Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 314
Filtrar
1.
Int J Biol Macromol ; 277(Pt 3): 134427, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39097050

RESUMEN

Salivary glands are the principal organs responsible for secreting saliva in the oral cavity. Tumors, trauma, inflammation, and other factors can cause functional or structural damage to the glands, leading to reduced saliva secretion. In this study, we innovatively prepared a acinar-mimetic silk fibroin-collagen-astragalus polysaccharide (SCA) scaffold using low-temperature three-dimensional (3D) printing and freeze-drying techniques. We evaluated the material properties and cell compatibility of the scaffold in vitro and implanted it into the damaged parotid glands (PG) of rats to assess its efficacy in tissue reconstruction and functional repair. The results demonstrated that the SCA scaffold featured a porous structure resembling natural acini, providing an environment conducive to cell growth and orderly aggregation. It exhibited excellent porosity, water absorption, mechanical properties, and biocompatibility, fulfilling the requirements for tissue engineering scaffolds. In vitro, the scaffold facilitated adhesion, proliferation, orderly polarization, and spherical aggregation of PG cells. In vivo, the SCA scaffold effectively recruited GECs locally, forming gland-like acinar structures that matured gradually, promoting the regeneration of damaged PGs. The SCA scaffold developed in this study supports tissue reconstruction and functional repair of damaged PGs, making it a promising implant material for salivary gland regeneration.


Asunto(s)
Colágeno , Fibroínas , Glándula Parótida , Polisacáridos , Impresión Tridimensional , Ingeniería de Tejidos , Andamios del Tejido , Fibroínas/química , Fibroínas/farmacología , Andamios del Tejido/química , Animales , Glándula Parótida/química , Ratas , Colágeno/química , Ingeniería de Tejidos/métodos , Polisacáridos/química , Polisacáridos/farmacología , Porosidad , Regeneración/efectos de los fármacos , Ratas Sprague-Dawley , Células Acinares/efectos de los fármacos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Proliferación Celular/efectos de los fármacos , Masculino
2.
ACS Nano ; 18(29): 19283-19302, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38990194

RESUMEN

Developing strategies to target injured pancreatic acinar cells (PACs) in conjunction with primary pathophysiology-specific pharmacological therapy presents a challenge in the management of acute pancreatitis (AP). We designed and synthesized a trypsin-cleavable organosilica precursor bridged by arginine-based amide bonds, leveraging trypsin's ability to selectively identify guanidino groups on arginine via Asp189 at the active S1 pocket and cleave the carboxy-terminal (C-terminal) amide bond via catalytic triads. The precursors were incorporated into the framework of mesoporous silica nanoparticles (MSNs) for encapsulating the membrane-permeable Ca2+ chelator BAPTA-AM with a high loading content (∼43.9%). Mesenchymal stem cell membrane coating and surface modification with PAC-targeting ligands endow MSNs with inflammation recruitment and precise PAC-targeting abilities, resulting in the highest distribution at 3 h in the pancreas with 4.7-fold more accumulation than that of naked MSNs. The outcomes transpired as follows: After bioinspired MSNs' skeleton biodegradation by prematurely and massively activated trypsin, BAPTA-AM was on-demand released in injured PACs, thereby effectively eliminating intracellular calcium overload (reduced Ca2+ level by 81.3%), restoring cellular redox status, blocking inflammatory cascades, and inhibiting cell necrosis by impeding the IκBα/NF-κB/TNF-α/IL-6 and CaMK-II/p-RIP3/p-MLKL/caspase-8,9 signaling pathways. In AP mice, a single dose of the formulation significantly restored pancreatic function (lipase and amylase reduced more by 60%) and improved the survival rate from 50 to 91.6%. The formulation offers a potentially effective strategy for clinical translation in AP treatment.


Asunto(s)
Pancreatitis , Tripsina , Animales , Pancreatitis/tratamiento farmacológico , Pancreatitis/patología , Pancreatitis/metabolismo , Tripsina/metabolismo , Tripsina/química , Ratones , Porosidad , Nanomedicina , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Nanopartículas/química , Dióxido de Silicio/química , Compuestos de Organosilicio/química , Compuestos de Organosilicio/farmacología , Masculino , Humanos , Células Acinares/efectos de los fármacos , Células Acinares/metabolismo , Células Acinares/patología , Ratones Endogámicos C57BL
3.
Front Immunol ; 15: 1418703, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39044831

RESUMEN

Introduction: Salivary gland dysfunction, often resulting from salivary gland obstruction-induced inflammation, is a prevalent condition. Corticosteroid, known for its anti-inflammatory and immunomodulatory properties, is commonly prescribed in clinics. This study investigates the therapeutic implications and potential side effects of dexamethasone on obstructive sialadenitis recovery using duct ligation mice and salivary gland organoid models. Methods: Functional and pathological changes were assessed after administering dexamethasone to the duct following deligation 2 weeks after maintaining ligation of the mouse submandibular duct. Additionally, lipopolysaccharide- and tumor necrosis factor-induced salivary gland organoid inflammation models were established to investigate the effects and underlying mechanisms of action of dexamethasone. Results: Dexamethasone administration facilitated SG function restoration, by increasing salivary gland weight and saliva volume while reducing saliva lag time. Histological evaluation revealed, reduced acinar cell atrophy and fibrosis with dexamethasone treatment. Additionally, dexamethasone suppressed pro-inflammatory cytokines IL-1ß and TNF expression. In a model of inflammation in salivary gland organoids induced by inflammatory substances, dexamethasone restored acinar markers such as AQP5 gene expression levels, while inhibiting pro-inflammatory cytokines TNF and IL6, as well as chemokines CCL2, CXCL5, and CXCL12 induction. Macrophages cultured in inflammatory substance-treated media from salivary gland organoid cultures exhibited pro-inflammatory polarization. However, treatment with dexamethasone shifted them towards an anti-inflammatory phenotype by reducing M1 markers (Tnf, Il6, Il1b, and Cd86) and elevating M2 markers (Ym1, Il10, Cd163, and Klf4). However, high-dose or prolonged dexamethasone treatment induced acino-ductal metaplasia and had side effects in both in vivo and in vitro models. Conclusions: Our findings suggest the effectiveness of corticosteroids in treating obstructive sialadenitis-induced salivary gland dysfunction by regulating pro-inflammatory cytokines.


Asunto(s)
Dexametasona , Factor 4 Similar a Kruppel , Sialadenitis , Animales , Dexametasona/farmacología , Dexametasona/uso terapéutico , Dexametasona/administración & dosificación , Ratones , Sialadenitis/tratamiento farmacológico , Sialadenitis/patología , Antiinflamatorios/farmacología , Antiinflamatorios/administración & dosificación , Antiinflamatorios/uso terapéutico , Modelos Animales de Enfermedad , Organoides/efectos de los fármacos , Citocinas/metabolismo , Ratones Endogámicos C57BL , Glándulas Salivales/efectos de los fármacos , Glándulas Salivales/patología , Glándulas Salivales/metabolismo , Glándulas Salivales/inmunología , Acuaporina 5/metabolismo , Acuaporina 5/genética , Masculino , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Células Acinares/efectos de los fármacos , Células Acinares/metabolismo , Células Acinares/patología , Humanos
4.
Biomolecules ; 14(6)2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38927047

RESUMEN

Acute pancreatitis (AP) entails pancreatic inflammation, tissue damage and dysregulated enzyme secretion, including pancreatic lipase (PL). The role of irisin, an anti-inflammatory and anti-apoptotic cytokine, in AP and exocrine pancreatic stress is unclear. We have previously shown that irisin regulates PL through the PPARγ-PGC1α-FNDC5 pathway. In this study, we investigated irisin and irisin's pathway on AP in in vitro (AR42J-B13) and ex vivo (rat primary acinar) models using molecular, biochemical and immunohistochemistry methodology. Pancreatitis induction (cerulein (cer)) resulted in a significant up-regulation of the PPARγ-PGC1α-FNDC5 axis, PL expression and secretion and endoplasmic reticulum (ER) stress unfolded protein response (UPR) signal-transduction markers (CHOP, XBP-1 and ATF6). Irisin addition in the cer-pancreatitis state resulted in a significant down-regulation of the PPARγ-PGC1α-FNDC5 axis, PPARγ nucleus-translocation and inflammatory state (TNFα and IL-6) in parallel to diminished PL expression and secretion (in vitro and ex vivo models). Irisin addition up-regulated the expression of pro-survival UPR markers (ATF6 and XBP-1) and reduced UPR pro-apoptotic markers (CHOP) under cer-pancreatitis and induced ER stress (tunicamycin), consequently increasing cells viability. Irisin's pro-survival effect under cer-pancreatitis state was abolished under PPARγ inhibition. Our findings suggest irisin as a potential therapeutic option for AP via its ability to up-regulate pro-survival UPR signals and activate the PPARγ-PGC1α-FNDC5 pathway.


Asunto(s)
Células Acinares , Supervivencia Celular , Fibronectinas , PPAR gamma , Pancreatitis , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Transducción de Señal , Respuesta de Proteína Desplegada , Fibronectinas/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Animales , Pancreatitis/metabolismo , Pancreatitis/patología , Pancreatitis/tratamiento farmacológico , PPAR gamma/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Ratas , Supervivencia Celular/efectos de los fármacos , Células Acinares/metabolismo , Células Acinares/efectos de los fármacos , Células Acinares/patología , Transducción de Señal/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Ceruletida , Masculino , Línea Celular , Lipasa/metabolismo
5.
Discov Med ; 36(185): 1162-1168, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38926102

RESUMEN

BACKGROUND: Atypical acinar cell foci (AACF) seen in pancreatic cancer are fatal and have been studied with some causative agents. However, for the first time, the effect of acetylsalicylic acid with nitric oxide (NO-ASA) on AACF was examined in this study. Although NO-ASA has very successful inhibitory effects against some types of cancer, it has not been investigated whether they can exert their inhibition effects on AACFs. METHODS: For experimental purposes, 21 14-day-old male Wistar albino rats were used. Azaserine (30 mg/kg) was dissolved in 0.9% NaCl solution and injected intraperitoneally (i.p.) into 14 rats, except for the Control group (Cont) rats, for three weeks. Rats that were injected with azaserine once a week for three weeks and those that did not receive treatment were divided into experimental groups. 15 days after the end of the azaserine injection protocol, NO-ASA was applied to azaserine with NO-ASA (Az+NO-ASA) group rats three consecutive times with an interval of 15 days by gavage. At the end of the 5-month period, pancreatic tissue was dissected and weighed. Pancreas preparations prepared from histological sections were examined for AACF burden and analyzed via a video image analyzer. One-way analysis of variance (ANOVA) non-parametric statistical analyses were performed to test whether there was a difference between the averages of the experimental and Control groups. RESULTS: AACF burden in both groups injected with azaserine was found to be statistically significant in all categories compared to that of the Control group (p < 0.05). The average Calculated Estimated average AACF volume (mm3) values, the Calculated estimated average AACF diameter (µm), the Estimated average number of AACF per unit volume, AACF rate as a % of Calculated Organ Volume were higher in the AzCont group rats than in the Az+NO-ASA group, when compared, and there was an important level statistical difference between the groups (p < 0.05). It was determined that for all parameters AACFs load in Az+NO-ASA group rats were significantly reduced compared to that of AzCont group rats (p < 0.05). CONCLUSIONS: We observed that, as a result of the NO-ASA application, the experimental AACF focus ratio created by azaserine injection was significantly inhibited. The inhibitory effect of AACFs in Az+NO-ASA group rats may have resulted from the significant and independent chemopreventive and/or chemotherapeutic activity of NO-ASA against exocrine pancreatic AACF foci.


Asunto(s)
Células Acinares , Aspirina , Óxido Nítrico , Páncreas Exocrino , Neoplasias Pancreáticas , Ratas Wistar , Animales , Masculino , Aspirina/farmacología , Aspirina/uso terapéutico , Aspirina/administración & dosificación , Óxido Nítrico/metabolismo , Ratas , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Células Acinares/efectos de los fármacos , Células Acinares/patología , Células Acinares/metabolismo , Páncreas Exocrino/efectos de los fármacos , Páncreas Exocrino/patología
6.
Biochem Pharmacol ; 225: 116279, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38740221

RESUMEN

Berberine, a natural isoquinoline alkaloid, exhibits a variety of pharmacological effects, but the pharmacological targets and mechanisms remain elusive. Here, we report a novel finding that berberine inhibits acetylcholine (ACh)-induced intracellular Ca2+ oscillations, mediated through an inhibition of the muscarinic subtype 3 (M3) receptor. Patch-clamp recordings and confocal Ca2+ imaging were applied to acute dissociated pancreatic acinar cells prepared from CD1 mice to examine the effects of berberine on ACh-induced Ca2+ oscillations. Whole-cell patch-clamp recordings showed that berberine (from 0.1 to 10 µM) reduced ACh-induced Ca2+ oscillations in a concentration-dependent manner, and this inhibition also depended on ACh concentrations. The inhibitory effect of berberine neither occurred in intracellular targets nor extracellular cholecystokinin (CCK) receptors, chloride (Cl-) channels, and store-operated Ca2+ channels. Together, the results demonstrate that berberine directly inhibits the muscarinic M3 receptors, further confirmed by evidence of the interaction between berberine and M3 receptors in pancreatic acinar cells.


Asunto(s)
Células Acinares , Berberina , Señalización del Calcio , Receptor Muscarínico M3 , Animales , Berberina/farmacología , Receptor Muscarínico M3/metabolismo , Receptor Muscarínico M3/antagonistas & inhibidores , Ratones , Células Acinares/efectos de los fármacos , Células Acinares/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Masculino , Acetilcolina/metabolismo , Calcio/metabolismo , Relación Dosis-Respuesta a Droga
7.
Pancreas ; 53(8): e662-e669, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38696385

RESUMEN

OBJECTIVES: Amino acids play an essential role in protein synthesis, metabolism, and survival of pancreatic acini. Adequate nutritional support is important for acute pancreatitis treatment. However, high concentrations of arginine and lysine may induce acute pancreatitis. The study aimed to identify the most suitable l -amino acids as safe energy sources for pancreatic acinar cells. MATERIALS AND METHODS: Pancreatic acini were isolated from male Wistar rats. Effects of amino acids (0.1-20 mM) on uncoupled respiration of isolated acini were studied with a Clark electrode. Cell death was evaluated with fluorescent microscopy and DNA gel electrophoresis. RESULTS: Among the tested amino acids, glutamate, glutamine, alanine, lysine, and aspartate were able to stimulate the uncoupled respiration rate of isolated pancreatic acini, whereas arginine, histidine, and asparagine were not. Lysine, arginine, and glutamine (20 mM) caused complete loss of plasma membrane integrity of acinar cells after 24 hours of incubation. Glutamine also caused early (2-4 hours) cell swelling and blebbing. Aspartate, asparagine, and glutamate only moderately decreased the number of viable cells, whereas alanine and histidine were not toxic. DNA fragmentation assay and microscopic analysis of nuclei showed no evidence of apoptosis in cells treated with amino acids. CONCLUSIONS: Alanine and glutamate are safe and effective energy sources for mitochondria of pancreatic acinar cells.


Asunto(s)
Células Acinares , Aminoácidos , Ratas Wistar , Animales , Masculino , Aminoácidos/metabolismo , Aminoácidos/farmacología , Células Acinares/metabolismo , Células Acinares/efectos de los fármacos , Ratas , Páncreas/metabolismo , Páncreas/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Fragmentación del ADN/efectos de los fármacos , Glutamina/metabolismo , Glutamina/farmacología , Arginina/farmacología , Supervivencia Celular/efectos de los fármacos , Ácido Glutámico/metabolismo , Ácido Glutámico/toxicidad , Ácido Glutámico/farmacología , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Apoptosis/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Histidina/farmacología
8.
ACS Nano ; 18(21): 13885-13898, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38757565

RESUMEN

Severe acute pancreatitis (SAP), characterized by pancreatic acinar cell death, currently lacks effective targeted therapies. Ellagic acid (EA), rich in pomegranate, shows promising anti-inflammatory and antioxidant effects in SAP treatment. However, the roles of other forms of EA, such as plant extracellular vesicles (EVs) extracted from pomegranate, and Urolithin A (UA), converted from EA through gut microbiota metabolism in vivo, have not been definitively elucidated. Our research aimed to compare the effects of pomegranate-derived EVs (P-EVs) and UA in the treatment of SAP to screen an effective formulation and to explore its mechanisms in protecting acinar cells in SAP. By comparing the protective effects of P-EVs and UA on injured acinar cells, UA showed superior therapeutic effects than P-EVs. Subsequently, we further discussed the mechanism of UA in alleviating SAP inflammation. In vivo animal experiments found that UA could not only improve the inflammatory environment of pancreatic tissue and peripheral blood circulation in SAP mice but also revealed that the mechanism of UA in improving SAP might be related to mitochondria and endoplasmic reticulum (ER) through the results including pancreatic tissue transcriptomics and transmission electron microscopy. Further research found that UA could regulate ER-mitochondrial calcium channels and reduce pancreatic tissue necroptosis. In vitro experiments of mouse pancreatic organoids and acinar cells also confirmed that UA could improve pancreatic inflammation by regulating the ER-mitochondrial calcium channel and necroptosis pathway proteins. This study not only explored the therapeutic effect of plant EVs on SAP but also revealed that UA could alleviate SAP by regulating ER-mitochondrial calcium channel and reducing acinar cell necroptosis, providing insights into the pathogenesis and potential treatment of SAP.


Asunto(s)
Cumarinas , Retículo Endoplásmico , Mitocondrias , Pancreatitis , Animales , Cumarinas/farmacología , Cumarinas/química , Pancreatitis/tratamiento farmacológico , Pancreatitis/metabolismo , Pancreatitis/patología , Ratones , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Canales de Calcio/metabolismo , Masculino , Ratones Endogámicos C57BL , Granada (Fruta)/química , Células Acinares/efectos de los fármacos , Células Acinares/metabolismo , Células Acinares/patología , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/química
9.
ACS Nano ; 18(18): 11778-11803, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38652869

RESUMEN

Severe acute pancreatitis (AP) is a life-threatening pancreatic inflammatory disease with a high mortality rate (∼40%). Existing pharmaceutical therapies in development or in clinical trials showed insufficient treatment efficacy due to their single molecular therapeutic target, poor water solubility, short half-life, limited pancreas-targeting specificity, etc. Herein, acid-responsive hollow mesoporous Prussian blue nanoparticles wrapped with neutrophil membranes and surface modified with the N,N-dimethyl-1,3-propanediamine moiety were developed for codelivering membrane-permeable calcium chelator BAPTA-AM (BA) and trypsin activity inhibitor gabexate mesylate (Ga). In the AP mouse model, the formulation exhibited efficient recruitment at the inflammatory endothelium, trans-endothelial migration, and precise acinar cell targeting, resulting in rapid pancreatic localization and higher accumulation. A single low dose of the formulation (BA: 200 µg kg-1, Ga: 0.75 mg kg-1) significantly reduced pancreas function indicators to close to normal levels at 24 h, effectively restored the cell redox status, reduced apoptotic cell proportion, and blocked the systemic inflammatory amplified cascade, resulting in a dramatic increase in the survival rate from 58.3 to even 100%. Mechanistically, the formulation inhibited endoplasmic reticulum stress (IRE1/XBP1 and ATF4/CHOP axis) and restored impaired autophagy (Beclin-1/p62/LC3 axis), thereby preserving dying acinar cells and restoring the cellular "health status". This formulation provides an upstream therapeutic strategy with clinical translation prospects for AP management through synergistic ion homeostasis regulation and pancreatic autodigestion inhibition.


Asunto(s)
Células Acinares , Calcio , Homeostasis , Nanomedicina , Pancreatitis , Animales , Pancreatitis/tratamiento farmacológico , Pancreatitis/patología , Pancreatitis/metabolismo , Células Acinares/efectos de los fármacos , Células Acinares/metabolismo , Células Acinares/patología , Ratones , Homeostasis/efectos de los fármacos , Calcio/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/patología , Inflamación/metabolismo , Nanopartículas/química , Páncreas/patología , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Ratones Endogámicos C57BL , Masculino , Humanos
10.
J Appl Toxicol ; 44(7): 1005-1013, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38462915

RESUMEN

Acute pancreatitis represents an inflammatory disease featuring pancreatic necrosis and inflammation. Inflammatory injury of pancreatic acinar cells (PACs) is critically involved in the initiation and progression of acute pancreatitis. Pyroptosis, a new kind of programmed cell death concomitant with a low-grade inflammatory reaction, plays a function in acute pancreatitis pathology. It is unclear whether saikosaponin d (SSd), a pharmacologically active natural product, could protect PACs by regulating pyroptosis. Here, we established a PAC injury model in vitro using cerulein to treat AR42J cells. SSd restored viability and proliferation and lowered the release of pancreatic enzymes and inflammatory interleukins in cerulein-treated AR42J cells. Cerulein-induced pyroptosis was evidenced by typical ultrastructural changes and NLRP3/caspase-1 activation in AR42J cells, but SSd attenuated cerulein-induced pyroptosis and inhibited NLRP3/caspase-1 pathway. Mechanically, SSd reduced mitochondrial damage and mtDNA release, and blocked cGAS-STING signaling in AR42J cells treated with cerulein, contributing to the inhibition of NLRP3-mediated pyroptosis. Furthermore, SSd abolished cerulein-elevated oxidative stress in AR42J cells, leading to the mitigation of mitochondrial damage and inhibition of cGAS-STING signaling and pyroptosis. In conclusion, SSd protected PACs against cerulein-induced pyroptosis by alleviating mitochondrial damage and inhibiting the cGAS-STING pathway, and it could be a therapeutic candidate for acute pancreatitis.


Asunto(s)
Células Acinares , Ceruletida , Mitocondrias , Ácido Oleanólico , Piroptosis , Saponinas , Transducción de Señal , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/farmacología , Saponinas/farmacología , Piroptosis/efectos de los fármacos , Ceruletida/toxicidad , Animales , Células Acinares/efectos de los fármacos , Células Acinares/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Páncreas/efectos de los fármacos , Páncreas/patología , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Línea Celular , Pancreatitis/inducido químicamente , Pancreatitis/prevención & control , Pancreatitis/tratamiento farmacológico , Pancreatitis/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Sustancias Protectoras/farmacología
11.
Naunyn Schmiedebergs Arch Pharmacol ; 397(8): 5989-5999, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38376541

RESUMEN

Ferroptosis, characterized by lipid peroxidation, plays a significant role in the pathogenesis of acute pancreatitis (AP). While sterol O-acyltransferase 2 (Soat2) is known for its crucial regulatory role in cholesterol homeostasis, its involvement in the development of AP remains unreported. We conducted this study to identify the pivotal role of Soat2 in AP using transcriptomic databases. Subsequently, we confirmed its alterations through both in vitro and in vivo experimental models. Furthermore, we performed intervention with the Soat2 inhibitor avasimibe to evaluate pancreatic tissue pathology and serum enzymatic levels and observe inflammatory cell infiltration through immunohistochemistry. Additionally, changes in indicators related to ferroptosis were also observed. The results showed that in the AP mouse model, the protein and mRNA levels of Soat2 were significantly increased. Following avasimibe administration, there was a decrease in serum amylase levels, reduction in pancreatic tissue pathological damage, and attenuation of inflammatory cell infiltration. Furthermore, avasimibe administration resulted in downregulation of ferroptosis-related indicators. In conclusion, our findings suggest that the Soat2 inhibitor avasimibe protects against AP in mice through inhibition of the ferroptosis.


Asunto(s)
Células Acinares , Ferroptosis , Pancreatitis , Esterol O-Aciltransferasa , Animales , Ferroptosis/efectos de los fármacos , Pancreatitis/tratamiento farmacológico , Pancreatitis/patología , Pancreatitis/metabolismo , Células Acinares/efectos de los fármacos , Células Acinares/metabolismo , Células Acinares/patología , Masculino , Ratones , Esterol O-Aciltransferasa/antagonistas & inhibidores , Esterol O-Aciltransferasa/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Páncreas/patología , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Humanos
12.
Toxicol Sci ; 199(1): 120-131, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38407484

RESUMEN

The effect of 2,2',4,4'-tetrabromodiphenyl ether (BDE-47), a persistent environmental pollutant commonly used as a flame retardant in various consumer products, on pancreatitis has not been clearly elucidated, although it has been reported to be toxic to the liver, nervous system, and reproductive system. Acute pancreatitis (AP) and chronic pancreatitis (CP) models were induced in this study by intraperitoneal injection of caerulein. The aim was to investigate the impact of BDE-47 on pancreatitis by exposing the animals to acute (1 week) or chronic (8 weeks) doses of BDE-47 (30 mg/kg in the low-concentration group and 100 mg/kg in the high-concentration group). Additionally, BDE-47 was utilized to stimulate mouse bone marrow-derived macrophages, pancreatic primary stellate cells, and acinar cells in order to investigate the impact of BDE-47 on pancreatitis. In vivo experiments conducted on mice revealed that chronic exposure to BDE-47, rather than acute exposure, exacerbated the histopathological damage of AP and CP, leading to elevated fibrosis in pancreatic tissue and increased infiltration of inflammatory cells in the pancreas. In vitro experiments showed that BDE-47 can promote the expression of the inflammatory cytokines Tnf-α and Il-6 in M1 macrophages, as well as promote acinar cell apoptosis through the activation of the PERK and JNK pathways via endoplasmic reticulum stress. The findings of this study imply chronic exposure to BDE-47 may exacerbate the progression of both AP and CP by inducing acinar cell apoptosis and dysregulating inflammatory responses.


Asunto(s)
Células Acinares , Apoptosis , Éteres Difenilos Halogenados , Pancreatitis Crónica , Pancreatitis , Animales , Éteres Difenilos Halogenados/toxicidad , Apoptosis/efectos de los fármacos , Pancreatitis Crónica/inducido químicamente , Pancreatitis Crónica/patología , Células Acinares/efectos de los fármacos , Células Acinares/patología , Células Acinares/metabolismo , Masculino , Pancreatitis/inducido químicamente , Pancreatitis/patología , Macrófagos/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones , Ceruletida/toxicidad , Páncreas/efectos de los fármacos , Páncreas/patología , Inflamación/inducido químicamente , Inflamación/patología , Células Estrelladas Pancreáticas/efectos de los fármacos , Células Estrelladas Pancreáticas/patología , Células Estrelladas Pancreáticas/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Retardadores de Llama/toxicidad , Células Cultivadas
13.
Cell Mol Biol (Noisy-le-grand) ; 67(2): 101-108, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34817332

RESUMEN

This research was carried out to investigate the effect and mechanism of Angelic Shaoyaosan mediated AMPK/SIRT1 positive feedback loop to promote autophagy and regulate systemic inflammatory response in acute pancreatitis. In this study, the rat pancreatic acini AR42J cells were chosen as the research object, the application of hyla induced pancreatic acinar cells made model for acute pancreatitis, application of different concentrations of angelica peony spread effect on building cells, thus divided into control group, built in the module, the low concentration group, concentration and high concentration groups, determined by MTT method was applied to explore the above categories in cell proliferation, cell apoptosis was measured by flow cytometry, the expression of inflammatory factors in cell supernatant was determined by enzyme-linked immunoassay, and the expression of autophagy marker proteins LC3- ? and P62 was determined by Western-Bolt method. In order to explore the relationship between AMPK and SIRT1, immunoco-precipitation method was used to determine the interaction between AMPK and SIRT1, and dual luciferase experiment was used to explore the effect of AMPK on SIRT1. The AICAR group, BLM-275 group and negative control group were established. To explore the effect of SIRT1 on AMPK, we established SRT 1720 group, EX-527 group and control group. Direct binding between AMPK and SIRT1 should be determined by chromatin co-precipitation assay. In order to further explore the effect of AMPK/SIRT1 positive feedback loop on the systemic inflammatory response of acute pancreatitis, this study selected the medium-concentration Danggui Shaoyajiao SAN group as the control group (group C), and applied AMPK inhibitor BLM-275 and SIRT1 inhibitor EX 527 to the effect of medium-concentration Danggui Shaoyajiao SAN cells, respectively. The expression of autophagy marker proteins LC3- ? and P62 in groups A and B were determined by the Western-Bolt method. Results showed that compared with the control group, the cell survival rate, the expression of AMPK, SIRT1 and LC3-II in the model group were decreased, and the apoptosis rate of iNOS, IL-2, TNF-?, P62 and apoptosis were increased in the model group (P<0.05). the levels of iNOS, IL-2, TNF-?, P62 and cell survival rate in low, medium and high concentration groups decreased gradually, while the expressions of AMPK, SIRT1, LC3-II and cell apoptosis rate increased (P<0.05). The levels of iNOS, IL-2 and TNF-? in the three groups were gradually decreased with the increase of the concentration (P<0.05). Immunoprecipitation showed that AMPK and SIRT1 could bind to each other in cells. The double luciferase experiment indicated that the reporter gene containing the SIRT1 binding site was constructed. The luciferase activity was increased in THE AICAR group and decreased in the BLM-275 group (P<0.05). The reporter gene containing the AMPK promoter binding site was constructed. The luciferase activity in SRT1720 group was increased, while that in EX-527 group was decreased. SIRT1 could directly bind to the AMPK promoter. SIRT1 and LC3- ? protein expressions in group A were down-regulated, and P62 protein was increased (P<0.05). The protein expressions of AMPK and LC3- ? in group B were down-regulated, and the protein expression of P62 was increased (P<0.05). It concluded that AMPK can directly bind to activate SIRT1 expression, and SIRT1 expression can also activate AMPK, forming a positive feedback loop between the two. Therefore, Angelic Shaoyaodong decoction can mediate AMPK/SIRT1 positive feedback pathway to promote autophagy and regulate systemic inflammatory response in acute pancreatitis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Angelica sinensis/química , Autofagia/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Retroalimentación Fisiológica/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Pancreatitis/metabolismo , Sirtuina 1/metabolismo , Células Acinares/citología , Células Acinares/efectos de los fármacos , Células Acinares/metabolismo , Enfermedad Aguda , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/química , Mediadores de Inflamación/sangre , Pancreatitis/patología , Ratas , Transducción de Señal/efectos de los fármacos
14.
Oxid Med Cell Longev ; 2021: 4187398, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34840668

RESUMEN

As a calcium-regulated protein, CaMK II is closely related to cell death, and it participates in the development of pathological processes such as reperfusion injury, myocardial infarction, and oligodendrocyte death. The function of CaMK II activation in acute pancreatitis (AP) remains unclear. In our study, we confirmed that the expression of p-CaMK II was increased significantly and consistently in injured pancreatic tissues after caerulein-induced AP. Then, we found that KN93, an inhibitor of CaMK II, could mitigate the histopathological manifestations in pancreatic tissues, reduce serum levels of enzymology, and decrease oxidative stress products. Accordingly, we elucidated the effect of KN93 in vitro and found that KN93 had a protective effect on the pancreatic acinar cell necroptosis pathway by inhibiting the production of ROS and decreasing the expression of RIP3 and p-MLKL. In addition, we identified the protective effect of KN93 on AP through another mouse model induced by pancreatic duct ligation (PDL). Together, these data demonstrated that CaMK II participates in the development of AP and that inhibiting CaMK II activation could protect against AP by reducing acinar cell necroptosis, which may provide a new idea target for the prevention and treatment of AP in the clinic.


Asunto(s)
Células Acinares/efectos de los fármacos , Bencilaminas/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Necroptosis , Pancreatitis/prevención & control , Sustancias Protectoras/farmacología , Especies Reactivas de Oxígeno/metabolismo , Sulfonamidas/farmacología , Células Acinares/metabolismo , Células Acinares/patología , Animales , Ceruletida/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Pancreatitis/inducido químicamente , Pancreatitis/metabolismo , Pancreatitis/patología
15.
Pancreas ; 50(7): 972-981, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34629447

RESUMEN

OBJECTIVES: The objective of this study was to test whether pyruvate and glutamine affect the ethanol and cholecystokinin (CCK) effects on the mitochondrial function, viability, and morphology of rat pancreatic acini. METHODS: Respiration was measured with Clark oxygen electrode. Mitochondrial membrane potential, reduced nicotinamide adenine dinucleotide (phosphate) (NAD(P)H), cell morphology, and viability were studied with fluorescence microscopy. RESULTS: In vitro, CCK (0.1 nM) caused pyruvate-dependent stimulation of basal and uncoupled respiration, and the effects were abolished by ethanol (20 mM). The combination of ethanol with CCK (2 hours) caused necrosis of approximately 40% acinar cells in medium with glucose, but not with pyruvate and/or glutamine. Cholecystokinin (10 nM) or ethanol with 0.1 nM CCK caused plasma membrane blebbing not related to apoptosis only when both glutamine and pyruvate were present. Glutamine, but not pyruvate, decreased NAD(P)H level and prevented the effects of ethanol with CCK on mitochondrial membrane potential and NAD(P)H, but, in combination with CCK and ethanol, decreased the uncoupled respiration. In vivo, the combination of ethanol (4 g/kg) and CCK (20 pmol/kg) suppressed basal and uncoupled respiration and caused acinar cell blebbing, but not necrosis. CONCLUSIONS: The lack of sufficient substrate supply in vitro makes pancreatic acinar cells susceptible to necrosis caused by ethanol and CCK in clinically relevant concentrations.


Asunto(s)
Células Acinares/efectos de los fármacos , Colecistoquinina/farmacología , Etanol/farmacología , Glutamina/metabolismo , Mitocondrias/efectos de los fármacos , Ácido Pirúvico/metabolismo , Células Acinares/metabolismo , Amilasas/sangre , Amilasas/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Etanol/sangre , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Microscopía Fluorescente , Mitocondrias/metabolismo , Necrosis , Oxidación-Reducción/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Páncreas/citología , Páncreas/metabolismo , Ratas Wistar
16.
Exp Eye Res ; 211: 108760, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34487726

RESUMEN

Little is known about the relationship between stimulation of lacrimal gland (LG) tear protein secretion by parasympathetic versus sympathetic nerves, particularly whether the spectrum of tear proteins evoked through each innervation pathway varies. We have previously shown that activity and abundance of cathepsin S (CTSS), a cysteine protease, is greatly increased in tears of Sjögren's syndrome (SS) patients and in tears from the male NOD mouse of autoimmune dacryoadenitis that recapitulates SS-associated dry eye disease. Beyond the increased synthesis of CTSS detected in the diseased NOD mouse LG, increased tear CTSS secretion in NOD mouse tears was recently linked to increased exocytosis from a novel endolysosomal secretory pathway. Here, we have compared secretion and trafficking of CTSS in healthy mouse LG acinar cells stimulated with either the parasympathetic acetylcholine receptor agonist, carbachol (CCh), or the sympathetic α1-adrenergic agonist, phenylephrine (PE). In situ secretion studies show that PE significantly increases CTSS activity and protein in tears relative to CCh stimulation by 1.2-fold (***, p = 0.0009) and ∼5-fold (*, p-0.0319), respectively. A similar significant increase in CTSS activity with PE relative to CCh is observed when cultured LGAC are stimulated in vitro. CCh stimulation significantly elevates intracellular [Ca2+], an effect associated with increases in the size of Rab3D-enriched vesicles consistent with compound fusion, and subsequently decreases in their intensity of labeling consistent with their exocytosis. PE stimulation induces a lower [Ca2+] response and has minimal effects on Rab3D-enriched SV diameter or the intensity of Rab3D-enriched SV labeling. LG deficient in Rab3D exhibit a higher sensitivity to PE stimulation, and secrete more CTSS activity. Significant increases in the colocalization of endolysosomal vesicle markers (Lamp1, Lamp2, Rab7) with the subapical actin suggestive of fusion of endolysosomal vesicles at the apical membrane occur both with CCh and PE stimulation, but PE demonstrates increased colocalization. In conclusion, the α1-adrenergic agonist, PE, increases CTSS secretion into tears through a pathway independent of the exocytosis of Rab3D-enriched mature SV, possibly representing an alternative endolysosomal secretory pathway.


Asunto(s)
Células Acinares/efectos de los fármacos , Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Catepsinas/metabolismo , Aparato Lagrimal/efectos de los fármacos , Fenilefrina/farmacología , Vías Secretoras/efectos de los fármacos , Lágrimas/metabolismo , Células Acinares/metabolismo , Animales , Western Blotting , Calcio/metabolismo , Carbacol/farmacología , Células Cultivadas , Agonistas Colinérgicos/farmacología , Modelos Animales de Enfermedad , Femenino , Silenciador del Gen , Aparato Lagrimal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , beta-N-Acetilhexosaminidasas/metabolismo , Proteínas de Unión al GTP rab3/genética
17.
Mol Med Rep ; 24(5)2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34498715

RESUMEN

Acute pancreatitis (AP) is one of the most frequent gastrointestinal diseases and has no specific treatment. It has been shown that dysfunction of pancreatic acinar cells can lead to AP progression. Emodin is a natural product, which can alleviate the symptoms of AP. However, the mechanism by which emodin regulates the function of pancreatic acinar cells remains unclear. Thus, the present study aimed to investigate the mechanism by which emodin modulates the function of pancreatic acinar cells. To mimic AP in vitro, pancreatic acinar cells were cotreated with caerulein and lipopolysaccharide (LPS). Exosomes were isolated using the ExoQuick precipitation kit. Western blot analysis, Nanosight Tracking analysis and transmission electron microscopy were performed to detect the efficiency of exosome separation. Gene expression was detected by reverse transcription­quantitative PCR. The levels of IL­1ß and TNF­α were detected by ELISA. The data indicated that emodin significantly decreased the levels of IL­1ß and TNF­α in the supernatant samples derived from AR42J cells cotreated with caerulein and LPS. In addition, emodin significantly promoted the proliferation of AR42J cells cotreated with caerulein and LPS, and inhibited apoptosis, while the effect of emodin was reversed by long non­coding (lnc)RNA taurine upregulated 1 (TUG1) overexpression. The expression level of TUG1 in AR42J cells or exosomes derived from AR42J cells was significantly increased following treatment of the cells with LPS and caerulein, while this effect was notably reversed by emodin treatment. In addition, exosomes derived from caerulein and LPS cotreated AR42J cells inhibited the differentiation and anti­inflammatory function of regulatory T cells, while treatment of the cells with emodin significantly decreased this effect. In conclusion, the data indicated that emodin inhibited the induction of inflammation in AR42J cells by regulating the expression of cellular and exosomal lncRNA. Therefore, emodin may be used as a potential agent for the treatment of AP.


Asunto(s)
Emodina/farmacología , Pancreatitis/tratamiento farmacológico , ARN Largo no Codificante/metabolismo , Células Acinares/efectos de los fármacos , Células Acinares/metabolismo , Células Acinares/patología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Exosomas/genética , Humanos , Masculino , Ratones , Pancreatitis/genética , Pancreatitis/metabolismo , Pancreatitis/patología , Inhibidores de Proteínas Quinasas/farmacología , ARN Largo no Codificante/genética , Ratas Wistar
18.
Am J Physiol Gastrointest Liver Physiol ; 321(5): G449-G460, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34523348

RESUMEN

Chronic pancreatitis (CP) is a complex inflammatory disorder with numerous associated genetic and environmental risk factors. The most distressing characteristic of CP is recalcitrant pain, often requiring surgical resection including total pancreatectomy with islet autotransplantation (TPIAT). We studied five consented subjects undergoing pancreatic resection and processed isolated cells for single-cell RNA sequencing (scRNA-Seq). Using high-dimensional transcriptomic cluster analysis, we identified 11 unique cell clusters in the pancreas tissue. These cell clusters include a cluster of undifferentiated/dedifferentiated cells and two unique clusters of acinar cells, one of which appears to be in a transitional stage. To determine the cellular response to protease inhibitor and stimulation, we treated aliquots of cells from one subject with a protease inhibitor cocktail with and without bethanechol (a muscarinic receptor agonist) at 100 and 400 µM and compared gene expression profiles. The protease inhibitors appeared to reduce cell stress. Pancreatic digestive enzymes and islet hormones were upregulated in both doses of bethanechol-treated cells compared with naïve cells. High-dose bethanechol appeared to be toxic and consistent with hyperstimulation. These studies demonstrate the feasibility of investigating human acinar cell physiology at the single-cell level and initial evidence that these cells retain responsiveness to agonist stimulation with predicted second messenger and transcriptomic responses.NEW & NOTEWORTHY We conducted single cell RNA sequencing on pancreas tissue from five individuals. We identified eleven unique cell clusters including a large population of dedifferentiated cells as well as two unique clusters of acinar cells, one of which appears to exist in a transitional state. We also examined the cellular response of pancreas tissue to stimulation and identified affected genes and pathways, including pancreatic digestive enzymes.


Asunto(s)
Células Acinares/metabolismo , Perfilación de la Expresión Génica , Páncreas/metabolismo , Pancreatitis Crónica/genética , RNA-Seq , Análisis de la Célula Individual , Transcriptoma , Células Acinares/efectos de los fármacos , Células Acinares/patología , Desdiferenciación Celular , Análisis por Conglomerados , Estudios de Factibilidad , Humanos , Agonistas Muscarínicos/farmacología , Páncreas/efectos de los fármacos , Páncreas/patología , Páncreas/cirugía , Pancreatectomía , Pancreaticoduodenectomía , Pancreatitis Crónica/metabolismo , Pancreatitis Crónica/patología , Pancreatitis Crónica/cirugía , Inhibidores de Proteasas/farmacología
19.
Oxid Med Cell Longev ; 2021: 9921839, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34422214

RESUMEN

Acute pancreatitis (AP), an inflammatory disorder of the pancreas, can cause systemic inflammatory responses. Escin Sodium (ES), a natural mixture of triterpene saponins extracted from the dry ripe fruit of Fructus Aesculi or horse chestnut crude, has been demonstrated to have antiedematous, anti-inflammatory, and antiexudative effects. We here aim to investigate the effects of ES pretreatment on AP in vivo and in vitro and explore its potential molecular mechanism. In the present study, we demonstrated that ES pretreatment could apparently decrease amylase and lipase, downregulate inflammatory cytokines, and attenuate pancreatic damage. Additionally, the increased expression of apoptotic-related proteins and the results of flow cytometry demonstrated the effects of ES on promoting apoptosis in acinar cells. Moreover, ES could enhance mitochondrial membrane potential (MMP, ΔΨm) and reactive oxygen species (ROS) level and reduce intracellular calcium concentration, which are closely related to mitochondrial-mediated death. The effect of ES pretreatment on acinar cell apoptosis was furtherly confirmed by the regulatory pathway of the ERK/STAT3 axis. These results suggest that ES attenuates the severity of AP by enhancing cell apoptosis via suppressing the ERK/STAT3 signaling pathway. These findings provide evidence for ES which is treated as a novel and potent therapeutic for the treatment of AP.


Asunto(s)
Apoptosis , Escina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Pancreatitis/tratamiento farmacológico , Factor de Transcripción STAT3/antagonistas & inhibidores , Células Acinares/efectos de los fármacos , Células Acinares/metabolismo , Células Acinares/patología , Animales , Calcio/metabolismo , Fármacos Cardiovasculares/farmacología , Masculino , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Pancreatitis/inducido químicamente , Pancreatitis/metabolismo , Pancreatitis/patología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
20.
Mediators Inflamm ; 2021: 5587297, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34349610

RESUMEN

Acute pancreatitis is a common clinical condition with increasing the proinflammatory mediators, including interleukin-6 (IL-6). Obesity is a negative prognostic factor in acute pancreatitis. Obese patients with acute pancreatitis have a higher systemic inflammatory response rate. Levels of serum resistin, an adipocytokine secreted by fat tissues, increase with obesity. Cerulein, a cholecystokinin analog, induces calcium (Ca2+) overload, oxidative stress, and IL-6 expression in pancreatic acinar cells, which are hallmarks of acute pancreatitis. A recent study showed that resistin aggravates the expression of inflammatory cytokines in cerulein-stimulated pancreatic acinar cells. We aimed to investigate whether resistin amplifies cerulein-induced IL-6 expression and whether astaxanthin (ASX), an antioxidant carotenoid with anti-inflammatory properties, inhibits ceruelin/resistin-induced IL-6 expression in pancreatic acinar AR42J cells. We found that resistin enhanced intracellular Ca2+ levels, NADPH oxidase activity, intracellular reactive oxygen species (ROS) production, NF-κB activity, and IL-6 expression in cerulein-stimulated AR42J cells, which were inhibited by ASX in a dose-dependent manner. The calcium chelator BAPTA-AM inhibited cerulein/resistin-induced NADPH oxidase activation and ROS production. Antioxidant N-acetyl cysteine (NAC) and ML171, a specific NADPH oxidase 1 inhibitor, suppressed cerulein/resistin-induced ROS production, NF-κB activation, and IL-6 expression. In conclusion, ASX inhibits IL-6 expression, by reducing Ca2+ overload, NADPH oxidase-mediated ROS production, and NF-κB activity in cerulein/resistin-stimulated pancreatic acinar cells. Consumption of ASX-rich foods could be beneficial for preventing or delaying the incidence of obesity-associated acute pancreatitis.


Asunto(s)
Células Acinares/metabolismo , Ceruletida/química , Interleucina-6/metabolismo , Páncreas/metabolismo , Resistina/química , Células Acinares/efectos de los fármacos , Adipoquinas/metabolismo , Animales , Antiinflamatorios/química , Calcio/química , Calcio/metabolismo , Línea Celular , Quelantes/química , NADPH Oxidasas/metabolismo , Obesidad/metabolismo , Estrés Oxidativo , Páncreas/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno , Xantófilas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA