Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 306
Filter
Add more filters

Publication year range
1.
FASEB J ; 37(7): e22994, 2023 07.
Article in English | MEDLINE | ID: mdl-37249555

ABSTRACT

Mucin-2 (MUC2) secreted by goblet cells participates in the intestinal barrier, but its mechanism in acute necrotizing pancreatitis (ANP) remains unclear. In acute pancreatitis (AP) patients, the functions of goblet cells (MUC2, FCGBP, CLCA1, and TFF3) decreased, and MUC2 was negatively correlated with AP severity. ANP rats treated with pilocarpine (PILO) (PILO+ANP rats) to deplete MUC2 showed more serious pancreatic and colonic injuries, goblet cell dysfunction, gut dysbiosis, and bacterial translocation than those of ANP rats. GC-MS analysis of feces showed that PILO+ANP rats had lower levels of butyric acid, isobutyric acid, isovaleric acid, and hexanoic acid than those of ANP rats. The expression of MUC2 was associated with colonic injury and gut dysbiosis. All these phenomena could be relieved, and goblet cell functions were also partially reversed by MUC2 supplementation in ANP rats. TNF-α-treated colonoids had exacerbated goblet cell dysfunction. MUC2 expression was negatively correlated with the levels of pro-inflammatory cytokines (IL-1ß and IL-6) (p < .05) and positively related to the expression of tight junction proteins (Claudin 1, Occludin, and ZO1) (p < .05). Downregulating MUC2 by siRNA increased the levels of the pro-inflammatory cytokines in colonoids. MUC2 might maintain intestinal homeostasis to alleviate ANP.


Subject(s)
Pancreatitis, Acute Necrotizing , Rats , Animals , Mucin-2/genetics , Mucin-2/metabolism , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/drug therapy , Pancreatitis, Acute Necrotizing/metabolism , Dysbiosis/metabolism , Acute Disease , Cytokines/metabolism , Homeostasis , Intestinal Mucosa/metabolism
2.
Br J Clin Pharmacol ; 89(9): 2788-2797, 2023 09.
Article in English | MEDLINE | ID: mdl-37161703

ABSTRACT

AIMS: To analyse the clinical characteristics and risk factors for tigecycline-induced pancreatitis (TIP) and evaluate the safety and efficiency of tigecycline use in non-TIP. METHODS: A retrospective case-control study was conducted on adult and juvenile patients administered tigecycline for >3 days. The adults were classified as TIP, non-TIP (pancreatitis with other causes) and non-pancreatitis. Univariate analyses were performed to compare TIP and non-pancreatitis, and multivariate analysis was used to identify risk factors for TIP. The clinical characteristics of TIP, and the safety and efficiency of tigecycline use in non-TIP were evaluated. RESULTS: A total of 3910 patients (3823 adults and 87 juveniles) were enrolled. The adult patients comprised 21 TIP, 82 non-TIP and 3720 non-pancreatitis. The TIP prevalence was 0.56% in adults and 1.15% in juveniles. The mean time from tigecycline use to symptom onset was 7.2 days, and all cases were mild pancreatitis. The mean time from tigecycline withdrawal to symptom relief was 3.6 days. The multivariate analysis identified comorbid renal insufficiency as an independent risk factor for TIP (odds ratio = 3.032). Among the 82 non-TIP patients, 81.7% had severe pancreatitis and 47.6% had necrotizing pancreatitis. The modified computed tomography severity score after tigecycline use was similar to that before tigecycline use, but the pancreatic enzymes and infection indices were significantly decreased. CONCLUSIONS: The prevalence of TIP was low. Comorbid renal insufficiency was as an independent risk factor for TIP. Tigecycline is safe and efficient for treatment of pancreatitis, especially necrotizing pancreatitis, with intra-abdominal infection.


Subject(s)
Anti-Bacterial Agents , Pancreatitis, Acute Necrotizing , Adult , Humans , Anti-Bacterial Agents/adverse effects , Tigecycline/adverse effects , Retrospective Studies , Case-Control Studies , Tertiary Care Centers , Risk Factors , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/drug therapy
3.
Cell Tissue Res ; 380(2): 207-222, 2020 May.
Article in English | MEDLINE | ID: mdl-32318810

ABSTRACT

Acute necrotizing pancreatitis (ANP) is a common gastrointestinal cause of emergency admissions in dogs and humans and can lead to a systemic inflammatory response syndrome resulting in multiple organ dysfunction syndrome. Among the various complications associated with ANP, acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) is a major contributor leading to high mortality rates associated with severe acute pancreatitis (AP) in human patients. The incidence of ALI/ARDS in ANP dogs is not well-characterized in spontaneous AP and there are no models to study it in rodent models. Most of the data related to AP comes from rodent models of AP, which may not always represent the true mechanisms occurring in the lungs of dogs or humans with ANP. Therefore, this manuscript provides a review of current and potential models to study the role of pulmonary intravascular macrophages (PIMs) in acute pancreatitis. Recently, we characterized lung inflammation in clinical cases of AP in dogs and found significant recruitment of PIMs which have been credited as pro-inflammatory cells in species such as cattle, horse, pigs, and sheep that normally have them. Considering the pro-inflammatory roles of constitutive or induced PIMs, we investigated whether a well-established mouse model of ANP has induced PIMs. We found induced PIMs in L-arginine-induced ANP in mouse and that MCP-1 is important in PIM induction in this model. Taken together, now we summarize information on spontaneous ANP in dog and a mouse model of induced ANP to study mechanisms of lung dysfunction and the role of PIMs during ANP.


Subject(s)
Macrophages, Alveolar/metabolism , Pancreatitis, Acute Necrotizing/physiopathology , Pancreatitis/physiopathology , Animals , Disease Models, Animal , Humans , Pancreatitis, Acute Necrotizing/chemically induced
4.
Int J Mol Sci ; 21(3)2020 Jan 30.
Article in English | MEDLINE | ID: mdl-32019239

ABSTRACT

Acute pancreatitis is an inflammatory disorder of the pancreas. Its presentation ranges from self-limiting disease to acute necrotizing pancreatitis (ANP) with multiorgan failure and a high mortality. Polyethylene glycols (PEGs) are non-immunogenic, non-toxic, and water-soluble chemicals composed of repeating units of ethylene glycol. The present article explores the effect of PEG35 administration on reducing the severity of ANP and associated lung injury. ANP was induced by injection of 5% sodium taurocholate into the biliopancreatic duct. PEG35 was administered intravenously either prophylactically or therapeutically. Three hours after ANP induction, pancreas and lung tissue samples and blood were collected and ANP severity was assessed. To evaluate the inflammatory response, gene expression of pro-inflammatory cytokines and chemokine and the changes in the presence of myeloperoxidase and adhesion molecule levels were determined in both the pancreas and the lung. To evaluate cell death, lactate dehydrogenase (LDH) activity and apoptotic cleaved caspase-3 localization were determined in plasma and in both the pancreatic and lung tissue respectively. ANP-associated local and systemic inflammatory processes were reduced when PEG35 was administered prophylactically. PEG35 pre-treatment also protected against acute pancreatitis-associated cell death. Notably, the therapeutic administration of PEG35 significantly decreased associated lung injury, even when the pancreatic lesion was equivalent to that in the untreated ANP-induced group. Our results support a protective role of PEG35 against the ANP-associated inflammatory process and identify PEG35 as a promising tool for the treatment of the potentially lethal complications of the disease.


Subject(s)
Inflammation/prevention & control , Lung Injury/drug therapy , Pancreatitis, Acute Necrotizing/drug therapy , Polyethylene Glycols/pharmacology , Taurocholic Acid/toxicity , Animals , Cholagogues and Choleretics/toxicity , Inflammation/etiology , Inflammation/pathology , Interleukin-6/metabolism , Lung Injury/chemically induced , Lung Injury/pathology , Male , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/pathology , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism
5.
Gastroenterology ; 154(3): 704-718.e10, 2018 02.
Article in English | MEDLINE | ID: mdl-29079517

ABSTRACT

BACKGROUND & AIMS: Acute pancreatitis is characterized by premature intracellular activation of digestive proteases within pancreatic acini and a consecutive systemic inflammatory response. We investigated how these processes interact during severe pancreatitis in mice. METHODS: Pancreatitis was induced in C57Bl/6 wild-type (control), cathepsin B (CTSB)-knockout, and cathepsin L-knockout mice by partial pancreatic duct ligation with supramaximal caerulein injection, or by repetitive supramaximal caerulein injections alone. Immune cells that infiltrated the pancreas were characterized by immunofluorescence detection of Ly6g, CD206, and CD68. Macrophages were isolated from bone marrow and incubated with bovine trypsinogen or isolated acinar cells; the macrophages were then transferred into pancreatitis control or cathepsin-knockout mice. Activities of proteases and nuclear factor (NF)-κB were determined using fluorogenic substrates and trypsin activity was blocked by nafamostat. Cytokine levels were measured using a cytometric bead array. We performed immunohistochemical analyses to detect trypsinogen, CD206, and CD68 in human chronic pancreatitis (n = 13) and acute necrotizing pancreatitis (n = 15) specimens. RESULTS: Macrophages were the predominant immune cell population that migrated into the pancreas during induction of pancreatitis in control mice. CD68-positive macrophages were found to phagocytose acinar cell components, including zymogen-containing vesicles, in pancreata from mice with pancreatitis, as well as human necrotic pancreatic tissues. Trypsinogen became activated in macrophages cultured with purified trypsinogen or co-cultured with pancreatic acini and in pancreata of mice with pancreatitis; trypsinogen activation required macrophage endocytosis and expression and activity of CTSB, and was sensitive to pH. Activation of trypsinogen in macrophages resulted in translocation of NF-kB and production of inflammatory cytokines; mice without trypsinogen activation (CTSB-knockout mice) in macrophages developed less severe pancreatitis compared with control mice. Transfer of macrophage from control mice to CTSB-knockout mice increased the severity of pancreatitis. Inhibition of trypsin activity in macrophages prevented translocation of NF-κB and production of inflammatory cytokines. CONCLUSIONS: Studying pancreatitis in mice, we found activation of digestive proteases to occur not only in acinar cells but also in macrophages that infiltrate pancreatic tissue. Activation of the proteases in macrophage occurs during endocytosis of zymogen-containing vesicles, and depends on pH and CTSB. This process involves macrophage activation via NF-κB-translocation, and contributes to systemic inflammation and severity of pancreatitis.


Subject(s)
Cathepsin B/metabolism , Endocytosis , Macrophages/enzymology , Pancreas/enzymology , Pancreatitis, Acute Necrotizing/enzymology , Trypsinogen/metabolism , Adoptive Transfer , Animals , Cathepsin B/deficiency , Cathepsin B/genetics , Cathepsin L/deficiency , Cathepsin L/genetics , Cells, Cultured , Ceruletide , Coculture Techniques , Cytokines/metabolism , Disease Models, Animal , Enzyme Activation , Genetic Predisposition to Disease , Humans , Hydrogen-Ion Concentration , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/pathology , Macrophages/transplantation , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Necrosis , Pancreas/immunology , Pancreas/pathology , Pancreatectomy , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/immunology , Pancreatitis, Acute Necrotizing/pathology , Phagocytosis , Phenotype , Severity of Illness Index , Time Factors
6.
Biochem Biophys Res Commun ; 520(1): 211-217, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31587872

ABSTRACT

NQDI-1, an inhibitor of ASK1, has been reported to have protective effects in several experimental human disease models. However, the role of NQDI-1 in acute pancreatitis (AP) has not been reported. In this study, we found that NQDI-1 could attenuate histological damage of pancreatic tissue as well as the levels of serum amylase and lipase in a mouse model of AP induced by caerulein. Moreover, the production of reactive oxygen species (ROS) and the expression of necrosis-related proteins (RIP3 and p-MLKL) were also reduced after NQDI-1 administration. Correspondingly, we elucidated the effect of NQDI-1 in vitro and found that NQDI-1 protected against pancreatic acinar cells necrosis via decreasing the ROS production and RIP3 and p-MLKL expression. In addition, we identified the protective effect of NQDI-1 on AP through two other mouse models induced by l-arginine and pancreatic duct ligation. Taken together, these findings showed that NQDI-1 could reduce the acinar cells necrosis and alleviate the severity of AP, which may afford a new therapeutic target on pancreatic necrosis in AP clinically.


Subject(s)
Acinar Cells/cytology , Aporphines/pharmacology , Necrosis/prevention & control , Pancreatitis/prevention & control , Quinolines/pharmacology , Acinar Cells/drug effects , Amylases/blood , Animals , Cell Survival , Disease Models, Animal , Down-Regulation , Lipase/blood , Male , Mice , Mice, Inbred ICR , Pancreas/metabolism , Pancreatitis/chemically induced , Pancreatitis, Acute Necrotizing/chemically induced , Protein Kinases/metabolism , Reactive Oxygen Species , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
7.
Biochem Biophys Res Commun ; 509(2): 421-428, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30594397

ABSTRACT

Acute pancreatitis (AP) is a severe and frequently lethal disorder, but the precise mechanisms are not well understood and there is lack of effective drugs. Therefore, our study examined the in vivo intervention effects of genistein and elucidated its mechanism in acute experimental pancreatitis models. We used cerulein or taurocholate to induce acute pancreatitis (AP) in Sprague-Dawley rats with prior genistein treatment. Histological examination of the pancreas was performed and the expression of unfolded protein response (UPR) components and apoptotic mediators like caspase 12 and c-Jun N-terminal protein kinase (JNK) were measured. The amount of apoptosis in pancreatic acinar cells was also determined. Our studies found that the severity of cerulein- or taurocholate-induced AP was rescued by prior genistein treatment. Genistein stimulated the activation of multiple endoplasmic reticulum (ER) stress-related regulators like GRP78, PERK, eIF2α, and upregulated the expression of the apoptotic genes, caspase 12 and CHOP. Moreover, TUNEL assays showed that genistein treatment promoted acinar cell apoptosis. Taken together, we speculated that ER stress-associated apoptotic pathways in AP are induced by genistein, which showed cytoprotective capacity in the exocrine pancreas. These data suggest novel therapeutic strategies that employ genistein in the prevention of AP.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Genistein/pharmacology , Pancreatitis, Acute Necrotizing/drug therapy , Acinar Cells/drug effects , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Apoptosis/genetics , Caspase 12/genetics , Caspase 12/metabolism , Ceruletide/administration & dosage , Endoplasmic Reticulum Stress/genetics , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/genetics , Pancreatitis, Acute Necrotizing/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Taurocholic Acid/administration & dosage , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Unfolded Protein Response/drug effects , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
8.
Appl Environ Microbiol ; 85(12)2019 06 15.
Article in English | MEDLINE | ID: mdl-30979838

ABSTRACT

An increase of Escherichia-Shigella was previously reported in acute necrotizing pancreatitis (ANP). We investigated whether Escherichia coli MG1655, an Escherichia commensal organism, increased intestinal injury and aggravated ANP in rats. ANP was induced by retrograde injection of 3.5% sodium taurocholate into the biliopancreatic duct. Using gut microbiota-depleted rats, we demonstrated that gut microbiota was involved in the pancreatic injury and intestinal barrier dysfunction in ANP. Using 16S rRNA gene sequencing and quantitative PCR, we found intestinal dysbiosis and a significant increase of E. coli MG1655 in ANP. Afterward, administration of E. coli MG1655 by gavage to gut microbiota-depleted rats with ANP was performed. We observed that after ANP induction, E. coli MG1655-monocolonized rats presented more severe injury in the pancreas and intestinal barrier function than gut microbiota-depleted rats. Furthermore, Toll-like receptor 4 (TLR4)/MyD88/p38 mitogen-activated protein (MAPK) and endoplasmic reticulum stress (ERS) activation in intestinal epithelial cells were also increased more significantly in the MG1655-monocolonized ANP rats. In vitro, the rat ileal epithelial cell line IEC-18 displayed aggravated tumor necrosis factor alpha-induced inflammation and loss of tight-junction proteins in coculture with E. coli MG1655, as well as TLR4, MyD88, and Bip upregulation. In conclusion, our study shows that commensal E. coli MG1655 increases TLR4/MyD88/p38 MAPK and ERS signaling-induced intestinal epithelial injury and aggravates ANP in rats. Our study also describes the harmful potential of commensal E. coli in ANP.IMPORTANCE This study describes the harmful potential of commensal E. coli in ANP, which has not been demonstrated in previous studies. Our work provides new insights into gut bacterium-ANP cross talk, suggesting that nonpathogenic commensals could also exhibit adverse effects in the context of diseases.


Subject(s)
Dysbiosis/physiopathology , Escherichia coli/physiology , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/microbiology , Pancreatitis, Acute Necrotizing/microbiology , Animals , Male , Pancreatitis, Acute Necrotizing/chemically induced , Rats , Rats, Sprague-Dawley , Symbiosis , Taurocholic Acid/pharmacology
9.
J Pediatr Hematol Oncol ; 41(2): e125-e128, 2019 03.
Article in English | MEDLINE | ID: mdl-29683955

ABSTRACT

Acute pancreatitis in children acute lymphoblastic leukemia is commonly caused by drugs, for example, L-Asparaginase, pegapargase, steroids. The incidence of this complication is estimated at 6.7% to 18%. Although the majority of drug-induced acute pancreatitis cases are mild, severe cases can rarely occur. This work presents a case of successful management of a child with drug-induced necrotizing pancreatitis during acute lymphoblastic leukemia therapy. This case illustrates that comprehensive care and immediate intensive treatment can rescue patient despite poor prognosis. Administration of octreotide may serve a role in limiting the severity of the disease.


Subject(s)
Antineoplastic Agents , Octreotide/administration & dosage , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Humans , Male
10.
Pancreatology ; 18(7): 742-752, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30115563

ABSTRACT

OBJECTIVE: To investigate the effects of quercetin on intestinal barrier disruption and inflammation in acute necrotizing pancreatitis (ANP) in rats, and its possible mechanism. METHODS: ANP was established by retrograde injection of 3.5% sodium taurocholate into the biliopancreatic duct, and quercetin (50 mg/kg × 3) was administered by intraperitoneal injection prior to and after ANP induction. Pancreatitis was assessed by pancreatic histopathology, plasma amylase, pancreatic myeloperoxidase (MPO) activity, IL-1ß, TNFα and IL-6 levels. Injury of the distal ileum was assessed by histological evaluation. The ultrastructural changes of ileal epithelial cells were examined by transmission electron microscope (TEM). Intestinal barrier function was estimated by plasma diamine oxidase (DAO), d-lactate, endotoxin; and intestinal tight junction proteins including zonula occludens-1 (ZO-1), claudin 1, occludin; and bacterial translocation. Intestinal inflammation was determined by IL-1ß, TNFα and IL-17 A expression. TLR4, MyD88, pp38 MAPK, and endoplasmic reticulum stress (ERS)-related molecules (Bip, p-IRE1α, sXBP1, p-eIF2α, ATF6) were measured by immunohistochemistry and WB. RESULTS: Quercetin intervention attenuated pancreatic and ileal pathological damages in ANP (P < 0.05), ameliorated intestinal barrier disruption and inflammation (P < 0.05). Meantime, QE significantly suppressed intestinal TLR4/MyD88/p38 MAPK pathway and ERS activation. CONCLUSIONS: Quercetin plays a protective role against intestinal barrier disruption and inflammation in ANP, probably partly by inhibiting TLR4/MyD88/p38 MAPK and ERS activation.


Subject(s)
Inflammation/prevention & control , Myeloid Differentiation Factor 88/metabolism , Pancreatitis, Acute Necrotizing/drug therapy , Quercetin/pharmacology , Toll-Like Receptor 4/metabolism , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation/drug effects , Humans , Intestines/drug effects , Myeloid Differentiation Factor 88/genetics , Pancreatitis, Acute Necrotizing/chemically induced , Taurocholic Acid/toxicity , Toll-Like Receptor 4/genetics , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
11.
J Immunol ; 197(10): 4137-4150, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27798150

ABSTRACT

Chromatin remodeling seems to regulate the patterns of proinflammatory genes. Our aim was to provide new insights into the epigenetic mechanisms that control transcriptional activation of early- and late-response genes in initiation and development of severe acute pancreatitis as a model of acute inflammation. Chromatin changes were studied by chromatin immunoprecipitation analysis, nucleosome positioning, and determination of histone modifications in promoters of proinflammatory genes in vivo in the course of taurocholate-induced necrotizing pancreatitis in rats and in vitro in rat pancreatic AR42J acinar cells stimulated with taurocholate or TNF-α. Here we show that the upregulation of early and late inflammatory genes rely on histone acetylation associated with recruitment of histone acetyltransferase CBP. Chromatin remodeling of early genes during the inflammatory response in vivo is characterized by a rapid and transient increase in H3K14ac, H3K27ac, and H4K5ac as well as by recruitment of chromatin-remodeling complex containing BRG-1. Chromatin remodeling in late genes is characterized by a late and marked increase in histone methylation, particularly in H3K4. JNK and p38 MAPK drive the recruitment of transcription factors and the subsequent upregulation of early and late inflammatory genes, which is associated with nuclear translocation of the early gene Egr-1 In conclusion, specific and strictly ordered epigenetic markers such as histone acetylation and methylation, as well as recruitment of BRG-1-containing remodeling complex are associated with the upregulation of both early and late proinflammatory genes in acute pancreatitis. Our findings highlight the importance of epigenetic regulatory mechanisms in the control of the inflammatory cascade.


Subject(s)
Chromatin Assembly and Disassembly , Epigenesis, Genetic , Gene Expression Regulation , Pancreatitis, Acute Necrotizing/genetics , Pancreatitis, Acute Necrotizing/immunology , Transcriptional Activation , Acetylation , Acinar Cells/drug effects , Animals , Chromatin Immunoprecipitation , DNA Helicases/genetics , Early Growth Response Protein 1/genetics , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histones/metabolism , Inflammation/genetics , Methylation , Nuclear Proteins/genetics , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/metabolism , Promoter Regions, Genetic , Protein Processing, Post-Translational , Rats , Taurocholic Acid/pharmacology , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/pharmacology
12.
Arch Gynecol Obstet ; 297(4): 933-942, 2018 04.
Article in English | MEDLINE | ID: mdl-29349553

ABSTRACT

OBJECTIVE: The objective of this study was to determine the mechanism of acute renal injury (ARI) in acute necrotizing pancreatitis in late pregnancy (ANPIP). METHODS: Pregnant Sprague-Dawley rats in the third trimester were used for this study, and an ANPIP model was induced by injecting 5% sodium taurocholate into the biliary pancreatic duct. The rats were randomly divided into three groups: the normal, sham-operated (SO) and acute necrotizing pancreatitis (ANP) groups. Rats were killed at 3, 6, 12 h after the operation, and blood, pancreatic and renal tissue samples were harvested. Differences were detected in the physiology, pathology and cellular and molecular responses among the different groups. RESULT: Serum amylase, lipase, urea and Cr levels were increased in rats with ANPIP. Additionally, expression of phosphorylation p38 and JNK as well as TNF-α and NF-κB were increased in the renal tissues of rats with ANPIP. The expression of phosphorylation ERK was decreased in the renal tissues of rats with ANPIP. CONCLUSIONS: Mitogen-activated protein kinases may play an important role in renal injury in rat models of ANPIP.


Subject(s)
Acute Kidney Injury/complications , JNK Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Pancreatitis, Acute Necrotizing/chemically induced , Taurocholic Acid/toxicity , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Female , Male , Mitogen-Activated Protein Kinases , Pancreatitis, Acute Necrotizing/complications , Phosphorylation , Pregnancy , Random Allocation , Rats , Rats, Sprague-Dawley
14.
J Clin Pharm Ther ; 42(4): 495-498, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28393377

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: Drug-induced acute pancreatitis comprises only 0.5%-2% of all cases of acute pancreatitis. Propofol is a potentially dangerous drug that can cause acute pancreatitis, but this complication is extremely rare. CASE SUMMARY: A 57-year-old patient developed acute pancreatitis after a planned thyroidectomy. As the common causes of acute pancreatitis were excluded, we believe that the pancreatitis was drug-induced, in this case by a single dose of propofol administered to the patient during the surgery. WHAT IS NEW AND CONCLUSION: We present a rare case of propofol-induced acute necrotising pancreatitis, which is to the best of our knowledge the first fatal case reported in an adult patient.


Subject(s)
Anesthetics, Intravenous/adverse effects , Pancreatitis, Acute Necrotizing/chemically induced , Propofol/adverse effects , Anesthetics, Intravenous/administration & dosage , Fatal Outcome , Female , Humans , Middle Aged , Propofol/administration & dosage , Thyroidectomy/methods
15.
Gut ; 65(8): 1333-46, 2016 08.
Article in English | MEDLINE | ID: mdl-26071131

ABSTRACT

OBJECTIVE: Acute pancreatitis is caused by toxins that induce acinar cell calcium overload, zymogen activation, cytokine release and cell death, yet is without specific drug therapy. Mitochondrial dysfunction has been implicated but the mechanism not established. DESIGN: We investigated the mechanism of induction and consequences of the mitochondrial permeability transition pore (MPTP) in the pancreas using cell biological methods including confocal microscopy, patch clamp technology and multiple clinically representative disease models. Effects of genetic and pharmacological inhibition of the MPTP were examined in isolated murine and human pancreatic acinar cells, and in hyperstimulation, bile acid, alcoholic and choline-deficient, ethionine-supplemented acute pancreatitis. RESULTS: MPTP opening was mediated by toxin-induced inositol trisphosphate and ryanodine receptor calcium channel release, and resulted in diminished ATP production, leading to impaired calcium clearance, defective autophagy, zymogen activation, cytokine production, phosphoglycerate mutase 5 activation and necrosis, which was prevented by intracellular ATP supplementation. When MPTP opening was inhibited genetically or pharmacologically, all biochemical, immunological and histopathological responses of acute pancreatitis in all four models were reduced or abolished. CONCLUSIONS: This work demonstrates the mechanism and consequences of MPTP opening to be fundamental to multiple forms of acute pancreatitis and validates the MPTP as a drug target for this disease.


Subject(s)
Acinar Cells , Mitochondrial Membrane Transport Proteins , Mitochondrial Proteins/metabolism , Pancreas , Pancreatitis, Acute Necrotizing , Phosphoprotein Phosphatases/metabolism , Acinar Cells/drug effects , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Autophagy/drug effects , Calcium/metabolism , Cell Culture Techniques , Disease Models, Animal , Humans , Inositol Phosphates/metabolism , Inositol Phosphates/pharmacology , Mice , Mitochondria/enzymology , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Necrosis , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/metabolism , Pancreatitis, Acute Necrotizing/pathology
16.
J Surg Res ; 203(2): 383-9, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27363647

ABSTRACT

BACKGROUND: Inflammatory explosion and oxidative stress are important mechanisms of injury in acute necrotizing pancreatitis (ANP). This study investigated the effects of N-acetylcysteine amid (NACA), a novel cell-permeant antioxidant with anti-inflammatory activity, on experimental ANP in rats. MATERIALS AND METHODS: Fifty-two adult male Sprague-Dawley rats were used, and ANP was induced by cerulein. The animals were divided into four groups which were sham + saline, sham + NACA, ANP + saline, and ANP + NACA. NACA (2.2 mg/kg, i.p) was administered for 6 h, after the induction of ANP. The extent of acinar cell injury, mortality, systemic cardiorespiratory variables, functional capillary density, renal/hepatic functions, and changes in some enzyme markers for pancreas and lung tissues were investigated. RESULTS: Induction of ANP increased mortality from 0% in the sham group to 43.75% in the ANP + saline group (P < 0.05), and administration of NACA significantly reduced mortality to 12.5% (P < 0.05). Induction of ANP also caused increases in pancreatic necrosis, serum amylase, alanine aminotransferase (ALT), interleukin-6, LDH in bronchoalveolar lavage fluid, serum urea, tissue myeloperoxidase in pancreas and lung tissues and malondialdehyde. There was less pronounced increase in these parameters in NACA treated group. Compared with ANP group, ANP + NACA group had lower levels of pancreatic necrosis (0.5 ± 0.2 versus 1.45 ± 0.2, P < 0.05) and inflammation (0.6 ± 0.2 versus 1.29 ± 00.3, P < 0.05) scores. CONCLUSIONS: Administration of NACA significantly decreased the ANP-induced mortality and also provided significant improvements in hemodynamic changes. The obtained positive effects of NACA on the course of pancreatitis indicates its potential usefulness in the management of ANP.


Subject(s)
Acetylcysteine/analogs & derivatives , Antioxidants/therapeutic use , Pancreatitis, Acute Necrotizing/drug therapy , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Animals , Antioxidants/pharmacology , Biomarkers/metabolism , Ceruletide , Hemodynamics/drug effects , Kidney/drug effects , Kidney/physiopathology , Liver/drug effects , Liver/physiopathology , Male , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Pancreas/physiopathology , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/mortality , Pancreatitis, Acute Necrotizing/physiopathology , Random Allocation , Rats , Rats, Sprague-Dawley , Treatment Outcome
17.
J Biochem Mol Toxicol ; 30(6): 302-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26862043

ABSTRACT

The aim of the study was to investigate the ameliorative effects of curcumin on fibrinogen like protein-2 (fgl-2), some oxido-inflammatory and apoptotic markers in rat-induced acute pancreatitis (AP). Seventy-five albino rats were divided into control group, l-arginine (l-Arg)-induced AP group, curcumin pre-treated group before AP induction, curcumin post-treated group after AP induction, and curcumin injected group only. AP group showed severe necrotizing pancreatitis confirmed by histopathological changes and elevations in serum amylase and lipase activities, levels of epithelial neutrophil-activating peptide 78, tissue content of protein carbonyls, levels of tumor necrosis factor α, and caspase-3 as well as myeloperoxidase activity. Significant elevation in pancreatic fgl-2 mRNA expression was detected in AP group. Improvement of all parameters was detected with increase of caspase-3 in both curcumin-treated groups that confirmed curcumin ameliorative effects against AP through induction of apoptosis and inhibition of micro-thrombosis, inflammation, and oxidative stress.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Curcumin/pharmacology , Fibrinogen/genetics , Pancreatitis, Acute Necrotizing/drug therapy , Animals , Arginine , Caspase 3/genetics , Caspase 3/metabolism , Disease Models, Animal , Fibrinogen/metabolism , Gene Expression Regulation , Inflammation/prevention & control , Injections, Intraperitoneal , Male , Oxidative Stress/drug effects , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/genetics , Pancreatitis, Acute Necrotizing/pathology , Peroxidase/genetics , Peroxidase/metabolism , Protein Carbonylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
Appl Microbiol Biotechnol ; 100(1): 337-46, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26411454

ABSTRACT

Hydrogen sulphide (H2S) is an endogenous inflammatory mediator produced by cystathionine-γ-lyase (CSE) in monocytes/macrophages. To determine the role of H2S and macrophages in inflammation, we used small interference RNA (siRNA) to target the CSE gene and investigated its effect in a mouse model of acute pancreatitis. Acute pancreatitis is characterised by increased levels of plasma amylase, myeloperoxidase (MPO) activity and pro-inflammatory cytokines and chemokines in the pancreas and lung. SiRNA treatment attenuated inflammation in the pancreas and lungs of mice following caerulein-induced acute pancreatitis. MPO activity increased in caerulein-induced acute pancreatitis (16.21 ± 3.571 SD fold increase over control) and treatment with siRNA significantly reduced this (mean 3.555 ± 2.522 SD fold increase over control) (p < 0.0001). Similarly, lung MPO activity increased following treatment with caerulein (3.56 ± 0.941 SD fold increase over control) while siRNA treatment significantly reduced MPO activity (0.8243 ± 0.4353 SD fold increase over control) (p < 0.0001). Caerulein treatment increased plasma amylase activity (7094 ± 207 U/l) and this significantly decreased following siRNA administration (5895 ± 115 U/l) (p < 0.0001). Cytokine and chemokine levels in caerulein-induced acute pancreatitis reduced following treatment with siRNA. For example, siRNA treatment significantly decreased pancreatic and lung monocyte chemoattractant protein (MCP)-1 (169.8 ± 59.75 SD; 90.01 ± 46.97 SD pg/ml, respectively) compared to caerulein-treated mice (324.7 ± 103.9 SD; 222.8 ± 85.37 SD pg/ml, pancreas and lun,g respectively) (p < 0.0001). These findings show a crucial pro-inflammatory role for H2S synthesised by CSE in macrophages in acute pancreatitis and suggest CSE gene silencing with siRNA as a potential therapeutic approach for this condition.


Subject(s)
Cystathionine gamma-Lyase/antagonists & inhibitors , Cystathionine gamma-Lyase/genetics , Hydrogen Sulfide/metabolism , Inflammation Mediators/metabolism , Monocytes/enzymology , Pancreatitis, Acute Necrotizing/prevention & control , RNA, Small Interfering/metabolism , Amylases/blood , Animals , Blood Chemical Analysis , Ceruletide/administration & dosage , Ceruletide/toxicity , Cytokines/blood , Disease Models, Animal , Gene Silencing , Lung/pathology , Mice , Monocytes/immunology , Pancreas/pathology , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/pathology , Peroxidase/analysis
19.
Pharm Biol ; 54(12): 2951-2959, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27339751

ABSTRACT

CONTEXT: Calendula officinalis L. (Asteraceae) has been traditionally used in treating inflammation of internal organs, gastrointestinal tract ulcers and wound healing. OBJECTIVE: The present study investigates the effect of ethanol extract (95%) of Calendula officinalis flowers in l-arginine induced acute necrotizing pancreatitis in rats. MATERIALS AND METHODS: Rats were divided into four groups: normal control, l-arginine control, Calendula officinalis extract (COE) treated and melatonin treated (positive control), which were further divided into subgroups (24 h, day 3 and 14) according to time points. Two injections of l-arginine 2 g/kg i.p. at 1 h intervals were administered in l-arginine control, COE and melatonin-treated groups to produce acute necrotizing pancreatitis. Biochemical parameters [serum amylase, lipase, pancreatic amylase, nucleic acid content, total proteins, transforming growth factor-ß1 (TGF-ß1), collagen content, lipid peroxidation, reduced glutathione and nitrite/nitrate] and histopathological studies were carried out. RESULTS: COE treatment (400 mg/kg p.o.) was found to be beneficial. This was evidenced by significantly lowered histopathological scores (2 at day 14). Nucleic acid content (DNA 21.1 and RNA 5.44 mg/g pancreas), total proteins (0.66 mg/mL pancreas) and pancreatic amylase (1031.3 100 SU/g pancreas) were significantly improved. Marked reduction in pancreatic oxidative and nitrosative stress; collagen (122 µmoles/100 mg pancreas) and TGF-ß1 (118.56 pg/mL) levels were noted. Results obtained were comparable to those of positive control. DISCUSSION AND CONCLUSION: The beneficial effect of COE may be attributed to its antioxidant, antinitrosative and antifibrotic actions. Hence, the study concludes that COE promotes spontaneous repair and regeneration of the pancreas.


Subject(s)
Arginine/toxicity , Calendula , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/drug therapy , Plant Extracts/therapeutic use , Animals , Female , Male , Pancreatitis, Acute Necrotizing/metabolism , Plant Extracts/isolation & purification , Rats , Rats, Sprague-Dawley
20.
Bratisl Lek Listy ; 117(10): 614-618, 2016.
Article in English | MEDLINE | ID: mdl-27826979

ABSTRACT

INTRODUCTION: The aim of this study is to investigate the potential antioxidant and anti-inflammatory effects of thymoquinone (TQ) on ceruleine induced acute pancreatitis. MATERIAL AND METHODS: A total of 14 male Wistar albino rats were divided into 2 groups as follows: (1) normal saline-treated group and (2) thymoquinone- treated groups. For achieving acute pancreatitis, intraperitoneal (IP) cerulein, a stable cholecystokinin (CCK) analogue, was applied in a 50 mcg/kg dose 2 times in one-hour interval in total. One hour after last ceruleine injection, IP 2 ml/kg isotonic saline solution was applied to the saline group and IP 5 mg/kg TQ was applied. The rats were sacrificed by decapitation 12 h after the last injection of last medication. Blood samples were taken, and serum interleukin-1ß (IL-1ß), amylase, lipase pancreatic, total antioxidant capacity (TAC), total oxidant status (TOS), and pancreatic Schoenberg scores were determined. Oxidative stress index (OSI) was calculated for each group. Results are given as mean ± SD. A value of p < 0.05 was accepted as statistically significant. SPSS for Windows v15.0 was used for statistical analyses. RESULTS: The increased serum amylase, lipase levels and histopathological scoring of pancreatic tissue showed that acute pancreatitis was present in both groups. Furthermore, serum IL-1ß level was significantly reduced in TQ administered group (p < 0.05). Although serum TAC level was high and TOS level was low, those changes were not statistically significant. Nevertheless, OSI index, which was driven from TOS/TAC, was significantly low in TQ groups (p < 0.05). Although TQ partially ameliorated the acute pancreatitis in terms of histopathological evaluations, the main effect of it was brought about by reducing the hemorrhage in acute pancreatitis (p < 0.05). CONCLUSION: In this study, it was shown that TQ can reduce the inflammation and has a positive effect on the oxidative status of organism in inflammatory cases such as acute pancreatitis. This is consistent with partial amelioration of acute pancreatitis in rats given TQ (Tab. 2, Fig. 4, Ref. 31).


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Benzoquinones/pharmacology , Pancreatitis, Acute Necrotizing/chemically induced , Animals , Ceruletide , Disease Models, Animal , Interleukin-1beta/blood , Male , Oxidative Stress/drug effects , Pancreas/drug effects , Pancreas/pathology , Pancreatitis, Acute Necrotizing/pathology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL