Icariin attenuates the severity of cerulein‑induced acute pancreatitis by inhibiting p38 activation in mice

  • Authors:
    • Dong‑Uk Kim
    • Gi‑Sang Bae
    • Myoung‑Jin Kim
    • Ji‑Won Choi
    • Dong‑Gu Kim
    • Ho‑Joon Song
    • Sung‑Joo Park
  • View Affiliations

  • Published online on: August 16, 2019     https://doi.org/10.3892/ijmm.2019.4312
  • Pages: 1563-1573
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Acute pancreatitis (AP) is an inflammatory disease of the pancreas. Icariin (ICA), a flavonoid glycoside, has been reported to have several pharmacological effects; however, the anti‑inflammatory effects of ICA against AP require further study. Therefore, we aimed to investigate the effect of ICA on cerulein‑induced AP. In the present study, AP was induced by intraperitoneally administering a supramaximal concentration of cerulein (50 µg/kg/h) for 6 h. ICA was also administered intraperitoneally, and mice were sacrificed 6 h after the final cerulein injection. Blood samples were collected to determine serum amylase and lipase levels. The pancreas and lung were rapidly removed for histological examination, and the analysis of myeloperoxidase activity. In addition, reverse transcription‑quantitative polymerase chain reaction was conducted to analyze the expression of inflammatory cytokines in pancreatic tissues. Our results revealed that the administration of ICA prevented an increase in the pancreas weight/body weight ratio of mice and serum digestive enzyme levels. ICA treatment also inhibited cerulein‑induced histological injury and neutrophil infiltration of the pancreas and lung. In addition, ICA suppressed the production of pro‑inflammatory cytokines, including interleukin (IL)‑1β, IL‑6 and tumor necrosis factor‑α in the pancreas. Furthermore, ICA administration was observed to inhibit p38 activation during cerulein‑induced AP. Inhibition of p38 activation resulted in alleviated pancreatitis. Collectively, our results suggested that ICA exhibits anti‑inflammatory effects in cerulein‑induced AP via the inhibition of p38.

Introduction

Acute pancreatitis (AP) is a sudden inflammation of the pancreas and the most common reason for hospitalization among non-malignant gastrointestinal disorders in the United States (1,2). AP is commonly triggered by various risk factors, such as alcohol abuse, duct obstruction, obesity, smoking, drug toxicity and metabolic disorders (3,4). This condition arises from pancreatic acinar cell injury by the activation of proteolytic pancreatic enzymes, including amylase, lipase, and trypsin (5). Injured acinar cells produce pro-inflammatory cytokines such as interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α (6). These cytokines damage the acinar cell, causing systemic inflammatory response syndrome, which is a subset of cytokine storm (5-8). Although AP is a critical disease, few studies have investigated effective therapeutic treatments for AP.

Icariin (ICA) is a natural flavonoid glycoside isolated from plants in the Epimedium genus (Yin-Yang-Huo) of the Berberidaceae family and a well-known traditional Chinese medicine for treating male sexual dysfunctions, such as erectile dysfunction and premature ejaculation (9,10). ICA, the major and active compound of Epimedium, has been associated with many biological and pharmacological effects, such as anti-osteoporotic (11), antioxidative (12), anti-inflammatory (13), anti-cancer (14), anti-depressant (15) and neuro-protective effects (16). Previous studies have reported that ICA ameliorates various diseases, including atherosclerosis, asthma and erectile dysfunction (17-19); however, the therapeutic effects of ICA in acute pancreatitis require further investigation.

The purpose of this study was to investigate the effects of ICA against cerulein-induced AP. Pancreas weight/body weight (PW/BW) ratio, serum amylase and lipase levels, histological appearance of the pancreas and lung, myeloperoxidase (MPO) activity and pro-inflammatory cytokine levels were evaluated to determine the attenuating effects of ICA on the severity of cerulein-induced AP. Furthermore, we evaluated the activation of mitogen activated protein kinases (MAPKs, indicated by MAPKs phosphorylation) and nuclear factor κB (NF-κB, indicated by Iκ-Bα degradation) to determine the inhibitory mechanisms of ICA on AP.

Materials and methods

Chemicals and reagents

ICA, dimethyl sulfoxide (DMSO), cerulein, NaCl, hexadecyl-trimethyl-ammonium bromide (HTAB), and Tris-HCl were purchased from Sigma-Aldrich (Merck KGaA). ICA stock solutions were using DMSO. Easy-Blue™ total RNA extraction kit was purchased from iNtRON Biotechnology. Anti-phosphorylated extracellular signal-regulated kinase (ERK)1/2 (1:1,000; cat. no. 9101L), phosphorylated c-Jun N-terminal kinase (JNK 1/3; 1:1,000; cat. no. 9251L), and p38 (1:1,000; cat. no. 9211L) antibodies and SB203580 (p38 inhibitor) were purchased from Cell signaling Technology, Inc. Total MAPK antibodies against ERK 1/2 (1:1,000; sc-93), JNK 1/3 (1:1,000; sc-474) and p38 (1:1,000; sc-535), inhibitory κ-Bα (Iκ-Bα; 1:1,000; sc-371), and β-actin antibody (1:1,000; sc-1615) were obtained from Santa Cruz Biotechnology, Inc. In addition, horseradish peroxidase (HRP)-conjugated secondary antibodies, including chicken anti-rabbit IgG-HRP (1:5.000; sc-516087) and chicken anti-goat IgG-HRP (1:10.000; sc-516086) were purchased from Santa Cruz Biotechnology, Inc.

Ethical aspects

All experiments were performed in accordance with the animal care regulations of Wonkwang University set forth and approved by the Wonkwang University Animal Ethics Committee.

Animal models

All experiments were performed according to the protocols approved by the Animal Care Committee of Wonkwang University. C57BL/6 mice (n=324, 6-8 weeks old, female, weighing 15-20 g), were purchased from Orient Bio. All animals were bred and housed in standard shoebox cages in a climate-controlled room with an ambient temperature of 23±2°C under a 12 h light/dark cycle for 7 days. Animals were fed standard laboratory chow, and water ad libitum. Mice were randomly assigned to control or experimental groups.

Experimental design

AP was induced via six intraperitoneal injections (i.p.) of cerulein (50 µg/kg) at intervals of 1 h. Control animals were administered DMSO under the same conditions. In the pre-treatment groups, ICA (5, 10 or 20 mg/kg, i.p.), SB203580 (1 mg/kg i.p., n=6) or DMSO (i.p.) were administered 1 h before the first cerulein injection, in the experimental (n=6) and the control group (n=6) respectively. Mice were sacrificed 6 h after the last cerulein injection. In the post-treatment groups, ICA (20 mg/kg, n=6) or DMSO (control group, n=6) were administered 1, 3 and 5 h after the first cerulein injection. Mice were sacrificed 6 h after the last cerulein injection. Blood (withdrawn from the heart), the pancreas, and the lung were rapidly removed and stored at −80°C for further analysis. For the detection of MAPKs and NF-κB, mice were pretreated with ICA (10 or 20 mg/kg) or DMSO 1 h before the intraperitoneal injection of cerulein (50 µg/kg), and sacrificed at 0, 15, 30 and 60 min after cerulein injection, respectively. For immunofluorescence, mice were pretreated with ICA (20 mg/kg) or DMSO 1 h before the intraperitoneal injection of cerulein (50 µg/kg), and sacrificed at 30 min after cerulein injection. Mice were sacrificed via CO2 asphyxiation. The CO2 flow rate displaced 20% of the cage volume per minute. To ensure death following CO2 asphyxiation, cervical dislocation was performed. The pancreas was rapidly removed and stored at −80°C for further analysis.

Histological analysis

The appearance of the entire pancreas and the semi-quantitative index based on edema and inflammation were examined from each treatment group. The pancreas and lung tissues were fixed in 4% formalin solution at room temperature overnight, embedded in paraffin, cut into 4 µm sections, stained with hematoxylin for 8 min and eosin for 2 min overnight. Samples were examined under a light microscope. Tissue sections from each sample, representing a minimum of 100 fields were examined and scored on a scale of 0-3 (0 corresponding to normal appearance and 3 corresponding to severe disease), based on the presence of interstitial edema and interstitial inflammation. In the assessment of the lung, the sections were examined for the presence of wall thickening and inflammation. The levels of edema, and pro-inflammatory cell infiltration were scored by pathologists in a blinded manner who were unware of the study design; scores were set based on a scale of 0 (normal) to 3 (severe) (20). In brief, a score of 0 presented the absence of edema and inflammatory cells, 1 indicated focal edema between lobules and 10 inflammatory cells; 2 suggested diffuse edema between lobules and 10-20 inflammatory cells, while a score of 3 reflected diffuse edema between acinar cells and >20 inflammatory cells.

Measurement of serum amylase and lipase levels

Blood samples for the determination of serum amylase and lipase levels were obtained 6 h after the last injection of cerulein. Mice were sacrificed via CO2 asphyxiation followed by cervical dislocation; blood samples were then withdrawn from the heart. Amylase and lipase activities were determined by an assay kit of Bio Assay Systems.

MPO activity

Sequestration of neutrophils within the pancreas and lung was evaluated by measuring the MPO activity within tissues. Briefly, tissue samples were weighed, homogenized with 20 mM phosphate buffer (pH 7.4), and centrifuged at 10,000 × g for 10 min, 4°C. The pellets were then re-suspended in 50 mM phosphate buffer (pH 6.0) containing 0.5 % HTAB. The samples were centrifuged at 10,000 × g for 5 min, 4°C, and mixed with 80 mM sodium phosphate buffer (pH 5.4) containing 1.6 mM TMB. The mixture was incubated at 37°C for 110 sec, and the reaction was terminated with 2 M H2SO4. Tissue MPO activity was determined by measuring the absorbance at 450 nm with a Versa Max microplate reader (Molecular Devices, LLC) and was expressed as U/mg protein.

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

mRNA transcripts of mouse pancreatic tissues were analyzed by RT-qPCR. Total RNA was isolated from the mouse pancreas using Easy-Blue™ and subjected to RT using ABI cDNA synthesis kit according to the manufacturer's protocols (Applied Biosystems; Thermo Fisher Scientific, Inc.). For each sample, triplicate test reactions and a control reaction lacking reverse transcriptase were analyzed for the expression of the gene of interest, and the results were normalized to those of the 'housekeeping' hypoxanthine phosphoribosyltransferase (HPRT) mRNA. qPCR was performed using a standard protocol from the TaqMan™ Universal Master Mix II, no UNG (Applied Biosystems; Thermo Fisher Scientific, Inc.) on StepOnePlus™ Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.). qPCR was performed at 50°C for 2 min and 95°C for 10 min, followed by 60 cycles of amplification at 95°C for 10 sec and 60°C for 30 sec. Forward, reverse, and probe oligonucleotide primers for multiplex real-time TaqMan PCR were purchased from Applied Biosystems (Thermo Fisher Scientific, Inc.). The forward, reverse and probe oligonucleotide primers for multiplex real-time TaqMan PCR were as follows: For mouse TNF-α forward, 5′-TCT CTT CAA GGG ACA AGG CTG-3′, reverse, 5′-ATA GCA AAT CGG CTG ACG GT-3′; mouse IL-1β forward, 5′-TTG ACG GAC CCC AAA AGA T-3′, reverse, 5′-GAA GCT GGA TGC TCT CAT CTG-3′; mouse IL-6 forward, 5′-TTC ATT CTC TTT GCT CTT GAA TTA GA-3′, reverse, 5′-GTC TGA CCT TTA GCT TCA AAT CCT-3′; mouse HPRT (forward, 5′-GAC CGG TCC CGT CAT GC-3′; reverse, 5′-CAT AAC CTG GTT CAT CAT CGC TAA-3′).

Western blotting

Pancreatic tissues were homogenized and lysed on ice with radioimmunoprecipitation assay lysis buffer (iNtRON Biotechnology). Then, the lysates were boiled in 62.5 mM Tris-HCl buffer, pH 6.8, containing 2% SDS, 20% glycerol, and 10% 2-mercaptoethanol. Proteins were separated on a 10% SDS-PAGE and transferred to a nitrocellulose membrane. The membrane was blocked with 5% skim milk in phosphate-buffered saline with Tween-20 for 2 h at room temperature, followed by incubation with primary antibodies overnight at 4°C. After washing three times, the membrane was incubated with secondary antibodies for 1 h at room temperature. The proteins were visualized using an enhanced chemiluminescence detection system (GE Healthcare) according to the manufacturer's protocols. The bands were detected and quantified by using Quantity One software (version 4.5.2; Bio-Rad Laboratories, Inc.).

Immunofluorescence

Immunofluorescence for the analysis of phosphorylated p38 was performed on pancreas tissue. The pancreas tissues were embedded in frozen section compound, cut into 9 µm sections, fixed in 100% methanol at −20°C for 5 min The tissues were incubated with the primary antibodies against phosphorylated p38 (1:250) at 4°C overnight followed by the fluorescence labeled secondary antibodies Alexa Fluor® 488 goat anti rabbit (1:2,000; A27034; Invitrogen; Thermo Fisher Scientific, Inc.) at room temperature for 2 h. Nuclei were counter stained with DAPI (5 ng/ml) at room temperature for 5 min. Stained sections were visualized using a confocal laser microscope (Olympus Corporation).

Statistical analysis

Data were expressed as the mean ± standard error of the mean. Statistical significance of intergroup differences was evaluated using two-way analysis of variance, with time and dose as variables, followed by a Duncan's post-hoc test. All statistical analyses were performed using SPSS version 10.0 statistical analysis software (SPSS, Inc.). P<0.05 was considered to indicate a statistically significant difference. All experiments were conducted in triplicate.

Results

Effects of ICA on the severity of cerulein-induced AP

To evaluate the prophylactic effects of ICA against cerulein-induced AP, histological examination of the pancreas and lung was carried out. In the DMSO-treated mice, histological features of the pancreas and lung showed typical normal architecture. In the DMSO-treated mice with AP, histological features of damaged pancreas tissue were observed, as characterized by interstitial edema and inflammatory cell infiltration. However, treatment with ICA ameliorated the pancreatic histological damage in a dose-dependent manner (Fig. 1B). In addition, histological features of the lung in cerulein-induced AP were observed; lung injury was characterized by alveolar wall thickening and inflammatory cell infiltration. Nevertheless, lung injury was significantly reduced by treatment with ICA compared with the control (Fig. 1D and E). In addition, to assess the effects of ICA on the severity of cerulein-induced AP, PW/BW ratio, serum amylase and lipase levels were measured. In the cerulein-induced AP mice, the PW/BW ratio was increased than in the control mice due to pancreatic edema; however, cerulein-induced increases in the PW/BW ratio was inhibited by ICA treatment in a dose-dependent manner (Fig. 1F). Furthermore, the levels of serum digestive enzymes, including amylase and lipase were significantly increased in the cerulein-induced AP mice than in the control mice; the levels of both enzymes were decreased by ICA treatment (Fig. 1G and H).

Effects of ICA on MPO activity and pancreatic cytokine production in cerulein-induced AP

Neutrophil migration into target tissue is increased when inflammation occurs (21). Therefore, we examined MPO activity in the pancreas and lung after the induction of AP, in order to measure neutrophil infiltration. As shown in Fig. 2A, MPO activity was significantly higher in cerulein-induced AP mice than in control mice; however, the increase of MPO activity was inhibited by ICA treatment in the pancreas and the lung in a dose dependent manner. Of note, the production of pro-inflammatory cytokines, including IL-1β, IL-6 and TNF-α were determined to increase during cerulein-induced AP (22). Thus, we examined the pancreatic mRNA levels of IL-1β, IL-6 and TNF-α. As shown in Fig. 2B, the expression levels of these pro-inflammatory cytokines significantly increased in cerulein-induced AP mice compared with the control; however, ICA treatment inhibited IL-1β, IL-6 and TNF-α expression in a dose-dependent manner.

Effects of ICA on the activation of MAPK and NF-κB in cerulein-induced AP

To investigate the inhibitory mechanisms of ICA on AP, the activation of MAPKs and NF-κB was examined in the pancreas. Mice were administered ICA (20 mg/kg) or DMSO for 1 h and then stimulated with cerulein for 0, 15, 30 and 60 min. Cerulein treatment triggered the phosphorylation of MAPKs and the degradation of Iκ-Bα. However, administration of ICA inhibited the phosphorylation of p38, but not of ERK 1/2, JNK, and degradation of Iκ-Bα (Fig. 3A). Consistent with Fig. 3A, p38 phosphorylation was also inhibited by ICA as determined via immunofluorescence staining (Fig. 3C). To determine whether the inhibition of p38 activation was responsible for the effects of ICA, we administered ICA at different concentrations (10 or 20 mg/kg). As shown in Fig. 3D and E, the phosphorylation of p38 was inhibited by ICA in a dose-dependent manner.

Effects of P38 on the severity in cerulein-induced AP

To determine whether inhibition of p38 could affect the reduction of inflammatory responses, a p38 inhibitor (SB203580, 1 mg/kg) was employed, after which the severity of AP was evaluated. As presented in Fig. 4, inhibition of p38 by SB203580 suppressed the histological damage to the pancreas and lung, and reduced the PW/BW ratio, and the levels of amylase and lipase compared with AP mice (Fig. 4). These observations were similar to the effects of ICA (Fig. 1). Additionally, administration of SB203580 significantly reduced MPO activity, and the mRNA levels of IL-1β, IL-6 and TNF-α compared with in AP mice (Fig. 5).

Therapeutic effects of ICA in cerulein-induced AP

To examine the therapeutic effects of ICA in cerulein-induced AP, we applied ICA after the onset of AP. Post-treatment of ICA at 1 h but not 3 and 5 h after first cerulein injection significantly reduced the PW/BW ratio, serum amylase and lipase activities, and histological injury of the pancreas and lung compared with the AP mice, which suggests that ICA could exhibit therapeutic properties in the early phase of AP (Fig. 6).

Discussion

AP is an acute inflammatory disease that occurs in the pancreas, and its incidence is ~300 in 1 million (23). The symptoms of AP vary from mild inflammation of the pancreas to severe inflammatory reactions with multiple organ failure (24). The mortality rate of mild AP is ~10% and most of them are self-healing without complications (25). The mortality rate of severe AP is about 20-30%, which is caused by complications of multiple organ failure, such as liver dysfunction and pulmonary dysfunction (26). In general, AP is treated via temporary symptomatic relief, including antibiotics or analgesics, and in severe cases, the necrotic pancreatic tissue is removed through surgery. However, current therapies have limitations such as AP-associated complications and high mortality (27). Therefore, agents for the prevention and treatment of AP are urgently required.

The diagnosis of AP is based on the increasing levels of serum amylase and lipase. This is due to the fact that the levels of these digestive enzymes, which increase during AP, contribute to acinar cell injury, further promoting the inflammatory process, including the release of cytokines and chemokines (28,29). Serum amylase concentration increases at least three times in comparison with the upper limit of the normal range (30). It starts to rise within a few hours after the onset of symptoms and reverts to the normal level within 3-5 days. Due to these features (serum amylase elevation in early AP and short half-life of amylase), the measurement of serum amylase level may be a suitable diagnosis criterion in early presentation. Additionally, serum lipase concentration is increased at least four times of the normal level in cerulein-induced AP (31). It remains high for a longer period of time than the levels of amylase at 8-14 days (31). Therefore, serum lipase analysis may have an advantage over serum amylase levels as of its delayed presentation. In the present study, serum amylase and lipase levels increased in mice with cerulein-induced AP; however, ICA treatment inhibited the elevation of serum amylase and lipase levels, which suggests the therapeutic effect of ICA on AP was mediated by the reduction in the levels of these enzymes.

Neutrophils have a central role in the development of AP by mediating local tissue damage and remote organ injury (32). They are recruited to sites of infection or inflammation, and in regions of pancreatitis (32). Infiltration and activated neutrophils prolong lifespan of neutrophils by several days and release inflammatory mediators, which further worsen the severity of AP (33,34). As inflammation continues, neutrophil accumulation increases in which 'neutrophil swarmimg' occurs; this promotes the progression of local pancreatic inflammation to multiple organ dysfunction syndrome (MODS) by overwhelming inflammatory responses (35). Neutrophil depletion by anti-neutrophil serum was reported to inhibit the severity of cerulein-induced AP (36). Thus, the measurement of neutrophil count may be regarded as a diagnosis index of AP. In this study, we assessed neutrophil infiltration into the pancreas and lung during cerulein-induced AP by measuring MPO activity. ICA treatment was determined to inhibit the activity of MPO in the pancreas and lung in AP. This suggested that the inhibitory effects of ICA on neutrophil infiltration could contribute to the amelioration of AP and AP-associated lung injury.

Severe AP often leads to MODS, and the most common and earliest target organ is the lung (37). Additionally, acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) the more severe form of ALI, are the common complications of AP (38). ALI occurred in 10-25% of AP patients and has been linked with ≤60% of AP-associated mortalities, particularly in elderly patients (39,40). Generally, AP-associated mild lung injury recovers rapidly; however, secondary lung infection, which leads to the release of lung-derived inflammatory mediators across the injured epithelial barrier into the circulation, gives rise to ARDS and mortality (41-43). The variety of effectors that originate from the inflammatory effects of AP are involved in the pathogenesis of secondary lung infection (44-46). Pancreatic mediators, such as pro-inflammatory cytokines and inflammatory cells, are representative of secondary lung infection pathogenesis (44-48). In the present study, our results showed that ICA treatment inhibited the elevation of pancreatic cytokines, which could be promote ALI. In addition, histological lung injury and lung MPO activity were decreased by ICA during AP in our study. Overall, ICA was proposed to exhibit protective activities in mice against AP-associated ALI via the inhibition of pancreatic mediators, such as pro-inflammatory cytokines and neutrophil infiltration.

In regards to the anti-inflammatory activities of ICA, which were determined to be mediated by MAPKs and the NF-κB pathway in a mouse model (49), we investigated the effects of ICA on the activation of MAPKs and NF-κB in cerulein-induced AP. The important signaling pathways, MAPKs and NF-κB, are known to involve various inflammatory regulators in the pancreas (50). In the present study, cerulein activated MAPK and NF-κB in the pancreas as expected, although, only cerulein-induced activation of p38 was inhibited by ICA treatment. These results are in line with that of a previous study where the anti-inflammatory activity of ICA was mainly mediated by the deactivation of p38 MAPK; however, the model of inflammation differed (13). In addition, we reported that the administration of SB203580, a p38 inhibitor, improved the severity of AP similar to ICA. These results suggest that the administration of ICA could reduce the severity of cerulein-induced AP via inhibition of p38.

In the present study, we have demonstrated that ICA exhibited protective and therapeutic effects against ceru-lein-induced AP and AP-associated lung injury. Furthermore, ICA inhibited the activation of p38 MAPK in cerulein-induced AP. Collectively, our findings suggest that ICA is a potential therapeutic agent for the treatment of AP.

Acknowledgments

Not applicable.

Funding

This study was supported by the National Research Foundation of Korea grant funded by the Korea government (MEST; grant nos. NRF-2017R1C1B2010031, NRF-2017R1D1A1B03032371 and NRF-2017R1A5A2015805).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

DUK, GSB, HJS and SJP made substantial contributions to the design of the study. DUK, GSB, MJK, JWC and DGK performed the experiments. MJK and DGK contributed to the statistical analysis of data. DUK and GSB wrote the manuscript. All authors reviewed the results and approved the final version of the manuscript.

Ethics approval and consent to participate

All experiments were carried out in accordance with the animal care regulations set forth and approved by the Wonkwang University Animal Ethics Committee.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Peery AF, Crockett SD, Barritt AS, Dellon ES, Eluri S, Gangarosa LM, Jensen ET, Lund JL, Pasricha S, Runge T, et al: Burden of gastrointestinal, liver, and pancreatic diseases in the United States. Gastroenterology. 149:1731–1741. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Maléth J and Hegyi P: Ca2+ toxicity and mitochondrial damage in acute pancreatitis: Translational overview. Philos Trans R Soc Lond B Biol Sci. 371:201504252016. View Article : Google Scholar : PubMed/NCBI

3 

Bettaieb A, Koike S, Chahed S, Bachaalany S, Griffey S, Sastre J and Haj FG: Pancreatic protein tyrosine phosphatase 1B deficiency exacerbates acute pancreatitis in mice. Am J Pathol. 186:2043–2054. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Yadav D and Lowenfels AB: The epidemiology of pancreatitis and pancreatic cancer. Gastroenterology. 144:1252–1261. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Minkov GA, Halacheva KS, Yovtchev YP and Gulubova MV: Pathophysiological mechanisms of acute pancreatitis define inflammatory markers of clinical prognosis. Pancreas. 44:713–717. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Büchler MW, Gloor B, Müller CA, Friess H, Seiler CA and Uhl W: Acute necrotizing pancreatitis: Treatment strategy according to the status of infection. Ann Surg. 232:619–626. 2000. View Article : Google Scholar : PubMed/NCBI

7 

Bae GS, Heo KH, Park KC, Choi SB, Jo IJ, Seo SH, Kim DG, Shin JY, Kang DG, Lee HS, et al: Apamin attenuated cerulein-induced acute pancreatitis by inhibition of JNK pathway in mice. Dig Dis Sci. 58:2908–2917. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Chakraborty M, Hickey AJ, Petrov MS, Macdonald JR, Thompson N, Newby L, Sim D, Windsor JA and Phillips AR: Mitochondrial dysfunction in peripheral blood mononuclear cells in early experimental and clinical acute pancreatitis. Pancreatology. 16:739–747. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Li LS, Luo YM, Liu J, Zhang Y, Fu XX and Yang DL: Icariin inhibits pulmonary hypertension induced by monocrotaline through enhancement of NO/cGMP signaling pathway in rats. Evid Based Complement Alternat Med. 2016:79154152016.PubMed/NCBI

10 

Hsueh TY, Ho JK, Lin LC, Chiu AW, Lin CH and Tsai TH: Herb-drug interaction of Epimedium extract on the pharmaco-kinetic of dapoxetine in rats. J Chromatogr B Analyt Technol Biomed Life Sci. 1014:64–69. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Chen G, Wang C, Wang J, Yin S, Gao H, Xiang LU, Liu H, Xiong Y, Wang P, Zhu X, et al: Antiosteoporotic effect of icariin in ovariectomized rats is mediated via the Wnt/β-catenin pathway. Exp Ther Med. 12:279–287. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Huang ZS, Xiao HJ, Qi T, Hu ZM, Li H, Chen DL, Xu YL and Chen J: Antioxidative protective effect of icariin on the FeSO4/H2O2-damaged human sperm based on confocal raman micro-spectroscopy. J Huazhong Univ Sci Technolog Med Sci. 34:755–760. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Kong L, Liu J, Wang J, Luo Q, Zhang H, Liu B, Xu F, Pang Q, Liu Y and Dong J: Icariin inhibits TNF-α/IFN-γ induced inflammatory response via inhibition of the substance P and p38-MAPK signaling pathway in human keratinocytes. Int Immunopharmacol. 29:401–407. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Tan HL, Chan KG, Pusparajah P, Saokaew S, Duangjai A, Lee LH and Goh BH: Anti-cancer properties of the naturally occurring aphrodisiacs: Icariin and its derivatives. Front Pharmacol. 7:1912016. View Article : Google Scholar : PubMed/NCBI

15 

Liu B, Xu C, Wu X, Liu F, Du Y, Sun J, Tao J and Dong J: Icariin exerts an antidepressant effect in an unpredictable chronic mild stress model of depression in rats and is associated with the regulation of hippocampal neuroinflammation. Neuroscience. 294:193–205. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Chen YJ, Zheng HY, Huang XX, Han SX, Zhang DS, Ni JZ and He XY: Neuroprotective effects of icariin on brain metabolism, mitochondrial functions, and cognition in triple-transgenic Alzheimer's disease mice. CNS Neurosci Ther. 22:63–73. 2016. View Article : Google Scholar

17 

Wang Y, Wang YS, Song SL, Liang H and Ji AG: Icariin inhibits atherosclerosis progress in Apoe null mice by downregulating CX3CR1 in macrophage. Biochem Biophys Res Commun. 470:845–850. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Wei Y, Liu B, Sun J, Lv Y, Luo Q, Liu F and Dong J: Regulation of Th17/Treg function contributes to the attenuation of chronic airway inflammation by icariin in ovalbumin-induced murine asthma model. Immunobiology. 220:789–797. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Liu T, Xin H, Li WR, Zhou F, Li GY, Gong YQ, Gao ZZ, Qin XC, Cui WS, Shindel AW and Xin ZC: Effects of icariin on improving erectile function in streptozotocin-induced diabetic rats. J Sex Med. 8:2761–2772. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Ethridge RT, Chung DH, Slogoff M, Ehlers RA, Hellmich MR, Rajaraman S, Saito H, Uchida T and Evers BM: Cyclooxygenase-2 gene disruption attenuates the severity of acute pancreatitis and pancreatitis-associated lung injury. Gastroenterology. 123:1311–1322. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Kim MJ, Bae GS, Choi SB, Jo IJ, Kim DG, Shin JY, Lee SK, Kim MJ, Shong HJ and Park SJ: Lupeol protects against cerulein-induced acute pancreatitis in mice. Phytother Res. 29:1634–1639. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Que RS, Cao LP, Ding GP, Hu JA, Mao KJ and Wang GF: Correlation of nitric oxide and other free radicals with the severity of acute pancreatitis and complicated systemic inflammatory response syndrome. Pancreas. 39:536–540. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Andersson R, Andersson B, Haraldsen P, Drewsen G and Eckerwall G: Incidence, management and recurrence rate of acute pancreatitis. Scand J Gastroenterol. 39:891–894. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Johnson CD, Besselink MG and Carter R: Acute pancreatitis. BMJ. 349:g48592014. View Article : Google Scholar : PubMed/NCBI

25 

Mossad DE, Dinh BV, Markert RJ, Musleh MN and Agrawal S: Predictors of in hospital mortality in acute pancreatitis. JOP J Pancreas. 18:465–469. 2017.

26 

Popa CC, Badiu DC, Rusu OC, Grigorean VT, Neagu SI and Strugaru CR: Mortality prognostic factors in acute pancreatitis. J Med Life. 9:413–418. 2016.PubMed/NCBI

27 

Kimura W and Mössner J: Role of hypertriglyceridemia in the pathogenesis of experimental acute pancreatitis in rats. Int J Pancreatol. 20:177–184. 1996. View Article : Google Scholar : PubMed/NCBI

28 

Raraty MG, Murphy JA, Mcloughlin E, Smith D, Criddle D and Sutton R: Mechanisms of acinar cell injury in acute pancreatitis. Scand J Surg. 94:89–96. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Cotton PB, Lehman G, Vennes J, Geenen JE, Russell RC, Meyers WC, Liqoury C and Nickl N: Endoscopic sphincterotomy complications and their management: An attempt at consensus. Gastrointest Endosc. 37:383–393. 1991. View Article : Google Scholar : PubMed/NCBI

30 

Winslet M, Hall C, London NJ and Neoptolemos JP: Relation of diagnostic serum amylase levels to aetiology and severity of acute pancreatitis. Gut. 33:982–986. 1992. View Article : Google Scholar : PubMed/NCBI

31 

Tietz NW and Shuey DF: Lipase in serum-the elusive enzyme: An overview. Clin Chem. 39:746–756. 1993.PubMed/NCBI

32 

Montecucco F, Mach F, Lenglet S, Vonlaufen A, Gomes Quinderé AL, Pelli G, Burger F, Galan K, Dallegri F, Carbone F, et al: Treatment with Evasin-3 abrogates neutrphil-mediated inflammation in mouse acute pancreatitis. Eur J Clin Invest. 44:940–950. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Merza M, Hartman H, Rahman M, Hwaiz R, Zhang E, Renström E, Luo L, Mörgelin M, Regner S and Thorlacius H: Neutrophil extracellular traps induce trypsin activation, inflammation, and tissue damage in mice with severe acute pancreatitis. Gastroenterology. 149:1920–1931. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Mantovani A, Cassatella MA, Costantini C and Jaillon S: Neutrophils in the activation and regulation of innate and adaptive immunity. Nat Rev Immunol. 11:519–531. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Yang ZW, Meng XX and Xu P: Central role of neutrophil in the pathogenesis of severe acute pancreatitis. J Cell Mol Med. 19:2513–2520. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Sandoval D, Gukovskaya A, Reavey P, Gukovsky S, Sisk A, Braquet P, Pandol SJ and Poucell-Hatton S: The role of neutrophils and platelet-activating factor in mediating experimental pancreatitis. Gastroenterology. 111:1081–1091. 1996. View Article : Google Scholar : PubMed/NCBI

37 

El-Menyar A, Thani E, Zakaria A, Zarour A, Tuma M, AbdulRahman H, Parchani A, Peralta R and Latifi R: Multiple organ dysfunction syndrome (MODS): Is it preventable or inevitable. Int J Clin Med. 3:722–730. 2012. View Article : Google Scholar

38 

Elder AS, Saccone GT and Dixon DL: Lung injury in acute pancreatitis: Mechanisms underlying augmented secondary injury. Pancreatology. 12:49–56. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Shields CJ, Winter DC and Redmond HP: Lung injury in acute pancreatitis: Mechanisms, prevention, and therapy. Curr Opin Crit Care. 8:158–163. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Rubenfeld GD, Caldwell E, Peabody E, Weaver J, Martin DP, Neff M, Stern EJ and Hudson LD: Incidence and outcomes of acute lung injury. N Engl J Med. 353:1685–1693. 2005. View Article : Google Scholar : PubMed/NCBI

41 

van Westerloo DJ, Schultz MJ, Bruno MJ, de Vos AF, Florquin S and van der Poll T: Acute pancreatitis in mice impairs bacterial clearance from the lungs, whereas concurrent pneumonia prolongs the course of pancreatitis. Crit Care Med. 32:1997–2001. 2004. View Article : Google Scholar : PubMed/NCBI

42 

Zhou MT, Chen CS, Chen BC, Zhang QY and Andersson R: Acute lung injury and ARDS in acute pancreatitis: Mechanisms and potential intervention. World J Gastroenterology. 16:2094–2099. 2010. View Article : Google Scholar

43 

Aeffner F, Bolon B and Davis IC: Mouse models of acute respiratory distress syndrome: A review of analytical approaches, pathologic features, and common measurements. Toxicol Pathol. 43:1074–1092. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Jaffray C, Yang J and Norman J: Elastase mimics pancreatitis-induced hepatic injury via inflammatory mediators. J Surg Res. 90:95–101. 2000. View Article : Google Scholar : PubMed/NCBI

45 

Lee WL and Downey GP: Neutrophil activation and acute lung injury. Curr Opin Crit Care. 7:1–7. 2001. View Article : Google Scholar : PubMed/NCBI

46 

Lopez-Font I, Gea-Sorlí S, de-Madaria E, Gutiérrez LM, Pérez-Mateo M and Closa D: Pancreatic and pulmonary mast cells activation during experimental acute pancreatitis. World J Gastroenterol. 16:3411–3417. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Fritz S, Hackert T, Hartwig W, Rossmanith F, Strobel O, Schneider L, Will-Schweiger K, Kommerell M, Büchler MW and Werner J: Bacterial translocation and infected pancreatic necrosis in acute necrotizing pancreatitis derives from small bowel rather than from colon. Am J Surg. 200:111–117. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Matull WR, Pereira SP and O'donohue JW: Biochemical markers of acute pancreatitis. J Clin Pathol. 59:340–344. 2006. View Article : Google Scholar : PubMed/NCBI

49 

Ma P, Zhang S, Su X, Qiu G and Wu Z: Protective effects of icariin on cisplatin-induced acute renal injury in mice. Am J Transl Res. 7:2105–2014. 2015.PubMed/NCBI

50 

Arthur JS and Ley SC: Mitogen-activated protein kinases in innate immunity. Nat Rev Immunol. 13:679–692. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2019
Volume 44 Issue 4

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
Kim DU, Bae GS, Kim MJ, Choi JW, Kim DG, Song HJ and Park SJ: Icariin attenuates the severity of cerulein‑induced acute pancreatitis by inhibiting p38 activation in mice. Int J Mol Med 44: 1563-1573, 2019
APA
Kim, D., Bae, G., Kim, M., Choi, J., Kim, D., Song, H., & Park, S. (2019). Icariin attenuates the severity of cerulein‑induced acute pancreatitis by inhibiting p38 activation in mice. International Journal of Molecular Medicine, 44, 1563-1573. https://doi.org/10.3892/ijmm.2019.4312
MLA
Kim, D., Bae, G., Kim, M., Choi, J., Kim, D., Song, H., Park, S."Icariin attenuates the severity of cerulein‑induced acute pancreatitis by inhibiting p38 activation in mice". International Journal of Molecular Medicine 44.4 (2019): 1563-1573.
Chicago
Kim, D., Bae, G., Kim, M., Choi, J., Kim, D., Song, H., Park, S."Icariin attenuates the severity of cerulein‑induced acute pancreatitis by inhibiting p38 activation in mice". International Journal of Molecular Medicine 44, no. 4 (2019): 1563-1573. https://doi.org/10.3892/ijmm.2019.4312