Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Diabet Med ; 41(6): e15279, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38185936

ABSTRACT

AIMS: Evidence is accumulating of the therapeutic benefits of mesenchymal stromal cells (MSCs) in diabetes-related conditions. We have identified a novel population of stromal cells within islets of Langerhans - islet stellate cells (ISCs) - which have a similar morphology to MSCs. In this study we characterize mouse ISCs and compare their morphology and function to MSCs to determine whether ISCs may also have therapeutic potential in diabetes. METHODS: ISCs isolated from mouse islets were compared to mouse bone marrow MSCs by analysis of cell morphology; expression of cell-surface markers and extracellular matrix (ECM) components; proliferation; apoptosis; paracrine activity; and differentiation into adipocytes, chondrocytes and osteocytes. We also assessed the effects of co-culture with ISCs or MSCs on the insulin secretory capacity of islet beta cells. RESULTS: Although morphological similar, ISCs were functionally distinct from MSCs. Thus, ISCs were less proliferative and more apoptotic; they had different expression levels of important paracrine factors; and they were less efficient at differentiation down multiple lineages. Co-culture of mouse islets with ISCs enhanced glucose induced insulin secretion more effectively than co-culture with MSCs. CONCLUSIONS: ISCs are a specific sub-type of islet-derived stromal cells that possess biological behaviors distinct from MSCs. The enhanced beneficial effects of ISCs on islet beta cell function suggests that they may offer a therapeutic target for enhancing beta cell functional survival in diabetes.


Subject(s)
Cell Differentiation , Coculture Techniques , Insulin-Secreting Cells , Islets of Langerhans , Mesenchymal Stem Cells , Animals , Mice , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Insulin-Secreting Cells/cytology , Cell Differentiation/physiology , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/physiology , Cell Proliferation/physiology , Insulin/metabolism , Cells, Cultured , Insulin Secretion/physiology , Mice, Inbred C57BL , Male , Apoptosis/physiology
2.
Exp Cell Res ; 411(1): 113000, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34958764

ABSTRACT

The accumulation of activated myofibroblastic pancreatic stellate cells (MF-PSCs) induces pancreatic cancer desmoplasia. These MF-PSCs are derived from quiescent pancreatic stellate cells (Q-PSCs). MF-PSCs in pancreatic cancer tend to glycolysis. However, increased glycolysis alone could not be sufficient for the increased metabolic demands of MF-PSCs. Yap and Myc signaling activation is involved in pancreatic cancer metabolism. Since elucidating the metabolic processes of MF-PSCs may be a promising strategy to suppress pancreatic cancer desmoplasia, we explored whether glutaminolysis meets the bioenergetic and biosynthetic demands of Q-PSCs converted into MF-PSCs and whether this is mediated by Yap signaling to Myc. In this study, we found that during the transdifferentiation of Q-PSCs into MF-PSCs, glutaminolysis regulatory genes were upregulated, and suppression of glutaminolysis inhibited transdifferentiation. Disrupting glutaminolysis in MF-PSCs inhibited cell growth, mitochondrial respiration, and fibrogenesis, while treatment of MF-PSCs with DKG (a glutaminolysis metabolite) reversed these activities. The expression of glutaminase (GLS1), a rate-limiting enzyme in glutaminolysis, was upregulated by Yap overexpression. Yap upregulates Myc to regulate the expression of GLS1 in MF-PSCs. Yap and Myc inhibitors disrupted glutaminolysis and inhibited myofibroblastic activities in PSCs. Thus, Yap-Myc signaling controls glutaminolysis to activate PSCs and might be a therapeutic target for pancreatic cancer desmoplasia.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Transdifferentiation , Glutamine/metabolism , Glycolysis , Pancreatic Stellate Cells/cytology , Pancreatic Stellate Cells/physiology , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , Cell Cycle Proteins/genetics , Cell Proliferation , Humans , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction , Transcription Factors/genetics
3.
Front Endocrinol (Lausanne) ; 12: 695467, 2021.
Article in English | MEDLINE | ID: mdl-34566887

ABSTRACT

Background: It has been demonstrated that activated islet stellate cells (ISCs) play a critical role in islet fibrogenesis and significantly contribute to the progression of type 2 diabetes mellitus. However, the key molecules responsible for ISCs activation have not yet been determined. This study aimed to identify the potential key genes involved in diabetes-induced activation of ISCs. Method: Stellate cells were isolated from three 10-week-old healthy male Wistar rats and three Goto-Kakizaki (GK) rats. Cells from each rat were primary cultured under the same condition. A Genome-wide transcriptional sequence of stellate cells was generated using the Hiseq3000 platform. The identified differentially expressed genes were validated using quantitative real-time PCR and western blotting in GK rats, high fat diet (HFD) rats, and their controls. Results: A total of 204 differentially expressed genes (DEGs) between GK. ISCs and Wistar ISCs (W.ISCs) were identified, accounting for 0.58% of all the 35,362 genes detected. After the Gene Ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) enrichment analyses, the mRNA levels of these genes were further confirmed by real-time PCR in cultured ISCs. We then selected Fos, Pdpn, Bad as the potential key genes for diabetes-induced activation of ISCs. Finally, we confirmed the protein expression levels of FOS, podoplanin, and Bad by western blotting and immunofluorescence in GK rats, HFD rats, and their controls. The results showed that the expression level of FOS was significantly decreased, while podoplanin and Bad were significantly increased in GK.ISCs and HFD rats compared with controls, which were consistent with the expression of α-smooth muscle actin. Conclusions: A total of 204 DEGs were found between the GK.ISCs and W.ISCs. After validating the expression of potential key genes from GK rats and HFD rats, Fos, Pdpn, and Bad might be potential key genes involved in diabetes-induced activation of ISCs.


Subject(s)
Islets of Langerhans/physiology , Pancreas/pathology , Pancreatic Stellate Cells/physiology , Transcriptome , Animals , Cell Proliferation/genetics , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Disease Progression , Fibrosis/genetics , Islets of Langerhans/metabolism , Male , Organ Specificity/genetics , Pancreas/metabolism , Pancreatic Stellate Cells/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar
4.
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
5.
Cells ; 10(8)2021 07 29.
Article in English | MEDLINE | ID: mdl-34440697

ABSTRACT

Pancreatic stellate cells (PSCs) are important pancreatic fibrogenic cells that interact with pancreatic cancer cells to promote the progression of pancreatic ductal adenocarcinoma (PDAC). In the tumor microenvironment (TME), several factors such as cytokines and nucleotides contribute to this interplay. Our aim was to investigate whether there is an interaction between IL-6 and nucleotide signaling, in particular, that mediated by the ATP-sensing P2X7 receptor (P2X7R). Using human cell lines of PSCs and cancer cells, as well as primary PSCs from mice, we show that ATP is released from both PSCs and cancer cells in response to mechanical and metabolic cues that may occur in the TME, and thus activate the P2X7R. Functional studies using P2X7R agonists and inhibitors show that the receptor is involved in PSC proliferation, collagen secretion and IL-6 secretion and it promotes cancer cell migration in a human PSC-cancer cell co-culture. Moreover, conditioned media from P2X7R-stimulated PSCs activated the JAK/STAT3 signaling pathway in cancer cells. The monoclonal antibody inhibiting the IL-6 receptor, Tocilizumab, inhibited this signaling. In conclusion, we show an important mechanism between PSC-cancer cell interaction involving ATP and IL-6, activating P2X7 and IL-6 receptors, respectively, both potential therapeutic targets in PDAC.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Carcinoma, Pancreatic Ductal/metabolism , Interleukin-6/metabolism , Pancreatic Stellate Cells/metabolism , Receptors, Purinergic P2X7/metabolism , STAT3 Transcription Factor/metabolism , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/physiopathology , Cell Communication , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Male , Mice , Pancreatic Stellate Cells/physiology , Signal Transduction , Tumor Microenvironment
6.
Pharmacol Res ; 168: 105595, 2021 06.
Article in English | MEDLINE | ID: mdl-33823219

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant lethal disease due to its asymptomatic at its early lesion of the disease and drug resistance. Target therapy associated with molecular pathways so far seems not to produce reasonable outcomes. Understanding of the molecular mechanisms underlying inflammation-initiated tumorigenesis may be helpful for development of an effective therapy of the disease. A line of studies showed that pancreatic tumorigenesis was resulted from pancreatitis, which was caused synergistically by various pancreatic cells. This review focuses on those players and their possible clinic implications, such as exocrine acinar cells, ductal cells, and various stromal cells, including pancreatic stellate cells (PSCs), macrophages, lymphocytes, neutrophils, mast cells, adipocytes and endothelial cells, working together with each other in an inflammation-mediated microenvironment governed by a myriad of cellular signaling networks towards PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/etiology , Pancreatic Neoplasms/etiology , Pancreatitis/complications , Acinar Cells/physiology , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/drug therapy , Hedgehog Proteins/physiology , Humans , MAP Kinase Signaling System/physiology , Macrophages/physiology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/drug therapy , Pancreatic Stellate Cells/physiology , Signal Transduction , Tumor Microenvironment
7.
J Diabetes Investig ; 11(2): 268-280, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31872946

ABSTRACT

There are numerous lines of clinical evidence that inhibition of the renin-angiotensin system (RAS) can prevent and delay the development of diabetes. Also, the role of RAS in the pathogenesis of diabetes, including insulin resistance and ß-cell dysfunction, has been extensively investigated. Nevertheless, this role had not yet been fully shown. A variety of possible protective mechanisms for RAS blockers in the regulation of glucose homeostasis have been suggested. However, the direct effect on pancreatic islet fibrosis has only recently been spotlighted. Various degrees of islet fibrosis are often observed in the islets of patients with type 2 diabetes mellitus, which can be associated with a decrease in ß-cell mass and function in these patients. Pancreatic stellate cells are thought to be deeply involved in this islet fibrosis. In this process, the activation of RAS in islets is shown to transform quiescent pancreatic stellate cells into the activated form, stimulates their proliferation and consequently leads to islet fibrotic destruction. In this article, we introduce existing clinical and experimental evidence for diabetes prevention through inhibition of RAS, and review the responsible local RAS signaling pathways in pancreatic stellate cells. Finally, we propose possible targets for the prevention of islet fibrosis.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Insulin-Secreting Cells/physiology , Islets of Langerhans/physiopathology , Pancreatic Stellate Cells/physiology , Renin-Angiotensin System , Animals , Diabetes Mellitus, Type 2/therapy , Humans
8.
PLoS Biol ; 17(9): e3000418, 2019 09.
Article in English | MEDLINE | ID: mdl-31513574

ABSTRACT

Damaged acinar cells play a passive role in activating pancreatic stellate cells (PSCs) via recruitment of immune cells that subsequently activate PSCs. However, whether acinar cells directly contribute to PSC activation is unknown. Here, we report that the Hippo pathway, a well-known regulator of proliferation, is essential for suppression of expression of inflammation and fibrosis-associated genes in adult pancreatic acinar cells. Hippo inactivation in acinar cells induced yes-associated protein 1 (YAP1)/transcriptional coactivator with PDZ binding motif (TAZ)-dependent, irreversible fibrosis and inflammation, which was initiated by Hippo-mediated acinar-stromal communications and ameliorated by blocking YAP1/TAZ target connective tissue growth factor (CTGF). Hippo disruption promotes acinar cells to secrete fibroinflammatory factors and induce stromal activation, which precedes acinar proliferation and metaplasia. We found that Hippo disruption did not induce cell-autonomous proliferation but primed acinar cells to exogenous pro-proliferative stimuli, implying a well-orchestrated scenario in which Hippo signaling acts as an intrinsic link to coordinate fibroinflammatory response and proliferation for maintenance of the tissue integrity. Our findings suggest that the fibroinflammatory program in pancreatic acinar cells is suppressed under normal physiological conditions. While transient activation of inflammatory gene expression during tissue injury may contribute to the control of damage and tissue repair, its persistent activation may result in tissue fibrosis and failure of regeneration.


Subject(s)
Acinar Cells/metabolism , Pancreas/metabolism , Pancreatitis/etiology , Protein Serine-Threonine Kinases/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Fibrosis , Hippo Signaling Pathway , Mice , Pancreas/pathology , Pancreatic Stellate Cells/physiology , Pancreatitis/metabolism , Pancreatitis/pathology , Signal Transduction , Transcription Factors/metabolism , YAP-Signaling Proteins
9.
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
10.
Exp Cell Res ; 384(1): 111606, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31493386

ABSTRACT

Pancreatic stellate cells (PSCs) have been recognized as key mediators of pancreatic fibrosis, a characteristic feature of chronic pancreatitis (CP). As a cullin-based E3 ubiquitin ligase, speckle-type POZ protein (SPOP) has been identified to participate in tumorigenesis and organ development. However, its biological role in CP remains unknown. Therefore, this study sought to investigate the changed expression of SPOP in CP and to examine the effect on mice PSCs activation of SPOP. We found that SPOP was downregulated in the pancreatic tissues of cerulein-induced CP mice. siRNA-mediated knockdown of SPOP led to significant promotion in primary PSCs activity by activating the nuclear factor-kappaB (NF-κB)/interleukin-6 (IL-6) signaling pathway. In addition, we examined the effects of Fas-associated death domain (FADD), a proven SPOP substrate that activates NF-κB, on the regulation of PSCs activation. We found that FADD was downregulated by SPOP via interaction-mediated degradation, and was upregulated during PSCs activation. The promotion of PSCs activation in knocking down SPOP with siSPOP-1 were counteracted by knocking down FADD. The results suggest that the SPOP-induced inhibition of PSCs activation partially depended on FADD. These results highlight the importance of SPOP in CP and provide a potential target for therapeutic intervention.


Subject(s)
Ceruletide/pharmacology , Fas-Associated Death Domain Protein/metabolism , Pancreas/metabolism , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/physiology , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/metabolism , Animals , Cells, Cultured , Death Domain/drug effects , Death Domain/physiology , Down-Regulation/drug effects , Fibrosis/metabolism , Interleukin-6/metabolism , Mice , NF-kappa B/metabolism , Pancreas/physiology , Pancreatic Stellate Cells/drug effects , Signal Transduction/drug effects , Up-Regulation/drug effects
11.
PLoS Genet ; 15(2): e1007971, 2019 02.
Article in English | MEDLINE | ID: mdl-30763305

ABSTRACT

The Wilms' tumor suppressor gene (Wt1) encodes a zinc finger transcription factor that plays an essential role in the development of kidneys, gonads, spleen, adrenals and heart. Recent findings suggest that WT1 could also be playing physiological roles in adults. Systemic deletion of WT1 in mice provokes a severe deterioration of the exocrine pancreas, with mesothelial disruption, E-cadherin downregulation, disorganization of acinar architecture and accumulation of ascitic transudate. Despite this extensive damage, pancreatic stellate cells do not become activated and lose their canonical markers. We observed that pharmacological induction of pancreatitis in normal mice provokes de novo expression of WT1 in pancreatic stellate cells, concomitant with their activation. When pancreatitis was induced in mice after WT1 ablation, pancreatic stellate cells expressed WT1 and became activated, leading to a partial rescue of the acinar structure and the quiescent pancreatic stellate cell population after recovery from pancreatitis. We propose that WT1 modulates through the RALDH2/retinoic acid axis the restabilization of a part of the pancreatic stellate cell population and, indirectly, the repair of the pancreatic architecture, since quiescent pancreatic stellate cells are required for pancreas stability and repair. Thus, we suggest that WT1 plays novel and essential roles for the homeostasis of the adult pancreas and, through its upregulation in pancreatic stellate cells after a damage, for pancreatic regeneration. Due to the growing importance of the pancreatic stellate cells in physiological and pathophysiological conditions, these novel roles can be of translational relevance.


Subject(s)
Genes, Wilms Tumor , Pancreas/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Aldehyde Oxidoreductases/metabolism , Animals , Cell Lineage/genetics , Ceruletide/toxicity , Disease Models, Animal , Epithelium/metabolism , Gene Expression , Homeostasis/genetics , Homeostasis/physiology , Humans , Mice , Mice, Knockout , Mice, Transgenic , Pancreatic Stellate Cells/pathology , Pancreatic Stellate Cells/physiology , Pancreatitis/chemically induced , Pancreatitis/genetics , Pancreatitis/physiopathology , Regeneration/genetics , Regeneration/physiology , Repressor Proteins/deficiency , Tissue Distribution , Translational Research, Biomedical , Tretinoin/metabolism , WT1 Proteins
12.
Am J Med Sci ; 357(1): 16-22, 2019 01.
Article in English | MEDLINE | ID: mdl-30466735

ABSTRACT

BACKGROUND: This study aimed to investigate the correlation between pancreatic stellate cell activation, matrix metallopeptidase 2 (MMP2) expression and lymph node metastasis in pancreatic carcinoma. METHODS: Alpha-smooth muscle actin (ACTA2), Desmin (DES) and MMP2 were detected in 40 pancreatic carcinoma patients and 10 cases of normal pancreas tissues using immunohistochemistry. Then MMP2 and ACTA2 expression profiles in pancreatic cancer were obtained from UCSC (University of California, Santa Cruz) and SurvExpress. RESULTS: A total of 67.5% and 55.0% of cases positively expressed ACTA2 and DES in pancreatic carcinoma, respectively. MMP2 in pancreatic carcinoma was expressed in 55.0% of cases, and there were significant differences between the lymph node metastasis group and the lymph node nonmetastasis group, as well as invasion and noninvasion to the peripheral tissue group (P < 0.01). High throughput sequencing databases verified that ACTA2 and MMP2 gene expression were both upregulated in pancreatic carcinoma tissues. CONCLUSIONS: The coexpression of ACTA2 and DES was related to the expression of MMP2, and positively correlated with lymph node metastasis. Activation of pancreatic stellate cells may promote the expression of MMP2 and enhance the invasion and metastasis of pancreatic carcinoma.


Subject(s)
Actins/genetics , Desmin/genetics , Lymphatic Metastasis/pathology , Matrix Metalloproteinase 2/genetics , Pancreas/pathology , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/physiology , Actins/metabolism , Adult , Aged , Desmin/metabolism , Female , Humans , Lymphatic Metastasis/genetics , Male , Matrix Metalloproteinase 2/metabolism , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms
13.
Biomed Res Int ; 2018: 4283673, 2018.
Article in English | MEDLINE | ID: mdl-30519576

ABSTRACT

BACKGROUND: Pancreatic cancer is a fatal malignancy with a poor prognosis. The interactions between tumor cells and stromal cells contribute to cancer progression. Pancreatic stellate cells (PSCs) play a key role in tumor-stroma crosstalk of pancreatic cancer. The in-depth exploration for tumor-stroma crosstalk is helpful to develop novel therapeutic strategies. Our aim was to identify the potential core genes and pathways in tumor-stroma crosstalk. METHODS: 3 microarray datasets were from Gene Expression Omnibus (GEO). Differentially expressed genes (DEGs) were screened through bioinformatics analysis. Gene Ontology (GO) enrichment, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment, and protein-protein interaction (PPI) network were used to obtain the biological roles of DEGs. The top 15 DEGs were explored by principal component analysis. We validated the top 15 DEGs expression in the tumor-stroma crosstalk model in which PSCs were treated with the mixture of Aspc-1 and Panc-1 supernatant. RESULTS: A total of 221 genes were filtered as DEGs for tumor-stroma crosstalk. The results of principal component analysis for the top 15 DEGs can distinguish three groups. According to the KEGG enrichment, there were 8, 7, and 7 DEGs enriched in cancer related pathway, PI3K-Akt signaling pathway, and microRNAs, respectively. In the tumor-stroma crosstalk model, significant differences can be validated in the AKAP12, CLDN1, CP, FKBP1A, LAMB3, LSM4, MTMR3, PRKARIA, YWHAZ, and JUND expressions. CONCLUSIONS: These results identified the potential core genes and pathways in pancreatic cancer for tumor-stroma crosstalk, which could provide potential targets for the treatment of pancreatic cancer.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Pancreatic Neoplasms/genetics , Stromal Cells/physiology , Animals , Biomarkers, Tumor/genetics , Computational Biology/methods , Databases, Genetic , Gene Expression Profiling/methods , Gene Ontology , Gene Regulatory Networks/genetics , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Pancreatic Stellate Cells/physiology , Phosphatidylinositol 3-Kinases/genetics , Principal Component Analysis/methods , Protein Interaction Maps/genetics , Signal Transduction/genetics
14.
Theranostics ; 8(18): 5072-5087, 2018.
Article in English | MEDLINE | ID: mdl-30429887

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) constitutes one of the most challenging lethal tumors and has a very poor prognosis. In addition to cancer cells, the tumor microenvironment created by a repertoire of resident and recruited cells and the extracellular matrix also contribute to the acquisition of hallmarks of cancer. Among these factors, cancer-associated fibroblasts (CAFs) are critical components of the tumor microenvironment. CAFs originate from the activation of resident fibroblasts and pancreatic stellate cells, the differentiation of bone marrow-derived mesenchymal stem cells and epithelial-to-mesenchymal transition. CAFs acquire an activated phenotype via various cytokines and promote tumor proliferation and growth, accelerate invasion and metastasis, induce angiogenesis, promote inflammation and immune destruction, regulate tumor metabolism, and induce chemoresistance; these factors contribute to the acquisition of major hallmarks of PDAC. Therefore, an improved understanding of the impact of CAFs on the major hallmarks of PDAC will highlight the diagnostic and therapeutic values of these targeted cells.


Subject(s)
Adenocarcinoma/pathology , Cancer-Associated Fibroblasts/pathology , Carcinoma, Ductal/pathology , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Adenocarcinoma/physiopathology , Animals , Carcinoma, Ductal/physiopathology , Disease Models, Animal , Epithelial-Mesenchymal Transition , Humans , Mesenchymal Stem Cells/physiology , Pancreatic Neoplasms/physiopathology , Pancreatic Stellate Cells/physiology
15.
Exp Cell Res ; 373(1-2): 132-144, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30321515

ABSTRACT

Pancreatic stellate cells (PSCs), when activated, are characterized by proliferation and collagen synthesis, and contribute to extracellular matrix deposition in pancreatic fibrosis. Concomitantly, fibrosis is linked with the loss of PTEN (phosphatase and tensin homolog) protein in several organs. This study investigated the association between PTEN protein levels and the activated or apoptotic status of PSCs in a rat model of chronic pancreatitis. In addition, the activation status and biological behaviors of culture-activated PSCs were analyzed after lentiviral transfection with wildtype or mutant (G129E) PTEN for upregulation, or PTEN short hairpin RNA for downregulation, of PTEN. In vivo, PTEN levels gradually decreased during pancreatic fibrosis, which positively correlated with apoptosis of activated PSCs, but negatively with PSC activation. In vitro, activated PSCs with wildtype PTEN showed less proliferation, migration, and collagen synthesis compared with control PSCs, and greater numbers were apoptotic; activated PSCs with mutant PTEN showed similar, but weaker, effects. Furthermore, AKT and FAK/ERK signaling was involved in this process. In summary, activated PSCs during pancreatic fibrosis in vivo have lower levels of PTEN. In vitro, PTEN appears to prevent PSCs from further activation and promotes apoptosis through regulation of the AKT and FAK/ERK pathways.


Subject(s)
PTEN Phosphohydrolase/metabolism , Pancreas/pathology , Pancreatic Stellate Cells/enzymology , Animals , Apoptosis , Cell Movement , Cell Proliferation , Cells, Cultured , Collagen/metabolism , Fibrosis , Humans , Male , Pancreas/enzymology , Pancreatic Stellate Cells/cytology , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/physiology , Rats, Wistar , Signal Transduction
16.
J Physiol ; 596(14): 2663-2678, 2018 07.
Article in English | MEDLINE | ID: mdl-29424931

ABSTRACT

KEY POINTS: Ca2+ signalling in different cell types in exocrine pancreatic lobules was monitored simultaneously and signalling responses to various stimuli were directly compared. Ca2+ signals evoked by K+ -induced depolarization were recorded from pancreatic nerve cells. Nerve cell stimulation evoked Ca2+ signals in acinar but not in stellate cells. Stellate cells are not electrically excitable as they, like acinar cells, did not generate Ca2+ signals in response to membrane depolarization. The responsiveness of the stellate cells to bradykinin was markedly reduced in experimental alcohol-related acute pancreatitis, but they became sensitive to stimulation with trypsin. Our results provide fresh evidence for an important role of stellate cells in acute pancreatitis. They seem to be a critical element in a vicious circle promoting necrotic acinar cell death. Initial trypsin release from a few dying acinar cells generates Ca2+ signals in the stellate cells, which then in turn damage more acinar cells causing further trypsin liberation. ABSTRACT: Physiological Ca2+ signals in pancreatic acinar cells control fluid and enzyme secretion, whereas excessive Ca2+ signals induced by pathological agents induce destructive processes leading to acute pancreatitis. Ca2+ signals in the peri-acinar stellate cells may also play a role in the development of acute pancreatitis. In this study, we explored Ca2+ signalling in the different cell types in the acinar environment of the pancreatic tissue. We have, for the first time, recorded depolarization-evoked Ca2+ signals in pancreatic nerves and shown that whereas acinar cells receive a functional cholinergic innervation, there is no evidence for functional innervation of the stellate cells. The stellate, like the acinar, cells are not electrically excitable as they do not generate Ca2+ signals in response to membrane depolarization. The principal agent evoking Ca2+ signals in the stellate cells is bradykinin, but in experimental alcohol-related acute pancreatitis, these cells become much less responsive to bradykinin and then acquire sensitivity to trypsin. Our new findings have implications for our understanding of the development of acute pancreatitis and we propose a scheme in which Ca2+ signals in stellate cells provide an amplification loop promoting acinar cell death. Initial release of the proteases kallikrein and trypsin from dying acinar cells can, via bradykinin generation and protease-activated receptors, induce Ca2+ signals in stellate cells which can then, possibly via nitric oxide generation, damage more acinar cells and thereby cause additional release of proteases, generating a vicious circle.


Subject(s)
Acinar Cells/physiology , Calcium Signaling , Calcium/metabolism , Pancreas, Exocrine/physiology , Pancreatic Stellate Cells/physiology , Pancreatitis/physiopathology , Acinar Cells/cytology , Acinar Cells/drug effects , Acinar Cells/metabolism , Alcohols/toxicity , Animals , Bradykinin/pharmacology , Cells, Cultured , Mice , Mice, Inbred C57BL , Pancreas, Exocrine/cytology , Pancreas, Exocrine/drug effects , Pancreas, Exocrine/metabolism , Pancreatic Stellate Cells/cytology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/metabolism , Pancreatitis/chemically induced , Pancreatitis/metabolism , Vasodilator Agents/pharmacology
17.
Cancer Lett ; 412: 143-154, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29061505

ABSTRACT

Specific cell populations leading the local invasion of cancer are called "leading cells". However, the underlying mechanisms are unclear. Here, we identified leading cells in pancreatic cancer and determined how these cells lead and promote cancer cell invasion in the extracellular matrix (ECM). Using three-dimensional matrix remodeling assay, we found that pancreatic stellate cells (PSCs) frequently invaded the collagen matrix with pancreatic cancer cells (PCCs), which invaded behind the invading PSCs. In addition, invading PSCs changed the alignment of collagen fibers, resulting in ECM remodeling and an increase in the parallel fibers along the direction of invading PSCs. Endo180 expression was higher in PSCs than in PCCs, Endo180 knockdown in PSCs attenuated the invasive abilities of PSCs and co-cultured PCCs, and decreased the expression level of phosphorylated myosin light chain 2 (MLC2). In mouse models, Endo180-knockdown PSCs suppressed tumor growth and changes in collagen fiber orientation in co-transplantation with PCCs. Our findings suggest that PSCs lead the local invasion of PCCs by physically remodeling the ECM, possibly via the function of Endo180, which reconstructs the actin cell skeleton by phosphorylation of MLC2.


Subject(s)
Extracellular Matrix/chemistry , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/physiology , Receptors, Mitogen/physiology , Cardiac Myosins/metabolism , Cell Line, Tumor , Collagen/chemistry , Humans , Myosin Light Chains/metabolism , Neoplasm Invasiveness , Phosphorylation
18.
J Chin Med Assoc ; 81(5): 437-443, 2018 May.
Article in English | MEDLINE | ID: mdl-29129515

ABSTRACT

BACKGROUND: The pathogenesis of chronic pancreatitis (CP) is a complex process of interaction between tissue injury and repair, which involves microcirculatory disturbance. Amygdalin, an effective component extracted from Semen Persicae (a kind of Chinese herbal medicine), can decrease blood viscosity and improve microcirculation. In this study, we investigated the therapeutic effects of amygdalin on pancreatic fibrosis in rats with CP. METHODS: The rat CP model was induced by injecting dibutyltin dichloride (DBTC) into the right caudal vein. Amygdalin was administrated via the penile vein at a dose of 10 mg/(kg d) from the next day, after the induction of CP, once a day for the previous 3 days, and then once every 2 days, until the end of the experiment. Body weight was observed every 7 days. Pancreatic blood flow and histopathological changes were assessed at 28 days. The activation of pancreatic stellate cells (PSCs) was estimated by the expression of α-smooth muscle actin (α-SMA). At the same time, the expression of platelet-derived growth factor-BB (PDGF-BB), transforming growth factor ß-1 (TGFß-1), endothelin-1 (ET-1), and calcitonin gene-related peptide (CGRP) of pancreatic tissues were detected. RESULTS: Treatment of CP rats with amygdalin improved body weight and pancreatic blood flow, as well as alleviated pancreatic fibrosis and acinar destruction, accompanied by the down-regulation of the expressions of α-SMA, PDGF-BB, TGFß-1, and ET-1, and the up-regulation of the CGRP's expression. CONCLUSION: Amygdalin could reduce the production of pro-fibrotic cytokines, inhibit the activation of PSCs, and attenuate pancreatic fibrosis in a rat with CP. The mechanism probably includes improving microcirculatory disturbance by regulating the production of ET-1 and CGRP.


Subject(s)
Amygdalin/pharmacology , Calcitonin Gene-Related Peptide/genetics , Endothelin-1/genetics , Gene Expression Regulation/drug effects , Microcirculation/drug effects , Pancreas/drug effects , Pancreatitis, Chronic/drug therapy , Actins/genetics , Amygdalin/therapeutic use , Animals , Becaplermin/genetics , Fibrosis , Male , Pancreas/blood supply , Pancreas/pathology , Pancreatic Stellate Cells/physiology , Rats , Rats, Wistar
19.
Sci Rep ; 7(1): 5219, 2017 07 12.
Article in English | MEDLINE | ID: mdl-28701767

ABSTRACT

The regulation of nuclear state by the cytoskeleton is an important part of cellular function. Actomyosin stress fibres, microtubules and intermediate filaments have distinct and complementary roles in integrating the nucleus into its environment and influencing its mechanical state. However, the interconnectedness of cytoskeletal networks makes it difficult to dissect their individual effects on the nucleus. We use simple image analysis approaches to characterize nuclear state, estimating nuclear volume, Poisson's ratio, apparent elastic modulus and chromatin condensation. By combining them with cytoskeletal quantification, we assess how cytoskeletal organization regulates nuclear state. We report for a number of cell types that nuclei display auxetic properties. Furthermore, stress fibres and intermediate filaments modulate the mechanical properties of the nucleus and also chromatin condensation. Conversely, nuclear volume and its gross morphology are regulated by intracellular outward pulling forces exerted by myosin. The modulation exerted by the cytoskeleton onto the nucleus results in changes that are of similar magnitude to those observed when the nucleus is altered intrinsically, inducing chromatin decondensation or cell differentiation. Our approach allows pinpointing the contribution of distinct cytoskeletal proteins to nuclear mechanical state in physio- and pathological conditions, furthering our understanding of a key aspect of cellular behaviour.


Subject(s)
Actomyosin/metabolism , Cell Nucleus/physiology , Chromatin/physiology , Cytoskeleton/physiology , Mesenchymal Stem Cells/physiology , Pancreatic Stellate Cells/physiology , Stress, Mechanical , Vimentin/metabolism , Animals , Bone Marrow/growth & development , Cell Differentiation , Cells, Cultured , Human Umbilical Vein Endothelial Cells , Humans , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Microtubules/physiology , Pancreatic Stellate Cells/cytology
20.
Curr Opin Gastroenterol ; 33(5): 383-389, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28682796

ABSTRACT

PURPOSE OF REVIEW: This report reviews recent aspects of pancreatitis immunology and environmental factors that link to development and progression of disease. RECENT FINDINGS: Limited human and animal model studies have recently attempted to understand immune mechanisms that lead to the pathogenesis of acute and chronic pancreatitis. Based on these studies innate immune responses emerge as critical elements in disease pathogenesis and severity of inflammation. The immune basis for environmental factors such as smoking, which are highly associated with disease progression highlight novel cross talk mechanisms between immune and nonimmune pancreatic cells such as the pancreatic stellate cells. SUMMARY: Better understanding of immune responses and signaling pathways are emerging as important contributors in pancreatitis development and progression. Such mechanisms are likely to offer future targetable therapies that can either halt or reverse disease progression.


Subject(s)
Environmental Exposure/adverse effects , Inflammation/immunology , Pancreatitis/immunology , Alcoholism/complications , Animals , Disease Models, Animal , Disease Progression , Humans , Immunity, Innate , Inflammation/physiopathology , Pancreatic Stellate Cells/physiology , Pancreatitis/etiology , Pancreatitis/physiopathology , Receptor Cross-Talk/physiology , Signal Transduction , Smoking/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...