Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Dis Colon Rectum ; 56(8): 945-51, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23838862

RESUMO

BACKGROUND: Laparoscopic rectal cancer surgery has limited short-term benefits in comparison with open surgery. Long-term measures of recovery are needed. OBJECTIVE: The aim of this study was to assess the impact of surgical approach (laparoscopic vs open) for the treatment of rectal cancer on the time to postoperative chemotherapy. DESIGN: This study is a retrospective review of 150 patients who underwent low anterior resection and received postoperative chemotherapy between 2005 and 2011. SETTINGS: This study was conducted at a tertiary care hospital. PATIENTS: One hundred fifty patients who had stage II or III rectal cancer who underwent low anterior resection were selected. All patients received postoperative chemotherapy, the timing of which was at the discretion of the oncologist. MAIN OUTCOME MEASURES: Patient demographics, clinicopathologic variables, and time to postoperative chemotherapy were compared. Multivariate analysis was performed to identify variables affecting the time to postoperative chemotherapy. RESULTS: There were no differences in clinicopathologic variables between cohorts including age, BMI, sex, ASA score, diverting ileostomy, preoperative radiotherapy, or pathologic stage. Univariate analysis demonstrated differences in intraoperative blood loss (300 vs 448 mL, p < 0.01), length of stay (7.6 vs 8.9 days, p < 0.05), wound infection (12.0 vs 24.0%, p < 0.05), and tumor location (8.0 vs 6.9 cm, p < 0.05) for laparoscopic vs open patients. There were more complications in the open vs laparoscopic group (47 vs 24, p < 0.001); however, the percentage of patients experiencing complications in the open vs laparoscopic cohorts did not reach statistical significance (32.0 vs 18.7%, p = 0.09). A decrease in mean time to postoperative chemotherapy was found for patients undergoing laparoscopic vs open surgery (50.1 vs 75.2 days, p < 0.0001). Multivariate analysis demonstrated that the approach of surgery was an independent predictor of time to postoperative chemotherapy (p < 0.01). LIMITATIONS: This study was limited by its retrospective design and selection bias. CONCLUSIONS: In selected patients, patients undergoing laparoscopic rectal cancer surgery receive postoperative chemotherapy 25 days earlier than patients undergoing open surgery. Time to postoperative chemotherapy serves as an outcome measure for improved recovery in laparoscopic rectal cancer surgery.


Assuntos
Antineoplásicos/uso terapêutico , Colectomia/métodos , Laparoscopia , Cuidados Pós-Operatórios/métodos , Neoplasias Retais/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/administração & dosagem , Esquema de Medicação , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Retais/diagnóstico , Neoplasias Retais/cirurgia , Estudos Retrospectivos , Fatores de Tempo , Resultado do Tratamento
2.
Proc Natl Acad Sci U S A ; 107(14): 6430-5, 2010 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-20308560

RESUMO

T-regulatory cells (Treg) and mast cells (MC) are abundant in colorectal cancer (CRC) tumors. Interaction between the two is known to promote immune suppression or loss of Treg functions and autoimmunity. Here, we demonstrate that in both human CRC and murine polyposis the outcome of this interaction is the generation of potently immune suppressive but proinflammatory Treg (DeltaTreg). These Treg shut down IL10, gain potential to express IL17, and switch from suppressing to promoting MC expansion and degranulation. This change is also brought about by direct coculture of MC and Treg, or culture of Treg in medium containing IL6 and IL2. IL6 deficiency in the bone marrow of mice susceptible to polyposis eliminated IL17 production by the polyp infiltrating Treg, but did not significantly affect the growth of polyps or the generation of proinflammatory Treg. IL6-deficient MC could generate proinflammatory Treg. Thus, MC induce Treg to switch function and escalate inflammation in CRC without losing T-cell-suppressive properties. IL6 and IL17 are not needed in this process.


Assuntos
Neoplasias Colorretais/imunologia , Mastócitos/imunologia , Linfócitos T Reguladores/imunologia , Animais , Técnicas de Cocultura , Humanos , Interleucinas/deficiência , Interleucinas/imunologia , Camundongos , Camundongos Knockout , Linfócitos T Auxiliares-Indutores/imunologia
3.
Cancer Metastasis Rev ; 30(1): 45-60, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21287360

RESUMO

Mast cells (MC) are a bone marrow-derived, long-lived, heterogeneous cellular population that function both as positive and negative regulators of immune responses. They are arguably the most productive chemical factory in the body and influence other cells through both soluble mediators and cell-to-cell interaction. MC are commonly seen in various tumors and have been attributed alternatively with tumor rejection or tumor promotion. Tumor-infiltrating MC are derived both from sentinel and recruited progenitor cells. MC can directly influence tumor cell proliferation and invasion but also help tumors indirectly by organizing its microenvironment and modulating immune responses to tumor cells. Best known for orchestrating inflammation and angiogenesis, the role of MC in shaping adaptive immune responses has become a focus of recent investigations. MC mobilize T cells and antigen-presenting dendritic cells. They function as intermediaries in regulatory T cells (Treg)-induced tolerance but can also modify or reverse Treg-suppressive properties. The central role of MC in the control of innate and adaptive immunity endows them with the ability to tune the nature of host responses to cancer and ultimately influence the outcome of disease and fate of the cancer patient.


Assuntos
Mastócitos/patologia , Neoplasias/patologia , Animais , Apresentação de Antígeno/imunologia , Modelos Animais de Doenças , Humanos , Imunidade Celular , Mastócitos/imunologia , Camundongos , Invasividade Neoplásica/imunologia , Neoplasias/diagnóstico , Neoplasias/imunologia , Prognóstico , Linfócitos T Reguladores/imunologia
4.
Int J Cancer ; 128(12): 2783-92, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20725998

RESUMO

Diets containing omega-6 (ω-6) fat have been associated with increased tumor development in carcinogen-induced pancreatic cancer models. However, the effects of ω-6 fatty acids and background strain on the development of genetically-induced pancreatic neoplasia is unknown. We assessed the effects of a diet rich in ω-6 fat on the development of pancreatic neoplasia in elastase (EL)-Kras(G12D) (EL-Kras) mice in two different backgrounds. EL-Kras FVB mice were crossed to C57BL/6 (B6) mice to produce EL-Kras FVB6 F1 (or EL-Kras F1) and EL-Kras B6 congenic mice. Age-matched EL-Kras mice from each strain were compared to one another on a standard chow. Two cohorts of EL-Kras FVB and EL-Kras F1 mice were fed a 23% corn oil diet and compared to age-matched mice fed a standard chow. Pancreata were scored for incidence, frequency, and size of neoplastic lesions, and stained for the presence of mast cells to evaluate changes in the inflammatory milieu secondary to a high fat diet. EL-Kras F1 mice had increased incidence, frequency, and size of pancreatic neoplasia compared to EL-Kras FVB mice. The frequency and size of neoplastic lesions and the weight and pancreatic mast cell densities in EL-Kras F1 mice were increased in mice fed a high ω-6 fatty acid diet compared to mice fed a standard chow. We herein introduce the EL-Kras B6 mouse model which presents with increased frequency of pancreatic neoplasia compared to EL-Kras F1 mice. The phenotype in EL-Kras F1 and FVB mice is promoted by a diet rich in ω-6 fatty acid.


Assuntos
Ácidos Graxos Ômega-6/administração & dosagem , Neoplasias Pancreáticas/etiologia , Animais , Apoptose , Sequência de Bases , Proliferação de Células , Primers do DNA , Genes ras , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Especificidade da Espécie
5.
J Surg Res ; 165(1): 75-81, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19631339

