Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Phytother Res ; 29(10): 1634-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26179197

RESUMEN

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


Asunto(s)
Antiinflamatorios/farmacología , Ceruletida , Pancreatitis/tratamiento farmacológico , Triterpenos Pentacíclicos/farmacología , Extractos Vegetales , Enfermedad Aguda , Amilasas , Animales , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Inyecciones Intraperitoneales , Lipasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/efectos de los fármacos , Páncreas/efectos de los fármacos , Pancreatitis/inducido químicamente , Peroxidasa/metabolismo , Extractos Vegetales/farmacología , Sustancias Protectoras/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
2.
Dig Dis Sci ; 58(10): 2908-17, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23918150

RESUMEN

BACKGROUND/AIM: We have previously reported that bee venom (BV) has a protective role against acute pancreatitis (AP). However, the effects of apamin, the major compound of BV, on AP have not been determined. The aim of this study was to evaluate the effects of apamin on cerulein-induced AP. METHODS: AP was induced via intraperitoneal injection of supramaximal concentrations of the stable cholecystokinin analogue cerulein (50 µg/kg) every hour for 6 times. In the apamin treatment group, apamin was administered subcutaneously (10, 50, or 100 µg/kg) at both 18 and 1 h before the first cerulein injection. The mice were sacrificed at 6 h after the final cerulein injection. Blood samples were obtained to determine serum amylase and lipase levels, as well as cytokine production. The pancreas and lung were rapidly removed for morphologic and histological examination, myeloperoxidase (MPO) assay, and real-time reverse transcription-polymerase chain reaction. Furthermore, we isolated the pancreatic acinar cells to specify the role of apamin in AP. RESULTS: Pre-treatment with apamin inhibited histological damage, pancreatic weight/body weight ratio, serum level of amylase and lipase, MPO activity, and cytokine production. In addition, apamin treatment significantly inhibited cerulein-induced pancreatic acinar cell death. Furthermore, apamin treatment inhibited the cerulein-induced activation of c-Jun NH2-terminal kinases (JNK). CONCLUSIONS: These results could suggest that apamin could protect against AP by inhibition of JNK activation.


Asunto(s)
Apamina/farmacología , Apamina/uso terapéutico , Ceruletida/efectos adversos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Pancreatitis/inducido químicamente , Pancreatitis/prevención & control , Enfermedad Aguda , Animales , Apamina/administración & dosificación , Ceruletida/administración & dosificación , Colecistoquinina/análogos & derivados , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inyecciones Intraperitoneales , Inyecciones Subcutáneas , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Páncreas/patología
3.
Phytother Res ; 26(12): 1893-7, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22430952

RESUMEN

Piperine, one of the main components of Piper longum Linn. and P. nigrum Linn., is a plant alkaloid with a long history of medicinal use. Piperine has been shown to modulate the immune response, but the mechanism underlying this modulation remains unknown. Here, we examined the effects of piperine on lipopolysaccharide (LPS)-induced inflammatory responses in bone-marrow-derived dendritic cells (BMDCs). Piperine significantly inhibited the expression of major histocompatibility complex class II, CD40 and CD86 in BMDCs in a dose-dependent manner. Furthermore, piperine treatment led to an increase in fluorescein-isothiocyanate-dextran uptake in LPS-treated dendritic cells and inhibited the production of tumour necrosis factor alpha and interleukin (IL)-12, but not IL-6. The inhibitory effects of piperine were mediated via suppression of extracellular signal-regulated kinases and c-Jun N-terminal kinases activation, but not p38 or nuclear factor-κB activation. These findings provide insight into the immunopharmacological role of piperine.


Asunto(s)
Alcaloides/farmacología , Benzodioxoles/farmacología , Diferenciación Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Piperidinas/farmacología , Alcamidas Poliinsaturadas/farmacología , Animales , Células de la Médula Ósea/efectos de los fármacos , Inflamación/metabolismo , Interleucina-12/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos , Ratones , Fosforilación , Factor de Necrosis Tumoral alfa/metabolismo
4.
Int Immunopharmacol ; 69: 225-234, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30738992

RESUMEN

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


Asunto(s)
Quimiocina CXCL2/metabolismo , Hemo-Oxigenasa 1/metabolismo , Neutrófilos/fisiología , Páncreas/metabolismo , Pancreatitis/metabolismo , Enfermedad Aguda , Amilasas/sangre , Animales , Ceruletida/administración & dosificación , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Naftoles/metabolismo , Infiltración Neutrófila , Páncreas/patología , Pancreatitis/patología
5.
Int Immunopharmacol ; 69: 169-177, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30716587

RESUMEN

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


Asunto(s)
Antiinflamatorios/uso terapéutico , Benzofuranos/uso terapéutico , Inflamasomas/metabolismo , Inflamación/tratamiento farmacológico , Macrófagos/inmunología , Neutrófilos/inmunología , Pancreatitis/tratamiento farmacológico , Enfermedad Aguda , Animales , Movimiento Celular/efectos de los fármacos , Ceruletida/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
6.
Pancreas ; 46(8): 1046-1055, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28797013

RESUMEN

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


Asunto(s)
Berberina/farmacología , Lesión Pulmonar/prevención & control , Pulmón/efectos de los fármacos , Páncreas/efectos de los fármacos , Pancreatitis Aguda Necrotizante/prevención & control , Amilasas/sangre , Animales , Colina/aislamiento & purificación , Dieta/efectos adversos , Etionina/administración & dosificación , Femenino , Lipasa/sangre , Pulmón/metabolismo , Pulmón/patología , Lesión Pulmonar/mortalidad , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Páncreas/metabolismo , Páncreas/patología , Pancreatitis Aguda Necrotizante/etiología , Pancreatitis Aguda Necrotizante/mortalidad , Fitoterapia/métodos , Tasa de Supervivencia
7.
Mol Immunol ; 74: 27-38, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27148818

RESUMEN

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


Asunto(s)
Antiinflamatorios/farmacología , Berberina/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Pancreatitis Aguda Necrotizante/patología , Animales , Western Blotting , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Técnica del Anticuerpo Fluorescente , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Endogámicos C57BL
8.
Inflammation ; 39(1): 87-95, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26260258

RESUMEN

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


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Antiinflamatorios/farmacología , Túbulos Renales Colectores/inmunología , Pregnenodionas/farmacología , Receptor Toll-Like 4/inmunología , Animales , Línea Celular , Ciclooxigenasa 2/biosíntesis , Femenino , Inflamación/tratamiento farmacológico , Interleucina-6/biosíntesis , Túbulos Renales Colectores/citología , Lipopolisacáridos , Macrófagos Peritoneales/inmunología , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/biosíntesis
9.
Eur J Pharmacol ; 765: 541-50, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26407655

RESUMEN

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


Asunto(s)
Iridoides/uso terapéutico , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Pancreatitis/metabolismo , Pancreatitis/prevención & control , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Relación Dosis-Respuesta a Droga , Femenino , Iridoides/farmacología , Ratones , Ratones Endogámicos C57BL , Sustancias Protectoras/farmacología , Sustancias Protectoras/uso terapéutico
10.
Pancreas ; 44(1): 31-40, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25102438

RESUMEN

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


Asunto(s)
Antiinflamatorios/farmacología , Ceruletida , Lithospermum/química , Páncreas/efectos de los fármacos , Pancreatitis/prevención & control , Extractos Vegetales/farmacología , Enfermedad Aguda , Animales , Antiinflamatorios/aislamiento & purificación , Biomarcadores/sangre , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocinas/sangre , Citoprotección , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Activación Enzimática , Femenino , Proteína HMGB1/metabolismo , Mediadores de Inflamación/sangre , Ratones Endogámicos C57BL , Infiltración Neutrófila/efectos de los fármacos , Páncreas/metabolismo , Páncreas/patología , Pancreatitis/sangre , Pancreatitis/inducido químicamente , Pancreatitis/patología , Fitoterapia , Extractos Vegetales/aislamiento & purificación , Plantas Medicinales , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Int Immunopharmacol ; 29(2): 730-738, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26371857

RESUMEN

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


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Lipopolisacáridos/antagonistas & inhibidores , Naftoles/farmacología , Nardostachys/química , Choque Séptico/prevención & control , Animales , Citocinas/biosíntesis , Activación Enzimática/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/efectos de los fármacos , Naftoles/aislamiento & purificación , Choque Séptico/inducido químicamente , Choque Séptico/patología , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos
12.
Int Immunopharmacol ; 26(1): 194-202, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25843255

RESUMEN

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


Asunto(s)
Antiinflamatorios/uso terapéutico , Ceruletida/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Pancreatitis/tratamiento farmacológico , Pregnenodionas/uso terapéutico , Enfermedad Aguda , Animales , Antiinflamatorios/administración & dosificación , Western Blotting , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Inyecciones Intraperitoneales , Lipasa/sangre , Ratones Endogámicos C57BL , Páncreas/efectos de los fármacos , Páncreas/inmunología , Páncreas/patología , Pancreatitis/enzimología , Pancreatitis/inmunología , Pregnenodionas/administración & dosificación
13.
Eur J Pharmacol ; 737: 149-58, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24861022

RESUMEN

Acute pancreatitis (AP) is a complicated disease which is largely undiscovered. Fisetin, a natural flavonoid from fruits and vegetables, has been shown to have anti-inflammatory, antioxidant, and anti-cancer activities in various disease models. However, the effects of fisetin on AP have not been determined. Pre- and post- treatment of mice with fisetin reduced the severity of AP and pancreatitis-associated lung injury and inhibited several biochemical parameters (pancreatic weight to body weight ratio, amylase, lipase, and myeloperoxidase activity) and production of inflammatory cytokines. In pancreatic acinar cells, fisetin also inhibited cell death and production of inflammatory cytokines. In addition, fisetin inhibited activation of c-Jun NH2-terminal kinase (JNK) and nuclear factor (NF)-κB in vivo and in vitro. In conclusion, these results suggest that fisetin exhibits anti-inflammatory effect on AP and could be a beneficial agent in the treatment of AP and its pulmonary complications.


Asunto(s)
Ceruletida/efectos adversos , Regulación hacia Abajo/efectos de los fármacos , Flavonoides/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Pancreatitis/patología , Transducción de Señal/efectos de los fármacos , Células Acinares/efectos de los fármacos , Células Acinares/patología , Enfermedad Aguda , Administración Oral , Animales , Activación Enzimática/efectos de los fármacos , Femenino , Flavonoides/administración & dosificación , Flavonoides/uso terapéutico , Flavonoles , Inyecciones Intraperitoneales , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Páncreas/efectos de los fármacos , Páncreas/patología , Pancreatitis/inducido químicamente , Pancreatitis/tratamiento farmacológico , Pancreatitis/prevención & control
14.
Pancreas ; 43(1): 118-27, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24326366

RESUMEN

OBJECTIVE: The aim of this study was to evaluate the effects of Opuntia humifusa (OH) on cerulein-induced acute pancreatitis (AP). METHODS: Acute pancreatitis was induced via intraperitoneal injection of cholecystokinin analog cerulein (50 µg/kg). In the OH pretreatment group, OH was administered intraperitoneally (100, 250, or 500 mg/kg) 1 hour before first cerulein injection. In the posttreatment group, OH was administered intraperitoneally (500 mg/kg) 1 hour after the first cerulein injection. Furthermore, we isolated the pancreatic acinar cells using collagenase method, then investigated the acinar cell viability, cytokine productions, and the regulating mechanisms. RESULTS: The both pretreatment and posttreatment of OH treatment attenuated the severity of AP, as shown by the histology of the pancreas and lung, and inhibited neutrophil infiltration; serum amylase and lipase activities; proinflammatory cytokine expression such as interleukin 1, interleukin 6, and tumor necrosis factor α; and cell death including apoptosis and necrosis. Furthermore, OH inhibited the activation of c-Jun N-terminal kinases. CONCLUSIONS: These results suggest that OH reduces the severity of AP by inhibiting acinar cell death through c-Jun N-terminal kinases.


