ArticlePublisher preview available

Mitogen-activated protein kinases are involved in hepatocanalicular dysfunction and cholestasis induced by oxidative stress

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

In previous studies, we showed that the pro-oxidant model agent tert-butyl hydroperoxide (tBuOOH) induces alterations in hepatocanalicular secretory function by activating Ca²⁺-dependent protein kinase C isoforms (cPKC), via F-actin disorganization followed by endocytic internalization of canalicular transporters relevant to bile formation (Mrp2, Bsep). Since mitogen-activated protein kinases (MAPKs) may be downstream effectors of cPKC, we investigated here the involvement of the MAPKs of the ERK1/2, JNK1/2, and p38MAPK types in these deleterious effects. tBuOOH (100 µM, 15 min) increased the proportion of the active, phosphorylated forms of ERK1/2, JNK1/2, and p38MAPK, and panspecific PKC inhibition with bisindolylmaleimide-1 (100 nM) or selective cPKC inhibition with Gö6976 (1 μM) prevented the latter two events. In isolated rat hepatocyte couplets, tBuOOH (100 µM, 15 min) decreased the canalicular vacuolar accumulation of the fluorescent Bsep and Mrp2 substrates, cholylglycylamido fluorescein, and glutathione-methylfluorescein, respectively, and selective inhibitors of ERK1/2 (PD098059), JNK1/2 (SP600125), and p38MAPK (SB203580) partially prevented these alterations. In in situ perfused rat livers, these three MAPK inhibitors prevented tBuOOH (75 µM)-induced impairment of bile flow and the decrease in the biliary output of the Bsep and Mrp2 substrates, taurocholate, and dinitrophenyl-S-glutathione, respectively. The changes in Bsep/Mrp2 and F-actin localization induced by tBuOOH, as assessed by (immuno)fluorescence staining followed by analysis of confocal images, were prevented total or partially by the MAPK inhibitors. We concluded that MAPKs of the ERK1/2, JNK1/2, and p38MAPK types are all involved in cholestasis induced by oxidative stress, by promoting F-actin rearrangement and further endocytic internalization of canalicular transporters critical for bile formation.
This content is subject to copyright. Terms and conditions apply.
1 3
Arch Toxicol (2017) 91:2391–2403
DOI 10.1007/s00204-016-1898-1
ORGAN TOXICITY AND MECHANISMS
Mitogen‑activated protein kinases are involved
in hepatocanalicular dysfunction and cholestasis induced
by oxidative stress
Flavia D. Toledo1 · Cecilia L. Basiglio1 · Ismael R. Barosso1 · Andrea C. Boaglio1 ·
Andrés E. Zucchetti1 · Enrique J. Sánchez Pozzi1 · Marcelo G. Roma1
Received: 26 May 2016 / Accepted: 24 November 2016 / Published online: 2 December 2016
© Springer-Verlag Berlin Heidelberg 2016
output of the Bsep and Mrp2 substrates, taurocholate, and
dinitrophenyl-S-glutathione, respectively. The changes in
Bsep/Mrp2 and F-actin localization induced by tBuOOH,
as assessed by (immuno)fluorescence staining followed by
analysis of confocal images, were prevented total or par-
tially by the MAPK inhibitors. We concluded that MAPKs
of the ERK1/2, JNK1/2, and p38MAPK types are all involved
in cholestasis induced by oxidative stress, by promoting
F-actin rearrangement and further endocytic internalization
of canalicular transporters critical for bile formation.
Keywords Oxidative stress · Hepatocellular cholestasis ·
Canalicular transporters · Mitogen-activated protein
kinases · Actin cytoskeleton
Introduction
Oxidative stress (OS) is a common feature in most
hepatopathies, including drug and environmental toxin-
induced hepatotoxicity, hepatic ischemia–reperfusion
injury, viral and autoimmune hepatitis, alcoholic and non-
alcoholic steatohepatitis, and pathologies leading to hepatic
accumulation of either heavy metals, such as iron (hemo-
chromatosis) and copper (Wilson’s disease), or bile acids
(obstructive or functional cholestasis) (Copple et al. 2010;
Jaeschke et al. 2002).
In recent years, evidence has accumulated that, in chole-
static hepatopathies, a vicious circle occurs, since OS is
cholestatic in nature, thus aggravating the initial secretory
failure (Roma and Sanchez Pozzi 2008). Indeed, our group
demonstrated in isolated rat hepatocyte couplets (IRHCs)
that OS induces rapid endocytic internalization of the
bile salt export pump (Bsep, AKA: Abcc11) (Perez et al.
2006b), the main canalicular bile salt transporter. A similar
Abstract In previous studies, we showed that the pro-
oxidant model agent tert-butyl hydroperoxide (tBuOOH)
induces alterations in hepatocanalicular secretory function
by activating Ca2+-dependent protein kinase C isoforms
(cPKC), via F-actin disorganization followed by endocytic
internalization of canalicular transporters relevant to bile
formation (Mrp2, Bsep). Since mitogen-activated protein
kinases (MAPKs) may be downstream effectors of cPKC,
we investigated here the involvement of the MAPKs of
the ERK1/2, JNK1/2, and p38MAPK types in these deleteri-
ous effects. tBuOOH (100 µM, 15 min) increased the pro-
portion of the active, phosphorylated forms of ERK1/2,
JNK1/2, and p38MAPK, and panspecific PKC inhibition with
bisindolylmaleimide-1 (100 nM) or selective cPKC inhibi-
tion with Gö6976 (1 μM) prevented the latter two events.
In isolated rat hepatocyte couplets, tBuOOH (100 µM, 15
min) decreased the canalicular vacuolar accumulation of the
fluorescent Bsep and Mrp2 substrates, cholylglycylamido
fluorescein, and glutathione-methylfluorescein, respectively,
and selective inhibitors of ERK1/2 (PD098059), JNK1/2
(SP600125), and p38MAPK (SB203580) partially prevented
these alterations. In in situ perfused rat livers, these three
MAPK inhibitors prevented tBuOOH (75 µM)-induced
impairment of bile flow and the decrease in the biliary
Electronic supplementary material The online version of this
article (doi:10.1007/s00204-016-1898-1) contains supplementary
material, which is available to authorized users.
* Marcelo G. Roma
mroma@fbioyf.unr.edu.ar
1 Instituto de Fisiología Experimental (IFISE-CONICET),
Facultad de Ciencias Bioquímicas y Farmacéuticas,
Universidad Nacional de Rosario (UNR), Suipacha 570,
2000, Rosario, Argentina
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
... In several physiological and pathological processes, different intracellular signaling pathways activated by the cytokine have been reported. Among these signaling proteins, TNFα is known to induce MEK/ERK kinase activation [18] which has been already implied in Abcc2 internalization induced by other cholestatic agents [19,20]. Hence, our aim was to evaluate the role of this kinase in TNFα-induced internalization of Abcc2. ...
... TNFα is capable of activating different signaling proteins. Among them, MEK-ERK [18], PI3K-AKT [41] and isoforms of PKC (α, δ) [42,43] have been already implied in other cholestasis models [44,45,20,46]. We proved the role of MEK-ERK pathway in TNFα-induced Abcc2 retrieval. ...
... The capacity of ROS, even when produced at very low levels, to induce biliary secretory failure and cholestasis is already known [49] as well as the mediatory action of ERK in ROS-induced Abcc2 internalization [20]. The low and short-term increase of intracellular ROS produced by TNFα is consistent with the role of oxidative stress previously described in LPS-induced cholestasis. ...
Article
TNFα is a cytokine whose levels are increased in inflammatory pathologies that are associated with cholestasis. Endocytic internalization of Abcc2 (multidrug resistance-associated protein 2), a canalicular transporter of organic anions that is implicated in the clearance of clinically important drugs, is a phenomenon that occurs in inflammatory liver diseases, and it has been established that cytokines act as mediators. However, the intracellular mechanism involved in this effect remains unknown. The aim of the present work was to characterize the internalization of Abcc2 induced by TNFα and to study the role of ERK1/2 and reactive oxygen species as signaling mediators of transporter internalization. Using rat hepatocyte couplets, we found that TNFα (6.25 pg/ml) induced a decrease in Abcc2 activity estimated by the accumulation of the Abcc2 substrate glutathione methylfluorescein in the canalicular vacuole that was accompanied by internalization of Abcc2 from the canalicular membrane. Inhibition of MEK1/2 (upstream of ERK1/2) partially prevented TNFα effects on Abcc2 internalization and activity impairment. Reactive oxygen species (ROS) scavengers such as vitamin C and mannitol partially prevented both TNFα-induced decrease in Abcc2 activity and ERK1/2 phosphorylation. Apocynin, a NADPH oxidase inhibitor, prevented the increase in ROS and the phosphorylation of ERK1/2 produced by TNFα. Taken together, these results indicate that TNFα activates a pathway involving NADPH oxidase, ROS and MEK1/2-ERK1/2 that is partially responsible for the internalization of Abcc2. This internalization leads to an altered transport activity of Abcc2 that could impair drug disposal, enhancing drug toxicity in patients suffering from inflammatory liver diseases.
... We found in isolated rat hepatocyte couplets that tBuOOH induces endocytic internalization of the bile salt export pump (Bsep, Abcb11), a main canalicular carrier that transports into the bile canaliculus osmotically active monoanionic bile salts, the main driving forces involved in the generation of the so-called 'bile salt-dependent bile flow' (19). The same holds true for the multidrug resistance-associated protein 2 (Mrp2, Abcc2), as has been shown in isolated rat hepatocyte couplets (20,21), and perfused rat livers exposed to the prooxidant agent tBuOOH (20,21); this transporter mediates the biliary excretion of glutathione, the main driving force of the so called 'bile salt-independent bile flow' (22). Bilirubin (BR), one of the end products of heme catabolic pathway, has been historically considered not only a waste product but also a potentially toxic compound. ...
... We found in isolated rat hepatocyte couplets that tBuOOH induces endocytic internalization of the bile salt export pump (Bsep, Abcb11), a main canalicular carrier that transports into the bile canaliculus osmotically active monoanionic bile salts, the main driving forces involved in the generation of the so-called 'bile salt-dependent bile flow' (19). The same holds true for the multidrug resistance-associated protein 2 (Mrp2, Abcc2), as has been shown in isolated rat hepatocyte couplets (20,21), and perfused rat livers exposed to the prooxidant agent tBuOOH (20,21); this transporter mediates the biliary excretion of glutathione, the main driving force of the so called 'bile salt-independent bile flow' (22). Bilirubin (BR), one of the end products of heme catabolic pathway, has been historically considered not only a waste product but also a potentially toxic compound. ...
... Importantly, they can be counteracted by 24 other HO-1 products, as for example, CO, which has potent anti-ischemic and antiinflammatory properties (74). Nevertheless, there is considerable evidence implicating ROS as another cause of the hepatic injury, since ischemia results in a build-up of NADH that, on reperfusion, generates a burst of ROS from the mitochondrial electron transport chain, which can be protected by HO-1-derived BR in the same manner it afforded full protection in our pro-oxidant model (21). ...
Article
We previously demonstrated in in vitro and ex vivo models that physiological concentrations of unconjugated bilirubin (BR) prevent oxidative stress (OS)-induced hepatocanalicular dysfunction and cholestasis. Here, we aimed to ascertain, in the whole rat, whether a similar cholestatic OS injury can be counteracted by heme oxygenase-1 (HO-1) induction that consequently elevates endogenous BR levels. This was achieved through the administration of hemin, an inducer of HO-1, the rate-limiting step in BR generation. We found that BR peaked between 6 and 8 h after hemin administration. During this time period, HO-1 induction fully prevented the pro-oxidant tert-butylhydroperoxide (tBuOOH)-induced drop in bile flow, and in the biliary excretion of bile salts and glutathione, the two main driving forces of bile flow; this was associated with preservation of the membrane localization of their respective canalicular transporters, bile salt export pump (Bsep) and multidrug resistance-associated protein 2 (Mrp2), which are otherwise endocytosed by OS. HO-1 induction counteracted the oxidation of intracellular proteins and membrane lipids induced by tBuOOH, and fully prevented the increase in the oxidized-to-total glutathione (GSHt) ratio, a sensitive parameter of hepatocellular OS. Compensatory elevations of the activity of the antioxidant enzymes catalase (CAT) and superoxide dismutase (SOD) were also prevented. We conclude that in vivo HO-1 induction protects the liver from acute oxidative injury, thus preventing consequent cholestasis. This reveals an important role for the induction of HO-1 and the consequently elevated levels of BR in preserving biliary secretory function under OS conditions, thus representing a novel therapeutic tool to limit the cholestatic injury that bears an oxidative background. © 2019 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.
... Previous studies have reported a crucial role for MAPK in cell apoptosis (Zheng et al., 2014;Yuan et al., 2017). In particular, the involvement of P38, ERK, and JNK have been verified in models of cholestasis induced by oxidative stress (Toledo et al., 2017). In the present study, changes in the levels of p-P38/P38, p-ERK/ERK, and p-JNK/JNK were detected in the FIGURE 14 | Effects of forsythoside-A, emodin and chlorogenic acid on CaSR expression in vitro. ...
... The mitochondria are the main cellular source of ROS. Accumulated bile acids have been reported to cause hepatocyte apoptosis in humans with cholestasis through ROS-mediated oxidative stress (Toledo et al., 2017). Meanwhile, calcium overload caused by CaSR is also implicated in cell apoptosis (Guo et al., 2012;Qi et al., 2013). ...
Article
Full-text available
Infantile cholestatic hepatopathy (ICH) is a clinical syndrome characterized by the accumulation of cytotoxic bile acids in infancy, leading to serious liver cirrhosis or liver failure. The aetiology of ICH is complicated and some of them is unknown. Regardless of the aetiology, the finial pathology of ICH is hepatocyte apoptosis caused by severe and persistent cholestasis. It is already known that activation of calcium-sensing receptor (CaSR) could lead to the apoptosis of cardiomyocytes. However, the mechanism by CaSR-mediated cholestasis-related hepatocyte apoptosis is not fully understood. Li-Dan-He-Ji (LDHJ), a Traditional Chinese Medicine prescription, was developed to treat ICH. Another aim of this study was to investigate the possible mechanisms of LDHJ in cholestasis-related hepatocyte apoptosis. Using the primary hepatocytes, we first investigated the molecular mechanism of CaSR-mediated hepatocyte apoptosis in cholestasis. Then we prepared LDHJ granules and used ultra-high-performance liquid chromatography to identify the predominant drugs; confirmed the stability of the main substances; and for cell experiments screened forsythoside-A, emodin and chlorogenic acid as the three active substances of LDHJ granules. In the young rats with ANIT-induced intrahepatic cholestasis and the primary hepatocytes with TCDC-induced cholestasis-related hepatocyte apoptosis, the levels of liver injury and cholestasis-related biomarkers, calcium-sensing receptor (CaSR), hepatocyte apoptosis, Bax/Bcl-2 ratio, Cytochrome-C, caspase-3, phosphorylated-c-Jun NH2-terminal kinase (p-JNK)/JNK, and p-P38/P38 were all increased, while the levels of p-extracellular signal-regulated kinase (p-ERK)/ERK were decreased. However, LDHJ granules and its three active substances effectively reversed these changes. Furthermore, the three active substances reduced the increases in the intracellular calcium concentration ([Ca²⁺]i) and ROS levels and attenuated the dissipation of the mitochondria membrane potential in the TCDC-induced primary hepatocytes. The opposite results were obtained from the TCDC-induced primary hepatocytes treated with an agonist of CaSR (GdCl3) plus forsythoside-A, emodin or chlorogenic acid. Based on the results from in vivo and in vitro studies, LDHJ functions as an antagonist of CaSR to regulate hepatocyte apoptosis in cholestasis through the mitochondrial pathway and mitogen-activated protein kinase pathway.
... E-17G-induced endocytosis of BSEP and MRP2 is associated with decreased bile flow and biliary excretion of BSEP/MRP2 substrates and p38 inhibition prevents the internalization of BSEP/MRP2 [95]. P38 MAPK plays a role in oxidative stress-induced retrieval of BSEP and MRP2 [100]. TLC activates p38 MAPK [101] and decreases PM localization of MRP2 in hepatocytes [27,102]. ...
... Researchers have considered redox status imbalance to be a common trigger of the signaling pathway leading to MRP2 internalization [27]. Previous studies showed that Ca 2+ -dependent PKC-MAPK pathways, including the ERK, JNK, and P38-type signaling pathways, participate in hepatocanalicular dysfunction and cholestasis by promoting F-actin rearrangement and further endocytic internalization of canalicular transporters in response to tert-butyl hydroperoxide-induced oxidative stress [28][29][30]. Moreover, estradiol-17β-D-glucuronide (E17G) activates the Fig. 3 Rifampicin (RFP) activated the PKC-ERK/JNK/p38 (protein kinase C-extracellular signal-regulated kinase/c-JUN N-terminal kinase/p38) and PI3K (phosphoinositide 3-kinase) signaling pathways via oxidative stress. ...
Article
It was reported that antituberculosis medicines could induce liver damage via oxidative stress. In this study, we investigated the effects of rifampicin (RFP) on the membrane expression of multidrug resistance-associated protein 2 (MRP2) and the relationship between oxidative stress and RFP-induced endocytosis of MRP2 in HepG2 cells. We found that RFP (12.5–50 μM) dose-dependently decreased the expression and membrane localization of MRP2 in HepG2 cells without changing the messenger RNA level. RFP (50 μM) induced oxidative stress responses that further activated the PKC-ERK/JNK/p38 (protein kinase C-extracellular signal-regulated kinase/c-JUN N-terminal kinase/p38) and PI3K (phosphoinositide 3-kinase) signaling pathways in HepG2 cells. Pretreatment with glutathione reduced ethyl ester (2 mM) not only reversed the changes in oxidative stress indicators and signaling molecules but also diminished RFP-induced reduction in green fluorescence intensity of MRP2. We conducted co-immunoprecipitation assays and revealed that a direct interaction existed among MRP2, clathrin, and adaptor protein 2 (AP2) in HepG2 cells, and their expression was clearly affected by the changes in intracellular redox levels. Knockdown of clathrin or AP2 with small interfering RNA attenuated RFP-induced decreases of membrane and total MRP2. We further demonstrated that RFP markedly increased the ubiquitin–proteasome degradation of MRP2 in HepG2 cells, which was mediated by the E3 ubiquitin ligase GP78, but not HRD1 or TEB4. In conclusion, this study demonstrates that RFP-induced oxidative stress activates the PKC-ERK/JNK/p38 and PI3K signaling pathways that leads to clathrin-dependent endocytosis and ubiquitination of MRP2 in HepG2 cells, which provides new insight into the mechanism of RFP-induced cholestasis.
... Cholestasis represents an intensively studied field of research in toxicological sciences (Starokozhko et al. 2017;Miszczuk et al. 2015;Toledo et al. 2017;Vinken et al. 2017;Leist et al. 2017). The biliary tract has been shown to consist of three anatomical domains that respond differently to cholestasis (Vartak et al. 2016): large ducts widen their diameters, interlobular ducts respond by branching and looping thereby forming a denser mesh, while the most upstream domain, the bile canaliculi, become wider and form spiny protrusions into hepatocytes (Vartak et al. 2016;Jansen et al. 2017). ...
Article
Drug-induced liver injury (DILI) is an adverse reaction to medications and other xenobiotics that leads to liver dysfunction. Based on differential clinical patterns of injury, DILI is classified into hepatocellular, cholestatic, and mixed types; although hepatocellular DILI is associated with inflammation, necrosis, and apoptosis, cholestatic DILI is associated with bile plugs and bile duct paucity. Ursodeoxycholic acid (UDCA) has been empirically used as a supportive drug mainly in cholestatic DILI, but both curative and prophylactic beneficial effects have been observed for hepatocellular DILI as well, according to preliminary clinical studies. This could reflect the fact that UDCA has a plethora of beneficial effects potentially useful to treat the wide range of injuries with different etiologies and pathomechanisms occurring in both types of DILI, including anticholestatic, antioxidant, anti-inflammatory, antiapoptotic, antinecrotic, mitoprotective, endoplasmic reticulum stress alleviating, and immunomodulatory properties. In this review, a revision of the literature has been performed to evaluate the efficacy of UDCA across the whole DILI spectrum, and these findings were associated with the multiple mechanisms of UDCA hepatoprotection. This should help better rationalize and systematize the use of this versatile and safe hepatoprotector in each type of DILI scenarios.
Article
Quantitative measurement of the degree of hepatic ischemia-reperfusion injury (IRI) is crucial for developing therapeutic strategies for its treatment. We hypothesized that clearance of fluorescent dye through bile metabolism may reflect the degree of hepatic IRI. In this study, we investigated sodium fluorescein clearance kinetics in blood and bile for quantifying the degree of hepatic IRI. Warm ischemia times (WITs) of 0, 30, or 60 min followed by 1 h or 4 h of reperfusion, were applied to the median and lateral lobes of the liver in Sprague Dawley rats. Subsequently, 2 mg/kg of sodium fluorescein was injected intravenously, and blood and bile samples were collected over 60 min to measure fluorescence intensities. The bile-to-plasma fluorescence ratios demonstrated an inverse correlation with WIT and were distinctly lower in the 60-min WIT group than in the control or 30-min WIT groups. Bile-to-plasma fluorescence ratios displayed superior discriminability for short versus long WITs when measured 1 h after reperfusion versus 4 h. We conclude that the bile-to-blood ratio of fluorescence after sodium fluorescein injection has the potential to enable quantification of hepatic IRI severity.
Article
Significance: Most hepatopathies are primarily or secondarily cholestatic in nature. Oxidative stress (OS) is a frequent trait among them, and impairs the machinery to generate bile by triggering endocytic internalization of hepatocellular transporters, thus causing cholestasis. This is critical, since it leads to accelerated transporter degradation, which could explain the common post-transcriptional downregulation of transporter expression in human cholestatic diseases. Recent Advances: The mechanisms involved in OS-induced hepatocellular transporter internalization are being revealed. Filamentous actin (F-actin) cytoskeleton disorganization and/or detachment of crosslinking actin proteins that afford transporter stability have been characterized as causal factors. Activation of redox-sensitive signaling pathways leading to changes in phosphorylation status of these structures is involved, including Ca2+-mediated activation of "classical" and "novel" protein kinase C (PKC) isoforms or redox-signaling cascades downstream of NADPH oxidase. Critical Issues: Despite the well-known occurrence of hepatocellular transporter internalization in human hepatopathies, the cholestatic implications of this phenomenon have been overlooked. Accordingly, no specific treatment has been established in the clinical practice for its prevention/reversion. Future Directions: We need to improve our knowledge on the pro-oxidant triggering factors and the multiple signaling pathways that mediate this oxidative injury in each cholestatic hepatopathy, so as to envisage tailor-made therapeutic strategies for each case. Meanwhile, administration of antioxidants or heme oxygenase-1 induction to elevate the hepatocellular levels of the endogenous scavenger bilirubin are promising alternatives that need to be re-evaluated and implemented. They may complement current treatments in cholestasis aimed to enhance transcriptional carrier expression, by providing membrane stability to the newly synthesized carriers. Antioxid. Redox Signal. 35, 808-831.
Article
The bile salt export pump (BSEP/ABCB11) is located on the apical membrane and mediates the secretion of bile salts from hepatocytes into the bile. BSEP-mediated bile salt efflux is the rate-limiting step of bile salt secretion and the main driving force of bile flow. BSEP drives and maintains the enterohepatic circulation of bile salts. In recent years, research efforts have been focused on understanding the physiological and pathological functions and regulatory mechanisms of BSEP. These studies elucidated the roles of farnesoid X receptor (FXR), AMP-activated protein kinase (AMPK), liver receptor homolog-1(LRH-1) and nuclear factor erythroid 2-related factor 2 (Nrf-2) in BSEP expression and discovered some regulatory factors which participate in its post-transcriptional regulation. A series of liver diseases have also been shown to be related to BSEP expression and dysfunction, such as cholestasis, drug-induced liver injury, and gallstones. Here, we systematically review and summarize recent literature on BSEP structure, physiological functions, regulatory mechanisms, and related diseases.
Article
Full-text available
Increased phosphorylation of myosin light chain (MLC) is necessary for the dynamic membrane blebbing that is observed at the onset of apoptosis. Here we identify ROCK I, an effector of the small GTPase Rho, as a new sub-strate for caspases. ROCK I is cleaved by caspase-3 at a conserved DETD1113/G sequence and its carboxy-terminal inhibitory domain is removed, resulting in deregulated and constitutive kinase activity. ROCK proteins are known to regulate MLC-phosphorylation, and apoptotic cells exhibit a gradual increase in levels of phosphorylated MLC concomitant with ROCK I cleavage. This phosphorylation, as well as membrane blebbing, is abrogated by inhibition of caspases or ROCK proteins, but both processes are independent of Rho activity. We also show that expression of active truncated ROCK I induces cell blebbing. Thus, activation of ROCK I by caspase-3 seems to be responsible for bleb formation in apoptotic cells.
Article
Full-text available
Background: Ethanol-induced gut barrier disruption is associated with several gastrointestinal and liver disorders. Aim: Since human data on effects of moderate ethanol consumption on intestinal barrier integrity and involved mechanisms are limited, the objectives of this study were to investigate effects of a single moderate ethanol dose on small and large intestinal permeability and to explore the role of mitogen activated protein kinase (MAPK) pathway as a primary signaling mechanism. Methods: Intestinal permeability was assessed in 12 healthy volunteers after intraduodenal administration of either placebo or 20 g ethanol in a randomised cross-over trial. Localization of the tight junction (TJ) and gene expression, phosphorylation of the MAPK isoforms p38, ERK and JNK as indicative of activation were analyzed in duodenal biopsies. The role of MAPK was further examined in vitro using Caco-2 monolayers. Results: Ethanol increased small and large intestinal permeability, paralleled by redistribution of ZO-1 and occludin, down-regulation of ZO-1 and up-regulation of myosin light chain kinase (MLCK) mRNA expression, and increased MAPK isoforms phosphorylation. In Caco-2 monolayers, ethanol increased permeability, induced redistribution of the junctional proteins and F-actin, and MAPK and MLCK activation, as indicated by phosphorylation of MAPK isoforms and myosin light chain (MLC), respectively, which could be reversed by pretreatment with either MAPK inhibitors or the anti-oxidant L-cysteine. Conclusions: Administration of moderate ethanol dosage can increase both small and colon permeability. Furthermore, the data indicate a pivotal role for MAPK and its crosstalk with MLCK in ethanol-induced intestinal barrier disruption. Trial registration: ClinicalTrials.gov NCT00928733.
Article
Full-text available
Oxidative stress (OS) is a common event in most hepatopathies, leading to mitochondrial permeability transition pore (MPTP) formation and further exacerbation of both OS from mitochondrial origin and cell death. Intracellular Ca(2+) increase plays a permissive role in these events, but the underlying mechanisms are poorly known. We examined in primary cultured rat hepatocytes whether the Ca(2+)/calmodulin (CaM)-dependent protein kinase II (CaMKII) signaling pathway is involved in this process, by using tert-butyl hydroperoxide (tBOOH) as a pro-oxidant, model compound. tBOOH (500 μM, 15 min) induced MPTP formation, as assessed by measuring mitochondrial membrane depolarization as a surrogate marker, and increased lipid peroxidation in a cyclosporin A (CsA)-sensitive manner, revealing the involvement of MPTPs in tBOOH-induced radical oxygen species (ROS) formation. Intracellular Ca(2+) sequestration with BAPTA/AM, CaM blockage with W7 or trifluoperazine, and CaMKII inhibition with KN-62 all fully prevented tBOOH-induced MPTP opening and reduced tBOOH-induced lipid peroxidation to a similar extent to CsA, suggesting that Ca(2+)/CaM/CaMKII signaling pathway fully mediates MPTP-mediated mitochondrial ROS generation. tBOOH-induced apoptosis, as shown by flow cytometry of annexin V/propidium iodide, mitochondrial release of cytochrome c, activation of caspase-3 and increase in the Bax-to-Bcl-xL ratio, and the Ca(2+)/CaM/CaMKII signaling antagonists fully prevented these effects. Intramitochondrial CaM and CaMKII were partially involved in tBOOH-induced MPTP formation, since W7 and KN-62 both attenuated the tBOOH-induced, MPTP-mediated swelling of isolated mitochondria. We concluded that Ca(2+)/CaM/CaMKII signaling pathway is a key mediator of OS-induced MPTP formation and the subsequent exacerbation of OS from mitochondrial origin and apoptotic cell death.
Chapter
Principles and ApplicationsExperimental ProceduresAppraisal of Enzymic Methods
Article
The transport properties of three different synthetically prepared fluorescent conjugated bile acid analogs (FBA), all with the fluorophore on the side chain,were determined using isolated rat hepatocytes and hepatocyte couplets. The compounds studied were cholylglycylamidofluorescein (CGamF), cholyl-(N epsilon-nitrobenzoxadiazolyl [NBD])-lysine (C-NBD-L), and chenodeoxycholyl-(N epsilon-NBD)-lysine (CDC-NBD-L). When hepatocytes were incubated at 37 degrees C with 0.3 mu mol/L of FBA and 0.15 mol/L of Na+, cell fluorescence increased linearly with time at a rate (U/min) of 7.8 +/- 0.5 for CGamF, 7.2 +/- 0.3 for C-NBD-L, and 13.7 +/- 1.0 for CDC-NBD-L (mean, +/- SE; n = 40 to 90). Uptake was concentration dependent for concentrations less than 20 mu mol/L and was saturable. The Michaelis constant (K-m) value (mu mol/L) for CGamF was 10.8, for C-NBD-L was 3.8, and for CDC-NSD-L was 3.0. In the absence of Na+, the uptake rate was decreased by 50% for CGamF and by 38% for C-NBD-L; but uptake of CDC-NBD-L was unchanged and thus Na+ independent. Cellular uptake of all three derivatives was specific to hepatocytes and was absent in several nonhepatocyte cell lines, For CGamF and C-NBD-L, both Na+-dependent and Na+-independent uptake was inhibited by 200-fold excess concentrations of cholyltaurine, dehydrocholyltaurine, and cholate, but for CDC-NBD-L, these nonfluorescent bile acids did not inhibit initial uptake. The intracellular fluorescence of CGamF was strongly pH dependent at an excitation wavelength of 495 nm, but pH independent at 440 nm excitation. In contrast, intracellular fluorescence of C-NBD-L and CDC-NBD-L was pH independent. All three FBA were secreted into the canalicular space of approximately 50% to 60% of couplets. Cellular adenosine triphosphate (ATP) depletion with either CN- or atractyloside inhibited secretion of ah three FBA. The multispecific organic anion transporter (MOAT) inhibitor, chlorodinitrobenzene, blocked secretion of fluorescent MOAT substrates at a concentration of 1 mu mol/L. At this concentration it did not affect secretion of the three FBA. At higher concentrations, chlorodinitrobenzene partially inhibited the canalicular secretion of CGamF but not the other two FBA. In conclusion, all three FBA are secreted by canalicular membrane bile acid transporters, but the sinusoidal uptake characteristics of CGamF and C-NBD-L are more similar than those of CDC-NBD-L to the transport properties of cholyltaurine. Therefore, C-NBD-L appears to be the best of the three for studies of conjugated trihydroxy-bile acid transport in hepatocytes.
Article
Bile acids, synthesized from cholesterol, are known to produce beneficial as well as toxic effects in the liver. The beneficial effects include choleresis, immunomodulation, cell survival, while the toxic effects include cholestasis, apoptosis and cellular toxicity. It is believed that bile acids produce many of these effects by activating intracellular signaling pathways. However, it has been a challenge to relate intracellular signaling to specific and at times opposing effects of bile acids. It is becoming evident that bile acids produce different effects by activating different isoforms of phosphoinositide 3-kinase (PI3K), Protein kinase Cs (PKCs), and mitogen activated protein kinases (MAPK). Thus, the apoptotic effect of bile acids may be mediated via PI3K-110γ, while cytoprotection induce by cAMP-GEF pathway involves activation of PI3K-p110α/β isoforms. Atypical PKCζ may mediate beneficial effects and nPKCε may mediate toxic effects, while cPKCα and nPKCδ may be involved in both beneficial and toxic effects of bile acids. The opposing effects of nPKCδ activation may depend on nPKCδ phosphorylation site(s). Activation of ERK1/2 and JNK1/2 pathway appears to mediate beneficial and toxic effects, respectively, of bile acids. Activation of p38α MAPK and p38β MAPK may mediate choleretic and cholestatic effects, respectively, of bile acids. Future studies clarifying the isoform specific effects on bile formation should allow us to define potential therapeutic targets in the treatment of cholestatic disorders.
Article
Severe influenza infections are featured by acute lung injury, a syndrome of pulmonary microvascular leak. A growing number of evidences have shown that the pulmonary microvascular endothelial cells (PMVEC) are critical target of influenza virus, promoting microvascular leak. It is reported that there are multiple mechanisms by which influenza virus could elicit increased pulmonary endothelial permeability, in both direct and indirect manners. Ezrin/radixin/moesin family proteins, the linkers between plasma membrane and actin cytoskeleton, have been reported to be involved in cell adhesion, motility and may modulate endothelial permeability. Studies have also shown that ERM is phosphorylated in response to various stimuli via p38MAPK, Rho/ROCK or PKC pathways. However, it is unclear that whether influenza infection could induce ERM phosphorylation and its relocalization. In the present study, we have found that there are cytoskeletal reorganization and permeability increases in the course of influenza virus infection, accompanied by upregulated levels of p-ERM. p-ERM's aggregation along the periphery of PMVEC upon influenza virus infection was detected via confocal microscopy. Furthermore, we sought to determine the role of p38MAPK, Rho/ROCK and PKC pathways in ERM phosphorylation as well as their involvement in influenza virus-induced endothelial malfunction. The activation of p38MAPK, Rho/ROCK and PKC pathways upon influenza virus stimulation were observed, as evidenced by the evaluation of phosphorylated p38 (p-p38), phosphorylated MKK (p-MKK) in p38MAPK pathway, ROCK1 in Rho/ROCK pathway and phosphorylated PKC (p-PKC) in PKC pathway. We also showed that virus-induced ERM phosphorylation was reduced by using p38MAPK inhibitor, SB203580 (20Um), Rho/ROCK inhibitor, Y27632 (20uM), PKC inhibitor, LY317615 (10uM). Additionally, influenza virus-induced F-actin reorganization and hyperpermeability were attenuated by pretreatment with SB203580, Y27632 and LY317615. Taken together, we provide the first evidence that p38MAPK, Rho/ROCK and PKC are involved in influenza-induced cytoskeletal changes and permeability increases in PMVEC via phosphorylating ERM.
Article
Bilirubin is an endogenous antioxidant with cytoprotective properties, and several studies highlight its potential in the treatment of pro-oxidant diseases. We demonstrated that oxidative stress (OS), a key feature in most hepatopathies, induces cholestasis by actin cytoskeleton disarrangement and further endocytic internalization of key canalicular transporters, such as the bile salt export pump (Bsep) and the multidrug resistance-associated protein 2 (Mrp2) . Here, we evaluated the capability of physiological concentrations of unconjugated bilirubin (UB) to limit OS and the impairment in biliary secretory function induced by the model pro-oxidant agent, tert-butylhydroperoxide (tBuOOH). UB fully prevented the formation of reactive oxygen species and membrane lipid peroxidation induced by tBuOOH in isolated rat hepatocytes. In the isolated rat hepatocyte couplet model, UB (17.1 μM) prevented the endocytic internalization of Bsep and Mrp2 and the impairment in their secretory function induced by tBuOOH. UB also prevented actin disarrangement, as evaluated by both plasma membrane bleb formation and actin fluorescent staining. Finally, UB prevented tBuOOH-induced cPKC activation. Experiments in isolated perfused rat livers showed that UB prevents the increase in oxidized glutathione biliary excretion and the drop in bile flow and the biliary excretion of specific Bsep and Mrp2 substrates. We conclude that physiological concentrations of UB are sufficient to prevent the biliary secretory failure induced by OS, by counteracting actin disarrangement and the consequent internalization of canalicular transporters relevant to normal bile formation. This reveals an important role for UB in preserving biliary secretory function under OS conditions.