RESUMEN
Regardless of its aetiology, sustained intracellular Ca2+ overload is a well-known hallmark of acute pancreatitis (AP). Toxic Ca2+ elevation induces pancreatic ductal cell damage characterized by impaired ion and fluid secretion - essential to wash out the protein-rich fluid secreted by acinar cells while maintaining the alkaline intra-ductal pH under physiological conditions - and mitochondrial dysfunction. While prevention of ductal cell injury decreases the severity of AP, no specific drug target has yet been identified in the ductal cells. Although Orai1, a store-operated Ca2+ influx channel, is known to contribute to sustained Ca2+ overload in acinar cells, details concerning its expression and function in ductal cells are currently lacking. In this study, we demonstrate that functionally active Orai1 channels reside predominantly in the apical plasma membrane of pancreatic ductal cells. Selective CM5480-mediated Orai1 inhibition impairs Stim1-dependent extracellular Ca2+ influx evoked by bile acids or ethanol combined with non-oxidative ethanol metabolites. Furthermore, prevention of sustained extracellular Ca2+ influx protects ductal cell secretory function in vitro and decreases pancreatic ductal cell death. Finally, Orai1 inhibition partially restores and maintains proper exocrine pancreatic secretion in in vivo AP models. In conclusion, our results indicate that Orai1 inhibition prevents AP-related ductal cell function impairment and holds the potential of improving disease outcome. KEY POINTS: Sustained intracellular Ca2+ overload in pancreatic acinar and ductal cells is a hallmark of biliary and alcohol-induced acute pancreatitis, which leads to impaired ductal ion and fluid secretion. Orai1 is a plasma membrane Ca2+ channel that mediates extracellular Ca2+ influx upon endoplasmic reticulum Ca2+ depletion. Results showed that Orai1 is expressed on the luminal plasma membrane of the ductal cells and selective Orai1 inhibition impaired Stim1-dependent extracellular Ca2+ influx evoked by bile acids or ethanol combined with non-oxidative ethanol metabolites. The prevention of sustained extracellular Ca2+ influx protected ductal cell secretory functions in in vitro models and maintained exocrine pancreatic secretion in in vivo acute pancreatitis models. Orai1 inhibition prevents the bile acid- and alcohol-induced damage of the pancreatic ductal secretion and holds the potential of improving the outcome of acute pancreatitis.
Asunto(s)
Pancreatitis , Enfermedad Aguda , Ácidos y Sales Biliares/toxicidad , Calcio/metabolismo , Señalización del Calcio , Etanol/toxicidad , Humanos , Proteína ORAI1/antagonistas & inhibidores , Pancreatitis/tratamiento farmacológico , Pancreatitis/etiología , Pancreatitis/metabolismo , Molécula de Interacción Estromal 1/metabolismoRESUMEN
BACKGROUND & AIMS: Metaplastic glands buried under squamous epithelium are frequently detected in patients with Barrett esophagus (BE). This subsquamous intestinal metaplasia might be responsible for cancers that develop despite endoscopic surveillance and for metaplasia recurrences after endoscopic ablation. To determine whether reflux induces BE cells to undergo an epithelial-to-mesenchymal transition (EMT) that produces subsquamous intestinal metaplasia, we assessed EMT in BE cells exposed to acidic bile salts and in rat and human esophageal tissues. METHODS: We compared markers of EMT and cell motility in trans-well and 3-dimensional organotypic culture systems among dysplastic BE epithelial cell lines, nondysplastic telomerase-immortalized BE cell lines (BAR-T), and BAR-T cells exposed acutely or for 20 weeks to acidic bile salts. Vascular endothelial growth factor (VEGF) A was inhibited with a neutralizing antibody or CRISPR-Cas9n and VEGF receptor 2 was inhibited with SU1498 or shRNA, and cells were analyzed by immunohistochemistry, quantitative polymerase chain reaction, or immunoblotting for markers of VEGF signaling and EMT; cell motility was assessed by trans-well assay. We used immunohistochemistry and quantitative polymerase chain reaction to assess EMT markers in the columnar-lined esophagus of rats with surgically induced reflux esophagitis and in esophagectomy specimens from patients with BE. RESULTS: We detected features of EMT (decreased cadherin 1 [CDH1]; increased fibronectin 1, vimentin, and matrix metalloproteinase 2; and increased motility) in dysplastic BE epithelial cell lines and in BAR-T cells exposed for 20 weeks, but not in unexposed BAR-T cells. Acute acidic bile salt exposure induced expression of zinc finger E-box binding homeobox 1 and 2 (ZEB1/2) in BAR-T cells, which decreased their expression of CDH1 and increased motility; inhibitors of VEGF signaling blocked these effects. Columnar-lined esophagus of rats with reflux esophagitis had increased expression of ZEB1/2 and decreased expression of CDH1 compared with controls. Dysplastic BE tissues also had significantly increased levels of ZEB1 and significantly decreased levels of CDH1 compared with nondysplastic BE tissues. CONCLUSIONS: In BE cell lines, acidic bile salts induce EMT by VEGF signaling, which increases expression of ZEB1/2, repressors of CDH1. These observations suggest that reflux induces EMT in metaplastic BE tissues, which promotes development of subsquamous intestinal metaplasia.
Asunto(s)
Esófago de Barrett/metabolismo , Ácidos y Sales Biliares/toxicidad , Transformación Celular Neoplásica/inducido químicamente , Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias Esofágicas/metabolismo , Esófago/efectos de los fármacos , Reflujo Gastroesofágico/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Esófago de Barrett/genética , Esófago de Barrett/patología , Línea Celular , Movimiento Celular/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Esófago/metabolismo , Esófago/patología , Reflujo Gastroesofágico/genética , Reflujo Gastroesofágico/patología , Humanos , Ratas , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismoRESUMEN
Increased colonic bile acid (BA) exposure, frequent in diarrhea-predominant irritable bowel syndrome (IBS-D), can affect gut function. Nerve growth factor (NGF) is implicated in the development of visceral hypersensitivity (VH). In this study, we tested the hypothesis that BAs cause VH via mucosal mast cell (MMC)-to-nociceptor signaling, which involves the farnesoid X receptor (FXR)/NGF/transient receptor potential vanilloid (TRPV)1 axis. BAs were intracolonically administered to rats for 15 d. Visceral sensitivity to colorectal distention and colonic NGF expression were examined. BAs caused VH, an effect that involved MMC-derived NGF and was accompanied by enhanced TRPV1 expression in the dorsal root ganglia. Anti-NGF treatment and TRPV1 antagonism inhibited BA-induced VH. BAs induced NGF mRNA and protein expression and release in cultured mast cells. Colonic supernatants from patients with IBS-D with elevated colonic BA content transcriptionally induced NGF expression. In FXR-/- mice, visceral sensitivity and colonic NGF expression were unaltered after BA treatment. Pharmacological antagonism and FXR silencing suppressed BA-induced NGF expression and release in mast cells. Mitogen-activated protein kinase kinase (MKK) 3/6/p38 MAPK/NF-κB signaling was mechanistically responsible for FXR-mediated NGF expression and secretion. The findings show an MMC-dependent and FXR-mediated pronociceptive effect of BAs and identify the BA/FXR/NGF/TRPV1 axis as a key player in MMC-to-neuron communication during pain processing in IBS.-Li, W.-T., Luo, Q.-Q., Wang, B., Chen, X., Yan, X.-J., Qiu, H.-Y., Chen, S.-L. Bile acids induce visceral hypersensitivity via mucosal mast cell-to-nociceptor signaling that involves the farnesoid X receptor/nerve growth factor/transient receptor potential vanilloid 1 axis.
Asunto(s)
Ácidos y Sales Biliares/toxicidad , Hipersensibilidad/patología , Síndrome del Colon Irritable/patología , Mastocitos/inmunología , Factor de Crecimiento Nervioso/metabolismo , Nociceptores/inmunología , Receptores Citoplasmáticos y Nucleares/metabolismo , Canales Catiónicos TRPV/metabolismo , Adulto , Animales , Estudios de Casos y Controles , Células Cultivadas , Femenino , Fármacos Gastrointestinales/toxicidad , Humanos , Hipersensibilidad/etiología , Hipersensibilidad/metabolismo , Síndrome del Colon Irritable/inducido químicamente , Síndrome del Colon Irritable/metabolismo , Masculino , Mastocitos/metabolismo , Mastocitos/patología , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Nociceptores/metabolismo , Nociceptores/patología , Ratas , Ratas Sprague-Dawley , Dolor Visceral/inducido químicamente , Dolor Visceral/metabolismo , Dolor Visceral/patologíaRESUMEN
BACKGROUND: Recent preclinical explorations strongly support the tumorigenic potential of bile on laryngopharyngeal mucosa. Herein, the authors describe, in bile-related human hypopharyngeal squamous cell carcinoma (HSCC), NF-κB-related messenger RNA (mRNA) and microRNA (miRNA) oncogenic phenotypes similar to those previously identified in acidic bile-exposed premalignant murine hypopharyngeal mucosa. METHODS: In this pilot study, the authors included human HSCC specimens paired with their adjacent normal tissue (ANT) derived from 3 representative patients with documented biliary laryngopharyngeal reflux (bile[+]) compared with 5 control patients without signs of bile reflux disease (bile[-]). Immunohistochemical, quantitative polymerase chain reaction, and miRNA analyses were used to detect the levels of activated NF-κB and expression levels of STAT3, EGFR, BCL2, WNT5A, IL-6, IL-1B, ΔNp63, cREL, TNF-α, TP53, NOTCH1, NOTCH2, NOTCH3, miR-21, miR-155, miR-192, miR-34a, miR-375, miR-451a, miR-489, miR-504, and miR-99a. RESULTS: Bile(+) HSCC demonstrated an intense NF-κB activation accompanied by significant overexpression of RELA(p65), EGFR, STAT3, BCL-2, cREL, ΔNp63, WNT5A, IL-6, and IL1B; upregulation of oncomir miR-21; and downregulation of tumor suppressor miR-375 compared with their respective ANTs. Bile(+) HSCC demonstrated significantly higher mRNA levels of all the analyzed genes, particularly RELA(p65), IL-6, EGFR, and TNF-α compared with bile(-) tumors. The miR-21/miR-375 ratio, which previously has been linked to tumor aggressiveness, was found to be >260-fold and >30-fold higher, respectively, in bile(+) HSCCs compared with their ANTs and bile(-) tumors. CONCLUSIONS: Although limitations apply to this pilot study due to the small number of patients with HSCC, the novel findings suggest that a history of bile as a component of esophageal reflux disease may represent an independent risk factor for hypopharyngeal carcinogenesis.
Asunto(s)
Reflujo Biliar/genética , Carcinoma de Células Escamosas/genética , Neoplasias Hipofaríngeas/genética , Proteínas de Neoplasias/genética , Anciano , Animales , Bilis/metabolismo , Ácidos y Sales Biliares/metabolismo , Ácidos y Sales Biliares/toxicidad , Reflujo Biliar/complicaciones , Reflujo Biliar/metabolismo , Reflujo Biliar/patología , Carcinoma de Células Escamosas/complicaciones , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hipofaríngeas/complicaciones , Neoplasias Hipofaríngeas/metabolismo , Neoplasias Hipofaríngeas/patología , Masculino , Ratones , MicroARNs/genética , Persona de Mediana Edad , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/patología , FN-kappa B/genética , ARN Mensajero/genéticaRESUMEN
We aimed at isolating and characterising microorganisms present in human breast milk with probiotic potential. In an 8-week postpartum sampling period, two strains of bifidobacteria (Bifidobacterium longum LM7a and Bifidobacterium dentium LM8a') and four strains of lactobacilli were isolated, all during the first 4-week postpartum. B. longum LM7a and B. dentium LM8a', together with four strains previously isolated from breast milk (Bifidobacterium lactis INL1, INL2, INL4 and INL5), were considered for further studies. Susceptibility of the strains to tetracycline, erythromycin, clindamycin, streptomycin, vancomycin and chloramphenicol was evaluated and the isolates exhibited, in general, the same properties as previously reported for bifidobacteria. All isolates showed low hydrophobicity and B. lactis and B. longum strains had satisfactory resistance to gastric digestion and bile shock, but not to pancreatin. B. lactis INL1, B. longum LM7a and B. dentium LM8a' were selected for some comparative technological studies. In particular, B. lactis INL1 displayed technological potential, with satisfactory growth in cheese whey-based media in biofermentor and resistance to freeze-drying, accelerated storage conditions and simulated gastric digestion.
Asunto(s)
Bifidobacterium/aislamiento & purificación , Medios de Cultivo/química , Lactobacillus/aislamiento & purificación , Leche Humana/microbiología , Probióticos/efectos adversos , Probióticos/aislamiento & purificación , Suero Lácteo/metabolismo , Antibacterianos/farmacología , Bifidobacterium/efectos de los fármacos , Bifidobacterium/crecimiento & desarrollo , Bifidobacterium/metabolismo , Ácidos y Sales Biliares/toxicidad , Femenino , Ácido Gástrico/metabolismo , Humanos , Lactobacillus/efectos de los fármacos , Lactobacillus/crecimiento & desarrollo , Lactobacillus/metabolismo , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Pancreatina/toxicidadRESUMEN
Biliary lipids consist mainly of bile salts, phospholipids and cholesterol, which form mixed micelles and vesicles. Bile salts play various physiological roles but have damaging effects on cell membranes due to their detergent properties. The cytotoxicity of bile salts on hepatocytes leads to liver injuries and is largely determined by the bile salt species, the concentrations of bile salts, phospholipids and cholesterol, and the lipid composition of cell membranes. In bile, monomers and simple micelles of bile salts coexist with mixed micelles and vesicles in dynamic equilibrium, and contribute to the cytotoxicity on hepatocytes. The ATP-binding cassette (ABC) transporter family members, ABCB11, ABCB4 and ABCG5/ABCG8, mediate the biliary secretion of bile salts, phospholipids and cholesterol, respectively. Mutations in ABCB4 result in severe cholestatic diseases, and the biliary phospholipids are necessary for the attenuation of bile salt cytotoxicity. On the other hand, cholesterol reverses the cytoprotective effects of phospholipids against bile salts. In addition, phosphatidylethanolamine N-methyltransferase increases the cell resistance to bile salts by changing the phospholipid composition and structures of the apical membranes. In this review, we focus on the molecular mechanisms for the protection of hepatocytes against bile salt cytotoxicity. Further understanding of these mechanisms will help to develop new therapeutic strategies for cholestatic liver diseases.
Asunto(s)
Ácidos y Sales Biliares/toxicidad , Hepatocitos/efectos de los fármacos , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Ácidos y Sales Biliares/química , Colesterol/química , Hepatocitos/citología , Hepatocitos/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Micelas , Fosfolípidos/químicaRESUMEN
The gastrointestinal (GI) tract is a highly complex organ composed of the intestinal epithelium layer, intestinal microbiota, and local immune system. Intestinal microbiota residing in the GI tract engages in a mutualistic relationship with the host. Different sections of the GI tract contain distinct proportions of the intestinal microbiota, resulting in the presence of unique bacterial products in each GI section. The intestinal microbiota converts ingested nutrients into metabolites that target either the intestinal microbiota population or host cells. Metabolites act as messengers of information between the intestinal microbiota and host cells. The intestinal microbiota composition and resulting metabolites thus impact host development, health, and pathogenesis. Many recent studies have focused on modulation of the gut microbiota and their metabolites to improve host health and prevent or treat diseases. In this review, we focus on the production of microbial metabolites, their biological impact on the intestinal microbiota composition and host cells, and the effect of microbial metabolites that contribute to improvements in inflammatory bowel diseases and metabolic diseases. Understanding the role of microbial metabolites in protection against disease might offer an intriguing approach to regulate disease.
Asunto(s)
Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino/patología , Enfermedades Metabólicas/patología , Bacterias/química , Bacterias/metabolismo , Ácidos y Sales Biliares/metabolismo , Ácidos y Sales Biliares/toxicidad , Ácidos Grasos Volátiles/metabolismo , Ácidos Grasos Volátiles/toxicidad , Flavonas/metabolismo , Flavonas/toxicidad , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/microbiología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/microbiología , Poliaminas/metabolismo , Poliaminas/toxicidadRESUMEN
Cholestatic liver injury results from impaired bile flow or metabolism and promotes hepatic inflammation and fibrogenesis. Toxic bile acids that accumulate in cholestasis induce apoptosis and contribute to early cholestatic liver injury, which is amplified by accompanying inflammation. The aim of the current study was to evaluate the role of the antiapoptotic caspase 8-homolog cellular FLICE-inhibitory (cFLIP) protein during acute cholestatic liver injury. Transgenic mice exhibiting hepatocyte-specific deletion of cFLIP (cFLIP-/-) were used for in vivo and in vitro analysis of cholestatic liver injury using bile duct ligation (BDL) and the addition of bile acids ex vivo. Loss of cFLIP in hepatocytes promoted acute cholestatic liver injury early after BDL, which was characterized by a rapid release of proinflammatory and chemotactic cytokines (TNF, IL-6, IL-1ß, CCL2, CXCL1, and CXCL2), an increased presence of CD68+ macrophages and an influx of neutrophils in the liver, and resulting apoptotic and necrotic hepatocyte cell death. Mechanistically, liver injury in cFLIP-/- mice was aggravated by reactive oxygen species, and sustained activation of the JNK signaling pathway. In parallel, cytoprotective NF-κB p65, A20, and the MAPK p38 were inhibited. Increased injury in cFLIP-/- mice was accompanied by activation of hepatic stellate cells and profibrogenic regulators. The antagonistic caspase 8-homolog cFLIP is a critical regulator of acute, cholestatic liver injury. NEW & NOTEWORTHY The current paper explores the role of a classical modulator of hepatocellular apoptosis in early, cholestatic liver injury. These include activation of NF-κB and MAPK signaling, production of inflammatory cytokines, and recruitment of neutrophils in response to cholestasis. Because these signaling pathways are currently exploited in clinical trials for the treatment of nonalcoholic steatohepatitis and cirrhosis, the current data will help in the development of novel pharmacological options in these indications.
Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/deficiencia , Coledocolitiasis/metabolismo , Conducto Colédoco/cirugía , Células Estrelladas Hepáticas/metabolismo , Hepatitis/metabolismo , Hepatocitos/metabolismo , Cirrosis Hepática/metabolismo , Hígado/metabolismo , Animales , Apoptosis , Ácidos y Sales Biliares/metabolismo , Ácidos y Sales Biliares/toxicidad , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Células Cultivadas , Coledocolitiasis/etiología , Coledocolitiasis/genética , Coledocolitiasis/patología , Citocinas/metabolismo , Predisposición Genética a la Enfermedad , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Hepatitis/etiología , Hepatitis/genética , Hepatitis/patología , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Mediadores de Inflamación/metabolismo , Ligadura , Hígado/efectos de los fármacos , Hígado/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Ratones Noqueados , Necrosis , Infiltración Neutrófila , Estrés Oxidativo , Fenotipo , Transducción de Señal , Factores de Tiempo , Factor de Transcripción ReIA/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismoRESUMEN
Drug-induced liver injury includes a spectrum of pathologies, some related to the mode of injury, some to the cell type primarily damaged. Among these, drug-induced bile duct injury is characterized by the destruction of the biliary epithelium following exposure to a drug. Most of the drugs associated with bile duct injury cause immune-mediated lesions to the epithelium of interlobular ducts. These share common histopathological features with primary biliary cholangitis, such as inflammation and necrosis at the expense of cholangiocytes and, if the insult persists, bile duct loss and biliary cirrhosis. Some drugs selectively target larger ducts. Such injury is often dose-dependent and thought to be the result of intrinsic drug toxicity. The histological changes resemble those seen in primary sclerosing cholangitis. This overview focuses on the clinical and pathological features of bile duct injury associated with drug treatment and on the immunological and biochemical effects that drugs exert on the biliary epithelium. This article is part of a Special Issue entitled: Cholangiocytes in Health and Disease edited by Jesus Banales, Marco Marzioni, Nicholas LaRusso and Peter Jansen.
Asunto(s)
Conductos Biliares/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Colangitis Esclerosante/inducido químicamente , Colestasis/inducido químicamente , Células Epiteliales/efectos de los fármacos , Combinación Amoxicilina-Clavulanato de Potasio/farmacocinética , Combinación Amoxicilina-Clavulanato de Potasio/toxicidad , Ácidos y Sales Biliares/toxicidad , Conductos Biliares/citología , Conductos Biliares/efectos de los fármacos , Conductos Biliares/metabolismo , Biomarcadores/análisis , Biotransformación , Carbamazepina/farmacocinética , Carbamazepina/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Colagogos y Coleréticos/uso terapéutico , Colangitis Esclerosante/diagnóstico , Colangitis Esclerosante/tratamiento farmacológico , Colangitis Esclerosante/patología , Colestasis/diagnóstico , Colestasis/tratamiento farmacológico , Colestasis/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patologíaRESUMEN
Acute kidney injury is common in patients with liver disease and associated with significant morbidity and mortality. Besides bacterial infections, fluid loss, and use of nephrotoxic drugs AKI in liver disease may be triggered by tubular toxicity of cholephiles. Cholemic nephropathy, also known as bile cast nephropathy, supposedly represents a widely underestimated but important cause of renal dysfunction in cholestasic or advanced liver diseases with jaundice. Cholemic nephropathy describes impaired renal function along with characteristic histomorphological changes consisting of intratubular cast formation and tubular epithelial cell injury directed towards distal nephron segments. The underlying pathophysiologic mechanisms are not entirely understood and clear defined diagnostic criteria are still missing. This review aims to summarize (i) the present knowledge on clinical and morphological characteristics of cholemic nephropathy, (ii) available preclinical models, (iii) potential pathomechanisms especially the potential role of bile acids, and (iv) future diagnostic and therapeutic strategies for cholemic nephropathy. This article is part of a Special Issue entitled: Cholangiocytes in Health and Disease edited by Jesus Banales, Marco Marzioni, Nicholas LaRusso and Peter Jansen.
Asunto(s)
Lesión Renal Aguda/etiología , Ácidos y Sales Biliares/toxicidad , Colestasis/complicaciones , Ictericia Obstructiva/complicaciones , Túbulos Renales/patología , Lesión Renal Aguda/diagnóstico , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/patología , Animales , Ácidos y Sales Biliares/metabolismo , Conductos Biliares/citología , Conductos Biliares/efectos de los fármacos , Conductos Biliares/metabolismo , Conductos Biliares/patología , Bilirrubina/metabolismo , Bilirrubina/toxicidad , Colagogos y Coleréticos/farmacología , Colagogos y Coleréticos/uso terapéutico , Colestasis/tratamiento farmacológico , Colestasis/patología , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Ictericia Obstructiva/tratamiento farmacológico , Ictericia Obstructiva/patología , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Eliminación Renal , Ácido Ursodesoxicólico/análogos & derivados , Ácido Ursodesoxicólico/farmacología , Ácido Ursodesoxicólico/uso terapéuticoRESUMEN
A new in vitro method was developed to determine the bile tolerance of potentially probiotic lactobacilli. The overnight culture of various lactobacilli strains was inoculated into sterile, half-strength MRS broth supplemented with and without 0.3% (wt/vol) oxgall, buffered with 0.1 M sodium phosphate buffer at a final pH of 7.3, and incubated at 37 °C for 12 h under anaerobic conditions. The bile tolerance ability of the lactobacilli strains was expressed as the percentage of the propagation generations of the bacterial cells in the presence of oxgall to those in the absence of oxgall. The bile tolerance ability of 11 strains of 8 Lactobacillus species, including 3 bile salt hydrolase (BSH)-negative strains and 8 BSH-positive strains, was analyzed using the newly developed method and two traditional methods. The results showed that bile tolerance ability of the strains was considerably different depending on the analysis method used. The newly developed method mimics the physiological environment of the human small intestine, and avoids changes in pH and bile salt composition during the incubation period, which are drawbacks of the traditional bile tolerance test methods. Therefore, the analysis method developed in this study is more suitable to screen or compare the bile tolerance ability of lactobacilli strains.
Asunto(s)
Ácidos y Sales Biliares/toxicidad , Tolerancia a Medicamentos , Lactobacillus/efectos de los fármacos , Pruebas de Sensibilidad Microbiana/métodos , Viabilidad Microbiana/efectos de los fármacos , Probióticos , Medios de Cultivo/química , Humanos , Concentración de Iones de Hidrógeno , Lactobacillus/fisiología , TemperaturaRESUMEN
Fatty acids (FAs) are one of the important factors that can influence cell growth and membrane composition. The aim of this study was to investigate the influence of supplementing MLM+ growth medium with C18 free fatty acids (C18-FFAs), including stearic (C18:0), oleic (C18:1), linoleic (C18:2), and linolenic (C18:3) acid, on the growth of Lactobacillus plantarum CICC21001 by forming ion pairs with lysine to increase the solubility of FAs in liquid medium. The utilization of C18-FFAs was further confirmed by GC-FID. The investigation of cell properties, including cell surface hydrophobicity and zeta potential, was carried out for the modified L. plantarum and control group (non-supplementation). Furthermore, cell survival was measured in real time under heat (at 55 and 62 °C for 5 min), acid (pH 2.2), and bile salt stress. Our results indicated that the action of L. plantarum was modulated by assimilating C18-FFAs. This study suggested that C18-FFAs altered the life cycles and physiochemical properties of L. plantarum, which provided a guideline for probiotics production and their medical application.
Asunto(s)
Medios de Cultivo/química , Ácidos Grasos no Esterificados/metabolismo , Lactobacillus plantarum/fisiología , Ácidos/toxicidad , Adhesión Bacteriana , Ácidos y Sales Biliares/toxicidad , Lactobacillus plantarum/crecimiento & desarrollo , Lactobacillus plantarum/metabolismo , Viabilidad Microbiana/efectos de los fármacos , Probióticos/metabolismo , Estrés Fisiológico , Propiedades de Superficie , TemperaturaRESUMEN
In cholestasis, increases in bile acid levels result in the generation of reactive oxygen species and the induction of DNA damage and mutation. It is believed that bile acid accumulation is associated with liver tumorigenesis. However, the mechanism that underpins this phenomenon remains to be elucidated. Mcl-1, which is overexpressed in hepatic cells, is a pro-survival member of the Bcl-2 family. In this study, we observed that Mcl-1 potently suppresses the repair of bile acid-induced abasic (apurinic/apyrimidinic) sites in DNA lesions. Upon exposure of hepatic cells to glycochenodeoxycholate, one of the major conjugated human bile acids, we observed an increase in AP site accumulation along with induction of poly(ADP-ribose) polymerase and XRCC1 ( X-Ray Repair Cross Complementing 1). In addition, accumulation of Mcl-1 was observed in the nuclei of QGY-7703 cells in response to glycochenodeoxycholate stimulation. Knockdown of endogenous Mcl-1 by RNA interference significantly accelerated the repair of DNA lesions in glycochenodeoxycholate-treated cells. However, unlike XRCC1, poly(ADP-ribose) polymerase was induced following Mcl-1 knockdown. Conversely, poly(ADP-ribose) polymerase suppression was observed following glycochenodeoxycholate treatment of cells overexpressing Mcl-1. Moreover, AP-site counting analyses revealed that DNA repair activity was enhanced in cells overexpressing poly(ADP-ribose) polymerase under glycochenodeoxycholate stress conditions. It is well known that poly(ADP-ribose) polymerase plays a crucial role in the base excision repair pathway. Thus, our findings suggest that Mcl-1 suppresses base excision repair by inhibiting poly(ADP-ribose) polymerase induction following glycochenodeoxycholate-induced DNA damage. These results potentially explain how bile acid accumulation results in genetic instability and carcinogenesis.
Asunto(s)
Colestasis/genética , Proteínas de Unión al ADN/genética , Neoplasias Hepáticas/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Poli(ADP-Ribosa) Polimerasas/genética , Ácido Apurínico/genética , Ácidos y Sales Biliares/normas , Ácidos y Sales Biliares/toxicidad , Colestasis/metabolismo , Colestasis/patología , Daño del ADN/efectos de los fármacos , Reparación del ADN/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Ácido Glicoquenodesoxicólico/toxicidad , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Especies Reactivas de Oxígeno/metabolismo , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos XRESUMEN
AM-19226 is a pathogenic, non-O1/non-O139 serogroup strain of Vibrio cholerae that uses a Type 3 Secretion System (T3SS) mediated mechanism to colonize host tissues and disrupt homeostasis, causing cholera. Co-culturing the Caco2-BBE human intestinal epithelial cell line with AM-19226 in the presence of bile results in rapid mammalian cell death that requires a functional T3SS. We examined the role of bile, sought to identify the mechanism, and evaluated the contributions of T3SS translocated effectors in in vitro cell death. Our results suggest that Caco2-BBE cytotoxicity does not proceed by apoptotic or necrotic mechanisms, but rather displays characteristics consistent with osmotic lysis. Cell death was preceded by disassembly of epithelial junctions and reorganization of the cortical membrane skeleton, although neither cell death nor cell-cell disruption required VopM or VopF, two effectors known to alter actin dynamics. Using deletion strains, we identified a subset of AM-19226 Vops that are required for host cell death, which were previously assigned roles in protein translocation and colonization, suggesting that they function other than to promote cytotoxicity. The collective results therefore suggest that cooperative Vop activities are required to achieve cytotoxicity in vitro, or alternatively, that translocon pores destabilize the membrane in a bile dependent manner.
Asunto(s)
Proteínas Bacterianas/genética , Ácidos y Sales Biliares/toxicidad , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno , Sistemas de Secreción Tipo III/genética , Vibrio cholerae/genética , Proteínas Bacterianas/metabolismo , Bilis/química , Células CACO-2 , Muerte Celular/efectos de los fármacos , Eliminación de Gen , Humanos , Presión Osmótica , Transducción de Señal , Sistemas de Secreción Tipo III/metabolismo , Vibrio cholerae/crecimiento & desarrollo , Vibrio cholerae/patogenicidad , VirulenciaRESUMEN
OBJECTIVES: To evaluate the therapeutic potential of I-BET-762, an inhibitor of the bromodomain and extra-terminal (BET) protein family, in experimental acute pancreatitis (AP). METHODS: AP was induced by retrograde infusion of taurolithocholic acid sulphate into the biliopancreatic duct (TLCS-AP) or 2 intraperitoneal (i.p.) injections of ethanol and palmitoleic acid 1 h apart (FAEE-AP) or 12 hourly i.p. injections of caerulein (CER-AP). In all treatment groups, I-BET-762 (30 mg/kg, i.p.) was administered at the time of disease induction and again 12 h later. AP severity was assessed at 24 h by serum biochemistry, multiple cytokines and histopathology. RESULTS: TLCS-AP, FAEE-AP and CER-AP resulted in characteristic elevations in serum amylase and cytokine levels, increased pancreatic trypsin and myeloperoxidase activity, typical pancreatic histopathological changes and lung injury. Treatment with I-BET-762 significantly reduced biochemical, cytokine and histopathological responses in TLCS-AP and FAEE-AP, but not CER-AP. CONCLUSIONS: These results suggest that in different forms of AP there are significant differences in the epigenetic control of gene transcription contributing to the severity of disease responses. There is therapeutic potential in targeting bromodomains for the treatment of gallstone- and alcohol-related pancreatitis.
Asunto(s)
Benzodiazepinas/farmacología , Ácidos y Sales Biliares/toxicidad , Ceruletida/toxicidad , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Pancreatitis/inducido químicamente , Receptores de Superficie Celular/antagonistas & inhibidores , Ácido Taurolitocólico/análogos & derivados , Enfermedad Aguda , Amilasas/sangre , Amilasas/metabolismo , Animales , Citocinas/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Inflamación/prevención & control , Pulmón/enzimología , Masculino , Ratones , Páncreas/enzimología , Páncreas/patología , Pancreatitis/terapia , Peroxidasa/genética , Peroxidasa/metabolismo , Ácido Taurolitocólico/toxicidad , Tripsina/metabolismoRESUMEN
It is known that ß-muricholic acid anions prevent membrane toxicity of hydrophobic bile acids, which are being used in therapy for solubilization of the cholesterol type bile stone. Better knowledge of these derivative micelles is very important for understanding their physiological and pharmacological effects. ß-Axial (a) oriented hydroxyl group from the steroid skeleton decreases the hydrophobic surface of the convex side of the steroid skeleton. Therefore, the critical micellization concentration (CMC) for steroid surfactants with ß-a-OH group should increase, but in the case of OH groups of different orientations forming H-bonds in the hydrophobic phase of the micelle, it has the opposite effect; the CMC decreses, and aggregation is more favored. The set of muricholic acids (MCs) is composed by α-MC, ß-MC, γ-MC, and ω-MC, where α-MC and ß-MC have ß-axial-OH groups. The aggregation numbers (n) are determined using the Moroi-Matsuoka-Sugioka thermodynamic method. CMC, enthalpy of demicellization, and ΔCp are determined by isothermal titration calorimetry (ITC). This report pioneers in the study of MC derivatives micellization. Micelles of ß-MC and γ-MC belong to the linear congeneric group (LCG) and their micelles above 85 mM have constant aggregation numbers n = 4-5. Micelles of α-MC and ω-MC are outliers in relation to the LCG, their aggregation number constantly increases; at 85 mM n = 6.8 (α-MC) and 6.5 (ω-MC). In micelles of derivatives ß-MC and γ-MC, there is a low probability for the existence of hydrogen bonds. A micelle of α-MC probably has hydrogen bonds in its hydrophobic domain.
Asunto(s)
Ácidos y Sales Biliares/química , Membrana Celular/efectos de los fármacos , Fármacos Gastrointestinales/química , Micelas , Ácidos y Sales Biliares/uso terapéutico , Ácidos y Sales Biliares/toxicidad , Calorimetría , Química Farmacéutica , Colelitiasis/terapia , Ácidos Cólicos/química , Fármacos Gastrointestinales/uso terapéutico , Fármacos Gastrointestinales/toxicidad , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Conformación Molecular , Sodio/química , Tensoactivos , TermodinámicaRESUMEN
Barrett's esophagus (BE) is considered to be the most severe complication of gastro-esophageal reflux disease (GERD), in which the prolonged, repetitive episodes of combined acidic and biliary reflux result in the replacement of the squamous esophageal lining by columnar epithelium. Therefore, the acid-extruding mechanisms of esophageal epithelial cells (EECs) may play an important role in the defense. Our aim was to identify the presence of acid/base transporters on EECs and to investigate the effect of bile acids on their expressions and functions. Human EEC lines (CP-A and CP-D) were acutely exposed to bile acid cocktail (BAC) and the changes in intracellular pH (pHi) and Ca(2+) concentration ([Ca(2+)]i) were measured by microfluorometry. mRNA and protein expression of ion transporters was investigated by RT-PCR, Western blot, and immunohistochemistry. We have identified the presence of a Na(+)/H(+) exchanger (NHE), Na(+)/HCO3 (-) cotransporter (NBC), and a Cl(-)-dependent HCO3 (-) secretory mechanism in CP-A and CP-D cells. Acute administration of BAC stimulated HCO3 (-) secretion in both cell lines and the NHE activity in CP-D cells by an inositol triphosphate-dependent calcium release. Chronic administration of BAC to EECs increased the expression of ion transporters compared with nontreated cells. A similar expression pattern was observed in biopsy samples from BE compared with normal epithelium. We have shown that acute administration of bile acids differently alters ion transport mechanisms of EECs, whereas chronic exposure to bile acids increases the expression of acid/base transporters. We speculate that these adaptive processes of EECs represent an important mucosal defense against the bile acid-induced epithelial injury.
Asunto(s)
Esófago de Barrett/metabolismo , Ácidos y Sales Biliares/toxicidad , Células Epiteliales/efectos de los fármacos , Mucosa Esofágica/efectos de los fármacos , Proteínas de Transporte de Membrana/metabolismo , Anciano , Anciano de 80 o más Años , Esófago de Barrett/patología , Ácidos y Sales Biliares/metabolismo , Calcio/metabolismo , Línea Celular , Antiportadores de Cloruro-Bicarbonato/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Mucosa Esofágica/metabolismo , Mucosa Esofágica/patología , Femenino , Regulación de la Expresión Génica , Humanos , Concentración de Iones de Hidrógeno , Fosfatos de Inositol/metabolismo , Transporte Iónico , Masculino , Proteínas de Transporte de Membrana/genética , Metaplasia , Persona de Mediana Edad , Simportadores de Sodio-Bicarbonato/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Factores de TiempoRESUMEN
BACKGROUND & AIMS: Sustained activation of the cytosolic calcium concentration induces injury to pancreatic acinar cells and necrosis. The calcium release-activated calcium modulator ORAI1 is the most abundant Ca(2+) entry channel in pancreatic acinar cells; it sustains calcium overload in mice exposed to toxins that induce pancreatitis. We investigated the roles of ORAI1 in pancreatic acinar cell injury and the development of acute pancreatitis in mice. METHODS: Mouse and human acinar cells, as well as HEK 293 cells transfected to express human ORAI1 with human stromal interaction molecule 1, were hyperstimulated or incubated with human bile acid, thapsigargin, or cyclopiazonic acid to induce calcium entry. GSK-7975A or CM_128 were added to some cells, which were analyzed by confocal and video microscopy and patch clamp recordings. Acute pancreatitis was induced in C57BL/6J mice by ductal injection of taurolithocholic acid 3-sulfate or intravenous' administration of cerulein or ethanol and palmitoleic acid. Some mice then were given GSK-7975A or CM_128, which inhibit ORAI1, at different time points to assess local and systemic effects. RESULTS: GSK-7975A and CM_128 each separately inhibited toxin-induced activation of ORAI1 and/or activation of Ca(2+) currents after Ca(2+) release, in a concentration-dependent manner, in mouse and human pancreatic acinar cells (inhibition >90% of the levels observed in control cells). The ORAI1 inhibitors also prevented activation of the necrotic cell death pathway in mouse and human pancreatic acinar cells. GSK-7975A and CM_128 each inhibited all local and systemic features of acute pancreatitis in all 3 models, in dose- and time-dependent manners. The agents were significantly more effective, in a range of parameters, when given at 1 vs 6 hours after induction of pancreatitis. CONCLUSIONS: Cytosolic calcium overload, mediated via ORAI1, contributes to the pathogenesis of acute pancreatitis. ORAI1 inhibitors might be developed for the treatment of patients with pancreatitis.
Asunto(s)
Células Acinares/efectos de los fármacos , Benzamidas/farmacología , Canales de Calcio/efectos de los fármacos , Canales de Calcio/metabolismo , Calcio/metabolismo , Pancreatitis/tratamiento farmacológico , Pirazoles/farmacología , Células Acinares/citología , Enfermedad Aguda , Animales , Ácidos y Sales Biliares/toxicidad , Calcio/toxicidad , Células Cultivadas , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Indoles/toxicidad , Ratones , Ratones Endogámicos C57BL , Proteína ORAI1 , Pancreatitis/inducido químicamente , Pancreatitis/metabolismo , Tapsigargina/toxicidad , Factores de Tiempo , Resultado del TratamientoRESUMEN
BACKGROUND/AIMS: In nucleated cells, bile acids may activate cation channels subsequently leading to entry of Ca2+. In erythrocytes, increase of cytosolic Ca2+ activity triggers eryptosis, the suicidal death of erythrocytes characterized by phosphatidylserine exposure at the cell surface and cell shrinkage. Eryptosis is triggered by bile duct ligation, an effect partially attributed to conjugated bilirubin. The present study explored, whether bile acids may stimulate eryptosis. METHODS: Phosphatidylserine exposing erythrocytes have been identified utilizing annexin V binding, cell volume estimated from forward scatter, cytosolic Ca2+ activity determined using Fluo-3 fluorescence, and ceramide abundance at the erythrocyte surface utilizing specific antibodies. RESULTS: The exposure of human erythrocytes to glycochenodesoxycholic (GCDC) and taurochenodesoxycholic (TCDC) acid was followed by a significant decrease of forward scatter and significant increase of Fluo-3 fluorescence, ceramide abundance as well as annexin V binding. The effect on annexin V binding was significantly blunted, but not abolished by removal of extracellular Ca2+. CONCLUSION: Bile acids stimulate suicidal cell death, an effect paralleled by and in part due to Ca2+ entry and ceramide. The bile acid induced eryptosis may in turn lead to accelerated clearance of circulating erythrocytes and, thus, may contribute to anemia in cholestatic patients.