Asunto(s)
Opuntia/química , Páncreas/efectos de los fármacos , Pancreatitis/prevención & control , Extractos Vegetales/farmacología , Células Acinares/efectos de los fármacos , Células Acinares/metabolismo , Enfermedad Aguda , Amilasas/sangre , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ceruletida , Citocinas/genética , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Expresión Génica/efectos de los fármacos , Proteína HMGB1/metabolismo , Inyecciones Intraperitoneales , Lipasa/sangre , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Páncreas/metabolismo , Páncreas/patología , Pancreatitis/sangre , Pancreatitis/inducido químicamente , Extractos Vegetales/administración & dosificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
15.
Artículo en Inglés | MEDLINE | ID: mdl-24795771

RESUMEN

It has been previously shown that Nardostachys jatamansi (NJ) exhibits anti-inflammatory properties against lipopolysaccharide (LPS) challenges. However, the potency of NJ constituents against LPS-induced inflammatory responses has not been examined. In this present study, we determined which NJ extract fractions exhibit inhibitory effects against LPS-induced inflammatory responses. Among the NJ fractions, NJ-1, NJ-3, NJ-4, and NJ-6 inhibited LPS-induced production of NO. The NJ-3, NJ-4, and NJ-6 fractions also inhibited the production of cytokines, such as IL-1 ß , IL-6, and TNF- α . However, NJ-1, NJ-3, NJ-4, and NJ-6 showed differential inhibitory mechanisms against LPS-induced inflammatory responses. NJ-1, NJ-3, and NJ-4 inhibited LPS-induced activation of c-jun NH2-terminal kinase (JNK) and p38 but did not affect activation of extracellular signal-regulated kinase (ERK) or NF- κ B. On the other hand, NJ-6 inhibited activation of MAPKs and NF- κ B. In addition, in vivo experiments revealed that administration of NJ-1, NJ-3, NJ-4, and NJ-6 reduced LPS-induced endotoxin shock, with NJ-6 especially showing a marked protective effect. Taken together, these results provide the evidence for the potential of selective NJ fractions against LPS-induced inflammation. Thus, it will be advantageous to further isolate and determine single effective compounds from these potent fractions.

16.
World J Gastroenterol ; 19(10): 1551-62, 2013 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-23539679

RESUMEN

AIM: To evaluate the inhibitory effects of Scolopendra subspinipes mutilans (SSM) on cerulein-induced acute pancreatitis (AP) in a mouse model. METHODS: SSM water extract (0.1, 0.5, or 1 g/kg) was administrated intraperitoneally 1 h prior to the first injection of cerulein. Once AP developed, the stable cholecystokinin analogue, cerulein was injected hourly, over a 6 h period. Blood samples were taken 6 h later to determine serum amylase, lipase, and cytokine levels. The pancreas and lungs were rapidly removed for morphological examination, myeloperoxidase assay, and real-time reverse transcription polymerase chain reaction. To specify the role of SSM in pancreatitis, the pancreatic acinar cells were isolated using collagenase method. Then the cells were pre-treated with SSM, then stimulated with cerulein. The cell viability, cytokine productions and high-mobility group box protein-1 (HMGB-1) were measured. Furthermore, the regulating mechanisms of SSM action were evaluated. RESULTS: The administration of SSM significantly attenuated the severity of pancreatitis and pancreatitis associated lung injury, as was shown by the reduction in pancreatic edema, neutrophil infiltration, vacuolization and necrosis. SSM treatment also reduced pancreatic weight/body weight ratio, serum amylase, lipase and cytokine levels, and mRNA expression of multiple inflammatory mediators such as tumor necrosis factor-α and interleukin-1ß. In addition, treatment with SSM inhibited HMGB-1 expression in the pancreas during AP. In accordance with in vivo data, SSM inhibited the cerulein-induced acinar cell death, cytokine, and HMGB-1 release. SSM also inhibited the activation of c-Jun NH2-terminal kinase, p38 and nuclear factor (NF)-κB. CONCLUSION: These results suggest that SSM plays a protective role during the development of AP and pancreatitis associated lung injury via deactivating c-Jun NH2-terminal kinase, p38 and NF-κB.


Asunto(s)
Antiinflamatorios/farmacología , Venenos de Artrópodos/farmacología , Proteína HMGB1/antagonistas & inhibidores , Páncreas/efectos de los fármacos , Pancreatitis/prevención & control , Enfermedad Aguda , Lesión Pulmonar Aguda/sangre , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/prevención & control , Amilasas/sangre , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ceruletida , Citocinas/sangre , Modelos Animales de Enfermedad , Activación Enzimática , Proteína HMGB1/metabolismo , Mediadores de Inflamación/sangre , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipasa/sangre , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Páncreas/metabolismo , Páncreas/patología , Pancreatitis/sangre , Pancreatitis/inducido químicamente , Pancreatitis/genética , Pancreatitis/patología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Exp Ther Med ; 4(3): 533-537, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23181131

RESUMEN

Previously, we reported that Nardostachys jatamansi (NJ) attenuated cerulein-induced mild acute pancreatitis (AP). In the present study, we investigated the ability of NJ to ameliorate severe acute pancreatitis (SAP) induced by a choline-deficient diet supplemented with ethionine (CDE). An NJ extract was orally administered ad libitum via the water during administration of the CDE. After three days, the CDE was replaced with a normal diet. After four days of normal feeding the mice were sacrificed and the blood and pancreas were obtained for further investigation. NJ treatment reduced SAP-induced pancreatic damage, as shown by histology. NJ treatment also inhibited neutrophil infiltration into the pancreas. NJ also inhibited the secretion of digestive enzymes and cytokine production, and inhibited the activation of mitogen-activated protein kinases (MAPKs) in the SAP-challenged pancreas. These data suggest that NJ protects against pancreatic injury in CDE-induced SAP by deactivating MAPKs.

18.
BMB Rep ; 45(7): 402-7, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22831975

RESUMEN

Nardostachys jatamansi (NJ) belonging to the Valerianaceae family has been used as a remedy for gastrointestinal inflammatory diseases for decades. However, the potential for NJ to ameliorate alcoholic chronic pancreatitis (ACP) is unknown. The aim of this study was to examine the inhibitory effects of NJ on ACP. C57black/6 mice received ethanol injections intraperitoneally for 3 weeks against a background of cerulein-induced acute pancreatitis. During ACP, NJ was ad libitum administrated orally with water. After 3 weeks of treatment, the pancreas was harvested for histological examination. NJ treatment increased the pancreatic acinar cell survival (confirmed by amylase level testing) and reduced collagen deposition and pancreatic stellate cell (PSC) activation. In addition, NJ treatment reduced the activation but not death of PSC. In conclusion, our results suggest that NJ attenuated ACP through the inhibition of PSC activation.


Asunto(s)
Alcoholismo/tratamiento farmacológico , Caprifoliaceae/química , Pancreatitis Crónica/tratamiento farmacológico , Extractos Vegetales/uso terapéutico , Animales , Ratones , Ratones Endogámicos C57BL
19.
Life Sci ; 91(17-18): 866-71, 2012 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-22982349

RESUMEN

AIMS: Acute pancreatitis (AP) is a complicated inflammatory disease that has an unknown underlying pathogenesis. Because alpha-pinene can modulate inflammation, we examined whether alpha-pinene plays a role in AP. MAIN METHODS: Alpha-pinene was administered intraperitoneally 1h prior to the first injection of cerulein. Once AP developed, cerulein, a stable cholecystokinin analog, was injected hourly over a 6-h period. Blood samples were taken 6h later to determine serum amylase and lipase levels. The pancreas and lungs were rapidly removed for morphological examination, myeloperoxidase assay, and real-time reverse transcription polymerase chain reaction. We also isolated the pancreatic acinar cells using a collagenase solution. Cell viability, and cytokine productions were measured in pancreatic acini. KEY FINDINGS: Intraperitoneal administration of alpha-pinene reduced the pancreatic weight (PW) to body weight (BW) ratio and the serum levels of amylase and lipase. Alpha-pinene treatment also reduced histological damage and myeloperoxidase activity in the pancreas and lungs. Furthermore, alpha-pinene pretreatment reduced the production of pancreatic tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6 during cerulein-induced AP. In vitro, alpha-pinene inhibited cerulein-induced cell death and cytokine production in isolated cerulein-treated pancreatic acinar cells. SIGNIFICANCE: These findings suggest that alpha-pinene has an anti-inflammatory effect during cerulein-induced AP.


Asunto(s)
Factores Inmunológicos/uso terapéutico , Monoterpenos/uso terapéutico , Páncreas/efectos de los fármacos , Páncreas/patología , Pancreatitis/inducido químicamente , Pancreatitis/prevención & control , Enfermedad Aguda , Amilasas/sangre , Animales , Monoterpenos Bicíclicos , Peso Corporal/efectos de los fármacos , Células Cultivadas , Ceruletida , Femenino , Factores Inmunológicos/administración & dosificación , Inyecciones Intraperitoneales , Lipasa/sangre , Pulmón/efectos de los fármacos , Pulmón/enzimología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Monoterpenos/administración & dosificación , Páncreas/enzimología , Pancreatitis/enzimología , Pancreatitis/patología , Peroxidasa/inmunología
20.
World J Gastroenterol ; 18(25): 3223-34, 2012 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-22783046

RESUMEN

AIM: To determine if the fraction of Nardostachys jatamansi (NJ) has the potential to ameliorate the severity of acute pancreatitis (AP). METHODS: Mice were administered the biologically active fraction of NJ, i.e., the 4th fraction (NJ4), intraperitoneally, and then injected with the stable cholecystokinin analogue cerulein hourly for 6 h. Six hours after the last cerulein injection, the pancreas, lung, and blood were harvested for morphological examination, measurement of cytokine expression, and examination of neutrophil infiltration. RESULTS: NJ4 administration attenuated the severity of AP and lung injury associated with AP. It also reduced cytokine production and neutrophil infiltration and resulted in the in vivo up-regulation of heme oxygenase-1 (HO-1). Furthermore, NJ4 and its biologically active fraction, NJ4-2 inhibited the cerulein-induced death of acinar cells by inducing HO-1 in isolated pancreatic acinar cells. CONCLUSION: These results suggest that NJ4 may be a candidate fraction offering protection in AP and NJ4 might ameliorate the severity of pancreatitis by inducing HO-1 expression.


Asunto(s)
Ceruletida , Nardostachys , Páncreas/efectos de los fármacos , Pancreatitis/prevención & control , Extractos Vegetales/farmacología , Enfermedad Aguda , Animales , Muerte Celular/efectos de los fármacos , Citocinas/metabolismo , Citoprotección , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Enzimas/sangre , Femenino , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Mediadores de Inflamación/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Nardostachys/química , Infiltración Neutrófila/efectos de los fármacos , Páncreas/metabolismo , Páncreas/patología , Pancreatitis/inducido químicamente , Pancreatitis/genética , Pancreatitis/metabolismo , Pancreatitis/patología , Raíces de Plantas , Índice de Severidad de la Enfermedad , Factores de Tiempo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA