1
|
Xie YJ, Gao WN, Wu QB, Yao XJ, Jiang ZB, Wang YW, Wang WJ, Li W, Hussain S, Liu L, Leung ELH, Fan XX. Chelidonine selectively inhibits the growth of gefitinib-resistant non-small cell lung cancer cells through the EGFR-AMPK pathway. Pharmacol Res 2020; 159:104934. [PMID: 32464330 DOI: 10.1016/j.phrs.2020.104934] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 02/08/2023]
Abstract
Tyrosine kinase inhibitors (TKIs) have been widely used for the clinical treatment of patients with non-small cell lung cancer (NSCLC) harboring mutations in the EGFR. Unfortunately, due to the secondary mutation in EGFR, eventual drug-resistance is inevitable. Therefore, to overcome the resistance, new agent is urgently required. Chelidonine, extracted from the roots of Chelidonium majus, was proved to effectively suppress the growth of NSCLC cells with EGFR double mutation. Proteomics analysis indicated that mitochondrial respiratory chain was significantly inhibited by chelidonine, and inhibitor of AMPK effectively blocked the apoptosis induced by chelidonine. Molecular dynamics simulations indicated that chelidonine could directly bind to EGFR and showed a much higher binding affinity to EGFRL858R/T790M than EGFRWT, which demonstrated that chelidonine could selectively inhibit the phosphorylation of EGFR in cells with EGFR double-mutation. In vivo study revealed that chelidonine has a similar inhibitory effect like second generation TKI Afatinib. In conclusion, targeting EGFR and inhibition of mitochondrial function is a promising anti-cancer therapeutic strategy for inhibiting NSCLC with EGFR mutation and TKI resistance.
Collapse
Affiliation(s)
- Ya-Jia Xie
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, PR China
| | - Wei-Na Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, PR China; Department of Chemistry, Southern University of Science and Technology, Shenzhen, Guangdong, PR China
| | - Qi-Biao Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, PR China
| | - Xiao-Jun Yao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, PR China
| | - Ze-Bo Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, PR China
| | - Yu-Wei Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, PR China
| | - Wen-Jun Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, PR China
| | - Wei Li
- TianJin NanKai Hospital, TianJin, PR China
| | - Shahid Hussain
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, PR China
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, PR China.
| | - Elaine Lai-Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, PR China.
| | - Xing-Xing Fan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, SAR, PR China.
| |
Collapse
|
2
|
Sommariva M, Gagliano N. E-Cadherin in Pancreatic Ductal Adenocarcinoma: A Multifaceted Actor during EMT. Cells 2020; 9:E1040. [PMID: 32331358 PMCID: PMC7226001 DOI: 10.3390/cells9041040] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/14/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a step-wise process observed in normal and tumor cells leading to a switch from epithelial to mesenchymal phenotype. In tumors, EMT provides cancer cells with a metastatic phenotype characterized by E-cadherin down-regulation, cytoskeleton reorganization, motile and invasive potential. E-cadherin down-regulation is known as a key event during EMT. However, E-cadherin expression can be influenced by the different experimental settings and environmental stimuli so that the paradigm of EMT based on the loss of E-cadherin determining tumor cell behavior and fate often becomes an open question. In this review, we aimed at focusing on some critical points in order to improve the knowledge of the dynamic role of epithelial cells plasticity in EMT and, specifically, address the role of E-cadherin as a marker for the EMT axis.
Collapse
Affiliation(s)
| | - Nicoletta Gagliano
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy;
| |
Collapse
|
3
|
Hesari A, Ghasemi F, Cicero AFG, Mohajeri M, Rezaei O, Hayat SMG, Sahebkar A. Berberine: A potential adjunct for the treatment of gastrointestinal cancers? J Cell Biochem 2018; 119:9655-9663. [DOI: 10.1002/jcb.27392] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/09/2018] [Indexed: 12/14/2022]
Affiliation(s)
- AmirReza Hesari
- Department of Biotechnology Faculty of Medicine, Arak University of Medical Sciences Arak Iran
| | - Faezeh Ghasemi
- Department of Biotechnology Faculty of Medicine, Arak University of Medical Sciences Arak Iran
| | - Arrigo F. G. Cicero
- Medical and Surgical Sciences Department University of Bologna Bologna Italy
| | - Mohammad Mohajeri
- Neurogenic Inflammation Research Center Mashhad University of Medical Sciences Mashhad Iran
- Department of Medical Biotechnology Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Omid Rezaei
- Faculty of Medicine, Arak University of Medical Sciences Arak Iran
| | | | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center Mashhad University of Medical Sciences Mashhad Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
- School of Pharmacy Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
4
|
Zielińska S, Jezierska-Domaradzka A, Wójciak-Kosior M, Sowa I, Junka A, Matkowski AM. Greater Celandine's Ups and Downs-21 Centuries of Medicinal Uses of Chelidonium majus From the Viewpoint of Today's Pharmacology. Front Pharmacol 2018; 9:299. [PMID: 29713277 PMCID: PMC5912214 DOI: 10.3389/fphar.2018.00299] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/15/2018] [Indexed: 12/20/2022] Open
Abstract
As antique as Dioscorides era are the first records on using Chelidonium as a remedy to several sicknesses. Inspired by the "signatura rerum" principle and an apparent ancient folk tradition, various indications were given, such as anti-jaundice and cholagogue, pain-relieving, and quite often mentioned-ophthalmological problems. Central and Eastern European folk medicine has always been using this herb extensively. In this region, the plant is known under many unique vernacular names, especially in Slavonic languages, associated or not with old Greek relation to "chelidon"-the swallow. Typically for Papaveroidae subfamily, yellow-colored latex is produced in abundance and leaks intensely upon injury. Major pharmacologically relevant components, most of which were first isolated over a century ago, are isoquinoline alkaloids-berberine, chelerythrine, chelidonine, coptisine, sanguinarine. Modern pharmacology took interest in this herb but it has not ended up in gaining an officially approved and evidence-based herbal medicine status. On the contrary, the number of relevant studies and publications tended to drop. Recently, some controversial reports and sometimes insufficiently proven studies appeared, suggesting anticancer properties. Anticancer potential was in line with anecdotical knowledge spread in East European countries, however, in the absence of directly-acting cytostatic compounds, some other mechanisms might be involved. Other properties that could boost the interest in this herb are antimicrobial and antiviral activities. Being a common synanthropic weed or ruderal plant, C. majus spreads in all temperate Eurasia and acclimates well to North America. Little is known about the natural variation of bioactive metabolites, including several aforementioned isoquinoline alkaloids. In this review, we put together older and recent literature data on phytochemistry, pharmacology, and clinical studies on C. majus aiming at a critical evaluation of state-of-the-art from the viewpoint of historical and folk indications. The controversies around this herb, the safety and drug quality issues and a prospective role in phytotherapy are discussed as well.
Collapse
Affiliation(s)
- Sylwia Zielińska
- Pharmaceutical Biology and Botany, Wrocław Medical University, Wrocław, Poland
| | - Anna Jezierska-Domaradzka
- Pharmaceutical Biology and Botany, Wrocław Medical University, Wrocław, Poland
- Botanical Garden of Medicinal Plants, Wrocław Medical University, Wrocław, Poland
| | | | - Ireneusz Sowa
- Analytical Chemistry, Medical University of Lublin, Lublin, Poland
| | - Adam Junka
- Pharmaceutical Microbiology and Parasitology, Wrocław Medical University, Wrocław, Poland
| | - Adam M. Matkowski
- Pharmaceutical Biology and Botany, Wrocław Medical University, Wrocław, Poland
- Botanical Garden of Medicinal Plants, Wrocław Medical University, Wrocław, Poland
| |
Collapse
|
5
|
Yu XZ, Guo ZY, Di Y, Yang F, Ouyang Q, Fu DL, Jin C. The relationship between SPARC expression in primary tumor and metastatic lymph node of resected pancreatic cancer patients and patients' survival. Hepatobiliary Pancreat Dis Int 2017; 16:104-109. [PMID: 28119265 DOI: 10.1016/s1499-3872(16)60168-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Previous researches in pancreatic cancer demonstrated a negative correlation between secreted protein acidic and rich in cysteine (SPARC) expression in primary tumor and survival, but not for SPARC expression in lymph node. In the present study, we aimed to evaluate the SPARC expression in various types of tissues and its impact on patients' prognosis. METHODS The expression of SPARC was examined by immunohistochemistry in resected pancreatic cancer specimens. Kaplan-Meier analyses and Cox proportional hazards regression were applied to assess the mortality risk. RESULTS A total of 222 tissue samples from 73 patients were collected to evaluate the SPARC expression, which included 73 paired primary tumor and adjacent normal tissues, 38 paired metastatic and normal lymph nodes. The proportion of positive SPARC expression in metastatic lymph node was high (32/38), whereas in normal lymph node it was negative (0/38). Positive SPARC expression in primary tumor cells was associated with a significantly decreased overall survival (P=0.007) and disease-free survival (P=0.003), whereas in other types of tissues it did not show a predictive role for prognosis. Univariate and multivariate analyses both confirmed this significance. CONCLUSION SPARC can serve a dual function role as both predictor for prognosis and potentially biomarker for lymph node metastasis in resected pancreatic cancer patients.
Collapse
Affiliation(s)
- Xin-Zhe Yu
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | | | | | | | | | | | | |
Collapse
|
6
|
Gagliano N, Celesti G, Tacchini L, Pluchino S, Sforza C, Rasile M, Valerio V, Laghi L, Conte V, Procacci P. Epithelial-to-mesenchymal transition in pancreatic ductal adenocarcinoma: Characterization in a 3D-cell culture model. World J Gastroenterol 2016; 22:4466-4483. [PMID: 27182158 PMCID: PMC4858630 DOI: 10.3748/wjg.v22.i18.4466] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/02/2016] [Accepted: 03/14/2016] [Indexed: 02/06/2023] Open
Abstract
AIM: To analyze the effect of three-dimensional (3D)-arrangement on the expression of epithelial-to-mesenchymal transition markers in pancreatic adenocarcinoma (PDAC) cells.
METHODS: HPAF-II, HPAC, and PL45 PDAC cells were cultured in either 2D-monolayers or 3D-spheroids. Ultrastructure was analyzed by transmission electron microscopy. The expression of E-cadherin, β-catenin, N-cadherin, collagen type I (COL-I), vimentin, α-smooth muscle actin (αSMA), and podoplanin was assayed by confocal microscopy in cells cultured on 12-mm diameter round coverslips and in 3D-spheroids. Gene expression for E-cadherin, Snail, Slug, Twist, Zeb1, and Zeb2 was quantified by real-time PCR. E-cadherin protein level and its electrophoretic pattern were studied by Western blot in cell lysates obtained from cells grown in 2D-monolayers and 3D-spheroids.
RESULTS: The E-cadherin/β-catenin complex was expressed in a similar way in plasma membrane cell boundaries in both 2D-monolayers and 3D-spheroids. E-cadherin increased in lysates obtained from 3D-spheroids, while cleavage fragments were more evident in 2D-monolayers. N-cadherin expression was observed in very few PDAC cells grown in 2D-monolayers, but was more evident in 3D-spheroids. Some cells expressing COL-I were observed in 3D-spheroids. Podoplanin, expressed in collectively migrating cells, and αSMA were similarly expressed in both experimental conditions. The concomitant maintenance of the E-cadherin/β-catenin complex at cell boundaries supports the hypothesis of a collective migration for these cells, which is consistent with podoplanin expression.
CONCLUSION: We show that a 3D-cell culture model could provide deeper insight into understanding the biology of PDAC and allow for the detection of marked differences in the phenotype of PDAC cells grown in 3D-spheroids.
Collapse
MESH Headings
- Antigens, CD
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Cadherins/genetics
- Cadherins/metabolism
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/ultrastructure
- Cell Line, Tumor
- Cell Shape
- Cell Survival
- Epithelial-Mesenchymal Transition
- Gene Expression Regulation, Neoplastic
- Humans
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Microscopy, Confocal
- Microscopy, Electron, Transmission
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/ultrastructure
- Phenotype
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Spheroids, Cellular
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
|
7
|
Felix K, Gaida MM. Neutrophil-Derived Proteases in the Microenvironment of Pancreatic Cancer -Active Players in Tumor Progression. Int J Biol Sci 2016; 12:302-13. [PMID: 26929737 PMCID: PMC4753159 DOI: 10.7150/ijbs.14996] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A hallmark of pancreatic ductal adenocarcinoma (PDAC) is the fibro-inflammatory microenvironment, consisting of activated pancreatic stellate cells, extracellular matrix proteins, and a variety of inflammatory cells, such as T cells, macrophages, or neutrophils. Tumor-infiltrating immune cells, which are found in nearly all cancers, including PDAC, often fail to eliminate the tumor, but conversely can promote its progression by altering the tumor microenvironment. Pancreatic cancer cells are able to attract polymorphonuclear neutrophils (PMN) via tumor secreted chemokines and in human PDAC, PMN infiltrates can be observed in the vicinity of tumor cells and in the desmoplastic tumor stroma, which correlate with undifferentiated tumor growth and poor prognosis. The behavior of tumor-infiltrating neutrophils in the tumor micromilieu is not yet understood at a mechanistic level. It has been shown that PMN have the potential to kill tumor cells, either directly or by antibody-dependent cell-mediated cytotoxicity, but on the other side various adverse effects of PMN, such as promotion of aggressive tumor growth with epithelial-to-mesenchymal transition and increased metastatic potential, have been described. Recent therapeutic approaches for PDAC focus not only the tumor cell itself, but also elements of the tumor microenvironment. Therefore, the role of PMN and their derived products (e.g. cytokines, proteases) as a new vein for a therapeutic target should be critically evaluated in this context. This review summarizes the current understanding of the interplay between proteases of tumor-infiltrating neutrophils and pancreatic tumor cells and elements of the desmoplastic stroma.
Collapse
Affiliation(s)
- Klaus Felix
- 1. Department of General Surgery, University of Heidelberg, INF 110, Heidelberg, Germany
| | - Matthias M Gaida
- 2. Institute of Pathology, University of Heidelberg, INF 224, Heidelberg, Germany
| |
Collapse
|
8
|
Jesionek W, Fornal E, Majer-Dziedzic B, Móricz ÁM, Nowicky W, Choma IM. Investigation of the composition and antibacterial activity of Ukrain™ drug using liquid chromatography techniques. J Chromatogr A 2016; 1429:340-7. [DOI: 10.1016/j.chroma.2015.12.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 12/02/2015] [Accepted: 12/03/2015] [Indexed: 12/19/2022]
|
9
|
Wang N, Tan HY, Li L, Yuen MF, Feng Y. Berberine and Coptidis Rhizoma as potential anticancer agents: Recent updates and future perspectives. JOURNAL OF ETHNOPHARMACOLOGY 2015; 176:35-48. [PMID: 26494507 DOI: 10.1016/j.jep.2015.10.028] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 10/17/2015] [Accepted: 10/17/2015] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The antineoplastic property of Coptidis Rhizoma and berberine was correlated with its traditional use of clearing internal fire, removing damp-heat and counteracting toxic pathogens. AIM OF THE STUDY The anti-tumor effect of Coptidis Rhizoma and berberine was extensively studied since our last comprehensive review in 2009. This study aims to summarize the recent updates and give rise to perspectives of Coptidis Rhizoma and berberine as potential novel antineoplastic agents. METHODS Quality studies in recent 5 years were retrieved from PubMed, Medline and CNKI with keywords including Coptis, Coptidis Rhizoma, huanglian, berberine, tumor and cancer. Studies were focused on the pharmacological actions of Coptidis Rhizoma and berberine in cancer progression. RESULTS It was shown that Coptidis Rhizoma extract and berberine may repress tumor progression by regressing abnormal cell proliferation, arresting cell cycle and inducing cell death. Studies also highlighted the actions of Coptidis Rhizoma extract and berberine in inhibiting tumor cell invasion and angiogenesis, which in turn abolish cancer metastasis. Some studies have also been conducted to reveal the potential effect of Coptidis Rhizoma extract and berberine in regulating tumor stromal microenvironment, as well as in preventing carcinogenesis. Most of the results have been demonstrated with in vivo models, but results of high-quality clinical trials are not yet available. Unspecified cancer type and staging, fluctuated dose information and variants of targets across studies of berberine/ Coptidis Rhizoma impede their clinical use for cancer treatment. CONCLUSION Recent advances highlighted by this review may shed light on future direction of studies featuring Coptidis Rhizoma and berberine as novel antineoplastic agents, which should be repeatedly proven in future animal and clinical studies. Although more evidences on its specificity and clinical efficacy are necessary to support its clinical use, Coptidis Rhizoma and berberine are highly expected to be effective, safe and affordable treatments for cancer patients.
Collapse
Affiliation(s)
- Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Hor-Yue Tan
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Lei Li
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Man-Fung Yuen
- Division of Gastroenterology and Hepatology, Queen Mary Hospital, and Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
10
|
Basso D, Bozzato D, Padoan A, Moz S, Zambon CF, Fogar P, Greco E, Scorzeto M, Simonato F, Navaglia F, Fassan M, Pelloso M, Dupont S, Pedrazzoli S, Fassina A, Plebani M. Inflammation and pancreatic cancer: molecular and functional interactions between S100A8, S100A9, NT-S100A8 and TGFβ1. Cell Commun Signal 2014; 12:20. [PMID: 24670043 PMCID: PMC4108065 DOI: 10.1186/1478-811x-12-20] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 03/08/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND In order to gain further insight on the crosstalk between pancreatic cancer (PDAC) and stromal cells, we investigated interactions occurring between TGFβ1 and the inflammatory proteins S100A8, S100A9 and NT-S100A8, a PDAC-associated S100A8 derived peptide, in cell signaling, intracellular calcium (Cai2+) and epithelial to mesenchymal transition (EMT). NF-κB, Akt and mTOR pathways, Cai2+ and EMT were studied in well (Capan1 and BxPC3) and poorly differentiated (Panc1 and MiaPaCa2) cell lines. RESULTS NT-S100A8, one of the low molecular weight N-terminal peptides from S100A8 to be released by PDAC-derived proteases, shared many effects on NF-κB, Akt and mTOR signaling with S100A8, but mainly with TGFβ1. The chief effects of S100A8, S100A9 and NT-S100A8 were to inhibit NF-κB and stimulate mTOR; the molecules inhibited Akt in Smad4-expressing, while stimulated Akt in Smad4 negative cells. By restoring Smad4 expression in BxPC3 and silencing it in MiaPaCa2, S100A8 and NT-S100A8 were shown to inhibit NF-κB and Akt in the presence of an intact TGFβ1 canonical signaling pathway. TGFβ1 counteracted S100A8, S100A9 and NT-S100A8 effects in Smad4 expressing, not in Smad4 negative cells, while it synergized with NT-S100A8 in altering Cai2+ and stimulating PDAC cell growth. The effects of TGFβ1 on both EMT (increased Twist and decreased N-Cadherin expression) and Cai2+ were antagonized by S100A9, which formed heterodimers with TGFβ1 (MALDI-TOF/MS and co-immuno-precipitation). CONCLUSIONS The effects of S100A8 and S100A9 on PDAC cell signaling appear to be cell-type and context dependent. NT-S100A8 mimics the effects of TGFβ1 on cell signaling, and the formation of complexes between TGFβ1 with S100A9 appears to be the molecular mechanism underlying the reciprocal antagonism of these molecules on cell signaling, Cai2+ and EMT.
Collapse
Affiliation(s)
- Daniela Basso
- Department of Laboratory Medicine, University-Hospital of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Dania Bozzato
- Department of Medicine – DIMED, University of Padova, Padova, Italy
| | - Andrea Padoan
- Department of Medicine – DIMED, University of Padova, Padova, Italy
| | - Stefania Moz
- Department of Medicine – DIMED, University of Padova, Padova, Italy
| | | | - Paola Fogar
- Department of Laboratory Medicine, University-Hospital of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Eliana Greco
- Department of Medicine – DIMED, University of Padova, Padova, Italy
| | - Michele Scorzeto
- Department of Biomedical Sciences – DSB, University of Padova, Padova, Italy
| | | | - Filippo Navaglia
- Department of Laboratory Medicine, University-Hospital of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Matteo Fassan
- Department of Medicine – DIMED, University of Padova, Padova, Italy
| | - Michela Pelloso
- Department of Medicine – DIMED, University of Padova, Padova, Italy
| | - Sirio Dupont
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | - Ambrogio Fassina
- Department of Medicine – DIMED, University of Padova, Padova, Italy
| | - Mario Plebani
- Department of Laboratory Medicine, University-Hospital of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Department of Medicine – DIMED, University of Padova, Padova, Italy
| |
Collapse
|
11
|
Ricci C, Mota C, Moscato S, D’Alessandro D, Ugel S, Sartoris S, Bronte V, Boggi U, Campani D, Funel N, Moroni L, Danti S. Interfacing polymeric scaffolds with primary pancreatic ductal adenocarcinoma cells to develop 3D cancer models. BIOMATTER 2014; 4:e955386. [PMID: 25482337 PMCID: PMC4578550 DOI: 10.4161/21592527.2014.955386] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 07/03/2014] [Accepted: 08/10/2014] [Indexed: 12/27/2022]
Abstract
We analyzed the interactions between human primary cells from pancreatic ductal adenocarcinoma (PDAC) and polymeric scaffolds to develop 3D cancer models useful for mimicking the biology of this tumor. Three scaffold types based on two biocompatible polymeric formulations, such as poly(vinyl alcohol)/gelatin (PVA/G) mixture and poly(ethylene oxide terephthalate)/poly(butylene terephthalate) (PEOT/PBT) copolymer, were obtained via different techniques, namely, emulsion and freeze-drying, compression molding followed by salt leaching, and electrospinning. In this way, primary PDAC cells interfaced with different pore topographies, such as sponge-like pores of different shape and size or nanofiber interspaces. The aim of this study was to investigate the influence played by the scaffold architecture over cancerous cell growth and function. In all scaffolds, primary PDAC cells showed good viability and synthesized tumor-specific metalloproteinases (MMPs) such as MMP-2, and MMP-9. However, only sponge-like pores, obtained via emulsion-based and salt leaching-based techniques allowed for an organized cellular aggregation very similar to the native PDAC morphological structure. Differently, these cell clusters were not observed on PEOT/PBT electrospun scaffolds. MMP-2 and MMP-9, as active enzymes, resulted to be increased in PVA/G and PEOT/PBT sponges, respectively. These findings suggested that spongy scaffolds supported the generation of pancreatic tumor models with enhanced aggressiveness. In conclusion, primary PDAC cells showed diverse behaviors while interacting with different scaffold types that can be potentially exploited to create stage-specific pancreatic cancer models likely to provide new knowledge on the modulation and drug susceptibility of MMPs.
Collapse
Key Words
- 2D, Bi-dimensional
- 3D, Three-dimensional
- BCA, Bicinchoninic acid
- BSA, Bovine serum albumin
- Dd, double distilled
- Ds, double stranded
- ECM, Extracellular matrix
- G, Gelatin
- HRP, Horseradish peroxidase
- K-ras, Kirsten rat sarcoma viral oncogene homolog
- MMP, Matrix metalloproteinase
- PBS, Phosphate buffer saline
- PCR, Polymer-chain reaction
- PDAC, Pancreatic ductal adenocarcinoma
- PEOT/PBT, Poly(ethylene oxide terephthalate)/poly(butylene terephthalate)
- PVA, Poly(vinyl alcohol)
- PanIN, Pancreatic intraepithelial neoplasia
- Pancreatic adenocarcinoma
- Smad4, Mothers against decapentaplegic homolog 4
- TME, Tumor microenvironment.
- cancer
- compression molding
- electrospinning
- emulsion and freeze-drying
- metalloproteinase 2 (MMP-2)
- metalloproteinase 9 (MMP-9)
- polyethylene oxide terephthalate (PEOT)
- polyvinyl alcohol (PVA)
- scaffold
Collapse
Affiliation(s)
- Claudio Ricci
- Department of Pathology and Diagnostics; University of Verona; Verona, Italy
- Department of Surgical, Medical, Molecular Pathology and Emergency Medicine; University of Pisa; Pisa, Italy
- Tissue Regeneration Department; University of Twente; Enschede, The Netherlands
| | - Carlos Mota
- Tissue Regeneration Department; University of Twente; Enschede, The Netherlands
| | - Stefania Moscato
- Department of Clinical and Experimental Medicine; University of Pisa; Pisa, Italy
| | - Delfo D’Alessandro
- Department of Surgical, Medical, Molecular Pathology and Emergency Medicine; University of Pisa; Pisa, Italy
| | - Stefano Ugel
- Department of Pathology and Diagnostics; University of Verona; Verona, Italy
| | - Silvia Sartoris
- Department of Pathology and Diagnostics; University of Verona; Verona, Italy
| | - Vincenzo Bronte
- Department of Pathology and Diagnostics; University of Verona; Verona, Italy
| | - Ugo Boggi
- Department of Translational Research and New Technologies in Medicine and Surgery; University of Pisa; Pisa, Italy
| | - Daniela Campani
- Department of Surgical, Medical, Molecular Pathology and Emergency Medicine; University of Pisa; Pisa, Italy
| | - Niccola Funel
- Department of Surgical, Medical, Molecular Pathology and Emergency Medicine; University of Pisa; Pisa, Italy
| | - Lorenzo Moroni
- Tissue Regeneration Department; University of Twente; Enschede, The Netherlands
| | - Serena Danti
- Department of Surgical, Medical, Molecular Pathology and Emergency Medicine; University of Pisa; Pisa, Italy
| |
Collapse
|
12
|
Pancreatic cancer cells retain the epithelial-related phenotype and modify mitotic spindle microtubules after the administration of ukrain in vitro. Anticancer Drugs 2013; 23:935-46. [PMID: 22700003 DOI: 10.1097/cad.0b013e32835507bc] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The aim of this study is to characterize the phenotype of pancreatic ductal adenocarcinoma (PDAC) cells in relation to the expression of epithelial-to-mesenchymal transition (EMT) markers and determine whether ukrain, an anticancer drug based on the alkaloids extracted from greater celandine, modulates in vitro the malignant behavior of PDAC cells in order to extend our understanding of its therapeutic potential. Three cell lines (HPAF-II, HPAC, and PL45) were treated with ukrain (5, 10, and 20 μmol/l) for 48 h or left untreated (control). Cell proliferation was assessed by growth curves. Apoptosis was determined by Hoechst nuclear staining and by cytochrome c and caspase-8 expressions. The EMT markers E-cadherin, β-catenin, and vimentin, as well as actin and tubulin cytoskeletons, were analyzed by immunofluorescence. Interphase and mitotic microtubules as well as abnormal mitotic figures were studied by fluorescence microscopy after tubulin immunolabeling. Ukrain strongly suppressed cell proliferation and induced apoptosis possibly through an extrinsic pathway as cytochrome c immunoreactivity suggested that the integrity of the mitochondria was not affected. Tubulin expression indicated an antiproliferative effect of ukrain on the basis of alterations in mitotic spindle microtubule dynamics, leading to abnormal mitosis. Membranous E-cadherin/β-catenin immunoreactivity was similarly expressed in control-treated and ukrain-treated cells, although the drug upregulated E-cadherin in cell lysates. Our results suggest that ukrain exerts its chemotherapeutic action on PDAC cells targeting mitotic spindle microtubules, leading to abnormal mitosis and apoptosis, and favoring cell cohesiveness. The differentiated epithelial phenotype of HPAF-II, HPAC, and PL45 cell lines concomitant with a highly invasive potential suggests that further experiments will be necessary to definitively clarify the role of EMT in PDAC progression.
Collapse
|
13
|
Li CL, Cui YF, Du XF, Tai S. Clinical significance of matrix metalloproteinases-3 and vascular endothelial growth factor expression in pancreatic carcinoma. Shijie Huaren Xiaohua Zazhi 2011; 19:1574-1578. [DOI: 10.11569/wcjd.v19.i15.1574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of vascular endothelial growth factor (VEGF) and matrix metalloproteinases-3 (MMP-3) in pancreatic carcinoma (PC) and to analyze their relationship with tumor angiogenesis, invasion and metastasis.
METHODS: The protein expression of VEGF and MMP-3 and microvessel density (MVD) were detected by immunohistochemistry in 56 PC specimens and 56 normal pancreatic tissue specimens.
RESULTS: The positive rates of both MMP-3 and VEGF were significantly higher in PC than in normal pancreatic tissue (75.00% vs 3.57%, 67.85% vs 1.78%, both P < 0.01). There was a positive correlation between the expression of VEGF and that of MMP-3. Higher expression of MMP-3 was detected in late-stage PC and PC with lymph node metastasis. The expression of VEGF was correlated with clinical stage (P < 0.05). MVD was correlated with expression of MMP-3 and VEGF in PC (both P < 0.05).
CONCLUSION: MMP-3 and VEGF may promote tumor angiogenesis and play an important role in the development and progression of PC. Detection of MMP-3 expression could be used to predict early metastasis of PC. Lower expression of VEGF may be related to lower arterial blood supply in PC.
Collapse
|