RESUMO

BACKGROUND: Diets containing omega-3 (ω-3) fat have been associated with decreased tumor development in the colon, breast, and prostate. We assessed the effects of a diet rich in ω-3 fat on the development of pancreatic precancer in elastase (EL)-Kras transgenic mice and examined the effect of an ω-3 fatty acid on pancreatic cancer cells in vitro. MATERIALS AND METHODS: Two cohorts of EL-Kras mice were fed a high ω-3 fat diet (23% menhaden oil) for 8 and 11 mo and compared with age-matched EL-Kras mice fed standard chow (5% fat). Pancreata from all mice were scored for incidence and frequency of precancerous lesions. Immunohistochemistry was performed for proliferating cell nuclear antigen (PCNA) to assess proliferative index in lesions of mice fed either a high ω-3 or standard diet. In vitro, the effect of the ω-3 fatty acid, docosahexaenoic acid (DHA), on two pancreatic cancer cell lines was assessed. Cancer cell proliferation was assessed with an MTT assay; cell cycle analysis was performed by flow cytometry; and apoptosis was assessed with annexin/PI staining. RESULTS: The incidence, frequency, and proliferative index of pancreatic precancer in EL-Kras mice was reduced in mice fed a high ω-3 fat diet compared with mice fed a standard chow. In vitro, DHA treatment resulted in a concentration-dependent decrease in proliferation through both G1/G0 cell cycle arrest and induction of apoptosis. CONCLUSIONS: A high ω-3 fat diet mitigates pancreatic precancer by inhibition of cellular proliferation through induction of cell cycle arrest and apoptosis.


Assuntos
Ácidos Graxos Ômega-3/administração & dosagem , Neoplasias Pancreáticas/prevenção & controle , Lesões Pré-Cancerosas/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Elastase Pancreática/fisiologia , Neoplasias Pancreáticas/patologia , Lesões Pré-Cancerosas/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética
6.
J Surg Res ; 167(2): 173-81, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21227456

RESUMO

BACKGROUND: The flavonoid apigenin exhibits anti-proliferative and anti-angiogenic activities. Our objective was to evaluate the effect of apigenin on hypoxia responsive genes important in pancreatic cancer cell proliferation. MATERIALS AND METHODS: Immunohistochemistry for GLUT-1 expression was conducted on human pancreatic cancer samples and adjacent controls. Real-time RT-PCR, Western blot analysis, and enzyme-linked immunosorbent assay (ELISA) were conducted on CD18 and S2-013 human pancreatic cancer cells treated with apigenin (0-50 µM) in normoxic and hypoxic conditions to evaluate HIF-1α, GLUT-1, and VEGF mRNA and protein expression and secretion. RESULTS: GLUT-1 expression was significantly increased in pancreatic adenocarcinoma samples versus adjacent controls (P < 0.001). Hypoxic conditions induced HIF-1α, GLUT-1, and VEGF protein expression in both CD18 and S2-013 pancreatic cancer cells. Apigenin (50 µM) blocked hypoxia induced up-regulation of all three proteins in both cell lines. Apigenin also impeded hypoxia-mediated induction of GLUT-1 and VEGF mRNA in both cell lines (P < 0.05). CONCLUSIONS: Apigenin inhibits HIF-1α, GLUT-1, and VEGF mRNA and protein expression in pancreatic cancer cells in both normoxic and hypoxic conditions. This may account for the mechanism of apigenin's anti-proliferative and anti-angiogenic effects and further supports the potential of apigenin as a future chemopreventive agent for pancreatic cancer.


Assuntos
Adenocarcinoma/metabolismo , Apigenina/farmacologia , Hipóxia Celular/genética , Regulação para Baixo/efeitos dos fármacos , Transportador de Glucose Tipo 1/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Pancreáticas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adenocarcinoma/patologia , Aminoácidos Dicarboxílicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Transportador de Glucose Tipo 1/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Pancreáticas/patologia , RNA Mensageiro/metabolismo , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/genética
7.
J Vasc Interv Radiol ; 20(8): 1075-82, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19560941

RESUMO

PURPOSE: An animal model of pancreatic cancer that is large enough to permit imaging and catheterization would be desirable for interventional radiologists to develop novel therapies for pancreatic cancer. The purpose of this study was to test the hypothesis that the VX2 rabbit model of pancreatic cancer could be developed as a suitable platform to test future interventional therapies. MATERIALS AND METHODS: The authors implanted and grew three pancreatic VX2 tumors per rabbit in six rabbits. Magnetic resonance (MR) imaging was performed at 2 weeks to confirm tumor growth. At 3 weeks, the authors selectively catheterized the gastroduodenal artery under guidance of x-ray digital subtraction angiography (DSA). T2-weighted anatomic imaging, diffusion-weighted MR imaging, and transcatheter intraarterial perfusion (TRIP) MR imaging were then performed. After imaging, tumors were confirmed at necropsy and histopathologically. Tumor sizes at 2 and 3 weeks were compared with a paired t test (P = .05). RESULTS: VX2 pancreatic tumors were grown in all six rabbits. The difference between tumor sizes at 2 and 3 weeks (1.29 cm +/- 0.39 vs 1.91 cm +/- 0.50, respectively) was significant (P < .001). All tumors were confirmed to be located within pancreatic tissue via histopathologic analysis. DSA and TRIP MR imaging were successful in five rabbits. Diffusion-weighted and anatomic MR imaging were successful in all six rabbits. CONCLUSIONS: The VX2 rabbit model of pancreatic cancer is feasible, as verified by imaging and pathologic correlation, and may be a suitable platform to test future interventional therapies.


Assuntos
Modelos Animais de Doenças , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/cirurgia , Radiografia Intervencionista/métodos , Animais , Linhagem Celular Tumoral , Humanos , Projetos Piloto , Coelhos
8.
Clin Cancer Res ; 14(20): 6525-30, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18927292

RESUMO

PURPOSE: Arachidonic acid metabolism via the cyclooxygenase (COX) and 5-lipoxygenase (5-LOX) pathways modulates cell growth and apoptosis. Many studies have examined the effects of COX inhibitors on human colorectal cancer, but the role of 5-LOX in colonic cancer development has not been well studied. The purpose of this study was to evaluate the expression of 5-LOX in colonic polyps and cancer and the effect of 5-LOX inhibition on colon cancer cell proliferation. EXPERIMENTAL DESIGN: Colonic polyps, cancer, and normal mucosa were evaluated for 5-LOX expression by immunohistochemistry. Reverse transcription-PCR was used to establish 5-LOX expression in colon cancer cells. Thymidine incorporation and cell counts were used to determine the effect of the nonspecific LOX inhibitor Nordihydroguaiaretic Acid and the 5-LOX inhibitor Rev5901 on DNA synthesis. A heterotopic xenograft model in athymic mice using HT29 and LoVo human colon cancer cells was used to evaluate the effect of the 5-LOX inhibitor zileuton on tumor growth. RESULTS: 5-LOX is overexpressed in adenomatous polyps and cancer compared with that of normal colonic mucosa. LOX inhibition and 5-LOX inhibition decreased DNA synthesis in a concentration- and time-dependent manner in the Lovo cell line (P < 0.05). Inhibition of 5-LOX in an in vivo colon cancer xenograft model inhibited tumor growth compared with that of controls (P < 0.05). CONCLUSIONS: This study showed that 5-LOX is up-regulated in adenomatous colon polyps and cancer compared with normal colonic mucosa. The blockade of 5-LOX inhibits colon cancer cell proliferation both in vitro and in vivo and may prove a beneficial chemopreventive therapy in colon cancer.


Assuntos
Araquidonato 5-Lipoxigenase/metabolismo , Neoplasias do Colo/enzimologia , Pólipos do Colo/enzimologia , Modelos Animais de Doenças , Inibidores de Lipoxigenase/farmacologia , Adenoma/tratamento farmacológico , Adenoma/enzimologia , Adenoma/patologia , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Pólipos do Colo/tratamento farmacológico , Pólipos do Colo/patologia , Feminino , Humanos , Técnicas Imunoenzimáticas , Técnicas In Vitro , Masoprocol/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Timidina/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Ann Surg ; 248(3): 438-46, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18791364

RESUMO

OBJECTIVES: To compare perioperative outcomes of laparoscopic left-sided pancreatectomy (LLP) with traditional open left-sided pancreatectomy (OLP) in a multicenter experience. SUMMARY AND BACKGROUND DATA: LLP is being performed more commonly with limited data comparing results with outcomes from OLP. METHODS: Data from 8 centers were combined for all cases performed between 2002-2006. OLP and LLP cohorts were matched by age, American Society of Anesthesiologists, resected pancreas length, tumor size, and diagnosis. Multivariate analysis was performed using binary logistic regression. RESULTS: Six hundred sixty-seven LPs were performed, with 159 (24%) attempted laparoscopically. Indications were solid lesion in 307 (46%), cystic in 295 (44%), and pancreatitis in 65 (10%) cases. Positive margins occurred in 51 (8%) cases, 335 (50%) had complications, and significant leaks occurred in 108 (16%). Conversion to OLP occurred in 20 (13%) of the LLPs. In the matched comparison, 200 OLPs were compared with 142 LLPs. There were no differences in positive margin rates (8% vs. 7%, P = 0.8), operative times (216 vs. 230 minutes, P = 0.3), or leak rates (18% vs. 11%, P = 0.1). LLP patients had lower average blood loss (357 vs. 588 mL, P < 0.01), fewer complications (40% vs. 57%, P < 0.01), and shorter hospital stays (5.9 vs. 9.0 days, P < 0.01). By MVA, LLP was an independent factor for shorter hospital stay (P < 0.01, odds ratio 0.33, 95% confidence interval 0.19-0.56). CONCLUSIONS: In selected patients, LLP is associated with less morbidity and shorter LOS than OLP. Pancreatic fistula rates are similar for OLP and LLP. LLP is appropriate for selected patients with left-sided pancreatic pathology.


Assuntos
Pancreatectomia/métodos , Pancreatopatias/cirurgia , Adulto , Idoso , Estudos de Coortes , Feminino , Humanos , Laparoscopia , Masculino , Pessoa de Meia-Idade , Pancreatectomia/efeitos adversos , Fístula Pancreática/etiologia , Estudos Retrospectivos
10.
Mol Carcinog ; 47(11): 835-44, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18404646

RESUMO

Pancreatic adeniocarcinoma is among the deadliest of human cancers. Apigenin, an antitumor flavonoid, inhibits pancreatic cancer cell proliferation in vitro. Geminin is a recently identified novel protein that plays a critical role in preventing abnormal DNA replication by binding to and inhibiting the essential replication factor Cdt1. Microarray analysis identified geminin to be downregulated in pancreatic cancer cells treated with apigenin. Therefore, we investigated the effects of apigenin on geminin expression and other proteins involved in replication (Cdc6, Cdt1, and MCM7) in pancreatic cancer cell lines CD18 and S2013. Real time RT-PCR and western blotting analysis showed that geminin expression is downregulated by apigenin at both mRNA and protein levels. Furthermore, treatment of cells with proteosome inhibitor MG132 reversed the downregulation of geminin by apigenin, supporting our hypothesis that the degradation pathway is another mechanism by which apigenin affects geminin expression. Apigenin treatment also resulted in downregulation of Cdc6 at both mRNA and protein levels. However, Cdt1 and MCM7 expression was not affected in apigenin-treated cells. The effect of apigenin treatment on geminin promoter activity was measured by transient transfection of Hela cells with a reporter gene, demonstrating that apigenin inhibited geminin promoter activity. Geminin expression was also evaluated in human pancreatic tissue (n = 15) by immunohistochemistry and showed that geminin is overexpressed in human pancreatic cancer compared to normal adjacent pancreatic tissue. In conclusion, our studies demonstrated that geminin is overexpressed in human pancreatic cancer and downregulated by apigenin which may contribute to the antitumor effect of this natural flavonoid.


Assuntos
Apigenina/farmacologia , Produtos Biológicos/farmacologia , Proteínas de Ciclo Celular/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pancreáticas/metabolismo , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Geminina , Humanos , Leupeptinas/farmacologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Regiões Promotoras Genéticas/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , RNA Mensageiro/genética
11.
Otolaryngol Clin North Am ; 37(2): 401-9, viii, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15064070

RESUMO

Minimally invasive sinus technique (MIST) is a targeted endoscopic intervention, introduced in 1994, with goals virtually identical to those originally reported for FESS but with distinct differences. The procedure, as outlined in the following section, is the only stepwise intranasal intervention with a defined beginning and end for all patients regardless of disease severity, thereby standardizing the procedure for surgeons and patients alike. This elegant, reproducible method avoids unnecessary disruption of normal mucosa while restoring mucociliary clearance through the primary birth ostia and reduces operative morbidity.


Assuntos
Endoscopia/métodos , Procedimentos Cirúrgicos Minimamente Invasivos/métodos , Doenças dos Seios Paranasais/terapia , Humanos
13.
J Gastrointest Surg ; 16(2): 389-93, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22072302

RESUMO

INTRODUCTION: The role of 5-lipoxygenase (5-LO) in colon cancer is unknown. Tumor-infiltrating macrophages, neutrophils, and mast cells have been shown to play important roles in colon tumorigenesis and are dependent on 5-LO for function. METHODS AND MATERIALS: Utilizing the APC(Δ468) polyposis model, we performed 5-LO gene knockouts and evaluated the subsequent changes in macrophage, neutrophil, and mast cell density at the tumor site. The proliferative and degranulation capacities of 5-LO-deficient mast cells were also measured, quantifying thymidine incorporation and ß-hexosaminidase release, respectively. RESULTS: APC(Δ468)/5LO(-/-) mice displayed increased tumor-infiltrating macrophages and decreased neutrophils at the polyp site. In vitro, mast cells deficient for 5-LO proliferated at a diminished rate while mast cell degranulation was unchanged. DISCUSSION: We provide evidence suggesting that 5-LO deficiency has differential effects on the infiltration of macrophages and neutrophils in adenomatous polyps, increasing and decreasing infiltration of these cells, respectively. Our observations are consistent with a protective role for tumor-infiltrating macrophages in the initiation of polyp formation. The mechanisms through which 5-LO deficiency negatively affects these cells are under investigation. CONCLUSIONS: These results provide evidence that 5-LO plays an important role in tumorigenesis and further indicate that 5-LO-selective inhibitors can be investigated as potential therapeutic agents for colorectal polyposis and cancer.


Assuntos
Araquidonato 5-Lipoxigenase/genética , Neoplasias do Colo/enzimologia , Pólipos do Colo/enzimologia , Macrófagos , Animais , Araquidonato 5-Lipoxigenase/fisiologia , Células Cultivadas , Neoplasias do Colo/etiologia , Neoplasias do Colo/patologia , Pólipos do Colo/patologia , Modelos Animais de Doenças , Macrófagos/enzimologia , Macrófagos/patologia , Mastócitos/enzimologia , Mastócitos/patologia , Camundongos , Camundongos Knockout , Neutrófilos/enzimologia , Neutrófilos/patologia
14.
Cancer Res ; 71(5): 1627-36, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21216893

RESUMO

Arachidonic acid metabolism has been implicated in colon carcinogenesis, but the role of hematopoietic 5-lipoxygenase (5LO) that may impact tumor immunity in development of colon cancer has not been explored. Here we show that tissue-specific deletion of the 5LO gene in hematopoietic cells profoundly attenuates polyp development in the APC(Δ468) murine model of colon polyposis. In vitro analyses indicated that mast cells in particular utilized 5LO to limit proliferation of intestinal epithelial cells and to mobilize myeloid-derived suppressor cells (MDSCs). Mice lacking hemapoietic expression of 5LO exhibited reduced recruitment of MDSCs to the spleen, mesenteric lymph nodes, and primary tumor site. 5LO deficiency also reduced the activity in MDSCs of arginase-1, which is thought to be critical for MDSC function. Together, our results establish a pro-tumorigenic role of hematopoietic 5LO in the immune microenvironment and suggest 5LO inhibition as an avenue for future investigation in treatment of colorectal polyposis and cancer.


Assuntos
Araquidonato 5-Lipoxigenase/metabolismo , Polipose Intestinal/enzimologia , Polipose Intestinal/genética , Mastócitos/enzimologia , Animais , Separação Celular , Citometria de Fluxo , Imunofluorescência , Genes APC , Polipose Intestinal/patologia , Camundongos , Camundongos Mutantes
15.
Anticancer Res ; 30(1): 73-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20150619

RESUMO

Survival of patients with pancreatic cancer remains poor due to inadequate chemotherapeutic options. Sansalvamide A, a cyclic depsipeptide produced by a marine fungus, has demonstrated significant anticancer activity. We previously observed antiproliferative effects in a series of sansalvamide A analogs in pancreatic cancer cells, one of which was further evaluated in this study. Two human pancreatic cancer cell lines (AsPC-1 and CD18) were incubated with increasing concentrations (10-50 muM) of the sansalvamide analog. Cell proliferation was then measured by thymidine incorporation and cell counting, and cell cycle analysis was determined by flow cytometry. Western blot analysis was used to evaluate expression of cyclin D1, cdk4, cdk6, cyclin E, cyclin A, cdk2, and p21. Sansalvamide caused G(1) phase cell cycle arrest in both cell lines, and Western blot analyses demonstrated up-regulation of p21, down-regulation of cyclins D1, E, and A, and cdk4, consistent with G(0)/G(1) cell cycle arrest. Cumulatively the results show that Sansalvamide A attenuates pancreatic cancer cell growth and represents a potential anticancer therapy.


Assuntos
Depsipeptídeos/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Ciclo Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ciclina A/biossíntese , Ciclina D1/biossíntese , Ciclina E/biossíntese , Quinase 4 Dependente de Ciclina/biossíntese , Quinase 6 Dependente de Ciclina/biossíntese , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Depsipeptídeos/química , Regulação para Baixo/efeitos dos fármacos , Humanos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia
16.
Clin Cancer Res ; 16(8): 2257-65, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20371681

RESUMO

PURPOSE: To assess the clinical and pathologic significance of mast cell infiltration in human pancreatic cancer and evaluate crosstalk between mast cells and cancer cells in vitro. EXPERIMENTAL DESIGN: Immunohistochemistry for tryptase was done on 53 pancreatic cancer specimens. Mast cell counts were correlated with clinical variables and survival. Serum tryptase activity from patients with cancer was compared with patients with benign pancreatic disease. In vitro, the effect of pancreatic cancer-conditioned medium on mast cell migration was assessed. The effect of conditioned medium from the human mast cell line, LAD-2, on cancer and normal ductal cell proliferation was assessed by thymidine incorporation. Matrigel invasion assays were used to evaluate the effect of mast cell-conditioned medium on cancer cell invasion in the presence and absence of a matrix metalloproteinase inhibitor, GM6001. RESULTS: Mast cell infiltration was significantly increased in pancreatic cancer compared with normal pancreatic tissue (11.4 +/- 6.7 versus 2.0 +/- 1.4, P < 0.001). Increased infiltrating mast cells correlated with higher grade tumors (P < 0.0001) and worse survival. Patients with pancreatic cancer had elevated serum tryptase activity (P < 0.05). In vitro, AsPC1 and PANC-1 cells induced mast cell migration. Mast cell-conditioned medium induced pancreatic cancer cell migration, proliferation, and invasion but had no effect on normal ductal cells. Furthermore, the effect of mast cells on cancer cell invasion was, in large part, matrix metalloproteinase-dependent. CONCLUSIONS: Tumor-infiltrating mast cells are associated with worse prognosis in pancreatic cancer. In vitro, the interaction between mast cells and pancreatic cancer cells promotes tumor growth and invasion.


Assuntos
Comunicação Celular/fisiologia , Mastócitos/fisiologia , Neoplasias Pancreáticas/patologia , Idoso , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Carcinoma Papilar/imunologia , Carcinoma Papilar/metabolismo , Carcinoma Papilar/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Meios de Cultivo Condicionados/farmacologia , Cistadenocarcinoma Mucinoso/imunologia , Cistadenocarcinoma Mucinoso/metabolismo , Cistadenocarcinoma Mucinoso/patologia , Progressão da Doença , Feminino , Humanos , Técnicas Imunoenzimáticas , Masculino , Invasividade Neoplásica , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/metabolismo , Prognóstico , Taxa de Sobrevida , Triptases/sangue
17.
Pancreas ; 38(4): 409-15, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19142175

RESUMO

OBJECTIVES: The aim of the study was to evaluate the effect of combination therapy of apigenin and gemcitabine on cell proliferation, the cell cycle, and gemcitabine resistance in human pancreatic cancer cells. METHODS: Cell counting was used to assess the effect of single-agent and combination treatment on the proliferation of CD18 and AsPC-1 pancreatic cancer cells. Flow cytometry was performed to assess the effect of combination treatment on cell cycle progression and induction of apoptosis. Western blot analysis was used to evaluate phosporylated AKT (pAkt) and cell cycle proteins. The effect of apigenin on gemcitabine-resistant AsPC-1 cells was assessed via thymidine incorporation. RESULTS: Apigenin in combination with gemcitabine inhibited pancreatic cancer cell proliferation more than either agent alone. Combination treatment induced both S and G2/M phase arrest and increased apoptosis. Apigenin down-regulated pAkt expression and abrogated gemcitabine-mediated pAkt induction. In gemcitabine-resistant AsPC-1 cells, apigenin significantly inhibited cell proliferation in a dose-dependent manner. CONCLUSION: Combination treatment with apigenin and gemcitabine inhibited pancreatic cancer cell growth via cell cycle arrest, down-regulation of the prosurvival factor pAkt, and induction of apoptosis. Combination therapy may prove useful for the treatment of pancreatic cancer.


Assuntos
Apigenina/farmacologia , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Antimetabólitos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Desoxicitidina/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Citometria de Fluxo , Fase G2/efeitos dos fármacos , Humanos , Mitose/efeitos dos fármacos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fase S/efeitos dos fármacos , Gencitabina
18.
Cancer Res ; 69(24): 9169-74, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19951995

RESUMO

To dissect the role of constitutively altered Tgfbr1 signaling in pancreatic cancer development, we crossed Elastase-Kras(G12D) (EL-Kras) mice with Tgfbr1 haploinsufficient mice to generate EL-Kras/Tgfbr1(+/-) mice. Mice were euthanized at 6 to 9 months to compare the incidence, frequency, and size of precancerous lesions in the pancreas. Only 50% of all EL-Kras/Tgfbr1(+/-) mice developed preinvasive lesions compared with 100% of EL-Kras (wild-type Tgfbr1) mice. The frequency of precancerous lesions was 4-fold lower in haploinsufficient than in control mice. Paradoxically, the precancerous lesions of EL-Kras/Tgfbr1(+/-) mice were considerably larger than those in EL-Kras mice. Yet, the mitotic index of precancerous cells and the observable levels of fibrosis, lipoatrophy, and lymphocytic infiltration were reduced in EL-Kras/Tgfbr1(+/-) mice. We conclude that Tgfbr1 signaling promotes the development of precancerous lesions in mice. These findings suggest that individuals with constitutively decreased TGFBR1 expression may have a decreased risk of pancreatic cancer.


Assuntos
Neoplasias Pancreáticas/genética , Lesões Pré-Cancerosas/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Animais , Apoptose/genética , Processos de Crescimento Celular/genética , Feminino , Haploidia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/patologia , Lesões Pré-Cancerosas/enzimologia , Lesões Pré-Cancerosas/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
19.
Pancreas ; 37(4): 426-31, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18953257

RESUMO

OBJECTIVES: The antiproliferative mechanisms of flavonoid drugs inpancreatic cancer cells remain unclear. In this study, we evaluated the effects of the flavonoid apigenin on glucose uptake, on the expression of the glucose transporter 1 (GLUT-1), and on the phosphoinositide 3-kinase (PI3K)/Akt pathway in human pancreatic cancer cells. METHODS: Human pancreatic cancer cells were treated with apigenin and then underwent glucose uptake assays. Real-time reverse transcription-polymerase chain reaction and Western blot analysis were conducted to evaluate GLUT-1 and pAkt expression in CD18 and S2-013 human pancreatic cancer cells after treatment with apigenin or PI3K inhibitors (LY294002 and wortmannin). RESULTS: Apigenin (0-100 microM) significantly inhibited, in a dose-dependent fashion, glucose uptake in CD18 and S2-013 human pancreatic cancer cell lines. Apigenin inhibited both GLUT-1 mRNA and protein expression in a concentration- and time-dependent fashion. The PI3K inhibitors, like apigenin, downregulated both GLUT-1 mRNA and protein expression. CONCLUSIONS: Our results demonstrate that the flavonoid apigenin decreases glucose uptake and downregulates the GLUT-1 glucose transporter in human pancreatic cancer cells. In addition, the inhibitory effects of apigenin and the PI3K inhibitors on GLUT-1 are similar, indicating that the PI3K/Akt pathway is involved in mediating apigenin's effects on downstream targets such as GLUT-1.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Antineoplásicos/farmacologia , Apigenina/farmacologia , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Transportador de Glucose Tipo 1/antagonistas & inibidores , Neoplasias Pancreáticas/enzimologia , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Humanos , Neoplasias Pancreáticas/patologia , Fosfatidilinositol 3-Quinases/metabolismo , RNA Mensageiro/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA