Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Surg Endosc ; 38(6): 3478-3485, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38769186

ABSTRACT

BACKGROUND: This study aims to report our surgical techniques for robot-assisted laparoscopic anterior resection, specifically focusing on mesorectal division using rolling division of the mesorectum, and to elucidate short-term outcomes at a single institution. Tumor-specific mesorectal excision (TSME) is commonly performed for resection of a tumor located in the upper rectum. However, especially in a narrow pelvis, it is difficult to perform appropriate mesorectal division at an adequate distance from the tumor in robot-assisted laparoscopic anterior resection. METHODS: Retrospective case series of patients with rectal cancer who underwent robot-assisted TSME using rolling division of mesorectum. Patient characteristics, perioperative clinical results, surgical and pathological details were recorded. RESULTS: A total of 198 patients underwent robot-assisted TSME for rectal cancer using rolling division of mesorectum between May 2019 and December 2023.The tumor was located in the upper rectum in 45 patients, middle rectum in 115 patients and lower rectum in 38 patients. The types of resections were 40 high anterior resection and 158 low anterior resections. The median operation time was 175 (range 109-310) min, and median mesorectal division time was 24 (range 15-45) min. Median blood loss was 3 (range 0-20) ml; no patients required blood transfusion. The overall complication rate of Clavien-Dindo classification grades I-IV was 7.1%. Anastomotic leakage was observed in two patients (1.0%) with grade III. There was no surgical mortality in this series. CONCLUSION: This robotic technique for anterior resection is a feasible and reliable procedure for achieving sufficient and safe TSME in this cohort.


Subject(s)
Rectal Neoplasms , Robotic Surgical Procedures , Humans , Rectal Neoplasms/surgery , Rectal Neoplasms/pathology , Robotic Surgical Procedures/methods , Male , Female , Middle Aged , Retrospective Studies , Aged , Adult , Aged, 80 and over , Proctectomy/methods , Treatment Outcome , Operative Time , Laparoscopy/methods , Rectum/surgery , Postoperative Complications/epidemiology , Postoperative Complications/etiology
2.
Gan To Kagaku Ryoho ; 47(6): 923-926, 2020 Jun.
Article in Japanese | MEDLINE | ID: mdl-32541169

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors(nivolumab)have been recommended as third-line chemotherapy for advanced gastric cancer(AGC)according to the Guidelines of Gastric Cancer(5th edition). Therefore, they have been used in daily clinical practice. On the other hand, the neutrophil-lymphocyte ratio(NLR)has been reported to be associated with the prognosis of cancer patients. METHODS: Twenty patients treated with nivolumab for AGC between January 2018 and November 2019 were retrospectively examined. RESULTS: Median age of the 20 patients(18 males, 2 females)was 70 years(55- 84 years). Nivolumab was administered as second-, third-, fourth-, and fifth-line therapy in 1, 11, 7, and 1 case, respectively. The best tumor response evaluation was observed in PR 1, SD 7 and PD 10 cases. Median overall survival(OS)was 10 months, and median progression-free survival(PFS)was 3 months. No serious adverse events occurred. Compared to the NLR>2.0 group, OS significantly prolonged(2.2 months vs 21.9 months)and PFS tended to prolong(1.4 months vs 6.2 months)in the NLR≤2.0 group. CONCLUSION: NLR may be an effective prognostic factor in patients with AGC receiving nivolumab treatment.


Subject(s)
Lymphocytes , Neutrophils , Nivolumab/therapeutic use , Stomach Neoplasms , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Stomach Neoplasms/drug therapy
3.
Gastroenterology ; 154(5): 1524-1537.e6, 2018 04.
Article in English | MEDLINE | ID: mdl-29274868

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) is characterized by activated pancreatic stellate cells (PSCs), abundance of extracellular matrix (ECM), and production of cytokines and chemokines. Galectin 3 (GAL3), a ß-galactoside-specific lectin, contributes to PDAC development but its effects on the stroma and cytokine production are unclear. METHODS: The effect of recombinant human GAL3 (rGAL3) on activation of PSCs, production of cytokines, and ECM proteins was determined by proliferation, invasion, cytokine array, and quantitative polymerase chain reaction. We assessed co-cultures of PDAC cells with GAL3 genetic alterations with PSCs. Production of interleukin 8 (IL8) and activities of nuclear factor (NF)-κB were determined by enzyme-linked immunosorbent assay and luciferase reporter analyses. We studied the effects of inhibitors of NF-κB and integrin-linked kinase (ILK) on pathways activated by rGAL3. RESULTS: In analyses of the Gene Expression Omnibus database and our dataset, we observed higher levels of GAL3, IL8, and other cytokines in PDAC than in nontumor tissues. Production of IL8, granulocyte-macrophage colony-stimulating factor, chemokine ligand 1, and C-C motif chemokine ligand 2 increased in PSCs exposed to rGAL3 compared with controls. Culture of PSCs with PDAC cells that express different levels of GAL3 resulted in proliferation and invasion of PSCs that increased with level of GAL3. GAL3 stimulated transcription of IL8 through integrin subunit beta 1 (ITGB1) on PSCs, which activates NF-κB through ILK. Inhibitors of ILK or NF-κB or a neutralizing antibody against ITGB1 blocked transcription and production of IL8 from PSCs induced by rGAL3. The GAL3 inhibitor significantly reduced growth and metastases of orthotopic tumors that formed from PDAC and PSC cells co-implanted in mice. CONCLUSION: GAL3 activates PSC cells to produce inflammatory cytokines via ITGB1signaling to ILK and activation of NF-κB. Inhibition of this pathway reduced growth and metastases of pancreatic orthotopic tumors in mice.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cytokines/metabolism , Galectin 3/metabolism , Integrin beta1/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/metabolism , Paracrine Communication , Stromal Cells/metabolism , Tumor Microenvironment , Animals , Antineoplastic Agents/pharmacology , Blood Proteins , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/secondary , Cell Line, Tumor , Cell Movement , Cell Proliferation , Coculture Techniques , Cytokines/genetics , Extracellular Matrix Proteins/metabolism , Galectin 3/antagonists & inhibitors , Galectins , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neoplasm Invasiveness , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/pathology , Paracrine Communication/drug effects , Phenotype , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Stromal Cells/drug effects , Stromal Cells/immunology , Stromal Cells/pathology , Time Factors , Transcription, Genetic , Xenograft Model Antitumor Assays
4.
Int J Surg Case Rep ; 36: 86-89, 2017.
Article in English | MEDLINE | ID: mdl-28550789

ABSTRACT

INTRODUCTION: Patients with esophageal cancer frequently cannot tolerate thoracotomy due to their overall debilitated condition. Moreover, some patients have severe adhesions in the thoracic cavity. Eversion stripping of the esophagus is an option for resection in these patients. PRESENTATION OF CASE: A 64-year-old man was admitted to our institution with the chief complaint of epigastric pain. Endoscopic examination showed a protruding lesion 22cm from the incisors, with a superficial and circumferential mucosal irregularity on the distal side of the lesion. Biopsy revealed squamous cell carcinoma. Clinical stage was T1b(sm)N0M0, cStage I. In addition to the poor pulmonary status of the patient, adhesions in the intrathoracic cavity were predicted. The decision was made to perform esophageal resection without a thoracotomy. In order to ensure complete invagination of the esophagus, the esophagus was insufflated prior to stripping. The stripping process was observed with a gastroscope. During the stripping, the esophagus did not bunch up, and stripping was smooth and with minimal resistance. DISCUSSION: The stripping resection of the esophagus is an important option for the esophageal surgeon. In this case report, we describe a new eversion stripping method of the esophagus. This easy and reliable stripping method incorporates intraesophageal insufflation. CONCLUSION: The indications for blunt esophageal dissection without thoracotomy have been decreasing. On the other hand, our method seems to be useful in optimal case of stripping of esophagus.

5.
Case Rep Surg ; 2017: 5862871, 2017.
Article in English | MEDLINE | ID: mdl-28299227

ABSTRACT

With advances of combined modality therapy, prognoses in esophageal cancer have been improving. After resection of esophageal cancer, the development of gastric tube cancer is a risk. While such cancer in an early stage can be cured endoscopically, total gastric tube resection is indicated in advanced stages. A 68-year-old man underwent subtotal esophagectomy reconstructed with a gastric tube through the retrosternal route. Gastric cancer was found one and a half years postoperatively. The gastric tube was resected without sternotomy. This is the first report of a patient undergoing resection of the gastric tube reconstructed through the retrosternal route without sternotomy.

6.
Pancreas ; 42(3): 494-501, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23146921

ABSTRACT

OBJECTIVE: Pancreatic diseases pose significant diagnostic challenge as signs and symptoms often overlap. We investigated the potential of pancreatic juice neutrophil gelatinase-associated lipocalin, macrophage inhibitory cytokine 1 (MIC-1), and carbohydrate antigen 19-9 (CA19-9) to aid in the diagnosis of patients with symptoms suggestive of pancreatic diseases. METHODS: A total of 105 chronic pancreatitis (CP), pancreatic cancer (PC), and nonpancreatic nonhealthy (patients with symptoms mimicking pancreatic disease but found to be free of any pancreatic disease) patients underwent endoscopic pancreatic juice collection after secretin stimulation. Neutrophil gelatinase-associated lipocalin and MIC-1 levels were measured by enzyme-linked immunosorbent assay, whereas CA19-9 was measured by radioimmunoassay. RESULTS: Neutrophil gelatinase-associated lipocalin, MIC-1, and CA19-9 were significantly elevated in the pancreatic juice of patients with CP and patients with PC as compared with nonpancreatic nonhealthy controls (P ≤ 0.034). Neutrophil gelatinase-associated lipocalin seemed most promising in differentiating diseased versus nondiseased pancreata (areas under the curve, 0.88-0.91), whereas MIC-1 was found to be higher in patients with PC than in patients with CP (P = 0.043). Interestingly, MIC-1 levels in diabetic patients with PC were higher than in nondiabetic patients with PC (P = 0.030) and diabetic patients with CP (P = 0.087). Carbohydrate antigen 19-9 showed the least ability to distinguish patient groups (areas under the curve, 0.61-0.76). CONCLUSIONS: Pancreatic juice neutrophil gelatinase-associated lipocalin shows potential utility in establishing pancreatic etiology in the context of nonspecific symptoms, whereas MIC-1 may aid in differentiating PC from CP.


Subject(s)
Acute-Phase Proteins/metabolism , CA-19-9 Antigen/metabolism , Growth Differentiation Factor 15/metabolism , Lipocalins/metabolism , Pancreatic Juice/metabolism , Proto-Oncogene Proteins/metabolism , Aged , Biomarkers/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diagnosis, Differential , Enzyme-Linked Immunosorbent Assay , Female , Humans , Lipocalin-2 , Male , Middle Aged , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/metabolism , Pancreatitis, Chronic/complications , Pancreatitis, Chronic/diagnosis , Pancreatitis, Chronic/metabolism , Prospective Studies , Radioimmunoassay , Sensitivity and Specificity
7.
Gastrointest Endosc ; 75(2): 242-53, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22115605

ABSTRACT

BACKGROUND: The prognosis of esophageal cancer (EC) depends on the depth of tumor invasion and lymph node metastasis. EC limited to the mucosa (T1a) can be treated effectively with minimally invasive endoscopic therapy, whereas submucosal (T1b) EC carries relatively high risk of lymph node metastasis and requires surgical resection. OBJECTIVE: To determine the diagnostic accuracy of EUS in differentiating T1a EC from T1b EC. DESIGN: We performed a comprehensive search of MEDLINE, SCOPUS, Cochrane, and CINAHL Plus databases to identify studies in which results of EUS-based staging of EC were compared with the results of histopathology of EMR or surgically resected esophageal lesions. DerSimonian-Laird random-effects model was used to estimate the pooled sensitivity, specificity, and likelihood ratio, and a summary receiver operating characteristic (SROC) curve was created. SETTING: Meta-analysis of 19 international studies. PATIENTS: Total of 1019 patients with superficial EC (SEC). INTERVENTIONS: EUS and EMR or surgical resection of SEC. MAIN OUTCOME MEASUREMENTS: Sensitivity and specificity of EUS in accurately staging SEC. RESULTS: The pooled sensitivity, specificity, and positive and negative likelihood ratio of EUS for T1a staging were 0.85 (95% CI, 0.82-0.88), 0.87 (95% CI, 0.84-0.90), 6.62 (95% CI, 3.61-12.12), and 0.20 (95% CI, 0.14-0.30), respectively. For T1b staging, these results were 0.86 (95% CI, 0.82-0.89), 0.86 (95% CI, 0.83-0.89), 5.13 (95% CI, 3.36-7.82), and 0.17 (95% CI, 0.09-0.30), respectively. The area under the curve was at least 0.93 for both mucosal and submucosal lesions. LIMITATIONS: Heterogeneity was present among the studies. CONCLUSION: Overall EUS has good accuracy (area under the curve ≥0.93) in staging SECs. Heterogeneity among the included studies suggests that multiple factors including the location and type of lesion, method and frequency of EUS probe, and the experience of the endosonographer can affect the diagnostic accuracy of EUS.


Subject(s)
Adenocarcinoma/diagnostic imaging , Adenocarcinoma/pathology , Endosonography , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/pathology , Humans , Neoplasm Invasiveness , Sensitivity and Specificity
8.
J Cell Physiol ; 226(4): 1074-81, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20857418

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal diseases. Novel molecularly targeted therapies are urgently needed. Here, we extended our studies on the role of protein kinase D1 (PKD1) in PDAC cell lines. Given that Panc-1 express moderate levels of PKD1, we used retroviral-mediated gene transfer to create a Panc-1 derivative that stably over-expresses PKD1 (Panc-1-PKD1). Reciprocally, we used shRNA targeting PKD1 in Panc-28 to produce a PKD1 under-expressing Panc-28 derivative (Panc-28-shPKD1). Our results demonstrate that Panc-1-PKD1 cells exhibit significantly increased anchorage-independent growth in soft agar and increased in vitro invasion compared with Panc-1-mock. Reciprocally, Panc-28-shPKD1 cells show a significant decrease in anchorage-independent growth and invasiveness, as compared with Panc-28-mock cells. The selective PKD family inhibitor CRT0066101 markedly decreased colony-forming ability and invasiveness by either Panc-1-PKD1 or Panc-28-mock cells. Secretion of the pro-angiogenic factors vascular endothelial growth factor (VEGF) and CXC chemokines (CXCL8) was significantly elevated by PKD1 over-expression in Panc-1 cells and reduced either by depletion of PKD1 via shRNA in Panc-28 cells or by addition of CRT0066101 to either Panc-1-PKD1 or Panc-28-mock cells. Furthermore, human umbilical vein endothelial cell (HUVEC) tube formation was significantly enhanced by co-culture with Panc-1-PKD1 compared with Panc-1-mock in an angiogenesis assay in vitro. Conversely, PKD1 depletion in Panc-28 cells decreased their ability to induce endotube formation by HUVECs. PDAC-induced angiogenesis in vitro and in vivo was markedly inhibited by CRT0066101. Our results lend further support to the hypothesis that PKD family members provide a novel target for PDAC therapy.


Subject(s)
Carcinoma, Pancreatic Ductal/blood supply , Carcinoma, Pancreatic Ductal/pathology , Neovascularization, Pathologic/enzymology , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/pathology , Protein Kinase C/metabolism , Animals , Carcinoma, Pancreatic Ductal/enzymology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Interleukin-8/biosynthesis , Mice , Microvessels/drug effects , Microvessels/pathology , Neoplasm Invasiveness , Pancreatic Neoplasms/enzymology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/metabolism
9.
Arch Med Sci ; 7(2): 353-5, 2011 Apr.
Article in English | MEDLINE | ID: mdl-22291781

ABSTRACT

We describe a case of a solid variant of serous cystadenoma of the pancreas. The preoperative examination results led to a diagnosis of a nonfunctional pancreatic islet cell tumour, and the patient underwent a pylorus-preserving pancreaticoduodenectomy. The tumour was diagnosed as a solid variant of serous cystadenoma by histopathological examination. Solid variant of serous cystadenoma of the pancreas is difficult to diagnose preoperatively. More cases must be accumulated and investigated to obtain clues for accurate diagnosis.

10.
J Surg Oncol ; 102(5): 469-77, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20872950

ABSTRACT

BACKGROUND AND OBJECTIVES: Interleukin (IL)-1α and hepatocyte growth factor (HGF) play an important role in pancreatic cancer proliferation, angiogenesis, and invasiveness. The aim of this study was to investigate the cooperative role of HGF and IL-1α in metastatic processes promoted by interactions between pancreatic cancer cells and stromal cells. METHODS: Expression of IL-1α and HGF mRNA and protein was determined by RT-PCR and ELISA. The effect of HGF on metastatic potential was evaluated by proliferation, invasion, and angiogenesis assays using an in vitro system consisting of co-cultured tumor cells and stromal cells. RESULTS: IL-1α expression was closely correlated with metastatic potential, and cancer cell-derived IL-1α significantly promoted HGF expression by fibroblasts (P < 0.01). HGF not only enhanced the invasiveness and proliferation of pancreatic cancer cells, but also enhanced migration and proliferation of human umbilical vein endothelial cells (HUVECs). HGF significantly enhanced HUVEC tube formation (P < 0.01). Furthermore, the high liver-metastatic pancreatic cancer cell line (BxPC-3), which secretes IL-1α, significantly enhanced HUVEC tube formation compared with the low liver-metastatic cell line (Capan-2), which does not produce IL-1α (P < 0.01). CONCLUSION: Autocrine IL-1α and paracrine HGF co-enhance the metastatic potential of pancreatic cancer cells via both IL-1α and HGF signaling pathways.


Subject(s)
Cell Communication , Hepatocyte Growth Factor/metabolism , Interleukin-1alpha/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/pathology , Cell Line, Tumor , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Humans , Neoplasm Metastasis , Pancreatic Neoplasms/metabolism , Polymerase Chain Reaction , RNA, Messenger/metabolism , Signal Transduction , Stromal Cells/metabolism
11.
Mol Cancer Ther ; 9(5): 1136-46, 2010 May.
Article in English | MEDLINE | ID: mdl-20442301

ABSTRACT

Protein kinase D (PKD) family members are increasingly implicated in multiple normal and abnormal biological functions, including signaling pathways that promote mitogenesis in pancreatic cancer. However, nothing is known about the effects of targeting PKD in pancreatic cancer. Our PKD inhibitor discovery program identified CRT0066101 as a specific inhibitor of all PKD isoforms. The aim of our study was to determine the effects of CRT0066101 in pancreatic cancer. Initially, we showed that autophosphorylated PKD1 and PKD2 (activated PKD1/2) are significantly upregulated in pancreatic cancer and that PKD1/2 are expressed in multiple pancreatic cancer cell lines. Using Panc-1 as a model system, we showed that CRT0066101 reduced bromodeoxyuridine incorporation; increased apoptosis; blocked neurotensin-induced PKD1/2 activation; reduced neurotensin-induced, PKD-mediated Hsp27 phosphorylation; attenuated PKD1-mediated NF-kappaB activation; and abrogated the expression of NF-kappaB-dependent proliferative and prosurvival proteins. We showed that CRT0066101 given orally (80 mg/kg/d) for 24 days significantly abrogated pancreatic cancer growth in Panc-1 subcutaneous xenograft model. Activated PKD1/2 expression in the treated tumor explants was significantly inhibited with peak tumor concentration (12 micromol/L) of CRT0066101 achieved within 2 hours after oral administration. Further, we showed that CRT0066101 given orally (80 mg/kg/d) for 21 days in Panc-1 orthotopic model potently blocked tumor growth in vivo. CRT0066101 significantly reduced Ki-67-positive proliferation index (P < 0.01), increased terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling-positive apoptotic cells (P < 0.05), and abrogated the expression of NF-kappaB-dependent proteins including cyclin D1, survivin, and cIAP-1. Our results show for the first time that a PKD-specific small-molecule inhibitor CRT0066101 blocks pancreatic cancer growth in vivo and show that PKD is a novel therapeutic target in pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation/drug effects , Pancreatic Neoplasms/pathology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , Molecular Weight , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Xenograft Model Antitumor Assays
12.
J Surg Res ; 160(1): 90-101, 2010 May 01.
Article in English | MEDLINE | ID: mdl-19560785

ABSTRACT

BACKGROUND: Type-1 insulin-like growth factor (IGF-1) up-regulates cell proliferation and invasiveness through activation of PI3K/Akt signaling pathway. IGF-1 also down-regulates the tumor suppressor chromosome 10 (PTEN). We investigated the mechanism by which IGF-1 affects cell proliferation and invasion by suppression of PTEN phosphorylation and interaction with PI3K/PTEN/Akt/NF-small ka, CyrillicB signaling pathway in pancreatic cancer. MATERIALS AND METHODS: The expression of IGF-1 receptor (IGF-1R) and PTEN in five pancreatic cancer cell lines was determined by RT-PCR and Western blot. Proliferation and invasion were investigated by WST-1 assay and Matrigel-double chamber assay. Pancreatic cancer cells were transfected with PTEN siRNA to investigate which signaling pathway correlates in regulation of cancer cell proliferation and invasion. RESULTS: Five pancreatic cancer cell lines expressed PTEN and IGF-1R in mRNA and protein levels. Suppression of PTEN phosphorylation strongly enhanced cell proliferation and invasion stimulated with IGF-1 via activation of PI3K/Akt/NF-small ka, CyrillicB signaling pathway. In addition, knockdown of PTEN by siRNA transfection also enhanced activation of PI3K/Akt/NF-small ka, CyrillicB pathway, subsequently up-regulating cell invasiveness and proliferation. CONCLUSIONS: The IGF-1/PI3K/PTEN/Akt/NF-small ka, CyrillicB cascade may be a key pathway stimulating metastasis of pancreatic cancer cells. We suggest that interfering with the functions of IGF-1/PI3K/Akt/NF-small ka, CyrillicB might be a novel therapeutic approach to inhibit aggressive spread of pancreatic cancer.


Subject(s)
Carcinoma/metabolism , Insulin-Like Growth Factor I/metabolism , PTEN Phosphohydrolase/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Antibodies/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation , Humans , Insulin-Like Growth Factor I/immunology , NF-kappa B/metabolism , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering , Receptor, IGF Type 1/metabolism , Signal Transduction
13.
Dig Dis Sci ; 55(4): 1167-76, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19399612

ABSTRACT

Since angiogenesis enables solid tumors, including pancreatic cancer (PaCa), to grow and metastasize, the development of anti-angiogenic agents is currently one of the urgent issues. Proteasome inhibitors are well known for inhibiting nuclear factor-kappa B (NF-kappaB) activity in various cancer cells, but little is known about their biologic mechanisms against angiogenesis in PaCa. We divided human PaCa cell lines into high-angiogenic (BxPC-3 and SW 1990) and low-angiogenic (MIA PaCa-2 and Capan-2) groups. The high-angiogenic PaCa cell lines constitutively expressed high NF-kappaB activity and produced high levels of vascular endothelial growth factor (VEGF) and interleukin 8 (IL-8). The conditioned media from BxPC-3 significantly enhanced both proliferation of and tube formation by human umbilical vein endothelial cells (HUVECs) and these enhancements were significantly inhibited by the proteasome inhibitor MG132 treatment. Collectively, MG132 blocked PaCa-derived VEGF and IL-8 production through inhibition of NF-kappaB activity. Thus, proteasome inhibitors may prove beneficial as anti-angiogenic therapy for PaCa. Our studies show that MG132, a proteasome inhibitor, significantly blocked pancreatic-cancer-associated angiogenesis through inhibition of NF-kappaB and NF-kappaB-dependent proangiogenic gene products VEGF and IL-8.


Subject(s)
Antineoplastic Agents/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , NF-kappa B/antagonists & inhibitors , Neovascularization, Pathologic/prevention & control , Pancreas/blood supply , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/drug therapy , Cell Division/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Endothelium, Vascular/drug effects , Humans , In Vitro Techniques , Interleukin-8/antagonists & inhibitors , Interleukin-8/metabolism , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/pathology , Umbilical Veins , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
14.
Mol Cell Biochem ; 331(1-2): 161-71, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19437103

ABSTRACT

Phosphoinositide 3-kinase (PI3K) pathway exerts its effects through Akt, its downstream target molecule, and thereby regulates various cell functions including cell proliferation, cell transformation, apoptosis, tumor growth, and angiogenesis. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) has been implicated in regulating cell survival signaling through the PI3K/Akt pathway. However, the mechanism by PI3K/PTEN signaling regulates angiogenesis and tumor growth in vivo remains to be elucidated. Vascular endothelial growth factor (VEGF) plays a pivotal role in tumor angiogenesis. The effect of PTEN on VEGF-mediated signal in pancreatic cancer is unknown. This study aimed to determine the effect of PTEN on both the expression of VEGF and angiogenesis. Toward that end, we used the siRNA knockdown method to specifically define the role of PTEN in the expression of VEGF and angiogenesis. We found that siRNA-mediated inhibition of PTEN gene expression in pancreatic cancer cells increase their VEGF secretion, up-modulated the proliferation, and migration of co-cultured vascular endothelial cell and enhanced tubule formation by HUVEC. In addition, PTEN modulated VEGF-mediated signaling and affected tumor angiogenesis through PI3K/Akt/VEGF/eNOS pathway.


Subject(s)
Neovascularization, Pathologic/enzymology , PTEN Phosphohydrolase/metabolism , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cinnamates/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Neovascularization, Pathologic/genetics , Nitric Oxide Synthase Type III/metabolism , PTEN Phosphohydrolase/genetics , Pancreatic Neoplasms/genetics , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Transfection , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/genetics
15.
J Surg Oncol ; 99(6): 361-7, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19204921

ABSTRACT

BACKGROUND AND OBJECTIVES: Interleukin (IL)-1alpha plays an important role in colon cancer progression and angiogenesis. We here asked whether IL-1alpha derived from cancer cells modulates vascular endothelial cell growth, migration and tubule formation. METHODS: The existence of IL-1alpha mRNA and protein in colon cancer cell lines (WiDr, HT-29, Caco-2, COLO 320) were investigated with RT-PCR and ELISA. Proliferation and invasion were investigated by MTS assay and Matrigel-double chamber assay. To answer our main question, we performed angiogenesis assay used an in vitro model consisting of co-cultivated tumor cells and stromal cells. RESULTS: IL-1alpha mRNA and protein were detected in highly metastatic colon cancer cells (WiDr and HT-29). Recombinant IL-1alpha significantly enhanced growth and invasiveness of human umbilical vein endothelial cells (HUVEC) (P < 0.01). Moreover, HUVEC growth and migration were significantly enhanced by WiDr compared to control (without co-culture) or Caco-2 (P < 0.05). Exogenous rIL-1alpha significantly enhanced HUVEC tube-like formation in a dose-dependent manner (P < 0.01) in a HUVEC/fibroblast co-cultivation system. Moreover, WiDr significantly enhanced HUVEC tubule formation compared with control or Caco-2 (P < 0.01). CONCLUSION: Based on these findings, we conclude that colon cancer cell-derived IL-1alpha up-regulates angiogenesis by modulating stromal cells within the tumor cells' microenvironment.


Subject(s)
Colonic Neoplasms/blood supply , Colonic Neoplasms/metabolism , Interleukin-1alpha/metabolism , Liver Neoplasms/blood supply , Liver Neoplasms/secondary , Neovascularization, Pathologic/metabolism , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/pathology , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-1alpha/genetics , Neoplasm Invasiveness , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
16.
J Surg Res ; 153(2): 274-81, 2009 May 15.
Article in English | MEDLINE | ID: mdl-18952231

ABSTRACT

BACKGROUND: Previously we reported the critical role of interleukin (IL)-1alpha in liver metastasis from pancreatic cancer (PaCa). However, its role in angiogenesis and metastasis, particularly as it relates to the interaction between tumor and stromal cells, was not clearly elucidated. To further investigate, we initially compared vascular endothelial cell migration and tube formation in human PaCa cell lines that differed in metastatic potential. We then compared the effects of IL-1alpha derived from PaCa cells on the same processes. MATERIALS AND METHODS: Expression of IL-1alpha mRNA and protein in PaCa cells was measured by reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay. IL-1alpha mRNA and protein were detected only in predominantly liver-metastatic PaCa cells. Next, we examined differences in migration and tube formation by human umbilical vein endothelial cells (HUVECs) in the various PaCa cells using invasion and angiogenesis assays, respectively. Furthermore, we determined the effects of IL-1alpha secreted by PaCa cells on migration and tube formation by HUVECs using coculture experiments. RESULTS: Expression of IL-1alpha mRNA and protein was observed only by the highly liver-metastatic PaCa cell lines BxPC-3 and SW 1990. Both HUVEC migration and tube formation were significantly enhanced by coculture with metastatic PaCa cells and IL-1alpha (P < 0.01). Similarly, blockade of IL-1alpha by its antagonist inhibited HUVEC migration and tube formation (P < 0.01). CONCLUSIONS: Our results indicate that IL-1alpha secreted by PaCa cells plays an important role in metastasis through vascular endothelial cell invasion and angiogenesis. Thus, blocking IL-1alpha is a potential novel therapeutic strategy in PaCa.


Subject(s)
Interleukin-1alpha/physiology , Neoplasm Metastasis/physiopathology , Neovascularization, Pathologic/physiopathology , Cell Line, Tumor , Cell Migration Assays , Coculture Techniques , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , RNA, Messenger/metabolism
17.
Int J Cancer ; 124(4): 853-61, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19035451

ABSTRACT

CXC-chemokines are involved in the chemotaxis of neutrophils, lymphocytes and monocytes. However, role of these chemokines in tumorigenesis, especially with regard to interaction between tumor and its microenvironment, has not been clearly elucidated. The purpose of this study was to analyze the co-operative role of CXCL8 and CXCL12 in the tumor-stromal interaction in pancreatic cancer (PaCa). Using enzyme-linked immunosorbent assay (ELISA) and reverse transcription polymerase chain reaction (RT-PCR), we initially confirmed the expression of ligands and receptors, respectively, of CXC-chemokines in PaCa and stromal cells. We examined the co-operative role of CXCL8 and CXCL12 in proliferation/invasion of PaCa and human umbilical vein endothelial cells (HUVECs), and in HUVEC tube-formations through tumor-stromal interaction by MTS, Matrigel invasion, and angiogenesis assays, respectively. We detected expression of CXCR4, but not CXCR2, in all PaCa cells and fibroblasts. PaCa cells secreted CXCL8, and fibroblast cells secreted CXCL12. CXCL8 production in PaCa was significantly enhanced by CXCL12, and CXCL12 production in fibroblasts was significantly enhanced by co-culturing with PaCa. CXCL8 enhanced proliferation/invasion of HUVECs but did not promote proliferation/invasion of PaCa. Both recombinant and PaCa-derived CXCL8 enhanced tube formation of HUVECs that were co-cultured with fibroblast cells. CXCL12 enhanced the proliferation/invasion of HUVECs and the invasion of PaCa cells but had no effect on tube formation of HUVEC. We showed that PaCa-derived CXCL8 and fibroblast-derived CXCL12 cooperatively induced angiogenesis in vitro by promoting HUVEC proliferation, invasion, and tube formation. Thus, corresponding receptors CXCR2 and CXCR4 are potential antiangiogenic and antimetastatic therapeutic targets in PaCa.


Subject(s)
Chemokine CXCL12/biosynthesis , Interleukin-8/biosynthesis , Neoplasm Invasiveness , Neovascularization, Pathologic , Pancreatic Neoplasms/pathology , Receptors, CXCR4/metabolism , Receptors, Interleukin-8B/metabolism , Cell Line, Tumor , Cell Proliferation , Chemokine CXCL12/metabolism , Coculture Techniques , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Fibroblasts/metabolism , Humans , Interleukin-8/metabolism , Pancreatic Neoplasms/metabolism , Recombinant Proteins/chemistry , Umbilical Veins/pathology
18.
BMC Gastroenterol ; 8: 56, 2008 Nov 26.
Article in English | MEDLINE | ID: mdl-19036165

ABSTRACT

BACKGROUND: The tumour suppressor phosphatase and tensin homolog (PTEN) is an important negative regulator of cell-survival signaling. To evaluate the correlation between PTEN expression and clinicopathological characteristics of colorectal cancer patients with and without liver metastases, we investigated PTEN expression in primary colorectal cancer and colorectal cancer liver metastases. METHODS: Sixty-nine pairs of primary colorectal cancer and corresponding liver metastasis specimens were analyzed immunohistochemically, and the correlation between immunohistochemical findings and clinicopathological factors was investigated. Seventy primary colorectal cancer specimens from patients without liver metastases were used as controls. RESULTS: PTEN was strongly expressed in 44 (62.9%) colorectal cancer specimens from patients without liver metastases. In contrast, PTEN was weakly expressed in 52 (75.4%) primary colorectal cancer specimens from patients with liver metastases, and was absent in liver metastases. Weak PTEN expression in colorectal cancer tissues was significantly associated with advanced TNM stage (p < 0.01) and lymph node metastasis (p < 0.05). PTEN expression was significantly stronger in primary colorectal cancer specimens from patients without liver metastases. Furthermore, among colorectal cancer patients with liver metastases, the 5-year survival rate was significantly higher in patients with positive PTEN expression compared to those with negative PTEN expression (p = 0.012). CONCLUSION: Our results suggest that loss of PTEN expression is involved with colorectal cancer aggressive capacity and that diagnostic evaluation of PTEN expression may provide valuable prognostic information to aid treatment strategies for colorectal cancer patients.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , PTEN Phosphohydrolase/metabolism , Aged , Case-Control Studies , Colorectal Neoplasms/diagnosis , Female , Humans , Kaplan-Meier Estimate , Liver Neoplasms/diagnosis , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Prognosis , Survival Rate
19.
J Surg Res ; 148(2): 197-204, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18395750

ABSTRACT

BACKGROUND: To better understand the underlying mechanism of liver metastasis formation in human gastric cancer, we evaluated the angiogenic capabilities of human gastric cancer cell lines with different metastatic potentials as well as the role of interleukin (IL)-1alpha in the angiogenic process. MATERIALS AND METHODS: Reverse transcription-polymerase chain reaction was used to detect the expression of IL-1alpha and vascular endothelial growth factor (VEGF) mRNA in gastric cancer cell lines with different liver metastatic potentials. Levels of VEGF secreted by human gastric cancer cells were measured by enzyme-linked immunosorbent assay. We also examined how gastric cancer cells with different metastatic potentials influence the proliferation and tube formation of human umbilical vein endothelial cells (HUVECs) using the Premix WST-1 cell proliferation assay system and an angiogenesis assay, respectively. RESULTS: IL-1alpha expression levels were significantly correlated with liver metastatic potential in gastric cancer cell lines. Levels of VEGF secreted by gastric cancer cells appear to be regulated by IL-1alpha through IL-1 receptor Type 1 and were correlated with liver metastatic potential. Both HUVEC proliferation and tube formation were strongly enhanced by coculture with high liver-metastatic gastric cancer cells and were enhanced to a similar extent by culture in the presence of IL-1alpha. In contrast, blockade of IL-1alpha inhibited both HUVEC proliferation and angiogenesis. CONCLUSIONS: IL-1alpha may play a role in liver metastasis of gastric cancer via enhanced vascular endothelial cell proliferation and angiogenesis.


Subject(s)
Interleukin-1alpha/physiology , Liver Neoplasms/secondary , Neovascularization, Pathologic/physiopathology , Stomach Neoplasms/blood supply , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , RNA, Messenger/metabolism , Receptors, Interleukin-1/metabolism , Stomach Neoplasms/pathology , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/metabolism
20.
J Surg Res ; 144(1): 117-23, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17688882

ABSTRACT

BACKGROUND: The invasive interaction between cells and their matrix has important roles in tumor cell invasion. This study investigated modulation of basement membrane (BM) proteins, especially collagen IV (Coll IV), laminin, and fibronectin (FN), in invasion of human pancreatic cancer cells. Furthermore, we examined the roles of beta(1)-integrins and arginine-glycine-aspartic (RGD)-containing oligopeptide in cell-matrix interactions. MATERIALS AND METHODS: Expression of integrins were examined by reverse transcriptase-polymerase chain reaction and flow-cytometric analysis in three human pancreatic cancer cell lines (BxPC-3, PANC-1, and SW1990), respectively. To determine the effect of BM proteins, invasion assays were performed. Western blot analysis for extracellular signal-regulated kinase (ERK) was performed to investigate the involvement of ERK1/2 signaling pathways. RESULTS: BM proteins significantly enhanced the invasive behavior of pancreatic cancer cells. Pretreatment with anti-beta(1)-integrin antibody suppressed invasion into Matrigel, but RGD-containing peptide inhibited invasion, which was enhanced by Coll IV and FN, not laminin. Treatment with both RGD-containing peptide and beta(1)-integrin antibody inhibited ERK1/2 phosphorylation activated by Coll IV and FN. CONCLUSIONS: BM proteins have positive actions on the processes of pancreatic cancer cell invasion and cross-talk between BM proteins and beta(1)-integrins widely participates in the multistep processes of pancreatic cancer invasion and metastasis formation.


Subject(s)
Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Neoplasm Invasiveness/physiopathology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/physiopathology , Basement Membrane/metabolism , Basement Membrane/pathology , Cell Line, Tumor , Collagen Type IV/genetics , Collagen Type IV/metabolism , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Integrin beta1/genetics , Integrin beta1/metabolism , Laminin/genetics , Laminin/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oligopeptides/genetics , Oligopeptides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...