Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
2.
Pancreatology ; 22(1): 9-19, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34642112

ABSTRACT

BACKGROUND/OBJECTIVES: Pancreatic ductal adenocarcinoma (PDAC) is characterized by excessive desmoplasia and autophagy-dependent tumorigenic growth. Pancreatic stellate cells (PSCs) as a predominant stromal cell type play a critical role in PDAC biology. We have previously reported that autophagy facilitates PSC activation, however, the mechanism remains unknown. We investigated the mechanism of autophagy in PSC activation. METHODS: We compared gene expression profiles between patient-derived PSCs from pancreatic cancer and chronic pancreatitis using a microarray. The stromal expression of target gene in specimen of PDAC patients (n = 63) was analyzed. The effect of target gene on autophagy and activation of PSCs was investigated by small interfering RNAs transfection, and the relationship between autophagy and ER stress was investigated. We analyzed the growth and fibrosis of xenografted tumor by orthotopic models. RESULTS: In analysis of gene expression microarray, endoplasmic reticulum aminopeptidase 2 (ERAP2) upregulated in cancer-associated PSCs was identified as the target gene. High stromal ERAP2 expression is associated with a poor prognosis of PDAC patients. Knockdown of ERAP2 inhibited unfolded protein response mediated autophagy, and led to inactivation of PSCs, thereby attenuating tumor-stromal interactions by inhibiting production of IL-6 and fibronectin. In vivo, the promoting effect of PSCs on xenografted tumor growth and fibrosis was inhibited by ERAP2 knockdown. CONCLUSIONS: Our findings demonstrate a novel mechanism of PSCs activation regulated by autophagy. ERAP2 as a promising therapeutic target may provide a novel strategy for the treatment of PDAC.


Subject(s)
Aminopeptidases , Autophagy , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Pancreatic Stellate Cells , Aminopeptidases/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Fibrosis , Gene Expression , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Pancreaticoduodenectomy , Signal Transduction , Pancreatic Neoplasms
3.
Oncol Lett ; 22(4): 744, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34466156

ABSTRACT

Pancreatic cancer cells (PCCs) are surrounded by an abundant stroma, which is produced by pancreatic stellate cells (PSCs). PSCs promote tumor cell proliferation and invasion. The objective of the current study was to identify compounds that suppress PSC activation. Gene expression profiles of cancer-derived fibroblasts and normal fibroblasts were used, and the pathway analysis suggested altered pathways that were chosen for validation. It was found that the 'neuroactive ligand-receptor interaction' pathway from the Kyoto Encyclopedia of Genes and Genomes pathway analysis was one of the altered pathways. Several compounds related with this pathway were chosen, and changes in PSC activity were investigated using fluorescence staining of lipid droplets, reverse transcription-quantitative PCR, western blotting, and invasion and migration assays. Among these candidates, duloxetine, a serotonin-noradrenaline reuptake inhibitor, was found to suppress PSC activation and disrupt tumor-stromal interaction. Thus, duloxetine may be a potential drug for suppressing PSC activation and pancreatic cancer growth.

4.
Oncol Lett ; 22(2): 633, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34267825

ABSTRACT

Pancreatic stellate cells (PSCs) play a key role in desmoplastic stroma, which is a characteristic of pancreatic ductal adenocarcinoma (PDAC), and they also enhance the malignancy of pancreatic cancer cells. Our previous study reported chloroquine's mitigating effects on PSC activation; however, the drug is known to induce adverse effects in clinical practice. The present study aimed to reduce chloroquine doses and develop a useful pre-treatment that targets PSCs using nanoparticles. Poly lactic-co-glycolic acid (PLGA) nanoparticles were used as carriers and loaded with indocyanine green (Nano-ICG) or chloroquine (Nano-CQ). Tumor accumulation of Nano-ICG was evaluated using an in vivo imaging system. The effects of chloroquine, Nano-CQ and/or chemotherapy drug gemcitabine were investigated in an orthotopic xenograft mouse model. Nano-ICG selectively accumulated in pancreatic tumors and persisted therein for over 7 days after administration. Additionally, Nano-ICG accumulated in the peritoneal metastasized regions, but not in the liver, kidney and normal pancreatic tissues. Nano-CQ reduced the density of activated PSCs at lower chloroquine doses and significantly restrained tumor progression in combination with gemcitabine. In conclusion, the PLGA nanosystem successfully delivered the drug to pancreatic tumors. Nano-CQ efficiently reduced PSC activation and may be a promising novel pre-treatment strategy for PDAC.

5.
Pancreatology ; 2021 Apr 20.
Article in English | MEDLINE | ID: mdl-33965328

ABSTRACT

BACKGROUND/OBJECTIVES: Pancreatic stellate cells (PSCs) are involved in abundant desmoplasia, which promotes cancer cell aggressiveness and resistance to anti-cancer drugs. Therefore, PSCs are suggested to be a promising therapeutic target by attenuating PSC activation to inhibit tumor-stromal interactions with pancreatic cancer cells. Here, we developed a screen to identify compounds that reduce the activity of PSCs and investigated the effect of candidates on pancreatic cancer. METHODS: Lipid droplet accumulation in PSCs was used to observe differences in PSC activity and a new high-throughput screening platform that quantified lipid droplets in PSCs was established. A library of 3398 Food and Drug Administration-approved drugs was screened by this platform. Validation assays were performed in vitro and in vivo. RESULTS: Thirty-two compounds were finally selected as candidate compounds by screening. These compounds decreased α-smooth muscle actin expression and inhibited autophagic flux in PSCs in vitro. Among the candidates, three drugs selected for validation assays inhibited the proliferation and migration of PSCs and invasion of cancer cells by disrupting tumor-stromal interactions. Production of extracellular matrix molecules was also decreased significantly by this treatment. In vivo testing in xenograft models showed that dopamine antagonist zuclopenthixol suppressed tumor growth; this suppression was significantly increased when combined with gemcitabine. CONCLUSIONS: A new screening platform that focused on the morphological features of PSCs was developed. Candidate drugs from this screening suppressed PSC activation and tumor growth. This screening system may be useful to discover new compounds that attenuate PSC activation.

6.
Int J Oncol ; 56(2): 596-605, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31894273

ABSTRACT

Cancer­associated fibroblasts (CAFs) promote the progression of pancreatic ductal adenocarcinoma (PDAC) via tumor­stromal interactions. Neutrophil extracellular traps (NETs) are extracellular DNA meshworks released from neutrophils together with proteolytic enzymes against foreign pathogens. Emerging studies suggest their contribution to liver metastasis in several types of cancer. Herein, in order to investigate the role of NETs in liver metastasis in PDAC, the effects of NET inhibitors on spontaneous PDAC mouse models were evaluated. It was demonstrated that DNase I, a NET inhibitor, suppressed liver metastasis. For further investigation, further attention was paid to liver micrometastasis and an experimental liver metastasis mouse model was used that was generated by intrasplenic tumor injection. Furthermore, DNase I also suppressed liver micrometastasis and notably, CAFs accumulated in metastatic foci were significantly decreased in number. In vitro experiments revealed that pancreatic cancer cells induced NET formation and consequently NETs enhanced the migration of hepatic stellate cells, which was the possible origin of CAFs in liver metastasis. On the whole, these results suggest that NETs promote liver micrometastasis in PDAC via the activation of CAFs.


Subject(s)
Cancer-Associated Fibroblasts/immunology , Carcinoma, Pancreatic Ductal/immunology , Liver Neoplasms/immunology , Neutrophils/immunology , Pancreatic Neoplasms/pathology , Aged , Animals , Carcinoma, Pancreatic Ductal/secondary , Carcinoma, Pancreatic Ductal/surgery , Cell Culture Techniques , Cell Line, Tumor/transplantation , Cell Movement/immunology , Cell Proliferation , Coculture Techniques , Deoxyribonuclease I/administration & dosage , Disease Models, Animal , Extracellular Traps/drug effects , Extracellular Traps/immunology , Extracellular Traps/metabolism , Hepatic Stellate Cells , Humans , Injections, Intraperitoneal , Liver Neoplasms/secondary , Male , Neoplasm Micrometastasis/immunology , Neoplasm Micrometastasis/prevention & control , Neutrophils/metabolism , Pancreas/immunology , Pancreas/pathology , Pancreas/surgery , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/surgery , Pancreaticoduodenectomy , Primary Cell Culture
7.
Surg Today ; 47(5): 555-567, 2017 May.
Article in English | MEDLINE | ID: mdl-27704248

ABSTRACT

PURPOSE: To investigate the impact of postoperative complications on survival after curative resection for pancreatic cancer. METHODS: We reviewed retrospectively the medical records of 122 patients who underwent curative R0 resection for pancreatic cancer. Major complications included pancreatic fistula and hemorrhage of grade B or C according to the International Study Group of Pancreatic Fistula or Surgery criteria, and other complications of grade ≥III according to the Clavien-Dindo classification. RESULTS: Thirty-eight patients (31 %) suffered major postoperative complications and 40 patients (33 %) suffered minor complications only. Univariate survival analysis showed that patients with major complications had a significantly worse prognosis than those without major complications, with regard to recurrence-free survival (RFS) (P < 0.01) and overall survival (OS) (P < 0.01), whereas minor complications did not affect survival. Major complications significantly inhibited or delayed adjuvant chemotherapy. Multivariate survival analysis showed that the absence of postoperative major complications was an independent favorable factor for RFS (hazard ratio 0.48; 95 % confidence interval: 0.28-0.85) and OS (hazard ratio 0.47; 95 % confidence interval 0.27-0.81). CONCLUSIONS: Postoperative major complications after pancreatectomy for pancreatic cancer affect the prognosis.


Subject(s)
Pancreatectomy , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/surgery , Postoperative Complications , Adult , Aged , Aged, 80 and over , Blood Loss, Surgical , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Humans , Male , Middle Aged , Pancreatic Fistula , Prognosis , Retrospective Studies , Severity of Illness Index , Survival Analysis , Survival Rate , Time Factors
8.
Jpn J Infect Dis ; 66(3): 207-15, 2013.
Article in English | MEDLINE | ID: mdl-23698481

ABSTRACT

Although IFN-γ release assays (IGRAs) provide increased specificity over tuberculin skin tests, the early and sensitive detection of reactivation of latently infected Mycobacterium tuberculosis is required to control tuberculosis (TB). Recently, a multicolor flow cytometry has been developed to study CD4(+) T cell cytokine responses (IFN-γ/IL-2/TNF-α) to purified protein derivatives (PPD) and M. tuberculosis-specific antigens (ESAT-6/CFP-10) and provided useful information regarding anti-TB immunity. However, the diagnostic relevancy remains uncertain. Here, we analyzed three additional CD4(+) T cell cytokine responses (IL-10/IL-13/IL-17) to latent mycobacterial antigens (α-crystallin, methylated heparin-binding hemagglutinin [HBHA], and mycobacterial DNA-binding protein 1 [MDP-1]) as well as PPD and ESAT-6/CFP-10 in 12 IGRA(+) TB cases and 8 healthy controls. No significant difference in IFN-γ response was observed between TB cases and controls, which was likely due to the high variation among the individuals. However, we found a significant increase over healthy controls in (i) the IL-2 response to HBHA in recovery stage TB cases, (ii) the number of M. tuberculosis-specific polyfunctional CD4(+) T cells in on-treatment and recovery stage cases, and (iii) the IL-17 response to HBHA and MDP-1 in on-treatment and recovery stage cases. These results suggest that a combination of these T cell cytokine parameters could aid in accurate diagnosis of latent TB infection.


Subject(s)
Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Flow Cytometry/methods , Latent Tuberculosis/diagnosis , Mycobacterium tuberculosis/immunology , Staining and Labeling/methods , Adult , Aged , Aged, 80 and over , Biomarkers , Color , Female , Fluorescent Dyes/analysis , Humans , Latent Tuberculosis/immunology , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...