may therefore serve as a promising therapeutic target in cholestasis. Figure 1. Inhibition of NET formation by PAD4 inhibitor (Cl-amidine) or DNase I protects cholestatic liver injury in BDL mice. Serum total bilirubin (A), total bile acids (B), and ALT levels (C) were assessed in control-, Cl-amidine-, or DNase I treated mice after either 3 days of sham laparotomy or BDL. (D) Quantification of necrotic hepatocytes in H&E stained liver sections from control-, Cl-amidine-, or DNase I treated mice 3 days after sham laparotomy or BDL. *P < 0.05 untreated BDL mice vs. sham mice. **P < 0.05 Cl-amidine or DNase I treated BDL mice vs. untreated BDL mice. Figure 1. Inhibition of NET formation by PAD4 inhibitor (Cl-amidine) or DNase I protects cholestatic liver injury in BDL mice. Serum total bilirubin (A), total bile acids (B), and ALT levels (C) were assessed in control-, Cl-amidine-, or DNase I treated mice after either 3 days of sham laparotomy or BDL. (D) Quantification of necrotic hepatocytes in H&E stained liver sections from control-, Cl-amidine-, or DNase I treated mice 3 days after sham laparotomy or BDL. *P < 0.05 untreated BDL mice vs. sham mice. **P < 0.05 Cl-amidine or DNase I treated BDL mice vs. untreated BDL mice. Disclosures No relevant conflicts of interest to declare.


2009 ◽  
Vol 47 (01) ◽  
Author(s):  
P Nalapareddy ◽  
S Schüngel ◽  
MP Manns ◽  
H Jaeschke ◽  
A Vogel

2018 ◽  
Vol 19 (9) ◽  
pp. 2509 ◽  
Author(s):  
Jing Zhang ◽  
Xin Guo ◽  
Taiji Hamada ◽  
Seiya Yokoyama ◽  
Yuka Nakamura ◽  
...  

Accumulating evidence indicates that oxidative stress plays a critical role in initiating the progression of inflammatory and fibrotic liver diseases, including cholestatic hepatitis. Peroxiredoxin 4 (PRDX4) is a secretory antioxidase that protects against oxidative damage by scavenging reactive oxygen species (ROS) in both the intracellular compartments and extracellular space. In this study, we examined the in vivo net effects of PRDX4 overexpression in a murine model of cholestasis. To induce cholestatic liver injury, we subjected C57BL/6J wild-type (WT) or human PRDX4 (hPRDX4) transgenic (Tg) mice to sham or bile duct ligation (BDL) surgery for seven days. Our results showed that the liver necrosis area was significantly suppressed in Tg BDL mice with a reduction in the severity of liver injuries. Furthermore, PRDX4 overexpression markedly reduced local and systemic oxidative stress generated by BDL. In addition, suppression of inflammatory cell infiltration, reduced proliferation of hepatocytes and intrahepatic bile ducts, and less fibrosis were also found in the liver of Tg BDL mice, along with a reduced mortality rate after BDL surgery. Interestingly, the composition of the hepatic bile acids (BAs) was more beneficial for Tg BDL mice than for WT BDL mice, suggesting that PRDX4 overexpression may affect BA metabolism during cholestasis. These features indicate that PRDX4 plays an important role in protecting against liver injury following BDL and might be a promising therapeutic modality for cholestatic diseases.


2019 ◽  
Vol 317 (6) ◽  
pp. G773-G783 ◽  
Author(s):  
Takanori Konishi ◽  
Rebecca M. Schuster ◽  
Holly S. Goetzman ◽  
Charles C. Caldwell ◽  
Alex B. Lentsch

The CXC chemokine receptor 2 (CXCR2) is critical for neutrophil recruitment and hepatocellular viability but has not been studied in the context of cholestatic liver injury following bile duct ligation (BDL). The present study sought to elucidate the cell-specific roles of CXCR2 on acute liver injury after BDL. Wild-type and CXCR2−/− mice were subjected BDL. CXCR2 chimeric mice were created to assess the cell-specific role of CXCR2 on liver injury after BDL. SB225002, a selective CXCR2 antagonist, was administrated intraperitoneally after BDL to investigate the potential of pharmacological inhibition. CXCR2−/− mice had significantly less liver injury than wild-type mice at 3 and 14 days after BDL. There was no difference in biliary fibrosis among groups. The chemokines CXCL1 and CXCL2 were induced around areas of necrosis and biliary structures, respectively, both areas where neutrophils accumulated after BDL. CXCR2−/− mice showed significantly less neutrophil accumulation in those injured areas. CXCR2Liver+/Myeloid+ and CXCR2Liver−/Myeloid− mice recapitulated the wild-type and CXCR2-knockout phenotypes, respectively. CXCR2Liver+/Myeloid+ mice suffered higher liver injury than CXCR2Liver+/Myeloid− and CXCR2Liver−/Myeloid+; however, only those chimeras with knockout of myeloid CXCR2 (CXCR2Liver+/Myeloid− and CXCR2Liver−/Myeloid−) showed reduction of neutrophil accumulation around areas of necrosis. Daily administration of SB225002 starting after 3 days of BDL reduced established liver injury at 6 days. In conclusion, neutrophil CXCR2 guides the cell to the site of injury, while CXCR2 on liver cells affects liver damage independent of neutrophil accumulation. CXCR2 appears to be a viable therapeutic target for cholestatic liver injury. NEW & NOTEWORTHY This study is the first to reveal cell-specific roles of the chemokine receptor CXCR2 in cholestatic liver injury caused by bile duct ligation. CXCR2 on neutrophils facilitates neutrophil recruitment to the liver, while CXCR2 on liver cells contributes to liver damage independent of neutrophils. CXCR2 may represent a viable therapeutic target for cholestatic liver injury.


Hepatology ◽  
2018 ◽  
Vol 67 (4) ◽  
pp. 1441-1457 ◽  
Author(s):  
Runping Liu ◽  
Xiaojiaoyang Li ◽  
Zhiming Huang ◽  
Derrick Zhao ◽  
Bhagyalaxmi Sukka Ganesh ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document