Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell Death Dis ; 15(9): 643, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39227564

RESUMO

This study investigates the potential anti-colorectal cancer (CRC) activity of IMT1, a novel specific inhibitor of mitochondrial RNA polymerase (POLRMT). Single-cell RNA sequencing data reveal that POLRMT is overexpressed in CRC cells. Additionally, elevated POLRMT expression was observed in local CRC tissues and cells, while its expression remained relatively low in colon epithelial tissues and cells. IMT1 significantly inhibited colony formation, cell viability, proliferation, cell cycle progression, and migration in both primary and immortalized CRC cells. Furthermore, IMT1 induced apoptosis and cell death in CRC cells. The inhibition of POLRMT by IMT1 disrupted mitochondrial functions in CRC cells, leading to mitochondrial depolarization, oxidative damage, and decreased ATP levels. Using targeted shRNA to silence POLRMT closely mirrored the effects of IMT1, showing robust anti-CRC cell activity. Crucially, the efficacy of IMT1 was diminished in CRC cells with silenced POLRMT. Contrarily, boosting POLRMT expression externally by a lentiviral construct promoted the proliferation and migration of CRC cells. Importantly, treatment with IMT1 or silencing POLRMT in primary colon cancer cells decreased the phosphorylation of Akt1-S6K1, whereas overexpression of POLRMT had the opposite effect. In nude mice, orally administering IMT1 potently restrained primary colon cancer xenograft growth. IMT1 suppressed POLRMT activity, disrupted mitochondrial function, hindered Akt-mTOR activation, and prompted apoptosis within the xenograft tissues. In addition, IMT1 administration suppressed lung metastasis of primary colon cancer cells in nude mice. These combined results highlight the robust anti-CRC activity of IMT1 by specifically targeting POLRMT.


Assuntos
Apoptose , Proliferação de Células , Neoplasias Colorretais , Camundongos Nus , Humanos , Neoplasias Colorretais/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Animais , Proliferação de Células/efeitos dos fármacos , Camundongos , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , RNA Polimerases Dirigidas por DNA/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Camundongos Endogâmicos BALB C , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Masculino
2.
Pancreatology ; 24(6): 863-869, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39174438

RESUMO

BACKGROUND: Pseudocyst formation is common in many patients with acute pancreatitis during follow-up. Many risk factors have been proposed to be associated with the development of PP, but the predictive factors are still underexplored. The focus of this study was to investigate whether early laboratory indicators could effectively predict the occurrence of PP. METHODS: 2811 AP patients hospitalized in the Second Affiliated Hospital of Soochow University between November 2008 and September 2020 were retrospectively studied. Univariate and multivariate analyses were used to screen the risk variables. The nomograms of those risk factors were validated and evaluated by logistic analysis. RESULTS: AP patients had a 6.1 % (172/2811) incidence of PP. In a univariate analysis, the development of PP was correlated with serum lactate dehydrogenase (LDH), albumin (ALB), calcium (Ca), hemoglobin (Hb), organ dysfunction, CT severity index (CTSI), etiology, age, etc. Further logistic regression analysis showed that the risk factors were different between hyperlipidemic pancreatitis patients (LDH, ALB and Ca) and non-hyperlipidemic pancreatitis patients (LDH, Hb, ALB and Ca). A nomogram based on the identified risk factors was developed. Our model showed good discrimination ability, with a boostrap - corrected C index of 0.905 (95 % CI = 0.875-0.935), and had well-fitted calibration curves. The area under the curve (AUC) of the nomogram were 0.905 (95 % CI = 0.875-0.935) and 0.933 (95 % CI = 0.890-0.975) in the training and validation groups, respectively. The results of DCA indicated that the nomogram may have clinic usefulness. CONCLUSIONS: The nomogram that incorporates early laboratory data (LDH, Hb, ALB, and Ca) in AP patients is able to predict the incidence of PP with greater accuracy than the CTSI and AP severity.


Assuntos
Nomogramas , Pseudocisto Pancreático , Pancreatite , Humanos , Feminino , Masculino , Pessoa de Meia-Idade , Pancreatite/complicações , Pancreatite/etiologia , Fatores de Risco , Pseudocisto Pancreático/diagnóstico por imagem , Pseudocisto Pancreático/complicações , Estudos Retrospectivos , Adulto , Idoso , Incidência , Doença Aguda
4.
Rev Esp Enferm Dig ; 112(12): 893-897, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33118356

RESUMO

INTRODUCTION: both percutaneous transhepatic cholangiography and drainage (PTCD) and endoscopic retrograde cholangiopancreatography (ERCP) with SEMS implantation have been used for unresectable hilar cholangiocarcinoma (HC) in the clinic for many years. However, which one is preferred is still unknown. OBJECTIVE: to study the effects of biliary drainage of self-expanding metal stents (SEMS) implantation under PTCD or ERCP to treat HC. METHODS: the clinical data of 82 patients with HC from January 2006 to January 2015 were recorded retrospectively. Patients were treated with biliary implantation of self-expanding metal stents (SEMS) under PTCD (PTCD group, 40 patients) or ERCP (ERCP group, 42 patients). Clinical data, including total bilirubin concentrations, complications and survival time were analyzed. RESULTS: the remission of jaundice was similar in both groups (p > 0.05). The median survival time of the ERCP group and PTCD group were 237 d and 252 d respectively, with no significant differences (p > 0.05). The biliary infection rates under ERCP and PTCD procedure were 52.4 % and 20.0 % respectively, with a significant statistical difference (p < 0.05). For those HC patients of Bismuth III/IV, the infection rates under ERCP and PTCD procedure were 58.3 % and 14.3 %, respectively (p < 0.05). CONCLUSIONS: both PTCD and ERCP with SEMS implantation were effective to prolong the survival time of HC patients. The biliary infection rates were higher in the ERCP group, especially for Bismuth III/IV HC patients.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Tumor de Klatskin , Neoplasias dos Ductos Biliares/diagnóstico por imagem , Neoplasias dos Ductos Biliares/cirurgia , Ductos Biliares Intra-Hepáticos , Colangiocarcinoma/diagnóstico por imagem , Colangiocarcinoma/terapia , Colangiografia , Colangiopancreatografia Retrógrada Endoscópica , Drenagem , Humanos , Estudos Retrospectivos , Stents
5.
Dig Dis Sci ; 64(10): 2955-2964, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31165380

RESUMO

BACKGROUND: There are many studies on submucosal injection materials, but their clinical use is restricted for various reasons. The objective of this study was to compare the feasibility and safety of injected EndoClot®SIS polysaccharide as a submucosal injection material (SFC) in ESD in the pig stomach to that of injected sigMAVisc™ or Eleview™. METHODS: Four pig stomachs were used for the ex vivo study. Eighteen pigs were used for the in vivo study. In the ex vivo study, four injections were made in the gastric submucosa to induce submucosal uplift and extend its duration. Tissue change was observed. The in vivo study was performed in 2 steps. First, 3 injections were made in the esophageal mucosa to induce submucosal uplift and extend its duration. Histological change was observed. Second, ESD was performed in the stomach by injecting EndoClot®SIS polysaccharide, sigMAVisc™, or Eleview™ (each, n = 6) as an SFC. The effects of these agents on wound healing were examined. We evaluated the efficacy and safety of endoscopic surgery after EndoClot®SIS polysaccharide injection. RESULTS: EndoClot®SIS polysaccharide produced a longer-lasting elevation with clearer margins than was achieved by sigMAVisc™, Eleview™, or 0.9% NaCl and thereby enabled precise ESD without complications, such as bleeding and perforation. No obvious histopathological damage was observed at the injection site on endoscopy and histology. CONCLUSION: Submucosally injected EndoClot®SIS polysaccharide increased the effective separation of the mucosa and submucosa and reduced surgical complications. Hence, EndoClot®SIS polysaccharide injection is a safe and effective submucosal injection material.


Assuntos
Ressecção Endoscópica de Mucosa , Mucosa Gástrica , Complicações Intraoperatórias/prevenção & controle , Polissacarídeos/farmacologia , Amido/análogos & derivados , Animais , Ressecção Endoscópica de Mucosa/efeitos adversos , Ressecção Endoscópica de Mucosa/métodos , Excipientes/farmacologia , Mucosa Gástrica/patologia , Mucosa Gástrica/cirurgia , Injeções/métodos , Masculino , Modelos Animais , Cuidados Pré-Operatórios/métodos , Amido/farmacologia , Suínos , Porco Miniatura
6.
J BUON ; 24(1): 77-83, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30941954

RESUMO

PURPOSE: To analyze relevant factors for pathological complete remission (pCR) after neoadjuvant therapy for locally advanced rectal cancer. METHODS: The clinical data of 531 patients with the American Joint Committee on Cancer (AJCC) stage II or III rectal cancer from January 2014 to December 2017 were retrospectively analyzed. Among these patients, 100 (18.83%) patients achieved pCR. Univariate and multivariate logistic regression analyses were applied to analyze the predictive factors for pCR after neoadjuvant therapy. RESULTS: According to univariate analysis, carcinoembryonic antigen (CEA) before chemo-radiotherapy (CRT) (p=0.021), tumor (T) stage before CRT (p=0.002), interval between the end of CRT and surgery (p<0.001) and maximum depth of tumor invasion before CRT (p=0.039) influenced significantly pCR. Multivariate analysis manifested that the CEA level before CRT [p=0.037, odds ratio (OR) =0.435] and the interval between the end of CRT and surgery (p=0.004, OR=2.864) were significant predictive factors for pCR. Stratified analysis showed that low-level CEA before CRT (p=0.029) affected pCR only in non-smoking group. CONCLUSIONS: pCR can be observed in some patients with locally advanced rectal cancer after neoadjuvant therapy. Low-level CEA before CRT and long interval between CRT and surgery are predictive factors for pCR of preoperative neoadjuvant therapy for locally advanced rectal cancer, but low-level CEA is effective in predicting pCR in non-smokers only.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais/metabolismo , Antígeno Carcinoembrionário/metabolismo , Quimioterapia Adjuvante/mortalidade , Terapia Neoadjuvante/mortalidade , Neoplasias Retais/patologia , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Retais/tratamento farmacológico , Neoplasias Retais/metabolismo , Indução de Remissão , Estudos Retrospectivos , Taxa de Sobrevida , Resultado do Tratamento
7.
Technol Cancer Res Treat ; 16(2): 141-149, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-26858085

RESUMO

Gastric cancer is a malignancy with high incidence and the second leading cause of cancer death worldwide. Development of efficient therapies against gastric cancer is urgent. Until now, the mechanisms of gastric cancer genesis remain elusive. The KDM5C is a histone demethylase that promotes cancer cell growth and is enriched in drug-resistant cancer cells. But the pathogenic breadth and mechanistic aspects of this effect relative to gastric cancer have not been defined. In present study, we found that KDM5C was overexpressed in gastric cancer cell lines and gastric cancer tissues but not in normal gastric tissues. The proliferation and invasive potential of gastric cancer cells was significantly increased by ectopic expression of KDM5C. Contrarily, RNA interference targeting KDM5C in gastric cancer cells significantly decreased the proliferation and invasive potential of cells. Moreover, we also found that the expression of p53 was modulated by KDM5C. Cells with overexpression of KDM5C exhibited greatly decreased p53 expression, whereas silencing of KDM5C expression dramatically increased p53 expression at both the messenger RNA and protein levels. Inhibition of p53 by small-interfering RNA reversed the shKDM5C-induced proliferation and invasion. Our results collectively suggested that KDM5C played a role in gastric cancer cells proliferation and invasion, which may be partly associated with the p53 expression.


Assuntos
Regulação Neoplásica da Expressão Gênica , Histona Desmetilases/genética , Neoplasias Gástricas/genética , Proteína Supressora de Tumor p53/genética , Biomarcadores , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Histona Desmetilases/metabolismo , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia
8.
Pancreas ; 45(9): 1282-1293, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27776048

RESUMO

OBJECTIVES: Mesenchymal stem cells (MSCs) have shown an obvious protective effect on acute pancreatitis (AP). The purpose of the present study was to analyze the effect of bone marrow MSC-derived microvesicles (bmMSC-MVs) on AP and explore the underlying mechanisms. METHODS: Bone marrow MSCs and bmMSC-MVs were isolated from Sprague-Dawley rats. Cerulein-induced mild AP (MAP) and sodium taurocholate-induced severe AP (SAP) were used as AP models in vivo and in vitro. Pancreatic injury was evaluated by measuring serum levels of amylase, lipase, chemokines, and interleukins, and by pancreatic histology, reverse transcription-polymerase chain reaction, Western blotting, and immunohistochemistry. The effects of bmMSC-MVs on the survival rates of pancreatic acinar cells in vitro were also assessed. RESULTS: Bone marrow MSC-MVs attenuated acute pancreatic injury in MAP and SAP by regulating IL-1α, IL-6, and TNF-α, and dramatically attenuated the nuclear translocation of NF-κB p65 in MAP and SAP. Bone marrow MSC-MVs improved the survival rates of pancreatic acinar cells in MAP and SAP models in vitro. CONCLUSIONS: Bone marrow MSC-MVs played a protective role in AP by reducing the levels of proinflammatory cytokines and regulating the nuclear translocation of NF-κB p65. Bone marrow MSC-MVs could be developed as a strategy for the clinical treatment of SAP.


Assuntos
Transplante de Células-Tronco Mesenquimais , Doença Aguda , Animais , Células da Medula Óssea , Modelos Animais de Doenças , Pancreatite , Ratos , Ratos Sprague-Dawley
9.
Medicine (Baltimore) ; 95(26): e3603, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27367977

RESUMO

The goal of this study is to evaluate how to predict high-risk nonvariceal upper gastrointestinal bleeding (NVUGIB) pre-endoscopically. A total of 569 NVUGIB patients between Match 2011 and January 2015 were retrospectively studied. The clinical characteristics and laboratory data were statistically analyzed. The severity of NVUGIB was based on high-risk NVUGIB (Forrest I-IIb), and low-risk NVUGIB (Forrest IIc and III). By logistic regression and receiver-operating characteristic curve, simple risk score systems were derived which predicted patients' risks of potentially needing endoscopic intervention to control bleeding. Risk score systems combined of patients' serum hemoglobin (Hb) ≤75 g/L, red hematemesis, red stool, shock, and blood urine nitrogen ≥8.5 mmol/L within 24 hours after admission were derived. As for each one of these clinical signs, the relatively high specificity was 97.9% for shock, 96.4% for red stool, 85.5% for red hematemesis, 76.7% for Hb ≤75 g/L, and the sensitivity was 50.8% for red hematemesis, 47.5% for Hb ≤75 g/L, 14.2% for red stool, and 10.9% for shock. When these 5 clinical signs were presented as a risk score system, the highest area of receiver-operating characteristic curve was 0.746, with sensitivity 0.675 and specificity 0.733, which discriminated well with high-risk NVUGIB. These simple risk factors identified patients with high-risk NVUGIB of needing treatment to manage their bleeding pre-endoscopically. Further validation in the clinic was required.


Assuntos
Endoscopia Gastrointestinal , Hemorragia Gastrointestinal/diagnóstico , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Emergências , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Adulto Jovem
10.
Am J Chin Med ; 43(6): 1117-35, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26364660

RESUMO

Rosmarinic Acid (RA), a caffeic acid ester, has been shown to exert anti-inflammation, anti-oxidant and antiallergic effects. Our study aimed to investigate the effect of RA in sodium taurocholate ( NaTC )-induced acute pancreatitis, both in vivo and in vitro. In vivo, RA (50 mg/kg) was administered intraperitoneally 2 h before sodium taurocholate injection. Rats were sacrificed 12 h, 24 h or 48 h after sodium taurocholate injection. Pretreatment with RA significantly ameliorated pancreas histopathological changes, decreased amylase and lipase activities in serum, lowered myeloperoxidase activity in the pancreas, reduced systematic and pancreatic interleukin-1 ß (IL-1ß), IL-6, and tumor necrosis factor-α (TNF-α) levels, and inhibited NF-κB translocation in pancreas. In vitro, pretreating the fresh rat pancreatic acinar cells with 80 µ mol/L RA 2 h before 3750 nmol/L sodium taurocholate or 10 ng/L TNF-α administration significantly attenuated the reduction of isolated pancreatic acinar cell viability and inhibited the nuclear activation and translocation of NF-κB. Based on our findings, RA appears to attenuate damage in sodium taurocholate-induced acute pancreatitis and reduce the release of inflammatory cytokines by inhibiting the activation of NF-κB. These findings might provide a basis for investigating the therapeutic role of RA in managing acute pancreatits.


Assuntos
Cinamatos/administração & dosagem , Depsídeos/administração & dosagem , Pancreatite/tratamento farmacológico , Extratos Vegetais/administração & dosagem , Fator de Necrose Tumoral alfa/imunologia , Animais , Humanos , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Masculino , NF-kappa B/genética , NF-kappa B/imunologia , Pâncreas/efeitos dos fármacos , Pâncreas/imunologia , Pancreatite/induzido quimicamente , Pancreatite/genética , Pancreatite/imunologia , Ratos , Ratos Sprague-Dawley , Ácido Taurocólico/efeitos adversos , Fator de Necrose Tumoral alfa/genética , Ácido Rosmarínico
11.
Int J Clin Exp Pathol ; 8(5): 4457-68, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26191136

RESUMO

OBJECTIVES: Mesenchymal stem cells (MSCs) have shown an obvious protective effect on systemic inflammation. The purpose of this study is to assess the effect and possible mechanism of bone marrow MSCs (bmMSCs) on acute pancreatitis (AP). METHODS: BmMSCs of SD rats were isolated and cultured in vitro. L-Arginine-induced acute pancreatitis was used as AP model in vivo. Pancreatic injury was assessed by serum amylase, lipase, cytokines and pancreatic histology. RT-PCR was applied to investigate mRNA expression of pancreas tissue. Western-blot and immunohistochemistry (IHC) were applied to test the role of NF-κB p65 signaling pathway. Tracking and Positioning of CM-Dil labeled bmMSCs in vivo was further studied. RESULTS: Treatment with bmMSCs attenuated acute pancreatic injury and AP-associated lung injury obviously, with decreased serum IL-1ß, IL-6, TNF-α, down-regulated expressions of IL-1α, IL-6, TNFα in pancreas tissue and reduced nuclear translocation of NF-κB p65 in AP. Localization of bmMSCs in vivo was due to being passively trapped in related organs, but not actively homing to inflammatory sites of pancreas during the early phase of AP. CONCLUSIONS: Taken together, the results showed that bmMSCs played a protective role in AP in many aspects, which might protect against experimental pancreatitis partly by regulating release of inflammatory cytokines by an exocrine secretion.


Assuntos
Células da Medula Óssea , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Pancreatite/patologia , Animais , Arginina/toxicidade , Western Blotting , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Imuno-Histoquímica , Pancreatite/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real
12.
Oncol Rep ; 34(1): 350-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25955689

RESUMO

The expression of CD9 has been shown to be inversely associated with pancreatic cancer metastasis but the underlying mechanism remains incompletely understood. Using the two closely associated pancreatic cancer cell lines, PaTu-8898s and PaTu-8898t, which are metastatic and non-metastatic, respectively, we showed that the PaTu-8988s cells expressed a lower level of CD9 but had higher proliferation and migration rates than the PaTu-8898t cells. An inverse correlation between CD9 expression and the cell surface level of epidermal growth factor receptor (EGFR) was observed in both cell lines. In the PaTu-8898s cells, overexpression of CD9 decreased the cell surface expression of EGFR, associated with increased expression of dynamin-2, whereas in the PaTu-8898t cells, knockdown of CD9 with RNA interference (RNAi) increased the cell surface expression of EGFR, associated with decreased expression of dynamin-2. However, the total EGFR level did not change by manipulation of CD9 expression, suggesting that CD9 plays a role in EGFR endocytosis. Furthermore, in the PaTu-8898ts cells, CD9 overexpression decreased the cell proliferation and migration, which were reversed by EGFR overexpression, whereas in the PaTu-8898t cells, CD9 knockdown enhanced the cell proliferation and migration which were blocked by EGFR RNAi both in vitro and in vivo. Thus, in pancreatic cancer cells, downregulation of CD9 may play a role in cancer growth and metastasis through, at least in part, enhancing cell surface expression of EGFR.


Assuntos
Receptores ErbB/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Tetraspanina 29/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação para Baixo , Feminino , Humanos , Camundongos , Metástase Neoplásica , Transplante de Neoplasias , Regulação para Cima
13.
Int J Clin Exp Pathol ; 7(5): 2133-42, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24966921

RESUMO

Diosmetin (3', 5, 7-trihydroxy-4'-methoxyflavone), the aglycone part of the flavonoid glycosides diosmin occurs naturally in citrus fruit, was considered to exhibit anti-inflammatory and antioxidant properties. Our study aimed to investigate the effect of diosmetin in a murine model of cerulein-induced acute pancreatitis (AP). Experimental AP was induced in mice by seven intraperitoneal injection of cerulein (50 ug/kg) at hourly intervals. Diosmetin (100 mg/kg) or vehicle was pretreated 2 h before the first cerulein injection. After 6 h, 9 h, 12 h of the first cerulein injection, the severity of acute pancreatitis was evaluated biochemically and morphologically. Pretreatment with diosmetin significantly reduced serum levels of amylase and lipase; the histological injury; the secretion of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6; myeloperoxidase (MPO) activity, trypsinogen activation peptide (TAP) level, the expression of inducible nitric oxide synthase (iNOS); and the nuclear factor (NF)-κB activation in cerulein-induced AP. This study showed that administration of diosmetin demonstrated a beneficial effect on the course of cerulein-induced AP in mice. Therefore, diosmetin may become a new therapeutic agent in future clinical trials for treatment of AP.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Ceruletídeo , Flavonoides/farmacologia , Mediadores da Inflamação/antagonistas & inibidores , NF-kappa B/antagonistas & inibidores , Pâncreas/efeitos dos fármacos , Pancreatite/prevenção & controle , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Doença Aguda , Animais , Citoproteção , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Masculino , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Pâncreas/metabolismo , Pâncreas/patologia , Pancreatite/induzido quimicamente , Pancreatite/metabolismo , Pancreatite/patologia , Índice de Gravidade de Doença , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
14.
Zhonghua Zhong Liu Za Zhi ; 34(2): 96-9, 2012 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-22780925

RESUMO

OBJECTIVE: To study the effect of the sphingosine kinase 1 (SphK1) inhibitor N,N-dimethylsphingosine (DMS) in combination with chemotherapeutic drugs (DDP, 5-Fu, MMC) on the proliferation of gastric cancer cells (SGC7901) in vitro, and to evaluate whether SphK1 inhibitors could be used as synergetic agents in chemotherapy. METHODS: SGC7901 cells were incubated in vitro with DMS (1 micromol/L) and 5-Fu, DDP, MMC at different concentrations in combination or separately for 24 h. The effects on the growth and survival of SGC7901 cells were determined by MTT assay. The inhibition rates were assessed by response surface analysis and the interactive relationships between the combined drugs were evaluated on the basis of positive/negative values of the cross product coefficients in the response surface equation. RESULTS: The growth inhibition rate of the gastric cancer cells by treatment with DMS (1 micromol/L) was (10.23 +/- 0.74)%. The growth inhibition rates of the gastric cancer cells treated with 5-Fu (1, 5 and 25 microg/ml) for 24 h were (9.95 +/- 3.24)%, (21.04 +/- 2.19)%, and (45.49 +/- 3.60)%, respectively. The growth inhibition rates of the gastric cancer cells treated with DDP (0.5, 2.5 and 12.5 microg/ml) for 24 h were (9.38 +/- 0.79)%, (19.61 +/- 0.90)%, and (29.83 +/- 0.54)%, respectively. The growth inhibition rates of the gastric cancer cells treated with MMC (0.1, 0.5 and 2.5 microg/ml) for 24 h were (15.35 +/- 0.77)%, (24.72 +/- 0.83)%, and (30.68 +/- 0.28)%, respectively. There were significant differences among the inhibition rates caused by different concentrations of the drugs (P < 0.05). When 1 micromol/L DMS was used in combination with 5-Fu (1, 5, and 25 microg/ml) for 24 h, the growth inhibition rates of the cancer cells were (16.76 +/- 0.41)%, (27.28 +/- 0.29)% and (52.56 +/- 3.60)%, respectively. When 1 micromol/L DMS was used in combination with DDP (0.5, 2.5, and 12.5 microg/ml) for 24 h, the growth inhibition rates of the cancer cells were (15.35 +/- 0.86)%, (25.57 +/- 0.27)%, (36.37 +/- 0.51)%, respectively. When 1 micromol/L DMS was used in combination with MMC (0.1, 0.5, and 2.5 microg/ml) for 24 h, the growth inhibition rates of the cancer cells were (21.02 +/- 0.28)%, (32.10 +/- 0.27)%, (36.36 +/- 0.28)%, respectively. There were also significant differences among the growth inhibition rates caused by different concentrations of the drugs alone and in combination groups (P < 0.05). CONCLUSIONS: DMS can suppress the proliferation of SGC7901 cells in vitro, and there are evident synergetic effects when it is used in combination with chemotherapeutic drugs. The results of this study indicate that SphK1 inhibitors may become novel and promising chemotherapeutic sensitizers.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Esfingosina/análogos & derivados , Neoplasias Gástricas/patologia , Antibióticos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Fluoruracila/farmacologia , Humanos , Mitomicina/farmacologia , Esfingosina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA