Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
1.
Cell Death Dis ; 13(8): 744, 2022 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-36038551

RESUMO

Alcohol abuse, an increasing problem in developed societies, is one of the leading causes of acute and chronic pancreatitis. Alcoholic pancreatitis is often associated with fibrosis mediated by activated pancreatic stellate cells (PSCs). Alcohol toxicity predominantly depends on its non-oxidative metabolites, fatty acid ethyl esters, generated from ethanol and fatty acids. Although the role of non-oxidative alcohol metabolites and dysregulated Ca2+ signalling in enzyme-storing pancreatic acinar cells is well established as the core mechanism of pancreatitis, signals in PSCs that trigger fibrogenesis are less clear. Here, we investigate real-time Ca2+ signalling, changes in mitochondrial potential and cell death induced by ethanol metabolites in quiescent vs TGF-ß-activated PSCs, compare the expression of Ca2+ channels and pumps between the two phenotypes and the consequences these differences have on the pathogenesis of alcoholic pancreatitis. The extent of PSC activation in the pancreatitis of different aetiologies has been investigated in three animal models. Unlike biliary pancreatitis, alcohol-induced pancreatitis results in the activation of PSCs throughout the entire tissue. Ethanol and palmitoleic acid (POA) or palmitoleic acid ethyl ester (POAEE) act directly on quiescent PSCs, inducing cytosolic Ca2+ overload, disrupting mitochondrial functions, and inducing cell death. However, activated PSCs acquire remarkable resistance against ethanol metabolites via enhanced Ca2+-handling capacity, predominantly due to the downregulation of the TRPA1 channel. Inhibition or knockdown of TRPA1 reduces EtOH/POA-induced cytosolic Ca2+ overload and protects quiescent PSCs from cell death, similarly to the activated phenotype. Our results lead us to review current dogmas on alcoholic pancreatitis. While acinar cells and quiescent PSCs are prone to cell death caused by ethanol metabolites, activated PSCs can withstand noxious signals and, despite ongoing inflammation, deposit extracellular matrix components. Modulation of Ca2+ signals in PSCs by TRPA1 agonists/antagonists could become a strategy to shift the balance of tissue PSCs towards quiescent cells, thus limiting pancreatic fibrosis.


Assuntos
Células Estreladas do Pâncreas , Pancreatite Alcoólica , Animais , Morte Celular , Regulação para Baixo/genética , Etanol/toxicidade , Ácidos Graxos/metabolismo , Fibrose , Pâncreas/patologia , Pancreatite Alcoólica/induzido quimicamente , Pancreatite Alcoólica/metabolismo , Pancreatite Alcoólica/patologia
2.
Biochim Biophys Acta Mol Basis Dis ; 1868(11): 166486, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-35835415

RESUMO

BACKGROUND: Alcohol abuse, a main cause of pancreatitis, has been known to augment NF-κB activation and cell necrosis in pancreatitis. However, the underlying mechanisms are unclear. We recently reported that inhibition of protein kinase D (PKD) alleviated NF-κB activation and severity of experimental pancreatitis. Here we investigated whether PKD signaling mediated the modulatory effects of alcohol abuse on pathological responses in alcoholic pancreatitis. METHODS: Alcoholic pancreatitis was provoked in two rodent models with pair-feeding control and ethanol-containing Lieber-DeCarli diets for up to 8 weeks followed by up to 7 hourly intraperitoneal injections of cerulein at 1 µg/kg (rats) or 3 µg/kg (mice). Effects of PKD inhibition by PKD inhibitors or genetic deletion of pancreatic PKD isoform (PKD3Δpanc mice) on alcoholic pancreatitis parameters were determined. RESULTS: Ethanol administration amplified PKD signaling by promoting expression and activation of pancreatic PKD, resulted in augmented/promoted pancreatitis responses. Pharmacological inhibition of PKD or with PKD3Δpanc mice prevented the augmenting/sensitizing effect of ethanol on NF-κB activation and inflammatory responses, cell necrotic death and the severity of disease in alcoholic pancreatitis. PKD inhibition prevented alcohol-enhanced trypsinogen activation, mRNA expression of multiple inflammatory molecules, the receptor-interacting protein kinase activation, ATP depletion, and downregulation of pro-survival Bcl-2 protein in alcoholic pancreatitis. Furthermore, PKD inhibitor CID755673 or CRT0066101, administrated after the induction of pancreatitis in mouse and rat alcoholic pancreatitis models, significantly mitigated the severity of pancreatitis. CONCLUSION: PKD mediates effect of alcohol abuse on pathological process of pancreatitis and constitutes a novel therapeutic target to treat this disease.


Assuntos
Alcoolismo , Pancreatite Alcoólica , Trifosfato de Adenosina , Alcoolismo/complicações , Alcoolismo/tratamento farmacológico , Alcoolismo/genética , Animais , Ceruletídeo , Etanol/toxicidade , Camundongos , NF-kappa B/metabolismo , Necrose , Pancreatite Alcoólica/tratamento farmacológico , Pancreatite Alcoólica/genética , Pancreatite Alcoólica/metabolismo , Proteína Quinase C/genética , Proteínas Proto-Oncogênicas c-bcl-2 , RNA Mensageiro , Ratos , Tripsinogênio
3.
Cell Mol Life Sci ; 79(5): 265, 2022 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-35484438

RESUMO

Alcoholic pancreatitis and hepatitis are frequent, potentially lethal diseases with limited treatment options. Our previous study reported that the expression of CFTR Cl- channel is impaired by ethanol in pancreatic ductal cells leading to more severe alcohol-induced pancreatitis. In addition to determining epithelial ion secretion, CFTR has multiple interactions with other proteins, which may influence intracellular Ca2+ signaling. Thus, we aimed to investigate the impact of ethanol-mediated CFTR damage on intracellular Ca2+ homeostasis in pancreatic ductal epithelial cells and cholangiocytes. Human and mouse pancreas and liver samples and organoids were used to study ion secretion, intracellular signaling, protein expression and interaction. The effect of PMCA4 inhibition was analyzed in a mouse model of alcohol-induced pancreatitis. The decreased CFTR expression impaired PMCA function and resulted in sustained intracellular Ca2+ elevation in ethanol-treated and mouse and human pancreatic organoids. Liver samples derived from alcoholic hepatitis patients and ethanol-treated mouse liver organoids showed decreased CFTR expression and function, and impaired PMCA4 activity. PMCA4 co-localizes and physically interacts with CFTR on the apical membrane of polarized epithelial cells, where CFTR-dependent calmodulin recruitment determines PMCA4 activity. The sustained intracellular Ca2+ elevation in the absence of CFTR inhibited mitochondrial function and was accompanied with increased apoptosis in pancreatic epithelial cells and PMCA4 inhibition increased the severity of alcohol-induced AP in mice. Our results suggest that improving Ca2+ extrusion in epithelial cells may be a potential novel therapeutic approach to protect the exocrine pancreatic function in alcoholic pancreatitis and prevent the development of cholestasis in alcoholic hepatitis.


Assuntos
Hepatite Alcoólica , Hepatite , Pancreatite Alcoólica , Animais , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Epiteliais/metabolismo , Etanol/toxicidade , Hepatite/metabolismo , Hepatite Alcoólica/genética , Hepatite Alcoólica/metabolismo , Humanos , Camundongos , Pancreatite Alcoólica/metabolismo
4.
Dig Dis Sci ; 67(2): 524-535, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-33555515

RESUMO

OBJECTIVE: Alcohol consumption is always the main cause of acute pancreatitis (AP). It has been reported that alcohol exerts direct damage to the pancreas. However, the specific role of alcohol during AP needs to be investigated. This study aims to examine the effects of alcohol in cerulein-induced AP and the role of the AMPK pathway. METHODS: Human subjects from operations, cerulein-induced AP rat, and cerulein-stimulated AR42J cell line were enrolled in this study. Electron microscopy was employed for observation of cell morphology, immunohistochemistry for identification of cells, ELISA for detection of inflammation factors, Annexin V/PI double staining for evaluation of cell apoptosis, immunofluorescence for assessment of autophagic flux, oil red O staining for examination of lipid droplet accumulation, and Western blot for measurement of expressions of proteins related to autophagy, apoptosis, and AMPK signal pathway. PI3K inhibitor 3-MA and AMPK inhibitor BML-275 were utilized for investigation of the relationship between impaired autophagic flux and the AMPK pathway by inhibiting or stimulating the formation of autophagosome. RESULTS: Alcohol consumption caused lipid droplet accumulation in the pancreas, and it also activated AMPK signaling pathway, thus aggravating the autophagic flux during AP. Alcohol up-regulated the expressions of anti-apoptotic proteins during the induction of AP to inhibit cell apoptosis and enhance cell necrosis. Inhibition of autophagosome formation by AMPK inhibitor BML-275 ameliorated the decreased cell viability caused by alcohol and cerulein in vitro. CONCLUSION: Alcohol aggravates AP progression by impairing autophagic flux and enhancing cell autophagy through the AMPK signaling pathway.


Assuntos
Adenilato Quinase/metabolismo , Autofagia/efeitos dos fármacos , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Pâncreas/efeitos dos fármacos , Pancreatite Alcoólica/metabolismo , Adenilato Quinase/antagonistas & inibidores , Adenilato Quinase/efeitos dos fármacos , Animais , Linhagem Celular , Ceruletídeo/toxicidade , Humanos , Pancreatite/induzido quimicamente , Pancreatite/metabolismo , Pancreatite/patologia , Pancreatite Alcoólica/patologia , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Ratos , Transdução de Sinais
5.
PLoS One ; 16(11): e0260309, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34807958

RESUMO

AIM: Liver cirrhosis and features of muscle or adipose tissues may affect the severity of acute pancreatitis (AP). We aimed to evaluate the impact of body composition parameters and liver cirrhosis on the severity of AP in patients with alcohol-induced AP (AAP). METHODS: Patients with presumed AAP who underwent CT within one week after admission were retrospectively enrolled. L3 sectional areas of abdominal fat and muscle, and mean muscle attenuations (MMAs) were quantified. The presence of liver cirrhosis was determined using clinical and CT findings. Factors potentially associated with moderately severe or severe AP were included in the multivariable logistic regression analysis. RESULTS: A total of 242 patients (47.0 ± 12.6 years, 215 males) with presumed AAP were included. The mild and moderately severe/severe (MSS) groups included 137 (56.6%) and 105 patients (43.4%), respectively. Patients in the MSS group had higher rates of liver cirrhosis, organ failure, and local complications. Among body composition parameters, mean MMA (33.4 vs 36.8 HU, P<0.0001) and abdominal muscle mass (126.5 vs 135.1 cm2, P = 0.029) were significantly lower in the MSS group. The presence of liver cirrhosis (OR, 4.192; 95% CI, 1.620-10.848) was found to be a significant risk factor for moderately severe or severe AP by multivariable analysis. CONCLUSION: The results of this study suggest that liver cirrhosis has a significant impact on the severity of AAP. Of the body composition parameters examined, MMA and abdominal muscle mass showed potential as promising predictors.


Assuntos
Cirrose Hepática/complicações , Pancreatite Alcoólica/complicações , Doença Aguda , Adulto , Idoso , Composição Corporal , Feminino , Humanos , Cirrose Hepática/metabolismo , Masculino , Pessoa de Meia-Idade , Pancreatite Alcoólica/metabolismo , Estudos Retrospectivos , Índice de Gravidade de Doença
6.
Gastroenterology ; 161(3): 982-995.e2, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34051238

RESUMO

BACKGROUND & AIMS: Heavy alcohol consumption is a common cause of acute pancreatitis; however, alcohol abuse does not always result in clinical pancreatitis. As a consequence, the factors responsible for alcohol-induced pancreatitis are not well understood. In experimental animals, it has been difficult to produce pancreatitis with alcohol. Clinically, alcohol use predisposes to hypophosphatemia, and hypophosphatemia has been observed in some patients with acute pancreatitis. Because of abundant protein synthesis, the pancreas has high metabolic demands, and reduced mitochondrial function leads to organelle dysfunction and pancreatitis. We proposed, therefore, that phosphate deficiency might limit adenosine triphosphate synthesis and thereby contribute to alcohol-induced pancreatitis. METHODS: Mice were fed a low-phosphate diet (LPD) before orogastric administration of ethanol. Direct effects of phosphate and ethanol were evaluated in vitro in isolated mouse pancreatic acini. RESULTS: LPD reduced serum phosphate levels. Intragastric administration of ethanol to animals maintained on an LPD caused severe pancreatitis that was ameliorated by phosphate repletion. In pancreatic acinar cells, low-phosphate conditions increased susceptibility to ethanol-induced cellular dysfunction through decreased bioenergetic stores, specifically affecting total cellular adenosine triphosphate and mitochondrial function. Phosphate supplementation prevented ethanol-associated cellular injury. CONCLUSIONS: Phosphate status plays a critical role in predisposition to and protection from alcohol-induced acinar cell dysfunction and the development of acute alcohol-induced pancreatitis. This finding may explain why pancreatitis develops in only some individuals with heavy alcohol use and suggests a potential novel therapeutic approach to pancreatitis. Finally, an LPD plus ethanol provides a new model for studying alcohol-associated pancreatic injury.


Assuntos
Metabolismo Energético , Hipofosfatemia/complicações , Mitocôndrias/metabolismo , Pâncreas/metabolismo , Pancreatite Alcoólica/metabolismo , Fosfatos/deficiência , Trifosfato de Adenosina/metabolismo , Animais , Modelos Animais de Doenças , Etanol , Hipofosfatemia/metabolismo , Hipofosfatemia/prevenção & controle , Masculino , Camundongos Endogâmicos C57BL , Mitocôndrias/patologia , Pâncreas/patologia , Pancreatite Alcoólica/induzido quimicamente , Pancreatite Alcoólica/patologia , Pancreatite Alcoólica/prevenção & controle , Fosfatos/administração & dosagem , Índice de Gravidade de Doença , Técnicas de Cultura de Tecidos
7.
Alcohol Res ; 41(1): 05, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33868869

RESUMO

Chronic, heavy alcohol consumption disrupts normal organ function and causes structural damage in virtually every tissue of the body. Current diagnostic terminology states that a person who drinks alcohol excessively has alcohol use disorder. The liver is especially susceptible to alcohol-induced damage. This review summarizes and describes the effects of chronic alcohol use not only on the liver, but also on other selected organs and systems affected by continual heavy drinking-including the gastrointestinal tract, pancreas, heart, and bone. Most significantly, the recovery process after cessation of alcohol consumption (abstinence) is explored. Depending on the organ and whether there is relapse, functional recovery is possible. Even after years of heavy alcohol use, the liver has a remarkable regenerative capacity and, following alcohol removal, can recover a significant portion of its original mass and function. Other organs show recovery after abstinence as well. Data on studies of both heavy alcohol use among humans and animal models of chronic ethanol feeding are discussed. This review describes how (or whether) each organ/tissue metabolizes ethanol, as metabolism influences the organ's degree of injury. Damage sustained by the organ/tissue is reviewed, and evidence for recovery during abstinence is presented.


Assuntos
Alcoolismo/metabolismo , Etanol/metabolismo , Hepatopatias Alcoólicas/metabolismo , Fígado/metabolismo , Abstinência de Álcool , Consumo de Bebidas Alcoólicas/metabolismo , Animais , Osso e Ossos/metabolismo , Trato Gastrointestinal/metabolismo , Coração/efeitos dos fármacos , Humanos , Camundongos , Pancreatite Alcoólica/metabolismo , Ratos
8.
Genes (Basel) ; 12(4)2021 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-33806082

RESUMO

BACKGROUND: Excessive alcohol consumption has long been known to be the primary cause of chronic pancreatitis (CP) but genetic risk factors have been increasingly identified over the past 25 years. The scale and scope of gene-alcohol interactions in CP nevertheless remain unclear. METHODS: All studies that had obtained genetic variant data concurrently on alcoholic CP (ACP) patients, non-ACP (NACP) patients and normal controls were collated. Employing normal controls as a common baseline, paired ORACP and ORNACP (odds ratios associated with ACP and NACP, respectively) values were calculated and used to assess gene-alcohol interactions. RESULTS: Thirteen variants involving PRSS1, SPINK1, CTRC, CLDN2, CPA1, CEL and CTRB1-CTRB2, and varying from very rare to common, were collated. Seven variants had an ORACP > ORNACP, which was regarded as an immediate indicator of gene-alcohol interactions in CP. Variants with an ORACP < ORNACP were also found to interact with alcohol consumption by virtue of their impact on age at first pancreatitis symptoms in ACP. CONCLUSIONS: This study revealed evidence for extensive gene-alcohol interactions in CP. Our findings lend support to the hypothesis that alcohol affects the expression of genetically determined CP and highlight a predominant role of weak-effect variants in the development of ACP.


Assuntos
Consumo de Bebidas Alcoólicas/efeitos adversos , Marcadores Genéticos , Predisposição Genética para Doença , Mutação , Pancreatite Alcoólica/patologia , Humanos , Pancreatite Alcoólica/etiologia , Pancreatite Alcoólica/metabolismo
9.
Alcohol Clin Exp Res ; 44(4): 777-789, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32056245

RESUMO

Alcohol is a major cause of acute and chronic pancreatitis. There have been some recent advances in the understanding of the mechanisms underlying alcoholic pancreatitis, which include perturbation in mitochondrial function and autophagy and ectopic exocytosis, with some of these cellular events involving membrane fusion soluble N-ethylmaleimide-sensitive factor receptor protein receptor proteins. Although new insights have been unraveled recently, the precise mechanisms remain complex, and their finer details have yet to be established. The overall pathophysiology of pancreatitis involves not only the pancreatic acinar cells but also the stellate cells and duct cells. Why only some are more susceptible to pancreatitis and with increased severity, while others are not, would suggest that there may be undefined protective factors or mechanisms that enhance recovery and regeneration after injury. Furthermore, there are confounding influences of lifestyle factors such as smoking and diet, and genetic background. Whereas alcohol and smoking cessation and a generally healthy lifestyle are intuitively the advice given to these patients afflicted with alcoholic pancreatitis in order to reduce disease recurrence and progression, there is as yet no specific treatment. A more complete understanding of the pathogenesis of pancreatitis from which novel therapeutic targets could be identified will have a great impact, particularly with the stubbornly high fatality (>30%) of severe pancreatitis. This review focuses on the susceptibility factors and underlying cellular mechanisms of alcohol injury on the exocrine pancreas.


Assuntos
Pancreatite Alcoólica/epidemiologia , Acetaldeído/metabolismo , Autofagia , Cálcio/metabolismo , Suscetibilidade a Doenças , Estresse do Retículo Endoplasmático , Etanol/metabolismo , Exocitose , Predisposição Genética para Doença , Humanos , Hiperlipidemias/epidemiologia , Infecções/epidemiologia , NAD/metabolismo , Obesidade/epidemiologia , Pancreatite Alcoólica/metabolismo , Fatores de Proteção , Espécies Reativas de Oxigênio/metabolismo , Fatores de Risco , Proteínas SNARE/metabolismo , Índice de Gravidade de Doença , Fumar/epidemiologia
10.
PLoS One ; 14(10): e0222927, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31661534

RESUMO

INTRODUCTION: Chronic pancreatitis (CP) may be caused by oxidative stress. An important source of reactive oxygen species (ROS) is the methylglyoxal-derived formation of advanced glycation endproducts (AGE). Methylglyoxal is detoxified by Glyoxalase I (GLO1). A reduction in GLO1 activity results in increased ROS. Single nucleotide polymorphisms (SNPs) of GLO1 have been linked to various inflammatory diseases. Here, we analyzed whether common GLO1 variants are associated with alcoholic (ACP) and non-alcoholic CP (NACP). METHODS: Using melting curve analysis, we genotyped a screening cohort of 223 ACP, 218 NACP patients, and 328 controls for 11 tagging SNPs defined by the SNPinfo LD TAG SNP Selection tool and the functionally relevant variant rs4746. For selected variants the cohorts were extended to up to 1,441 patient samples. RESULTS: In the ACP cohort, comparison of genotypes for rs1937780 between patients and controls displayed an ambiguous result in the screening cohort (p = 0.08). However, in the extended cohort of 1,441 patients no statistically significant association was found for the comparison of genotypes (p = 0.11), nor in logistic regression analysis (p = 0.214, OR 1.072, 95% CI 0.961-1.196). In the NACP screening cohort SNPs rs937662, rs1699012, and rs4746 displayed an ambiguous result when patients were compared to controls in the recessive or dominant model (p = 0.08, 0.08, and 0.07, respectively). Again, these associations were not confirmed in the extended cohorts (rs937662, dominant model: p = 0.07, logistic regression: p = 0.07, OR 1.207, 95% CI 0.985-1.480) or in the replication cohorts for rs4746 (Germany, p = 0.42, OR 1.080, 95% CI 0.673-1.124; France, p = 0.19, OR 0.90, 95% CI 0.76-1.06; China, p = 0.24, OR 1.18, 95% CI 0.90-1.54) and rs1699012 (Germany, Munich; p = 0.279, OR 0.903, 95% CI 0.750-1.087). CONCLUSIONS: Common GLO1 variants do not increase chronic pancreatitis risk.


Assuntos
Predisposição Genética para Doença , Lactoilglutationa Liase/genética , Pancreatite Alcoólica/genética , Pancreatite Crônica/genética , Feminino , Estudos de Associação Genética , Genótipo , Produtos Finais de Glicação Avançada/genética , Humanos , Masculino , Pessoa de Meia-Idade , Estresse Oxidativo/genética , Pancreatite Alcoólica/metabolismo , Pancreatite Alcoólica/patologia , Pancreatite Crônica/metabolismo , Pancreatite Crônica/patologia , Polimorfismo de Nucleotídeo Único/genética , Aldeído Pirúvico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Risco
11.
Dig Dis Sci ; 64(12): 3610-3615, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31286346

RESUMO

BACKGROUND: Modified Marshall Score is one of the severity scores for acute pancreatitis (AP) and is included in the Revised Atlanta Classification, but given its utilization of a set serum creatinine level (sCr), it may misclassify stable patients with chronic kidney disease (CKD) to a more severe class just due to their elevated sCr. AIMS: Our study aims to evaluate the role of CKD in AP and the possibility of utilizing acute kidney injury (AKI) into developing a new scoring system. METHODS: We retrospectively reviewed the electronic medical records of three hundred consecutive patients who were diagnosed with AP during hospitalization. Multiple demographic variables and clinical course indices were collected. Univariate logistic regression was then applied to predict mortality and ICU admission. Finally, receiver operating curve was utilized to compare original versus New Revised Marshall Score. RESULTS: Two hundred and eight-four (284) patients had a definitive diagnosis of AP. When comparing patients who had AKI on admission to those without AKI, the AKI group showed statistically significant higher mortality rate (5.6% vs. 1.1%, p = 0.04). Finally, we substituted the renal part of Marshall Score with our AKIN and we plotted the New "Revised" Marshall Score, which showed a higher AUROC compared to the original modified version (C-statistics 0.93 vs. 0.89, p < 0.05). CONCLUSION: We found that AKI predicts mortality and outperforms the use of a fixed sCr value alone. The use of our New Revised Marshall Score can accurately classify AP severity, avoiding misclassification of AP severity and providing better patient care.


Assuntos
Injúria Renal Aguda/epidemiologia , Pancreatite/mortalidade , Insuficiência Renal Crônica/epidemiologia , Injúria Renal Aguda/metabolismo , Adulto , Creatinina/metabolismo , Feminino , Cálculos Biliares/complicações , Humanos , Hipertrigliceridemia/complicações , Unidades de Terapia Intensiva/estatística & dados numéricos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Mortalidade , Pancreatite/etiologia , Pancreatite/metabolismo , Pancreatite Alcoólica/metabolismo , Pancreatite Alcoólica/mortalidade , Prognóstico , Curva ROC , Insuficiência Renal Crônica/metabolismo , Estudos Retrospectivos , Medição de Risco , Índice de Gravidade de Doença
12.
Eur J Trauma Emerg Surg ; 45(3): 539-543, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29525967

RESUMO

INTRODUCTION: Acute pancreatitis (AP) is a severe disease associated with significant morbidity and mortality. The overall outcome has improved, but specific treatment(s) remains elusive. The challenge is the early identification and treatment of patients who will develop severe acute pancreatitis. Therefore, the aim of the present study is to investigate plasma levels of tumor necrosis factor-like weak inducer of apoptosis (TWEAK) in the initial phase of predicted severe acute pancreatitis. METHODS: Between June 2014 and January 2016, 64 patients with acute pancreatitis and 36 healthy individuals were included to study. Four blood samples, for serum TWEAK measurement, were taken from each individual in each group. The first measurement was taken from the admission blood sample. The subsequent three samples were taken at 12, 24, and 48 h after the hospital admission. RESULTS: Serum TWEAK levels were significantly higher in patients with acute pancreatitis when compared with healthy controls. TWEAK plasma concentrations in severe pancreatitis patients were significantly higher than in mild pancreatitis patients. CONCLUSION: Serum TWEAK levels increase progressively with the severity of acute pancreatitis and TWEAK might be a novel early marker of severity in acute pancreatitis.


Assuntos
Citocina TWEAK/sangue , Pancreatite/sangue , Adulto , Idoso , Proteína C-Reativa/metabolismo , Estudos de Casos e Controles , Diagnóstico Precoce , Intervenção Médica Precoce , Feminino , Cálculos Biliares/complicações , Humanos , Lipase/sangue , Masculino , Pessoa de Meia-Idade , Pancreatite/etiologia , Pancreatite/metabolismo , Pancreatite Alcoólica/sangue , Pancreatite Alcoólica/metabolismo , Prognóstico , Índice de Gravidade de Doença
13.
Pancreas ; 47(1): 18-24, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29200128

RESUMO

OBJECTIVES: Mitochondrial permeability transition pore inhibition is a promising approach to treat acute pancreatitis (AP). We sought to determine (i) the effects of the mitochondrial permeability transition pore inhibitor 3,5-seco-4-nor-cholestan-5-one oxime-3-ol (TRO40303) on murine and human pancreatic acinar cell (PAC) injury induced by fatty acid ethyl esters (FAEEs) or taurolithocholic acid-3-sulfate and (ii) TRO40303 pharmacokinetics and efficacy in experimental alcoholic AP (FAEE-AP). METHODS: Changes in mitochondrial membrane potential (Δψm), cytosolic Ca ([Ca]c), and cell fate were examined in freshly isolated murine or human PACs by confocal microscopy. TRO40303 pharmacokinetics were assessed in cerulein-induced AP and therapeutic efficacy in FAEE-AP induced with palmitoleic acid and ethanol. Severity of AP was assessed by standard biomarkers and blinded histopathology. RESULTS: TRO40303 prevented loss of Δψm and necrosis induced by 100 µM palmitoleic acid ethyl ester or 500 µM taurolithocholic acid-3-sulfate in murine and human PACs. Pharmacokinetic analysis found TRO40303 accumulated in the pancreas. A single dose of 3 mg/kg TRO40303 significantly reduced serum amylase (P = 0.043), pancreatic trypsin (P = 0.018), and histopathology scores (P = 0.0058) in FAEE-AP. CONCLUSIONS: TRO40303 protects mitochondria and prevents necrotic cell death pathway activation in murine and human PACs, ameliorates the severity of FAEE-AP, and is a candidate drug for human AP.


Assuntos
Ésteres/farmacologia , Ácidos Graxos/farmacologia , Mitocôndrias/efeitos dos fármacos , Oximas/farmacologia , Pancreatite Alcoólica/prevenção & controle , Secoesteroides/farmacologia , Células Acinares/efeitos dos fármacos , Células Acinares/metabolismo , Doença Aguda , Animais , Ceruletídeo , Ésteres/metabolismo , Ácidos Graxos/metabolismo , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Necrose/prevenção & controle , Oximas/farmacocinética , Pancreatite/induzido quimicamente , Pancreatite/prevenção & controle , Pancreatite Alcoólica/metabolismo , Pancreatite Alcoólica/patologia , Secoesteroides/farmacocinética , Ácido Taurolitocólico/análogos & derivados , Ácido Taurolitocólico/farmacologia
14.
Alcohol Clin Exp Res ; 42(2): 306-314, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29121396

RESUMO

BACKGROUND: Pancreatic fibrosis is a key pathological feature of alcoholic chronic pancreatitis (ACP). Bacterial endotoxin lipopolysaccharide (LPS) is considered as an important cofactor in the fibrogenesis of ACP. However, there are limitations in the use of exogenous LPS for evaluating the role of endotoxin in ACP pathogenesis. In this study, we determined the relationship between the concentration of LPS in the portal vein and pancreatic type I collagen (Col1) content in chronic alcohol-fed rats. METHODS: Male Sprague Dawley rats were divided into 2 groups and fed with Lieber-DeCarli isocaloric control (CON) liquid diet or ethanol (EtOH) (15 g/kg/d) liquid diet. Eleven CON or EtOH rats were euthanized at the end of week 8, 9, or 10. The plasma LPS from portal vein was determined. Pancreatic inflammatory injury and fibrosis were assessed. Pancreatic stellate cells (PSCs) and macrophages were identified; pancreatic type I collagen alpha 1 (Col1A1) and Toll-like receptor (TLR4) mRNA and protein were examined; pancreatic chemokines and transforming growth factor-beta1 (TGF-ß1) were determined. RESULTS: Pancreatic inflammatory scores were increased in 10-week EtOH rats compared with CON rats, but there was no significant difference in collagen deposition between 2 groups. The levels of portal vein LPS and pancreatic TLR4 and Col1A1 mRNA and protein were increased in a time-dependent fashion in EtOH rats, with the highest levels occurring at 10 weeks. Additionally, by 8 weeks, pancreatic TLR4 and Col1A1 mRNA in EtOH rats were statistically increased as compared to CON rats, whereas portal vein LPS remained unchanged. The number of PSCs and macrophages and expression of chemokines (MCP-1, MIP-1α, and RANTES), TGF-ß1, or Col1A1 were significantly increased, each of which was positively correlated with the level of portal vein LPS in 10-week EtOH rats. CONCLUSIONS: These results suggest that LPS is associated with alcohol-induced fibrosis in pancreatitis and targeting of bacterial endotoxin may be a promising therapeutic strategy for ACP.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Colágeno Tipo I/efeitos dos fármacos , Etanol/farmacologia , Lipopolissacarídeos/farmacologia , Pâncreas/efeitos dos fármacos , Pancreatite Alcoólica/metabolismo , Animais , Quimiocinas/efeitos dos fármacos , Quimiocinas/metabolismo , Colágeno Tipo I/biossíntese , Cadeia alfa 1 do Colágeno Tipo I , Fibrose , Macrófagos/efeitos dos fármacos , Masculino , Pâncreas/metabolismo , Pâncreas/patologia , Células Estreladas do Pâncreas/efeitos dos fármacos , Pancreatite Alcoólica/patologia , Veia Porta , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor 4 Toll-Like/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Fator de Crescimento Transformador beta1/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo
15.
South Med J ; 110(11): 727-732, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29100225

RESUMO

Acute pancreatitis is a fascinating disease. In the United States, the two most common etiologies of acute pancreatitis are gallstones and excessive alcohol consumption. The diagnosis of acute pancreatitis is made with a combination of history, physical examination, computed tomography scan, and laboratory evaluation. Differentiating patients who will have a benign course of their pancreatitis from patients who will have severe pancreatitis is challenging to the clinician. C-reactive protein, pro-calcitonin, and the Bedside Index for Severity of Acute Pancreatitis appeared to be the best tools for the early and accurate diagnosis of severe pancreatitis. Early laparoscopic cholecystectomy is indicated for patients with mild gallstone pancreatitis. For patients who are going to have a prolonged hospitalization, enteral nutrition is preferred. Total parenteral nutrition should be reserved for patients who cannot tolerate enteral nutrition. Prophylactic antibiotics are not indicated for patients with pancreatic necrosis. Surgical intervention for infected pancreatic necrosis should be delayed as long as possible to improve patient outcomes.


Assuntos
Colecistectomia Laparoscópica , Colelitíase/cirurgia , Nutrição Enteral , Hidratação/métodos , Pseudocisto Pancreático/cirurgia , Pancreatite/terapia , Antibacterianos/uso terapêutico , Proteína C-Reativa/metabolismo , Calcitonina/metabolismo , Colelitíase/complicações , Drenagem , Hospitalização , Humanos , Tempo de Internação , Pseudocisto Pancreático/etiologia , Pancreatite/diagnóstico , Pancreatite/etiologia , Pancreatite/metabolismo , Pancreatite Alcoólica/diagnóstico , Pancreatite Alcoólica/metabolismo , Pancreatite Alcoólica/terapia , Nutrição Parenteral Total , Índice de Gravidade de Doença , Tomografia Computadorizada por Raios X
16.
Alcohol Res ; 38(2): 173-182, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28988572

RESUMO

Findings from epidemiologic studies and research with experimental animal models provide insights into alcohol-related disease pathogeneses. Epidemiologic data indicate that heavy drinking and smoking are associated with high rates of pancreatic disease. Less clear is the association between lower levels of drinking and pancreatitis. Intriguingly, a very low percentage of drinkers develop clinical pancreatitis. Experimental models demonstrate that alcohol administration alone does not initiate pancreatitis but does sensitize the pancreas to disease. Understanding the effects of alcohol use on the pancreas may prove beneficial in the prevention of both pancreatitis and pancreatic cancer.


Assuntos
Modelos Animais de Doenças , Pancreatite Alcoólica , Animais , Humanos , Pancreatite Alcoólica/epidemiologia , Pancreatite Alcoólica/etiologia , Pancreatite Alcoólica/metabolismo , Pancreatite Alcoólica/patologia
17.
Gastroenterology ; 153(6): 1674-1686, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28847752

RESUMO

BACKGROUND & AIMS: Smoking, an independent risk factor for pancreatitis, accelerates the development of alcoholic pancreatitis. Alcohol feeding of mice induces up-regulation of spliced X-box binding protein 1 (XBP1s), which regulates the endoplasmic reticulum (ER) unfolded protein response and promotes cell survival upon ER stress. We examined whether smoking affects the adaptive mechanisms induced by alcohol and accelerates disorders of the ER in pancreatic acinar cells. METHODS: We studied the combined effects of ethanol (EtOH) and cigarette smoke extract (CSE) on ER stress and cell death responses in mouse and human primary acini and the acinar cell line AR42J. Cells were incubated with EtOH (50 mmol/L), CSE (20-40 µg/mL), or both (CSE+EtOH), and analyzed by immunoblotting, quantitative reverse-transcription polymerase chain reaction, and cell death assays. Some cells were incubated with MKC-3946, an inhibitor of endoplasmic reticulum to nucleus signaling 1 (ERN1, also called IRE1) that blocks XBP1s formation. Male Sprague-Dawley rats were fed isocaloric amounts of an EtOH-containing (Lieber-DeCarli) or control diet for 11 weeks and exposed to cigarette smoke or room air in an exposure chamber for 2 hours each day. During the last 3 weeks, a subset of rats received intravenous injections of lipopolysaccharide (LPS, 3 mg/kg per week) to induce pancreatitis or saline (control). Pancreatic tissues were collected and analyzed by histology and immunostaining techniques. RESULTS: In AR42J and primary acini, CSE+EtOH induced cell death (necrosis and apoptosis), but neither agent alone had this effect. Cell death was associated with a significant decrease in expression of XBP1s. CSE+EtOH, but neither agent alone, slightly decreased adenosine triphosphate levels in AR42J cells, but induced oxidative stress and sustained activation (phosphorylation) of eukaryotic translation initiation factor 2 alpha kinase 3 (EIF2AK3, also called PERK) and increased protein levels of DNA damage inducible transcript 3 (DDIT3, also called CHOP). CHOP regulates transcription to promote apoptosis. Incubation of AR42J or primary mouse or human acinar cells with MKC-3946 reduced expression of XBP1s, increased levels of CHOP, and induced cell death. In rats fed an EtOH diet, exposure to cigarette smoke increased ER stress in acinar cells and sensitized the pancreas to LPS-induced pathology. CONCLUSIONS: Cigarette smoke promotes cell death and features of pancreatitis in EtOH-sensitized acinar cells by suppressing the adaptive unfolded protein response signaling pathway. It also activates ER stress pathways that promote acinar cell death.


Assuntos
Células Acinares/efeitos dos fármacos , Consumo de Bebidas Alcoólicas/efeitos adversos , Fumar Cigarros/efeitos adversos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Etanol/toxicidade , Pâncreas Exócrino/efeitos dos fármacos , Pancreatite Alcoólica/etiologia , Fumaça/efeitos adversos , Células Acinares/metabolismo , Células Acinares/patologia , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Humanos , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Necrose , Estresse Oxidativo/efeitos dos fármacos , Pâncreas Exócrino/metabolismo , Pâncreas Exócrino/patologia , Pancreatite Alcoólica/metabolismo , Pancreatite Alcoólica/patologia , Ratos Sprague-Dawley , Fatores de Risco , Fatores de Tempo , Técnicas de Cultura de Tecidos , Resposta a Proteínas não Dobradas/efeitos dos fármacos
18.
Pancreas ; 46(6): 806-812, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28609370

RESUMO

OBJECTIVES: The aim of this study was to identify differentially expressed proteins in the pancreatic tissue of hepatic alcohol dehydrogenase-deficient deer mice fed ethanol to understand metabolic basis and mechanism of alcoholic chronic pancreatitis. METHODS: Mice were fed liquid diet containing 3.5 g% ethanol daily for 3 months, and differentially expressed pancreatic proteins were identified by protein separation using 2-dimensional gel electrophoresis and identification by mass spectrometry. RESULTS: Nineteen differentially expressed proteins were identified by applying criteria established for protein identification in proteomics. An increased abundance was found for ribosome-binding protein 1, 60S ribosomal protein L31-like isoform 1, histone 4, calcium, and adenosine triphosphate (ATP) binding proteins and the proteins involved in antiapoptotic processes and endoplasmic reticulum function, stress, and/or homeostasis. Low abundance was found for endoA cytokeratin, 40S ribosomal protein SA, amylase 2b isoform precursor, serum albumin, and ATP synthase subunit ß and the proteins involved in cell motility, structure, and conformation. CONCLUSIONS: Chronic ethanol feeding in alcohol dehydrogenase-deficient deer mice differentially expresses pancreatic functional and structural proteins, which can be used to develop biomarker(s) of alcoholic chronic pancreatitis, particularly amylase 2b precursor, and 60 kDa heat shock protein and those involved in ATP synthesis and blood osmotic pressure.


Assuntos
Álcool Desidrogenase/deficiência , Consumo de Bebidas Alcoólicas , Etanol , Fígado/enzimologia , Pâncreas/metabolismo , Pancreatite Alcoólica/metabolismo , Proteínas/metabolismo , Álcool Desidrogenase/genética , Animais , Modelos Animais de Doenças , Genótipo , Masculino , Camundongos Knockout , Pancreatite Alcoólica/genética , Peromyscus , Fenótipo , Proteômica/métodos , Fatores de Tempo
19.
Eur J Intern Med ; 32: 79-83, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27157403

RESUMO

AIM: The aim of our study was to investigate the influence of metabolic syndrome on the course of acute pancreatitis determined by disease severity, the presence of local and systemic complications and survival rate. PATIENTS AND METHODS: 609 patients admitted to our hospital in the period from January 1, 2008 up to June 31, 2015 with the diagnosis of acute pancreatitis were analyzed. The diagnosis and the severity of acute pancreatitis were made according to the revised Atlanta classification criteria from 2012. RESULTS: Of 609 patients with acute pancreatitis, 110 fulfilled the criteria for metabolic syndrome. Patients with metabolic syndrome had statistically significantly higher incidence of moderately severe (38.2% vs. 28.5%; p=0.05) and severe (22.7% vs. 12.8%; p=0.01) acute pancreatitis in comparison to those without metabolic syndrome, while patients without metabolic syndrome had higher incidence of mild acute pancreatitis in comparison to those patients with metabolic syndrome (58.7% vs. 39.1%; p<0.001). Patients with metabolic syndrome had a higher number of local and systemic complications, and higher APACHE II score in comparison to patients without metabolic syndrome. In multivariable logistic regression analysis, the presence of metabolic syndrome was independently associated with moderately severe and severe acute pancreatitis. Comparing survival rates, patients suffering from metabolic syndrome had a higher death rate compared to patients without metabolic syndrome (16% vs. 4.5%; p<0.001). CONCLUSION: The presence of metabolic syndrome at admission portends a higher risk of moderately severe and severe acute pancreatitis, as well as higher mortality rate.


Assuntos
Síndrome Metabólica/epidemiologia , Pancreatite/epidemiologia , APACHE , Doença Aguda , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Proteína C-Reativa/metabolismo , Feminino , Cálculos Biliares/complicações , Humanos , Hipertrigliceridemia/complicações , Incidência , Modelos Logísticos , Masculino , Síndrome Metabólica/metabolismo , Pessoa de Meia-Idade , Análise Multivariada , Pancreatite/etiologia , Pancreatite/metabolismo , Pancreatite Alcoólica/epidemiologia , Pancreatite Alcoólica/metabolismo , Estudos Retrospectivos , Índice de Gravidade de Doença
20.
Biomolecules ; 5(4): 3309-38, 2015 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-26610589

RESUMO

Alcohol consumption and its abuse is a major health problem resulting in significant healthcare cost in the United States. Chronic alcoholism results in damage to most of the vital organs in the human body. Among the alcohol-induced injuries, alcoholic liver disease is one of the most prevalent in the United States. Remarkably, ethanol alters expression of a wide variety of microRNAs that can regulate alcohol-induced complications or dysfunctions. In this review, we will discuss the role of microRNAs in alcoholic pancreatitis, alcohol-induced liver damage, intestinal epithelial barrier dysfunction, and brain damage including altered hippocampus structure and function, and neuronal loss, alcoholic cardiomyopathy, and muscle damage. Further, we have reviewed the role of altered microRNAs in the circulation, teratogenic effects of alcohol, and during maternal or paternal alcohol consumption.


Assuntos
Transtornos do Sistema Nervoso Induzidos por Álcool/genética , Cardiomiopatia Alcoólica/genética , Hepatopatias Alcoólicas/genética , MicroRNAs/genética , Pancreatite Alcoólica/genética , Transtornos do Sistema Nervoso Induzidos por Álcool/metabolismo , Animais , Cardiomiopatia Alcoólica/metabolismo , Humanos , Hepatopatias Alcoólicas/metabolismo , Pancreatite Alcoólica/metabolismo , RNA Longo não Codificante/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA