Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
J Investig Med ; 70(5): 1285-1292, 2022 06.
Article in English | MEDLINE | ID: mdl-35078865

ABSTRACT

Chronic pancreatitis (CP) is a pathological fibroinflammatory syndrome of the pancreas. Currently, there are no therapeutic agents available for treating CP-associated pancreatic fibrosis. Fraxinus rhynchophylla (FR) reportedly exhibits anti-inflammatory, antioxidative and antitumor activities. Although FR possesses numerous properties associated with the regulation of diverse diseases, the effects of FR on CP remain unknown. Herein, we examined the effects of FR on CP. For CP induction, mice were intraperitoneally administered cerulein (50 µg/kg) 6 times a day, 4 days per week for 3 weeks. FR extract (100 or 400 mg/kg) or saline (control group) was intraperitoneally injected 1 hour before the first cerulein injection. After 3 weeks, the pancreas was harvested for histological analysis. In addition, pancreatic stellate cells (PSCs) were isolated to examine the antifibrogenic effects and regulatory mechanisms of FR. Administration of FR significantly inhibited histological damage in the pancreas, increased pancreatic acinar cell survival, decreased PSC activation and collagen deposition, and decreased pro-inflammatory cytokines. Moreover, FR treatment inhibited the expression of fibrotic mediators, such as α-smooth muscle actin (α-SMA), collagen, fibronectin 1, and decreased pro-inflammatory cytokines in isolated PSCs stimulated with transforming growth factor (TGF)-ß. Furthermore, FR treatment suppressed the phosphorylation of Smad 2/3 but not of Smad 1/5 in TGF-ß-stimulated PSCs. Collectively, these results suggest that FR ameliorates pancreatic fibrosis by inhibiting PSC activation during CP.


Subject(s)
Fraxinus , Pancreatitis, Chronic , Animals , Ceruletide/metabolism , Ceruletide/pharmacology , Ceruletide/therapeutic use , Collagen/metabolism , Collagen/pharmacology , Collagen/therapeutic use , Fibrosis , Humans , Mice , Pancreas/pathology , Pancreatitis, Chronic/drug therapy , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/pathology , Plant Bark/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
2.
Int J Mol Sci ; 22(13)2021 Jun 26.
Article in English | MEDLINE | ID: mdl-34206763

ABSTRACT

Acute pancreatitis (AP) is an inflammatory disorder, involving acinar cell death and the release of inflammatory cytokines. Currently, there are limited effective therapeutic agents for AP. Betulinic acid (BA) is a pentacyclic triterpenoid extracted from Betula platyphylla that has been shown to have anti-inflammatory effects. In this study, we aimed to investigate the effects of BA on AP and elucidate the potential underlying mechanisms. AP was induced in mice through six intraperitoneal injections of cerulein. After the last cerulein injection, the mice were sacrificed. Our results revealed that pre- and post-treatment with BA significantly reduced the severity of pancreatitis, as evidenced by a decrease in histological damage in the pancreas and lung, serum amylase and lipase activity and pancreatic myeloperoxidase activity. Furthermore, BA pretreatment reduced proinflammatory cytokine production, augmentation of chemokines, and infiltration of macrophages and neutrophils in the pancreas of AP mice. In addition, mice that were pretreated with BA showed a reduction in Iκ-Bα degradation and nuclear factor-kappa B (NF-κB) binding activity in the pancreas. Moreover, BA reduced the production of proinflammatory cytokines and NF-κB activation in pancreatic acinar cells (PACs). These findings suggest that BA may have prophylactic and therapeutic effects on AP via inhibition of the NF-κB signaling pathway.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , NF-kappa B/metabolism , Pancreatitis/drug therapy , Pentacyclic Triterpenes/therapeutic use , Amylases/blood , Animals , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Cytokines/drug effects , Cytokines/metabolism , Female , Lipase/blood , Lung/drug effects , Lung/metabolism , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Neutrophils , Pancreas/drug effects , Pancreas/metabolism , Pentacyclic Triterpenes/pharmacology , Peroxidase/metabolism , Signal Transduction , Betulinic Acid
3.
Int J Mol Sci ; 22(3)2021 Jan 31.
Article in English | MEDLINE | ID: mdl-33572597

ABSTRACT

Cisplatin is the most widely used chemotherapeutic agent. However, it often causes nephrotoxicity, which results in acute kidney injury (AKI). Therefore, we urgently need a drug that can reduce the nephrotoxicity induced by cisplatin. Loganin is a major iridoid glycoside isolated from Corni fructus that has been used as an anti-inflammatory agent in various pathological models. However, the renal protective activity of loganin remains unclear. In this study, to examine the protective effect of loganin on cisplatin-induced AKI, male C57BL/6 mice were orally administered with loganin (1, 10, and 20 mg/kg) 1 h before intraperitoneal injection of cisplatin (10 mg/kg) and sacrificed at three days after the injection. The administration of loganin inhibited the elevation of blood urea nitrogen (BUN) and creatinine (CREA) in serum, which are used as biomarkers of AKI. Moreover, histological kidney injury, proximal tubule damages, and renal cell death, such as apoptosis and ferroptosis, were reduced by loganin treatment. Also, pro-inflammatory cytokines, such as interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α, reduced by loganin treatment. Furthermore, loganin deactivated the extracellular signal-regulated kinases (ERK) 1 and 2 during AKI. Taken together, our results suggest that loganin may attenuate cisplatin-induced AKI through the inhibition of ERK1/2.


Subject(s)
Acute Kidney Injury/drug therapy , Cisplatin/adverse effects , Iridoids/administration & dosage , MAP Kinase Signaling System/drug effects , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Animals , Apoptosis/drug effects , Blood Urea Nitrogen , Cell Death/drug effects , Creatinine/blood , Cytokines/metabolism , Kidney/drug effects , Kidney/pathology , Male , Mice , Mice, Inbred C57BL
4.
Int Immunopharmacol ; 88: 106900, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32829089

ABSTRACT

Acute pancreatitis (AP) refers to inflammation in the pancreas, which may lead to death in severe cases. Coenzyme Q10 (Q10), generally known to generate energy, plays an important role as an anti-oxidant and anti-inflammatory effector. Here, we showed the effect of Q10 on inflammatory response in murine AP model. For this study, we induced AP by injection of cerulein intraperitoneally or pancreatic duct ligation (PDL) in mice. The level of cytokines and digestive enzymes were measured in pancreas, and blood. All pancreatic tissues were excised for investigation such as histological changes, infiltration of immune cells. Administration of Q10 attenuated the severity of AP and its associated pulmonary complication as shown by reduction of acinar cell death, parenchymal edema, inflammatory cell infiltration and alveolar thickening in both cerulein-induced AP and PDL-induced AP. Moreover, reduction of the cytokines such as interleukin (IL)-1ß, IL-6 and tumor necrosis factor (TNF)-α were observed in pancreas and pancreatic acinar cells by Q10. Furthermore, Q10 reduced the infiltration of immune cells such as monocytes and neutrophils and augmentation of chemokines such as CC chemokine-2 (CCL2) and C-X-C chemokine-2 (CXCL2) in pancreas of AP mice. In addition, Q10 deactivates the phosphorylation of extracellular signal-regulated kinase (ERK) and c-jun NH2-terminal kinase (JNK) in pancreas. In conclusion, these observations suggest that Q10 could attenuate the pancreatic damage and its associated pulmonary complications via inhibition of inflammatory cytokines and inflammatory cell infiltration and that the deactivation of ERK and JNK by Q10 might contribute to the attenuation of AP.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Pancreatitis/drug therapy , Ubiquinone/analogs & derivatives , Animals , Anti-Inflammatory Agents/pharmacology , Ceruletide , Cytokines/genetics , Cytokines/immunology , Female , Lung/drug effects , Lung/pathology , MAP Kinase Signaling System/drug effects , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/immunology , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/immunology , Pancreas/drug effects , Pancreas/immunology , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/immunology , Pancreatitis/pathology , Ubiquinone/pharmacology , Ubiquinone/therapeutic use
5.
Am J Chin Med ; 48(4): 987-1003, 2020.
Article in English | MEDLINE | ID: mdl-32431181

ABSTRACT

Our previous report revealed that Gardenia jasminoides (GJ) has protective effects against acute pancreatitis. So, we examined whether aqueous extract of GJ has anti-inflammation and antifibrotic effects even against cerulein-induced chronic pancreatitis (CP). CP was induced in mice by an intraperitoneal injection of a stable cholecystokinin (CCK) analogue, cerulein, six times a day, four days per week for three weeks. GJ extract (0.1 or 1[Formula: see text]g/kg) or saline (control group) were intraperitoneally injected 1[Formula: see text]h before first cerulein injection. After three weeks of stimulation, the pancreas was harvested for the examination of several fibrotic parameters. In addition, pancreatic stellate cells (PSCs) were isolated using gradient methods to examine the antifibrogenic effects of GJ. In the cerulein-induced CP mice, the histological features of the pancreas showed severe tissue damage such as enlarged interstitial spaces, inflammatory cell infiltrate and glandular atrophy, and tissue fibrosis. However, treatment of GJ reduced the severity of CP such as pancreatic edema and inflammatory cell infiltration. Furthermore, treatment of GJ increased pancreatic acinar cell survival, and reduced pancreatic fibrosis and activation of PSC in vivo and in vitro. In addition, GJ treatment inhibited the activation of c-Jun N-terminal kinase (JNK) and extracellular signal-regulated protein kinase (ERK) in the PSCs. These results suggest that GJ attenuated the severity of CP and the pancreatic fibrosis by inhibiting JNK and ERK activation during CP.


Subject(s)
Ceruletide/adverse effects , Gardenia/chemistry , Pancreatitis, Chronic/drug therapy , Pancreatitis, Chronic/prevention & control , Phytotherapy , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Animals , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibrosis , Injections, Intraperitoneal , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Pancreas/pathology , Pancreatic Stellate Cells/pathology , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/pathology , Plant Extracts/isolation & purification
6.
Pancreas ; 49(1): 89-95, 2020 01.
Article in English | MEDLINE | ID: mdl-31856083

ABSTRACT

OBJECTIVES: In this study, we investigated the anti-inflammatory effects of silymarin on cerulein-induced acute pancreatitis (AP) in mice. METHODS: Cerulein (50 µg/kg) was injected intraperitoneally once hourly for 6 hours to induce AP. To investigate the prophylactic effects of silymarin, dimethyl sulfoxide or silymarin (25, 50, and 100 mg/kg) was injected intraperitoneally 1 hour before cerulein injection. To investigate the therapeutic effects of silymarin, dimethyl sulfoxide or silymarin (100 mg/kg) was injected intraperitoneally 1, 3, or 5 hours after the first cerulein injection. Blood, pancreas, and lungs were harvested 6 hours after the last cerulein injection. RESULTS: Pre- and posttreatment with silymarin decreased the pancreas weight/body weight ratio and serum amylase activity. Furthermore, silymarin treatment inhibited pancreas and lung injury and neutrophil infiltration during cerulein-induced AP. In addition, silymarin inhibited increased secretion of proinflammatory cytokines such as interleukin 1ß, interleukin 6, and tumor necrosis factor α. Finally, mitogen-activated protein kinases (MAPKs) and nuclear factor-κB were activated by cerulein, and only p38 in MAPK was inhibited by silymarin. CONCLUSIONS: These findings suggest that silymarin attenuates the severity of AP through inhibition of p38 MAPKs and that silymarin could be a potential prophylactic and therapeutic agent for the treatment of AP.


Subject(s)
Pancreas/drug effects , Pancreatitis/prevention & control , Severity of Illness Index , Silymarin/pharmacology , Acute Disease , Amylases/blood , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacology , Ceruletide , Cytokines/metabolism , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Organ Size/drug effects , Pancreas/metabolism , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/metabolism , Protective Agents/administration & dosage , Protective Agents/pharmacology , Silymarin/administration & dosage
7.
Mol Med Rep ; 21(1): 258-266, 2020 01.
Article in English | MEDLINE | ID: mdl-31746359

ABSTRACT

The major role of inner medullary collecting duct (IMCD) cells is to maintain water and sodium homeostasis. In addition to the major role, it also participates in the protection of renal and systemic inflammation. Although IMCD cells could take part in renal and systemic inflammation, investigations on renal inflammation in IMCD cells have rarely been reported. Although berberine (BBR) has been reported to show diverse pharmacological effects, its anti­inflammatory and protective effects on IMCD cells have not been studied. Therefore, in the present study, we examined the anti­inflammatory and protective effects of BBR in mouse IMCD­3 (mIMCD­3) cells against lipopolysaccharide (LPS). An MTT assay was carried out to investigate the toxicity of BBR on mIMCD­3 cells. Reverse transcription quantitative­PCR and western blotting were performed to analysis pro­inflammatory molecules and cytokines. Mechanisms of BBR were examined by western blotting and immunocytochemistry. According to previous studies, pro­inflammatory molecules, such as inducible nitric oxide synthase and cyclooxygenase­2, and pro­inflammatory cytokines, such as interleukin (IL)­1ß, IL­6 and tumor necrosis factor­α are increased in LPS­exposed mIMCD­3 cells. However, the production of these pro­inflammatory molecules is significantly inhibited by treatment with BBR. In addition, BBR inhibited translocation of nuclear factor (NF)­κB p65 from the cytosol to the nucleus, and degradation of inhibitory κ­Bα in LPS­exposed mIMCD­3 cells. In conclusion, BBR could inhibit renal inflammatory responses via inhibition of NF­κB signaling and ultimately contribute to amelioration of renal injury during systemic inflammation.


Subject(s)
Berberine/pharmacology , Inflammation/drug therapy , Kidney/drug effects , Transcription Factor RelA/genetics , Animals , Cell Line , Cyclooxygenase 2/genetics , Gene Expression Regulation/drug effects , Humans , Inflammation/chemically induced , Inflammation/pathology , Interleukin-1beta/genetics , Interleukin-6/genetics , Kidney/pathology , Lipopolysaccharides/toxicity , Mice , NF-kappa B/genetics , Nitric Oxide Synthase/genetics , Signal Transduction/drug effects
8.
Mol Immunol ; 114: 620-628, 2019 10.
Article in English | MEDLINE | ID: mdl-31542607

ABSTRACT

Acute pancreatitis (AP) is a severe inflammatory condition of the pancreas, with no specific treatment available. We have previously reported that Nardostachys jatamansi (NJ) ameliorates cerulein-induced AP. However, the specific compound responsible for this inhibitory effect has not been identified. Therefore, in the present study, we focused on a single compound, 8α-hydroxypinoresinol (HP), from NJ. The aim of this study was to determine the effect of HP on the development of pancreatitis in mice and to explore the underlying mechanism(s). AP was induced by the injection of cerulein (50 µg/kg/h) for 6 h. HP (0.5, 5 or 10 mg/kg, i.p.) was administered 1 h prior to and 1, 3 or 5 h after the first cerulein injection, with vehicle- and DMSO-treated groups as controls. Blood samples were collected to determine serum levels of amylase, lipase, and cytokines. The pancreas was removed for morphological examination, myeloperoxidase (MPO) assays, cytokine assays, and assessment of nuclear factor (NF)-κB activation. The lungs were removed for morphological examination and MPO assays. Administration of HP dramatically improved pancreatic damage and pancreatitis-associated lung damage and also reduced amylase and lipase activities in serum. Moreover, administration of HP reduced the production of pro-inflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6 in the pancreas and serum during AP. In addition, the administration of HP inhibited degradation of inhibitory κ-Bα (Iκ-Bα), NF-κB p65 translocation into nucleus and NF-κB binding activity in the pancreas. Our results suggest that HP exerted therapeutic effects on pancreatitis and these beneficial effects may be due to the inhibition of NF-κB activation.


Subject(s)
Ceruletide/pharmacology , Furans/pharmacology , Lignans/pharmacology , Nardostachys/chemistry , Pancreas/drug effects , Pancreatitis/chemically induced , Pancreatitis/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Animals , Cytokines/metabolism , Female , Inflammation/drug therapy , Inflammation/metabolism , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , Pancreas/metabolism , Pancreatitis/metabolism , Signal Transduction/drug effects
9.
Mol Med Rep ; 20(4): 3709-3718, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31485676

ABSTRACT

Chronic pancreatitis (CP) is characterized by recurrent pancreatic injury, resulting in inflammation and fibrosis. Currently, there are no drugs for the treatment of pancreatic fibrosis associated with CP. Piperine, a natural alkaloid found in black pepper, has been reported to show anti­inflammatory, anti­oxidative, and antitumor activities. Although piperine exhibits numerous properties in regards to the regulation of diverse diseases, the effects of piperine on CP have not been established. To investigate the effects of piperine on CP in vivo, we induced CP in mice through the repetitive administration of cerulein (50 µg/kg) six times at 1­h intervals, 5 times per week, for a total of 3 weeks. In the pre­treatment groups, piperine (1, 5, or 10 mg/kg) or corn oil were administrated orally at 1 h before the first cerulein injection, once a day, 5 times a week, for a total of 3 weeks. In the post­treatment groups, piperine (10 mg/kg) or corn oil was administered orally at 1 or 2 week after the first cerulein injection. Pancreases were collected for histological analysis. In addition, pancreatic stellate cells (PSCs) were isolated to examine the anti­fibrogenic effects and regulatory mechanisms of piperine. Piperine treatment significantly inhibited histological damage in the pancreas, increased the pancreatic acinar cell survival, reduced collagen deposition and reduced pro­inflammatory cytokines and chemokines. In addition, piperine treatment reduced the expression of fibrotic mediators, such as α­smooth muscle actin (α­SMA), collagen, and fibronectin 1 in the pancreas and PSCs. Moreover, piperine treatment reduced the production of transforming growth factor (TGF)­ß in the pancreas and PSCs. Furthermore, piperine treatment inhibited TGF­ß­induced pSMAD2/3 activation but not pSMAD1/5 in the PSCs. These findings suggest that piperine treatment ameliorates pancreatic fibrosis by inhibiting TGF­ß/SMAD2/3 signaling during CP.


Subject(s)
Alkaloids/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Benzodioxoles/therapeutic use , Pancreatitis, Chronic/drug therapy , Piperidines/therapeutic use , Polyunsaturated Alkamides/therapeutic use , Smad Proteins/immunology , Transforming Growth Factor beta/immunology , Animals , Disease Models, Animal , Female , Fibrosis , Mice , Mice, Inbred C57BL , Pancreas/drug effects , Pancreas/immunology , Pancreas/pathology , Pancreatitis, Chronic/immunology , Pancreatitis, Chronic/pathology , Signal Transduction/drug effects
10.
Int J Mol Med ; 44(4): 1563-1573, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31432106

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 anti­inflammatory effects of ICA against AP require further study. Therefore, we aimed to investigate the effect of ICA on cerulein­induced 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 transcription­quantitative 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 cerulein­induced histological injury and neutrophil infiltration of the pancreas and lung. In addition, ICA suppressed the production of pro­inflammatory cytokines, including interleukin (IL)­1ß, IL­6 and tumor necrosis factor­α in the pancreas. Furthermore, ICA administration was observed to inhibit p38 activation during cerulein­induced AP. Inhibition of p38 activation resulted in alleviated pancreatitis. Collectively, our results suggested that ICA exhibits anti­inflammatory effects in cerulein­induced AP via the inhibition of p38.


Subject(s)
Ceruletide/adverse effects , Flavonoids/pharmacology , Pancreatitis/etiology , Pancreatitis/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism , Amylases/blood , Animals , Biomarkers , Disease Models, Animal , Female , Lipase/blood , Lipase/metabolism , Mice , NF-kappa B/metabolism , Pancreatitis/diagnosis , Pancreatitis/drug therapy , Severity of Illness Index
11.
Int Immunopharmacol ; 69: 225-234, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30738992

ABSTRACT

Heme oxygenase-1 (HO-1) has an anti-inflammatory action in acute pancreatitis (AP). However, its mechanism of action and natural compounds/drugs to induce HO-1 in pancreas are not well understood. In this study, we investigated the regulatory mechanisms of HO-1 during AP using desoxo-narchinol-A (DN), the natural compound inducing HO-1 in the pancreas. Female C57/BL6 Mice were intraperitoneally injected with supramaximal concentrations of cerulein (50 µg/kg) hourly for 6 h to induce AP. DMSO or DN was administered intraperitoneally, then mice were sacrificed 6 h after the final cerulein injection. Administration of DN increased pancreatic HO-1 expression through activation of activating protein-1, mediated by mitogen-activated protein kinases. Furthermore, DN treatment reduced the pancreatic weight-to-body weight ratio as well as production of digestive enzymes and pro-inflammatory cytokines. Inhibition of HO-1 by tin protoporphyrin IX abolished the protective effects of DN on pancreatic damage. Additionally, DN treatment inhibited neutrophil infiltration into the pancreas via regulation of chemokine (C-X-C motif) ligand 2 (CXCL2) by HO-1. Our results suggest that DN is an effective inducer of HO-1 in the pancreas, and that HO-1 regulates neutrophil infiltration in AP via CXCL2 inhibition.


Subject(s)
Chemokine CXCL2/metabolism , Heme Oxygenase-1/metabolism , Neutrophils/physiology , Pancreas/metabolism , Pancreatitis/metabolism , Acute Disease , Amylases/blood , Animals , Ceruletide/administration & dosage , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Naphthols/metabolism , Neutrophil Infiltration , Pancreas/pathology , Pancreatitis/pathology
12.
Int Immunopharmacol ; 69: 169-177, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30716587

ABSTRACT

Inflammasomes promote the production of pro-inflammatory cytokines, such as interleukin (IL)-1ß and IL-18, which are the representative mediators of inflammation. Abnormal activation of inflammasomes leads to the development of inflammatory diseases such as acute pancreatitis (AP). In this study, we demonstrate the inhibitory effects of a new natural compound fraxinellone on inflammasome formation and examine the role of inflammasomes in a mouse model of AP. AP was induced with hourly intraperitoneal injections of supramaximal concentrations of the stable cholecystokinin analogue cerulein (50 µg/kg) for 6 h. Mice were sacrificed 6 h after the final cerulein injection. Blood and pancreas samples were obtained for further experiments. Intraperitoneal injection of fraxinellone significantly inhibited the pancreatic activation of multiple inflammasome molecules such as NACHT, LRR and PYD domains-containing protein 3 (NLRP3), PY-CARD, caspase-1, IL-18, and IL-1ß during AP. In addition, fraxinellone treatment inhibited pancreatic injury, elevation in serum amylase and lipase activities, and infiltration of inflammatory cells such as neutrophils and macrophages but had no effect on pancreatic edema. To investigate whether inflammasome activation leads to the infiltration of inflammatory cells, we used parthenolide, a well-known natural inhibitor, and IL-1 receptor antagonist mice. The inhibition of inflammasome activation by pharmacological/or genetic modification restricted the infiltration of inflammatory cells, but not edema, consistent with the results observed with fraxinellone. Taken together, our study highlights fraxinellone as a natural inhibitor of inflammasomes and that inflammasome inhibition may lead to the suppression of inflammatory cells during AP.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Benzofurans/therapeutic use , Inflammasomes/metabolism , Inflammation/drug therapy , Macrophages/immunology , Neutrophils/immunology , Pancreatitis/drug therapy , Acute Disease , Animals , Cell Movement/drug effects , Ceruletide/administration & dosage , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL
13.
Pancreas ; 46(8): 1046-1055, 2017 09.
Article in English | MEDLINE | ID: mdl-28797013

ABSTRACT

OBJECTIVES: We set out to examine whether berberine (BBR) might affect the severity of pancreatitis and pancreatitis-associated lung injury in choline-deficient ethionine-supplemented (CDE) diet-induced severe acute pancreatitis. METHODS: Severe acute pancreatitis was induced by feeding a CDE diet for 3 days. Berberine was administered intraperitoneally during CDE diet. Mice were killed on days 1, 2, and 3 after the onset of CDE diet. The severity of pancreatitis was assessed by evaluating changes to the pancreas and lung and survival rate. Blood, pancreas, and lung were harvested for further examination. Furthermore, the regulating mechanisms of BBR were evaluated on the pancreas. RESULTS: Administration of BBR significantly inhibited histological damage to the pancreas and lung and decreased serum level of amylase and lipase, myeloperoxidase activity, cytokine production, and the mortality rate. Furthermore, administration of BBR inhibited activation of nuclear factor kappa B, c-Jun N-terminal kinases, and p38 in the pancreas during CDE diet. CONCLUSIONS: These findings suggest that BBR attenuates the severity of pancreatitis by inhibiting activation of nuclear factor kappa B, c-Jun N-terminal kinase, and p38 and that BBR could be used as a beneficial agent to regulate AP.


Subject(s)
Berberine/pharmacology , Lung Injury/prevention & control , Lung/drug effects , Pancreas/drug effects , Pancreatitis, Acute Necrotizing/prevention & control , Amylases/blood , Animals , Choline/isolation & purification , Diet/adverse effects , Ethionine/administration & dosage , Female , Lipase/blood , Lung/metabolism , Lung/pathology , Lung Injury/mortality , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Pancreas/metabolism , Pancreas/pathology , Pancreatitis, Acute Necrotizing/etiology , Pancreatitis, Acute Necrotizing/mortality , Phytotherapy/methods , Survival Rate
14.
Mol Immunol ; 74: 27-38, 2016 06.
Article in English | MEDLINE | ID: mdl-27148818

ABSTRACT

Acute pancreatitis (AP) is a life-threatening disease. Berberine (BBR), a well-known plant alkaloid, is reported to have anti-inflammatory activity in many diseases. However, the effects of BBR on AP have not been clearly elucidated. Therefore, the present study aimed to investigate the effects of BBR on cerulein-induced AP in mice. AP was induced by either cerulein or l-arginine. In the BBR treated group, BBR was administered intraperitoneally 1h before the first cerulein or l-arginine injection. Blood samples were obtained to determine serum amylase and lipase activities and nitric oxide production. The pancreas and lung were rapidly removed for examination of histologic changes, myeloperoxidase (MPO) activity, and real-time reverse transcription-polymerase chain reaction. Furthermore, the regulating mechanisms of BBR were evaluated. Treatment of mice with BBR reduced pancreatic injury and activities of amylase, lipase, and pancreatitis-associated lung injury, as well as inhibited several inflammatory parameters such as the expression of pro-inflammatory cytokines and inducible nitric oxide synthesis (iNOS). Furthermore, BBR administration significantly inhibited c-Jun N-terminal kinase (JNK) activation in the cerulein-induced AP. Deactivation of JNK resulted in amelioration of pancreatitis and the inhibition of inflammatory mediators. These results suggest that BBR exerts anti-inflammatory effects on AP via JNK deactivation on mild and severe acute pancreatitis model, and could be a beneficial target in the management of AP.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Berberine/pharmacology , MAP Kinase Signaling System/drug effects , Pancreatitis, Acute Necrotizing/pathology , Animals , Blotting, Western , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL
15.
Inflammation ; 39(1): 87-95, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26260258

ABSTRACT

Guggulsterone (GS) is a phytosterol that has been used to treat inflammatory diseases such as colitis, obesity, and thrombosis. Although many previous studies have examined activities of GS, the effect of GS on lipopolysaccharide (LPS)-induced inflammatory responses in mouse inner medullary collecting duct-3 (mIMCD-3) cells have not been examined. Therefore, here, we investigated the anti-inflammatory action of GS on mIMCD-3 cells exposed to LPS. LPS treatment on mIMCD-3 cells produced pro-inflammatory molecules such as inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α) significantly; however, GS treatment significantly inhibited the production of pro-inflammatory molecules. In addition, GS inhibited the degradation of Iκ-Bα and translocation of NF-κB on mIMCD-3 cells. These results suggest that GS could inhibit inflammatory responses in collecting duct cells which could contribute to kidney injury during systemic infection.


Subject(s)
Acute Kidney Injury/drug therapy , Anti-Inflammatory Agents/pharmacology , Kidney Tubules, Collecting/immunology , Pregnenediones/pharmacology , Toll-Like Receptor 4/immunology , Animals , Cell Line , Cyclooxygenase 2/biosynthesis , Female , Inflammation/drug therapy , Interleukin-6/biosynthesis , Kidney Tubules, Collecting/cytology , Lipopolysaccharides , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/biosynthesis
16.
Int Immunopharmacol ; 29(2): 730-738, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26371857

ABSTRACT

We previously reported that Nardostachys jatamansi (NJ) exhibits anti-inflammatory activity against lipopolysaccharide (LPS). However, the active compound in NJ is unknown. Therefore, here, we examined the effects of desoxo-narchinol-A (DN) isolated from NJ against LPS-induced inflammation. To demonstrate the anti-inflammatory effect of DN against LPS, we used two models; murine endotoxin shock model for in vivo model, and peritoneal macrophage responses for in vitro. In endotoxin shock model, DN was administrated intraperitoneally 1h before LPS challenge, then we evaluated mice survival rates and organ damages. Pretreatment with DN (0.05mg/kg, 0.1mg/kg, or 0.5mg/kg) dramatically reduced mortality in a murine LPS-induced endotoxin shock model. Furthermore, DN inhibited tissue injury and production of pro-inflammatory cytokines, such as interleukin (IL)-1ß, IL-6, and tumor necrosis factor alpha (TNF-α), in the liver and lung. In in vitro macrophage model, we examined the inflammatory mediators and regulatory mechanisms such as mitogen-activated protein kinases (MAPKs) and nuclear factor kappa B (NF-κB). DN inhibited the production of inflammatory mediators, such as inducible nitric oxide synthase (iNOS) and its derivative nitric oxide (NO), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), IL-1ß, IL-6 and TNF-α and H3 protein acetylation in murine peritoneal macrophages. DN also inhibited p38 activation, but not extracellular signal-regulated kinase (ERK), c-jun NH2-terminal kinase (JNK), and NF-κB. These results suggest that DN from NJ exhibits protective effects against LPS-induced endotoxin shock and inflammation through p38 deactivation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Naphthols/pharmacology , Nardostachys/chemistry , Shock, Septic/prevention & control , Animals , Cytokines/biosynthesis , Enzyme Activation/drug effects , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/drug effects , Naphthols/isolation & purification , Shock, Septic/chemically induced , Shock, Septic/pathology , p38 Mitogen-Activated Protein Kinases/drug effects
17.
Eur J Pharmacol ; 765: 541-50, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26407655

ABSTRACT

Acute pancreatitis (AP) is an inflammatory disease of the pancreas, which, in its most severe form, is associated with multi-organ failure and death. Loganin, a major iridoid glycoside obtained from Corni fructus, has been shown to have anti-inflammatory and anti-shock effects. However, the effects of loganin on AP have not been determined. Pre-treatment of loganin reduced pancreatic damage and AP-associated lung injury and attenuated the severity of AP, as evidenced by (1) a reduction in several biochemical parameters (pancreatic weight to body weight ratio, myeloperoxidase activity, and level of amylase) and (2) production of pro-inflammatory cytokines such as interleukin (IL)-1ß and tumor necrosis factor (TNF)-α. However, post-treatment of loganin failed to improve pancreatic damage and biochemical parameters of AP, but could inhibit the AP-induced elevation of IL-1ß and TNF-α significantly. In addition, cerulein-induced activation of nuclear factor (NF)-κB was inhibited in the pancreas by administration of loganin. In conclusion, these results suggest that loganin exhibits an anti-inflammatory effect in cases of AP and its pulmonary complications through inhibition of NF-κB activation.


Subject(s)
Iridoids/therapeutic use , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Pancreatitis/metabolism , Pancreatitis/prevention & control , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Dose-Response Relationship, Drug , Female , Iridoids/pharmacology , Mice , Mice, Inbred C57BL , Protective Agents/pharmacology , Protective Agents/therapeutic use
18.
Phytother Res ; 29(10): 1634-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26179197

ABSTRACT

Lupeol is a triterpenoid commonly found in fruits and vegetables and is known to exhibit a wide range of biological activities, including antiinflammatory and anti-cancer effects. However, the effects of lupeol on acute pancreatitis specifically have not been well characterized. Here, we investigated the effects of lupeol on cerulein-induced acute pancreatitis in mice. Acute pancreatitis was induced via an intraperitoneal injection of cerulein (50 µg/kg). In the lupeol treatment group, lupeol was administered intraperitoneally (10, 25, or 50 mg/kg) 1 h before the first cerulein injection. Blood samples were taken to determine serum cytokine and amylase levels. The pancreas was rapidly removed for morphological examination and used in the myeloperoxidase assay, trypsin activity assay, and real-time reverse transcription polymerase chain reaction. In addition, we isolated pancreatic acinar cells using a collagenase method to examine the acinar cell viability. Lupeol administration significantly attenuated the severity of pancreatitis, as was shown by reduced pancreatic edema, and neutrophil infiltration. In addition, lupeol inhibited elevation of digestive enzymes and cytokine levels, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1, and interleukin (IL)-6. Furthermore, lupeol inhibited the cerulein-induced acinar cell death. In conclusion, these results suggest that lupeol exhibits protective effects on cerulein-induced acute pancreatitis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Ceruletide , Pancreatitis/drug therapy , Pentacyclic Triterpenes/pharmacology , Plant Extracts , Acute Disease , Amylases , Animals , Cell Survival/drug effects , Cytokines/metabolism , Injections, Intraperitoneal , Lipase/metabolism , Male , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Pancreas/drug effects , Pancreatitis/chemically induced , Peroxidase/metabolism , Plant Extracts/pharmacology , Protective Agents/pharmacology , Tumor Necrosis Factor-alpha/metabolism
19.
Int Immunopharmacol ; 26(1): 194-202, 2015 May.
Article in English | MEDLINE | ID: mdl-25843255

ABSTRACT

Guggulsterone (GS), a plant steroid and a compound found at high levels in Commiphora myrrha, exhibits anti-inflammatory, anti-cancer, and cholesterol-lowering effects. However, the potential of GS to ameliorate acute pancreatitis (AP) is unknown. The aim of this study was to evaluate the effects of GS on cerulein-induced AP. AP was induced by intraperitoneally injecting supramaximal concentrations of the stable cholecystokinin analog cerulein (50 µg/kg) hourly for 6 h. In the GS-treated group, GS was administered intraperitoneally (10, 25, or 50mg/kg) 1 h before the first cerulein injection. Mice were sacrificed 6 h after the final cerulein injection. Blood samples were collected to measure serum lipase levels and evaluate cytokine production. The pancreas and lung were rapidly removed for morphologic and histological examinations, flow cytometry analysis, myeloperoxidase (MPO) assay, and real-time reverse transcription-polymerase chain reaction analysis. Pre-treatment with GS attenuated cerulein-induced histological damage, reduced pancreas weight/body weight ratio, decreased serum lipase levels, inhibited infiltrations of macrophages and neutrophils, and suppressed cytokine production. Additionally, GS treatment suppressed the activation of extracellular signal-regulated protein kinase (ERK) and c-Jun N-terminal kinase (JNK) in the pancreas in cerulein-induced pancreatitis. In conclusion, our results suggest that GS attenuates AP via deactivation of ERK and JNK.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Ceruletide/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pancreatitis/drug therapy , Pregnenediones/therapeutic use , Acute Disease , Animals , Anti-Inflammatory Agents/administration & dosage , Blotting, Western , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Injections, Intraperitoneal , Lipase/blood , Mice, Inbred C57BL , Pancreas/drug effects , Pancreas/immunology , Pancreas/pathology , Pancreatitis/enzymology , Pancreatitis/immunology , Pregnenediones/administration & dosage
20.
Pancreas ; 44(1): 31-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25102438

ABSTRACT

OBJECTIVES: We aimed to evaluate the anti-inflammatory and inhibitory effects of Lithospermum erythrorhizon (LE) on cerulein-induced acute pancreatitis (AP) in a mouse model. METHODS: Acute pancreatitis was induced via intraperitoneal injection of cerulein (50 µg/kg) every hour for 6 times. In the LE, water extract (100, 250, or 500 mg/kg) was administered intraperitoneally 1 hour before the first injection of cerulein. Six hours after AP, blood, the pancreas, and the lung were harvested for further examination. In addition, pancreatic acinar cells were isolated using a collagenase method, and then, we investigated the acinar cell viability and cytokine productions. RESULTS: Treatment with LE reduced pancreatic damage and AP-associated lung injury and attenuated the severity of AP, as evidenced by the reduction in neutrophil infiltration, serum amylase and lipase levels, trypsin activity, and proinflammatory cytokine expression. In addition, treatment with LE inhibited high mobility group box 1 expression in the pancreas during AP. In accordance with in vivo data, LE inhibited the cerulein-induced acinar cell death, cytokine productions, and high-mobility group box 1 expression. Furthermore, LE also inhibited the activation of p38 mitogen-activated protein kinases. CONCLUSIONS: These results suggest that LE plays a protective role during the development of AP by inhibiting the activation of p38.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Ceruletide , Lithospermum/chemistry , Pancreas/drug effects , Pancreatitis/prevention & control , Plant Extracts/pharmacology , Acute Disease , Animals , Anti-Inflammatory Agents/isolation & purification , Biomarkers/blood , Cell Survival/drug effects , Cells, Cultured , Cytokines/blood , Cytoprotection , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation , Female , HMGB1 Protein/metabolism , Inflammation Mediators/blood , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Pancreas/metabolism , Pancreas/pathology , Pancreatitis/blood , Pancreatitis/chemically induced , Pancreatitis/pathology , Phytotherapy , Plant Extracts/isolation & purification , Plants, Medicinal , Severity of Illness Index , Signal Transduction/drug effects , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...