Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Publication year range
1.
J Pathol ; 262(1): 61-75, 2024 01.
Article in English | MEDLINE | ID: mdl-37796386

ABSTRACT

Pancreatic stellate cells (PSCs) are stromal cells in the pancreas that play an important role in pancreatic pathology. In chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC), PSCs are known to get activated to form myofibroblasts or cancer-associated fibroblasts (CAFs) that promote stromal fibroinflammatory reactions. However, previous studies on PSCs were mainly based on the findings obtained using ex vivo expanded PSCs, with few studies that addressed the significance of in situ tissue-resident PSCs using animal models. Their contributions to fibrotic reactions in CP and PDAC are also lesser-known. These limitations in our understanding of PSC biology have been attributed to the lack of specific molecular markers of PSCs. Herein, we established Meflin (Islr), a glycosylphosphatidylinositol-anchored membrane protein, as a PSC-specific marker in both mouse and human by using human pancreatic tissue samples and Meflin reporter mice. Meflin-positive (Meflin+ ) cells contain lipid droplets and express the conventional PSC marker Desmin in normal mouse pancreas, with some cells also positive for Gli1, the marker of pancreatic tissue-resident fibroblasts. Three-dimensional analysis of the cleared pancreas of Meflin reporter mice showed that Meflin+ PSCs have long and thin cytoplasmic protrusions, and are localised on the abluminal side of vessels in the normal pancreas. Lineage tracing experiments revealed that Meflin+ PSCs constitute one of the origins of fibroblasts and CAFs in CP and PDAC, respectively. In these diseases, Meflin+ PSC-derived fibroblasts showed a distinctive morphology and distribution from Meflin+ PSCs in the normal pancreas. Furthermore, we showed that the genetic depletion of Meflin+ PSCs accelerated fibrosis and attenuated epithelial regeneration and stromal R-spondin 3 expression, thereby implying that Meflin+ PSCs and their lineage cells may support tissue recovery and Wnt/R-spondin signalling after pancreatic injury and PDAC development. Together, these data indicate that Meflin may be a marker specific to tissue-resident PSCs and useful for studying their biology in both health and disease. © 2023 The Pathological Society of Great Britain and Ireland.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Pancreatitis, Chronic , Animals , Humans , Mice , Carcinoma, Pancreatic Ductal/pathology , Fibrosis , Pancreas/pathology , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Pancreatitis, Chronic/genetics , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/pathology , Regeneration
2.
Toxicol Appl Pharmacol ; 491: 117078, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39214171

ABSTRACT

RUNX1 with CBFß functions as an activator or repressor of critical mediators regulating cellular function. The aims of this study were to clarify the role of RUNX1 on regulating TGF-ß1-induced COL1 synthesis and the mechanism of calcipotriol (Cal) on antagonizing COL1 synthesis in PSCs. RT-qPCR and Western Blot for determining the mRNAs and proteins of RUNX1 and COL1A1/1A2 in rat PSC line (RP-2 cell). Luciferase activities driven by RUNX1 or COL1A1 or COL1A2 promoter, co-immunoprecipitation and immunoblotting for pSmad3/RUNX1 or CBFß/RUNX1, and knockdown or upregulation of Smad3 and RUNX1 were used. RUNX1 production was regulated by TGF-ß1/pSmad3 signaling pathway in RP-2 cells. RUNX1 formed a coactivator with CBFß in TGF-ß1-treated RP-2 cells to regulate the transcriptions of COL1A1/1A2 mRNAs under a fashion of pSmad3/RUNX1/CBFß complex. However, Cal effectively abrogated the levels of COL1A1/1A2 transcripts in TGF-ß1-treated RP-2 cells by downregulating RUNX1 production and hindering the formation of pSmad3/RUNX1/CBFß complexes. This study suggests that RUNX1 may be a promising antifibrotic target for the treatment of chronic pancreatitis.


Subject(s)
Calcitriol , Collagen Type I , Core Binding Factor Alpha 2 Subunit , Down-Regulation , Pancreatic Stellate Cells , Smad3 Protein , Transforming Growth Factor beta1 , Animals , Calcitriol/pharmacology , Calcitriol/analogs & derivatives , Transforming Growth Factor beta1/metabolism , Smad3 Protein/metabolism , Rats , Down-Regulation/drug effects , Collagen Type I/metabolism , Collagen Type I/biosynthesis , Collagen Type I/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/metabolism , Cell Line , Signal Transduction/drug effects
3.
Cancer Cell Int ; 23(1): 219, 2023 Sep 27.
Article in English | MEDLINE | ID: mdl-37759302

ABSTRACT

BACKGROUND: Recently, natural killer (NK) cells emerged as a treatment option for various solid tumors. However, the immunosuppressive tumor immune microenvironment (TIME) can reduce the cytotoxic ability of NK cells in pancreatic ductal adenocarcinoma. Cancer-associated fibroblasts within the tumor stroma can suppress immune surveillance by dysregulating factors involved in the cellular activity of NK cells. Herein, the effect of activated pancreatic stellate cells (aPSCs) on NK cell-mediated anticancer efficacy under three-dimensional (3D) coculture conditions was investigated. METHODS: 3D cocultures of PANC-1 tumor spheroids (TSs) with aPSCs and NK-92 cells in a collagen matrix were optimized to identify the occurring cellular interactions and differential cytokine profiles in conditioned media using microchannel chips. PANC-1 TSs and aPSCs were indirectly cocultured, whereas NK-92 cells were allowed to infiltrate the TS channel using convective medium flow. RESULTS: Coculture with aPSCs promoted PANC-1 TSs growth and suppressed the antitumor cytotoxic effects of NK-92 cells. Mutual inhibition of cellular activity without compromising migration ability was observed between aPSCs and NK-92 cells. Moreover, the reduced killing activity of NK-92 cells was found to be related with reduced granzyme B expression in NK cells. CONCLUSIONS: Herein, a novel TIME-on-chip model based on the coculture of PANC-1 TSs, aPSCs, and NK-92 cells was described. This model may be useful for studying the detailed mechanisms underlying NK cells dysregulation and for exploring future therapeutic interventions to restore NK cell activity in the tumor microenvironment.

4.
Dig Dis Sci ; 68(4): 1339-1350, 2023 04.
Article in English | MEDLINE | ID: mdl-36002675

ABSTRACT

AIM: Pancreatic fibrosis is the main pathological characteristic of chronic pancreatitis (CP) and pancreatic cancer. Pancreatic stellate cells (PSCs) play a critical role in pancreatic fibrosis. Any targets that may have an impact on the activation of PSCs could become potential treatment candidates for CP and pancreatic cancer. Our goal was to investigate the effect of P-element-induced wimpy-testis (PIWI) protein 1 (PIWIL1) on PSC activation. METHODS: Lentivirus-based RNA interference (RNAi) and overexpression vector construction were used to knock down and over-express the PIWIL1 protein. Immunocytofluorescent staining, western blotting, wound healing assay, transwell assay, and phalloidin staining were used to investigate the effects of PIWIL1 on the secretion of extracellular matrix components (EMC), actin cytoskeleton, and on the invasion and migration abilities of primary PSCs isolated from C57BL/6 mice. Moreover, pancreatic fibrosis was induced by L-arginine in C57BL/6 mice. The expression of PIWIL1 and collagen deposition in vivo were tested by western blotting and Sirius red staining. RESULTS: Expression levels of collagen I, collagen III, and α-smooth muscle actin were significantly decreased in the LV-PIWIL1 group. Compared with the si-PIWIL1 group, significant differences were observed in the expression of desmin, p-PI3K, p-AKT, and p-mTOR in the LV-PIWIL1 group. Furthermore, PIWIL1 suppressed the PSCs' invasion and migration abilities. In a rescue experiment, the PI3K/AKT/mTOR signaling pathway was found to be the underlying mechanism in PSCs activation mediated by PIWIL1. CONCLUSIONS: Our findings suggest that PIWIL1 inhibits the activation of PSCs via the PI3K/AKT/mTOR signaling pathway. PIWIL1 is a potential therapeutic target for pancreatic fibrosis.


Subject(s)
Pancreatic Diseases , Pancreatic Neoplasms , Pancreatitis, Chronic , Male , Mice , Animals , Pancreas/pathology , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pancreatic Stellate Cells/pathology , Testis/metabolism , Testis/pathology , Cells, Cultured , Mice, Inbred C57BL , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Pancreatic Neoplasms/pathology , Pancreatitis, Chronic/chemically induced , Pancreatic Diseases/pathology , Collagen/metabolism , Fibrosis , Pancreatic Neoplasms
5.
Acta Biochim Biophys Sin (Shanghai) ; 55(9): 1393-1403, 2023 Jun 19.
Article in English | MEDLINE | ID: mdl-37337632

ABSTRACT

Since the prognosis of patients with pancreatic cancer is very poor and there is a lack of treatment methods, this study is performed to investigate the function of PITX2 in pancreatic stellate cells (PSCs) in the progression of pancreatic cancer. Scientific hypotheses are proposed according to bioinformatics analysis and tissue microarray analysis. Stable knockdown of PITX2 in PSCs is achieved through lentiviral infection. The relative expressions of PITX2, α-SMA, vimentin, CTNNB1, AXIN1 and LEF1 are measured in wild-type PSCs and PITX2-knockdown PSCs. Proliferative capacity is measured by EdU assay. After coculture with PSCs, the proliferation, invasion and migration capacity of pancreatic cancer cells are tested. EMT and Wnt/ß-catenin downstream genes of pancreatic cancer cells are investigated to reveal the potential mechanism. Bioinformatics analysis reveals that the PITX2 gene is highly expressed in stromal cells in pancreatic cancer and is correlated with squamous-type PDAC. Analysis of PDAC tissue microarray further demonstrates that high PITX2 level in stromal cells is correlated with poor prognosis in PDAC. After stable knockdown of PITX2 in PSCs, the relative protein levels of α-SMA, vimentin, CTNNB1, AXIN1 and LEF1 are decreased, and the proliferative capacity of PSCs is also decreased. After coculture with PSCs, in which PITX2 expression is downregulated, the proliferation, invasion and migration capacities of pancreatic cancer cells are inhibited. Thus, our results show that PITX2-silenced PSCs inhibit the growth, migration and invasion of pancreatic cancer cells via reduced EMT and Wnt/ß-catenin signaling.


Subject(s)
Pancreatic Neoplasms , beta Catenin , Humans , beta Catenin/genetics , beta Catenin/metabolism , Vimentin/genetics , Vimentin/metabolism , Pancreatic Stellate Cells/metabolism , Cell Movement/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Wnt Signaling Pathway/genetics , Cell Line, Tumor , Cell Proliferation/genetics
6.
Exp Cell Res ; 404(2): 112634, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34004193

ABSTRACT

NLRP3 inflammasome activation plays an important role in the development of pancreatic fibrosis. However, it is unclear whether the activation of the NLRP3 inflammasome is directly involved in the activation of Pancreatic stellate cells (PSCs). The aim of this study was to investigate the role and mechanism of the NLRP3 inflammasome in the activation of PSCs. In vivo, a rat model of chronic pancreatitis (CP) was induced by intravenous injection of dibutyltin dichloride (DBTC). In vitro, rat primary PSCs were isolated from pancreatic tissues and incubated with the NLRP3 inflammasome activator LPS, the NLRP3 inhibitor MCC950, or NLRP3 siRNA. The results showed that the expression of NLRP3, pro-Caspase-1, Caspase-1 and IL-18 was increased in the rat model of CP and during PSCs activation. LPS increased the protein levels of NLRP3, ASC, Caspase-1, IL-1ß and IL-18 accompanied by the upregulation of α-SMA, Col I and FN expression. Moreover, MCC950 or NLPR3 siRNA decreased the expression of α-SMA, Col I, FN, TGF-ß1 and p-Smad3. Furthermore, MCC950 reversed the LPS-induced upregulation of α-SMA, FN and Col Ⅰ expression in PSCs. This study revealed that the NLRP3 inflammasome is directly involved in the activation of PSCs in vivo and in vitro. Inhibiting NLRP3 suppresses the activation of PSCs through the TGF-ß1/Smad3 pathway.


Subject(s)
Fibrosis/metabolism , Inflammasomes/metabolism , Lipopolysaccharides/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pancreatic Stellate Cells/metabolism , Animals , Caspase 1/metabolism , Cells, Cultured , Fibrosis/chemically induced , Rats, Wistar , Transforming Growth Factor beta1/metabolism
7.
J Biol Chem ; 295(25): 8413-8424, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32358063

ABSTRACT

Pancreatic cancer (PC) remains a therapeutic challenge because of its intrinsic and extrinsic chemoresistance mechanisms. Here, we report that C-X-C motif chemokine receptor 4 (CXCR4) and hedgehog pathways cooperate in PC chemoresistance via bidirectional tumor-stromal crosstalk. We show that when PC cells are co-cultured with pancreatic stellate cells (PSCs) they are significantly more resistant to gemcitabine toxicity than those grown in monoculture. We also demonstrate that this co-culture-induced chemoresistance is abrogated by inhibition of the CXCR4 and hedgehog pathways. Similarly, the co-culture-induced altered expression of genes in PC cells associated with gemcitabine metabolism, antioxidant defense, and cancer stemness is also reversed upon CXCR4 and hedgehog inhibition. We have confirmed the functional impact of these genetic alterations by measuring gemcitabine metabolites, reactive oxygen species production, and sphere formation in vehicle- or gemcitabine-treated monocultures and co-cultured PC cells. Treatment of orthotopic pancreatic tumor-bearing mice with gemcitabine alone or in combination with a CXCR4 antagonist (AMD3100) or hedgehog inhibitor (GDC-0449) displays reduced tumor growth. Notably, we show that the triple combination treatment is the most effective, resulting in nearly complete suppression of tumor growth. Immunohistochemical analysis of Ki67 and cleaved caspase-3 confirm these findings from in vivo imaging and tumor measurements. Our findings provide preclinical and mechanistic evidence that a combination of gemcitabine treatment with targeted inhibition of both the CXCR4 and hedgehog pathways improves outcomes in a PC mouse model.


Subject(s)
Hedgehog Proteins/metabolism , Receptors, CXCR4/metabolism , Anilides/pharmacology , Anilides/therapeutic use , Animals , Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Benzylamines , Cell Communication , Cell Survival/drug effects , Coculture Techniques , Cyclams , Deoxycytidine/analogs & derivatives , Deoxycytidine/metabolism , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Resistance, Neoplasm/genetics , Hedgehog Proteins/antagonists & inhibitors , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/therapeutic use , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/cytology , Pancreatic Stellate Cells/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Signal Transduction/drug effects , Gemcitabine
8.
Am J Physiol Gastrointest Liver Physiol ; 321(4): G378-G388, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34405729

ABSTRACT

It was previously identified that systemic Nrf2 deletion attenuates pancreatic cancer progression in a mutant K-ras/p53-expressing mouse model (KPC mouse). In this study, the type of cell that is responsible for the retarded cancer progression was elucidated. Human pancreatic cancers were first examined, and elevated expression of NRF2-target gene products in α-smooth muscle actin-positive cells was found, suggesting that pancreatic stellate cells (PSCs) are involved in this process. Closer examination of primary cultured PSCs from Nrf2-deleted mice revealed that the cells were less proliferative and retained a lower migration capacity. The conditioned medium of Nrf2-deleted PSCs exhibited reduced growth-stimulating effects in pancreatic cancer cells. KPC mouse-derived pancreatic cancer cells coinjected with wild-type PSCs developed significantly larger subcutaneous tumors in immunodeficient mice than those coinjected with Nrf2-deleted PSCs. These results demonstrate that Nrf2 actively contributes to the function of PSCs to sustain KPC cancer progression, thus, suggesting that Nrf2 inhibition in PSCs may be therapeutically important in pancreatic cancer.NEW & NOTEWORTHY This study identified that Nrf2 contributes to PSC activation. Nrf2 deletion in PSCs resulted in attenuation of cancer-promoting role. Nrf2 in PSCs could be an attractive therapeutic target in pancreatic cancer.


Subject(s)
NF-E2-Related Factor 2/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/metabolism , Animals , Cell Line , Cell Movement , Cell Proliferation , Cells, Cultured , Humans , Mice , NF-E2-Related Factor 2/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/physiology , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/genetics
9.
Cytokine ; 143: 155536, 2021 07.
Article in English | MEDLINE | ID: mdl-33893003

ABSTRACT

Pancreatic stellate cells (PSCs) play a key role in fibrogenesis during alcoholic chronic pancreatitis (ACP). Transforming growth factor-ß1 (TGF-ß1) is a major regulator of PSC activation and extracellular matrix production. Interleukin-6 (IL-6) has shown to participate in TGF-ß1 production and rat PSC activation. This study aimed to investigate whether IL-6 promotes human PSC activation and collagen 1(Col1) production through the TGF-ß1/Smad pathway. Our results showed that the expression of IL-6 and IL-6R in activated PSCs and macrophages (Mφs) were enhanced in the pancreas of ACP compared to healthy controls and that the mRNA expression of IL-6, IL-6R, TGF-ß1, α-SMA or Col1a1 were significantly increased in the pancreas of ACP, showing positive correlations between elevated IL-6 levels and either TGF-ß1 or α-SMA or Col1a1 levels and between elevated TGF-ß1 levels and α-SMA or Col1a1 levels. In in vitro studies, we identified that IL-6R expression or IL-6 and TGF-ß1 secretions were significantly increased in, respectively, Mφs and PSCs by ethanol (EtOH) or lipopolysaccharide (LPS) stimulation while EtOH- or LPS-induced α-SMA or Col1a1 mRNA and protein production in PSCs were partially blocked by IL-6 antibody. IL-6-induced TGF-ß1 production in PSCs was antagonized by si-IL-6R RNA or by an inhibitor of STAT3. Additionally, IL-6-promoted α-SMA or Col1a1 protein production was blocked by TGF-ß1 antibody and IL-6-induced phosphorylation of Smad2/3 and transcription of α-SMA and Col1a1 mRNA were antagonized by si-TGF-ß1 RNA. Our findings indicate that IL-6 contributes to PSC activation and Col1 production through up-regulation of TGF-ß1/Smad2/3 pathway.


Subject(s)
Collagen Type I, alpha 1 Chain/metabolism , Interleukin-6/metabolism , Pancreatic Stellate Cells/metabolism , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Actins/metabolism , Cells, Cultured , Collagen Type I, alpha 1 Chain/genetics , Ethanol/pharmacology , Humans , Interleukin-6/genetics , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Naphthols/pharmacology , Pancreatic Stellate Cells/drug effects , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology , Up-Regulation/drug effects
10.
Strahlenther Onkol ; 197(1): 27-38, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32705304

ABSTRACT

INTRODUCTION: Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase protein frequently overexpressed in cancer and has been linked to an increase in the stem cell population of tumors, resistance to therapy, and metastatic spread. Pharmacological FAK inhibition in pancreatic cancer has received increased attention over the last few years, either alone or in combination with other therapeutics including chemotherapy and immunotherapy. However, its prognostic value and its role in radioresistance of pancreatic ducal adenocarcinoma (PDAC) is unknown. METHODS AND MATERIALS: Using the TCGA and GTEx databases, we investigated the genetic alterations and mRNA expression levels of PTK2 (the encoding-gene for FAK) in normal pancreatic tissue and pancreatic cancer and its impact on patient survival. Furthermore, we evaluated the expression of FAK and its tyrosine domain Ty-397 in three pancreatic cancer cell lines. We went further and evaluated the role of a commercial FAK tyrosine kinase inhibitor VS-4718 on the viability and radiosensitization of the pancreatic cell lines as well as its effect on the extracellular matrix (ECM) production from the pancreatic stellate cells. Furthermore, we tested the effect of combining radiation with VS-4718 in a three-dimensional (3D) multicellular pancreatic tumor spheroid model. RESULTS: A database analysis revealed a relevant increase in genetic alterations and mRNA expression of the PTK2 in PDAC, which were associated with lower progression-free survival. In vitro, there was only variation in the basal phosphorylation level of FAK in cell lines. VS-4718 radiosensitized pancreatic cell lines only in the presence of ECM-producing pancreatic stellate cells and markedly reduced the ECM production in the stromal cells. Finally, using a 3D multicellular tumor model, the combination of VS-4718 and radiotherapy significantly reduced the growth of tumor aggregates. CONCLUSION: Pharmacological inhibition of FAK in pancreatic cancer could be a novel therapeutic strategy as our results show a radiosensitization effect of VS-4718 in vitro in a multicellular 2D- and in a 3D-model of pancreatic cancer.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Focal Adhesion Kinase 1/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Radiation-Sensitizing Agents/pharmacology , Carcinoma, Pancreatic Ductal/enzymology , Cell Cycle/drug effects , Cell Line, Tumor , Coculture Techniques , Collagen/metabolism , Extracellular Matrix Proteins/metabolism , Histones/analysis , Humans , Kaplan-Meier Estimate , Pancreatic Neoplasms/enzymology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/metabolism , Progression-Free Survival , RNA Interference , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Radiation Tolerance/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/radiation effects , Stromal Cells/drug effects , Tumor Stem Cell Assay
11.
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.

12.
Pancreatology ; 20(7): 1368-1378, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32967795

ABSTRACT

BACKGROUND: Chronic pancreatitis (CP) is a complex inflammatory disorder of the pancreas affecting acinar cells, duct cells, islet cells and inflammatory cells including fibrosis-producing stellate cells. Serum trypsinogen is a biomarkers of acinar cell function. AIM: To define the degree of correlation between low trypsinogen levels as a marker of acinar cell function and variable features of CP. METHODS: Serum samples from previously ascertained and well phenotyped case and control subjects from the North American Pancreatitis Study II (NAPS2) were used to measure serum trypsinogen levels in a commercial laboratory. Control samples were used to define normal ranges and compared with levels in CP patients with defined features. RESULTS: A final cohort of 279 CP patients and 262 controls from the NAPS2 studies were evaluated. In controls trypsinogen had a mean of 34.96 ng/ml and SD = 11.99. Cut-off values for low trypsinogen ranged from <20 to 10 ng/ml and very low trypsinogen at <10 ng/ml. Compared to controls, CP was associated with very low trypsinogen levels (p < 0.0001). Within CP, very low trypsinogen levels correlated with parenchymal loss (pancreatic surgery [p < 0.05]; atrophy with calcifications, [p < 0.001]), EPI (p < 0.01, trend p < 0.001) and diabetes (trend p < 0.01) but not CT-based criteria for fibrosis (pancreatic duct dilation, irregularity, strictures). CONCLUSIONS: Very low serum trypsinogen levels correlate with measures of acinar cell loss including surgical resection, atrophic-calcific CP, diabetes and functional symptoms EPI but not duct morphology criteria. Serum trypsinogen levels correlate with decreased acinar cell function and therefore have biomarker utility clinical management.


Subject(s)
Diabetes Complications/blood , Exocrine Pancreatic Insufficiency/blood , Pancreatitis, Chronic/blood , Pancreatitis, Chronic/diagnostic imaging , Trypsinogen/blood , Acinar Cells , Adult , Aged , Atrophy , Biomarkers/blood , Calcinosis/pathology , Cohort Studies , Exocrine Pancreatic Insufficiency/pathology , Female , Fibrosis , Humans , Male , Middle Aged , Pancreas/pathology , Pancreatic Ducts/pathology , Pancreatitis, Chronic/pathology , Severity of Illness Index , Surveys and Questionnaires , Tomography, X-Ray Computed
13.
Alcohol Clin Exp Res ; 44(4): 777-789, 2020 04.
Article in English | MEDLINE | ID: mdl-32056245

ABSTRACT

Alcohol is a major cause of acute and chronic pancreatitis. There have been some recent advances in the understanding of the mechanisms underlying alcoholic pancreatitis, which include perturbation in mitochondrial function and autophagy and ectopic exocytosis, with some of these cellular events involving membrane fusion soluble N-ethylmaleimide-sensitive factor receptor protein receptor proteins. Although new insights have been unraveled recently, the precise mechanisms remain complex, and their finer details have yet to be established. The overall pathophysiology of pancreatitis involves not only the pancreatic acinar cells but also the stellate cells and duct cells. Why only some are more susceptible to pancreatitis and with increased severity, while others are not, would suggest that there may be undefined protective factors or mechanisms that enhance recovery and regeneration after injury. Furthermore, there are confounding influences of lifestyle factors such as smoking and diet, and genetic background. Whereas alcohol and smoking cessation and a generally healthy lifestyle are intuitively the advice given to these patients afflicted with alcoholic pancreatitis in order to reduce disease recurrence and progression, there is as yet no specific treatment. A more complete understanding of the pathogenesis of pancreatitis from which novel therapeutic targets could be identified will have a great impact, particularly with the stubbornly high fatality (>30%) of severe pancreatitis. This review focuses on the susceptibility factors and underlying cellular mechanisms of alcohol injury on the exocrine pancreas.


Subject(s)
Pancreatitis, Alcoholic/epidemiology , Acetaldehyde/metabolism , Autophagy , Calcium/metabolism , Disease Susceptibility , Endoplasmic Reticulum Stress , Ethanol/metabolism , Exocytosis , Genetic Predisposition to Disease , Humans , Hyperlipidemias/epidemiology , Infections/epidemiology , NAD/metabolism , Obesity/epidemiology , Pancreatitis, Alcoholic/metabolism , Protective Factors , Reactive Oxygen Species/metabolism , Risk Factors , SNARE Proteins/metabolism , Severity of Illness Index , Smoking/epidemiology
14.
J Pathol ; 248(1): 51-65, 2019 05.
Article in English | MEDLINE | ID: mdl-30575030

ABSTRACT

Cancer-associated fibroblasts (CAF) are orchestrators of the pancreatic ductal adenocarcinoma (PDAC) microenvironment. Stromal heterogeneity may explain differential pathophysiological roles of the stroma (pro- versus anti-tumoural) in PDAC. We hypothesised that multiple CAF functional subtypes exist in PDAC, that contribute to stromal heterogeneity through interactions with cancer cells. Using molecular and functional analysis of patient-derived CAF primary cultures, we demonstrated that human PDAC-derived CAFs display a high level of inter- and intra-tumour heterogeneity. We identified at least four subtypes of CAFs based on transcriptomic analysis, and propose a classification for human PDAC-derived CAFs (pCAFassigner). Multiple CAF subtypes co-existed in individual patient samples. The presence of these CAF subtypes in bulk tumours was confirmed using publicly available gene expression profiles, and immunostainings of CAF subtype markers. Each subtype displayed specific phenotypic features (matrix- and immune-related signatures, vimentin and α-smooth muscle actin expression, proliferation rate), and was associated with an assessable prognostic impact. A prolonged exposure of non-tumoural pancreatic stellate cells to conditioned media from cancer cell lines (cancer education experiment) induced a CAF-like phenotype, including loss of capacity to revert to quiescence and an increase in the expression of genes related to CAF subtypes B and C. This classification demonstrates molecular and functional inter- and intra-tumoural heterogeneity of CAFs in human PDAC. Our subtypes overlap with those identified from single-cell analyses in other cancers, and pave the way for the development of therapies targeting specific CAF subpopulations in PDAC. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Humans , Kaplan-Meier Estimate , Pancreatic Neoplasms/genetics , Pancreatic Stellate Cells/pathology , Phenotype , Prognosis , Stromal Cells/pathology , Tumor Cells, Cultured , Tumor Microenvironment
15.
Int J Med Sci ; 17(1): 137-144, 2020.
Article in English | MEDLINE | ID: mdl-31929747

ABSTRACT

Human pancreatic stellate cells (PSCs) play a critical role in fibrogenesis during chronic pancreatitis (CP). However, primary PSCs have a short lifespan in vitro, which seriously affects their use in various applications. We have established a stable immortalized human PSC line (HP-1) by RSV promoter/enhancer-driven SV40 T antigen expression in primary activated human PSCs. HP-1 cells express cytoskeleton proteins including glial fibrillary acidic protein (GFAP), α-smooth muscle actin (α-SMA), vimentin and desmin, and are typical of PSCs, which are high transfeciability and viable in 0.5% serum. The cells express receptors such as TGFßR2, PDGFR, TGF-ß pseudoreceptor Bambi and PPRPγ that are commonly found in PSCs. HP-1 cells are similar to activated human PSCs in that they have enhanced expression of α-SMA, CTGF, Col1 and TIMP-2 mRNAs or proteins, as well as decreased expression of MMP-1/2 mRNAs or proteins in response to TGF-ß1 stimulation. Comparative proteomics revealed 4,537 shared proteins between HP-1 cells and PSCs and no single protein in HP-1 cells versus PSCs. Statistical analysis reveals no significantly difference between HP-1 cells and PSCs in their expression of proteins associated with matrix and matrix remodeling. The similarity between HP-1 cell and PSC is further shown by the finding that only 9 proteins are differentially up-regulated > ± 2-fold in HP-1 cells and 13 proteins are up-regulated > ± 2-fold in PSCs and none of these proteins include ECM proteins, cytokines, growth factors or matrix remodeling regulatory proteins. Therefore, HP-1 cells can be used as an effective tool for the study of PSC-mediated pancreatic fibrosis.


Subject(s)
Cell Proliferation/genetics , Pancreatic Stellate Cells/metabolism , Pancreatitis, Chronic/genetics , Proteomics , Actins/genetics , Cell Line , Connective Tissue Growth Factor/genetics , Fibrosis/genetics , Fibrosis/pathology , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/genetics , Humans , Matrix Metalloproteinase 2/genetics , Pancreas/metabolism , Pancreatic Stellate Cells/pathology , Pancreatitis, Chronic/pathology , Tissue Inhibitor of Metalloproteinase-2/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1/genetics
16.
Adv Exp Med Biol ; 1263: 67-84, 2020.
Article in English | MEDLINE | ID: mdl-32588324

ABSTRACT

As tumor microenvironments share many of the same qualities as chronic wounds, attention is turning to the wound-repair cells that support the growth of cancerous cells. Stellate cells are star-shaped cells that were first discovered in the perisinusoidal spaces in the liver and have been found to support wound healing by the secretion of growth factors and extracellular matrix. They have since been also found to serve a similar function in the pancreas. In both organs, the wound-healing process may become dysregulated and lead to pathological fibrosis (also known as cirrhosis in the liver). In recent years there has been increasing attention paid to the role of these cells in tumor formation and progression. They may be a factor in initiating the first steps of carcinogenesis such as with liver cirrhosis and hepatocellular carcinoma and also contribute to continued tumor growth, invasion, metastasis, evasion of the immune system, and resistance to chemotherapy, in cancers of both the liver and pancreas. In this chapter we aim to review the structure and function of hepatic and pancreatic stellate cells and their contributions to the tumor microenvironment in their respective cancers and also discuss potential new targets for cancer therapy based on our new understanding of these vital components of the tumor stroma.


Subject(s)
Hepatic Stellate Cells/pathology , Liver Neoplasms/pathology , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Tumor Microenvironment , Carcinoma, Hepatocellular/pathology , Humans
17.
Biochem Biophys Res Commun ; 515(3): 493-498, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31171359

ABSTRACT

BACKGROUND: The polysaccharide hyaluronan (HA) is abundant in pancreatic cancer (PC) tissue and promotes pancreatic cancer cell (PCC) motility in vitro. However, it is controversial as to whether high-molecular-weight HA (HMW-HA) or low-molecular weight HA(LMW-HA) is present in the pancreatic cancer stroma and whether PCC or pancreatic stellate cell (PSC) in PC tissue produces HA. We thereby aim to characterize the molecular weight and source of HA in PC tissue that promotes cancer cell motility. METHODS: We analyzed the expression of hyaluronan synthase 2 (HAS2) and the hydrolyzing enzyme hyaluronidase 1 (HYAL1) in PCC lines and pancreatic stellate cells (PSCs) using real-time PCR. HA production in the supernatant of PCC lines and PSCs and in PC tissues was quantitatively and qualitatively examined. Finally, we knocked down HYAL1 expression in one of the PCC line PANC-1 cells and analyzed the impact on cell migration. RESULTS: HAS2 was abundantly expressed in activated PSCs (aPSCs) but less so in quiescent PSCs (qPSCs) and PCC lines. The baseline expression of HYAL1 did not differ among the cell types. The concentration of HMW-HA was higher in the supernatant of aPSCs than in that of PCC lines. Treatment with exogenous HMW-HA promoted PANC-1 cell motility. Knockdown of HYAL1 decreased HMW-HA-promoted PANC-1 cell migration, which was accompanied by a decrease in intracellular HA levels. CONCLUSION: aPSCs are an important source of stromal HMW-HA, which promotes PCC migration in an HYAL1-dependent manner in PC.


Subject(s)
Cell Movement , Hyaluronic Acid/pharmacology , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Paracrine Communication , Signal Transduction , Cell Line, Tumor , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronan Synthases/genetics , Hyaluronan Synthases/metabolism , Hyaluronoglucosaminidase/metabolism , Molecular Weight , Pancreatic Neoplasms/genetics , Pancreatic Stellate Cells/drug effects , Paracrine Communication/drug effects , Signal Transduction/drug effects , Up-Regulation/drug effects
18.
Pancreatology ; 19(1): 196-203, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30393009

ABSTRACT

OBJECTIVES: The microRNA (miRNA) let-7d is linked to the formation of pancreatic cancer-related fibrosis. In this study, the mechanism by which let-7d regulates the activation of the human pancreatic stellate cell (hPSC) was evaluated. METHODS: The transient transfection of a let-7d mimic in the hPSCs was performed, and the altered thrombospondin 1 (THBS1) expression was confirmed by western blotting and real-time qPCR. Targeting of the 3'-untranslated region (UTR) of THBS1 by let-7d was investigated by the luciferase assays. After hPSC transfection using THBS1 siRNA, the fibrosis markers (α-SMA and collagen 1A1) were evaluated by western blotting and real-time qPCR. The correlation between tumor fibrosis and let-7d or THBS1 was estimated using the data from The Cancer Genome Atlas project. Finally, the effects of genistein on the hPSCs were evaluated. RESULTS: We found that a let-7d mimic inhibits THBS1 expression by targeting its 3'-UTR. THBS1 inhibition by siRNA inhibited hPSC activation. An in silico analysis revealed that let-7d and THBS1 expression are negatively correlated. Additionally, let-7d was negatively correlated with the stromal score, while THBS1 was positively correlated with this score. Genistein substantially induced let-7d and decreased the expression of fibrosis marker along with the inhibition of THBS1. CONCLUSIONS: Let-7d inhibited hPSC activation by targeting THBS1. Genistein induced the expression of let-7d and might modulate pancreatic fibrosis.


Subject(s)
MicroRNAs/metabolism , Pancreatic Stellate Cells/metabolism , Thrombospondin 1/metabolism , Biomarkers/metabolism , Down-Regulation , Fibrosis/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Genistein/pharmacology , Humans , MicroRNAs/genetics , Phosphatidylethanolamines , RNA, Messenger , RNA, Small Interfering , Thrombospondin 1/genetics
19.
Pharmacol Res ; 147: 104357, 2019 09.
Article in English | MEDLINE | ID: mdl-31356863

ABSTRACT

Chronic pancreatitis (CP) is characterized by persistent inflammation and fibrosis of the pancreas. To date, no clinical therapy is available to reverse the inflammatory damage or pancreatic fibrosis associated with CP. This study systematically investigated the effect of Dasatinib, a multiple tyrosine kinases (TKs) inhibitor, on pancreatic fibrosis and inflammation in vivo and in vitro. We found that Dasatinib notably ameliorated pancreatic fibrosis and infiltration of macrophages in a model of caerulein-induced murine CP. Further RNA-seq and phosphoproteomic analysis and in vitro validation assays indicated that Dasatinib exerted a marked inhibition on the proliferation and activation of PSCs, which may be resulted from increased GSK3ß-mediated ß-catenin cytosol retention by inhibiting upstream multiple TKs (such as PDGFR and Src) and MAPK cascades (including ERK1/2 and p38 MAPK). In addition, Dasatinib significantly restrained both the M1 and M2 polarization of macrophages, and impeded its recruitment and crosstalk with PSCs. Our findings indicated that Dasatinib is a potential anti-inflammatory and anti-fibrotic therapeutic strategy for CP.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Dasatinib/therapeutic use , Pancreatitis, Chronic/drug therapy , Animals , Cell Movement/drug effects , Ceruletide , Fibrosis , Male , Mice , Mice, Inbred C57BL , Pancreas/drug effects , Pancreas/pathology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/metabolism , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/pathology , RAW 264.7 Cells , Wound Healing/drug effects
20.
Endocr J ; 66(12): 1053-1062, 2019 Dec 25.
Article in English | MEDLINE | ID: mdl-31474673

ABSTRACT

In this study, we aim to explore the anti-tumor effect of liraglutide (Lira), an anti-diabetic medicine, on pancreatic cancer cell PANC-1 co-cultured with or without pancreatic stellate cells (PSCs). The chemical count kit-8 and Annexin V-FITC apoptosis detection were conducted to investigate the effect of Lira on cell viability and proliferation of PANC-1 with or without PSCs co-culture. Then, the wound healing and transwell experiments were performed to explore the influence of Lira on PANC-1 cells' migration and invasion capabilities. To identify the potential action mechanism of Lira on PANC-1, the expression of E-cadherin and N-cadherin and the intracellular calcium content in PANC-1, after Lira administration, were detected. The results indicated that Lira in 100 and 1,000 nmol/L, effectively decreased the cell viability and dose-dependently promoted cell apoptosis of PANC-1 co-cultured with or without PSCs. Lira significantly reduced the migration and invasion of PANC-1 and also reduced the inducing effect of PSCs to PANC-1. Lira effectively induced the expression of E-cadherin and suppressed the expression of N-cadherin with a dose-dependent manner. Otherwise, Lira significantly reduced the abnormal high content of calcium in PANC-1 and also weakened the elevation of calcium in PANC-1 induced by cell-cell interaction. The current study firstly indicated that Lira suppressed the cell proliferation, migration and invasion of PANC-1 with or without PSCs co-culture. This effect was partially due to the calcium modulation of Lira and its influence on Ca2+-binding proteins, such as E-cadherin and N-cadherin.


Subject(s)
Calcium/analysis , Liraglutide/pharmacology , Neoplasm Metastasis/prevention & control , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/physiology , Antineoplastic Agents , Apoptosis/drug effects , Cadherins/analysis , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Coculture Techniques , Humans , Hypoglycemic Agents/pharmacology , Neoplasm Invasiveness/prevention & control , Pancreatic Stellate Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL