1
|
Combined chemotherapy based on bioactive black phosphorus for pancreatic cancer therapy. J Control Release 2023; 354:889-901. [PMID: 36586672 DOI: 10.1016/j.jconrel.2022.12.054] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 01/02/2023]
Abstract
Pancreatic cancer is the most aggressive malignant tumor with difficulty in early diagnosis, very short survival time in advanced stage, and lack of effective treatment options. In this work, a novel combination chemotherapy strategy based on bioactive black phosphorus (BP) and gemcitabine (GEM) is developed for efficient treatment of pancreatic cancer. The combined cell cycle blockage in G2/M phase induced by BP and G0/G1 phase by GEM results in synergistic killing of pancreatic cancer cells with the combination index (CI) < 1. The iRGD modified zein nanoparticles co-loaded with BP quantum dots (BPQDs) and GEM are designed and prepared as a targeted nanoplatform (BP-GEM@NPs). After intravenous injection, the in vivo distribution and pharmacokinetics results demonstrate that BP-GEM@NPs shows excellent tumor targeting capability and significantly prolonged blood circulation time. The targeted co-delivery of BPQDs and GEM induces much more pancreatic tumor cell apoptosis and synergistically inhibits tumor growth in both subcutaneous xenograft and orthotopic models. Meanwhile, BP-GEM@NPs exhibit good biocompatibility without bring adverse effects. This work indicates the great potential of BP-GEM@NPs as a combination chemotherapy for pancreatic cancer and provides insights into development of biomedicine by exploring the intrinsic bioactivities of nanomaterials.
Collapse
|
2
|
Bitter apricot ethanolic extract induces apoptosis through increasing expression of Bax/Bcl-2 ratio and caspase-3 in PANC-1 pancreatic cancer cells. Mol Biol Rep 2020; 47:1895-1904. [PMID: 32026321 DOI: 10.1007/s11033-020-05286-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/27/2020] [Indexed: 10/25/2022]
Abstract
Pancreatic cancer is the fourth common cause of cancer death. Surgery and chemotherapy are the common treatment strategies for pancreatic cancer patients; however, the response rate is less than 20% at advanced stages. In recent years, growing interest has been dedicated to natural products. Bitter apricot seeds possess a number of pharmacological properties including antitumor activity and amygdalin from bitter apricot seeds can induce apoptosis. In this study, we investigated the cyto/genotoxic effects of bitter apricot ethanolic extract (BAEE) and amygdalin on human pancreatic cancer PANC-1 and normal epithelial 293/KDR cells. BAEE was assessed using high-performance liquid chromatography for the confirmation of the structure. The biological impacts of BAEE and amygdalin on PANC-1 and 293/KDR cells were evaluated by MTT assay, DAPI staining, AnnexinV/PI and Real-time qPCR analysis. BAEE and amygdalin inhibited cancer cell growth in a dose- and time-dependent manner. DAPI staining and flow cytometric analysis revealed fragmented nuclei and elevated numbers of early and late apoptotic cells, respectively. Also, increased Bax/Bcl-2 ratio and upregulation of caspase-3 further confirmed the occurrence of apoptosis in PANC-1 cells, but not in non-cancerous 293/KDR cells. These results indicate that BAEE could mediate apoptosis induction in cancer cells through a mitochondria dependent pathway. These findings suggest that BAEE functions as a potent pro-apoptotic factor for human pancreatic cancer cells without a significant effect on 293/KDR cells. Though, the potent anti-cancer components of BAEE should be further identified. Moreover, in vivo investigations are required to confirm bitter apricot ethanolic extract's clinical value as an anti-tumor drug.
Collapse
|
3
|
Liu QH, Yong HM, Zhuang QX, Zhang XP, Hou PF, Chen YS, Zhu MH, Bai J. Reduced expression of annexin A1 promotes gemcitabine and 5-fluorouracil drug resistance of human pancreatic cancer. Invest New Drugs 2019; 38:350-359. [PMID: 31124054 DOI: 10.1007/s10637-019-00785-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/18/2019] [Indexed: 12/24/2022]
Abstract
Intrinsic chemoresistance is the main reason for the failure of human pancreatic ductal adenocarcinoma (PDAC) therapy. To identify the candidate protein, we compared the protein expression profiling of PDAC cells and its distinct surviving cells following primary treatment with gemcitabine (GEM) and 5-fluorouracil (5-FU) by two-dimensional electrophoresis combined with liquid chromatography-mass spectrometry or mass spectrometry. A total of 20 differentially expressed proteins were identified, and annexin A1 (ANXA1) was analyzed for further validation. The functional validation showed that the downregulation of ANXA1 contributes to GEM and 5-FU resistance in PDAC cells through protein kinase C/c-Jun N-terminal kinase/P-glycoprotein signaling pathway. Our findings provide a platform for the further elucidation of the underlying mechanisms of PDAC intrinsic chemoresistance and demonstrated that ANXA1 may be a valid marker for anticancer drug development.
Collapse
Affiliation(s)
- Qing-Hua Liu
- Cancer Institute, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu Province, China.,Department of Pathology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Hong-Mei Yong
- Department of Medical Oncology, Huai'an Hospital to Xuzhou Medical University, Huai'an, Jiangsu Province, China
| | - Qing-Xin Zhuang
- Department of Medical Oncology, People's Hospital of Ningxia Hui Autonomous Region/The First Affiliated Hospital of Northwest University of Nationalities, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xu-Ping Zhang
- Cancer Institute, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu Province, China
| | - Ping-Fu Hou
- Cancer Institute, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu Province, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yan-Su Chen
- Cancer Institute, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu Province, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Ming-Hua Zhu
- Department of Pathology, Changhai Hospital, Secondary Military Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu Province, China. .,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, China.
| |
Collapse
|
4
|
Oluwasanmi A, Al-Shakarchi W, Manzur A, Aldebasi MH, Elsini RS, Albusair MK, Haxton KJ, Curtis ADM, Hoskins C. Diels Alder-mediated release of gemcitabine from hybrid nanoparticles for enhanced pancreatic cancer therapy. J Control Release 2017; 266:355-364. [PMID: 28943195 DOI: 10.1016/j.jconrel.2017.09.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/13/2017] [Accepted: 09/18/2017] [Indexed: 12/11/2022]
Abstract
Hybrid nanoparticles (HNPs) have shown huge potential as drug delivery vehicles for pancreatic cancer. Currently, the first line treatment, gemcitabine, is only effective in 23.8% of patients. To improve this, a thermally activated system was developed by introducing a linker between HNPs and gemcitabine. Whereby, heat generation resulting from laser irradiation of the HNPs promoted linker breakdown resulting in prodrug liberation. In vitro evaluation in pancreatic adenocarcinoma cells, showed the prodrug was 4.3 times less cytotoxic than gemcitabine, but exhibited 11-fold improvement in cellular uptake. Heat activation of the formulation led to a 56% rise in cytotoxicity causing it to outperform gemcitabine by 26%. In vivo the formulation outperformed free gemcitabine with a 62% reduction in tumor weight in pancreatic xenografts. This HNP formulation is the first of its kind and has displayed superior anti-cancer activity as compared to the current first line drug gemcitabine after heat mediated controlled release.
Collapse
Affiliation(s)
- Adeolu Oluwasanmi
- Institute of Science and Technology in Medicine, School of Pharmacy, Keele University, Keele ST5 5BG, UK
| | - Wejdan Al-Shakarchi
- Institute of Science and Technology in Medicine, School of Pharmacy, Keele University, Keele ST5 5BG, UK
| | - Ayesha Manzur
- Institute of Science and Technology in Medicine, School of Pharmacy, Keele University, Keele ST5 5BG, UK
| | - Mohammed H Aldebasi
- College of Medicine, Al Imam Mohammad Ibn, Saud Islamic University, Riyadh, Saudi Arabia
| | - Rayan S Elsini
- College of Medicine, Al Imam Mohammad Ibn, Saud Islamic University, Riyadh, Saudi Arabia
| | - Malek K Albusair
- College of Medicine, Al Imam Mohammad Ibn, Saud Islamic University, Riyadh, Saudi Arabia
| | - Katherine J Haxton
- School of Physical and Geographical Sciences, Faculty of Natural Sciences, Keele University, Keele ST5 5BG, UK
| | - Anthony D M Curtis
- Institute of Science and Technology in Medicine, School of Pharmacy, Keele University, Keele ST5 5BG, UK
| | - Clare Hoskins
- Institute of Science and Technology in Medicine, School of Pharmacy, Keele University, Keele ST5 5BG, UK.
| |
Collapse
|
5
|
Li T, Wang Z, Hou YF, Li YY. Pim-3 Regulates Stemness of Pancreatic Cancer Cells via Activating STAT3 Signaling Pathway. J Cancer 2017; 8:1530-1541. [PMID: 28775772 PMCID: PMC5535708 DOI: 10.7150/jca.18628] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/27/2017] [Indexed: 02/06/2023] Open
Abstract
Due to its aggressiveness and unusual resistance to conventional therapies, pancreatic cancer is a highly lethal gastrointestinal malignancy with poor prognosis. According to the cancer stem cell hypothesis, there exists a fraction of cancer cells, that is, cancer stem cells, responsible for tumor maintenance and therapeutic failure. Herein we investigated the involvement of proto-oncogene Pim-3 in driving the stemness properties in pancreatic cancer. Expression levels of several stemness-associated markers were examined in several pancreatic cancer cell lines. The double positive (CD24+ESA+) and double negative (CD24-ESA-) pancreatic cancer cells were isolated from PANC-1 and L3.6pl, and their self-renewal ability, tumorigenicity as well as sensitivity to gemcitabine were then evaluated. Results showed that there existed heterogeneity in expression levels of stemness-associated surface markers among pancreatic cancer cell lines. CD24+ESA+ pancreatic cancer cells exhibited increased tumorigenicity and decreased chemosensitivity to gemcitabine as compared to CD24-ESA- cells. Besides, the double positive (CD24+ESA+) subpopulation also exhibited greater expression level of Pim-3 when compared with the double negative (CD24-ESA-) ones. Furthermore, silencing of Pim-3 in pancreatic cancer cells leads to decreased proportions of both single positive (CD24+ and ESA+) and double positive (CD24+ESA+) pancreatic cancer cells. Overexpression of Pim-3 was associated with increased levels of some stemness-associated transcription factors (STAT3, etc.). Moreover, the phosphorylation level and transcriptional activity of STAT3 were decreased in Pim-3 silenced pancreatic cancer cells and restoration of its activity results in restitution of stem cell-like phenotypes. Therefore, Pim-3 maintains stemness of pancreatic cancer cells via activating STAT3 signaling pathway and might be used as a novel therapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Ting Li
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhen Wang
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Feng Hou
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying-Yi Li
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Wylie PG, Onley DJ, Hammerstein AF, Bowen WP. Advances in Laser Scanning Imaging Cytometry for High-Content Screening. Assay Drug Dev Technol 2015; 13:66-78. [DOI: 10.1089/adt.2014.607] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Paul G. Wylie
- TTP Labtech Limited, Melbourn, Hertfordshire, United Kingdom
| | - David J. Onley
- TTP Labtech Limited, Melbourn, Hertfordshire, United Kingdom
| | | | - Wayne P. Bowen
- TTP Labtech Limited, Melbourn, Hertfordshire, United Kingdom
| |
Collapse
|
7
|
Aznar MÁ, Lasa-Saracíbar B, Blanco-Prieto MJ. Edelfosine lipid nanoparticles overcome multidrug resistance in K-562 leukemia cells by a caspase-independent mechanism. Mol Pharm 2014; 11:2650-8. [PMID: 24865362 DOI: 10.1021/mp5000696] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The antitumor ether lipid edelfosine is the prototype of a novel generation of promising anticancer drugs that has been shown to be an effective antitumor agent in numerous malignancies. However, several cancer types display resistance to different antitumoral compounds due to multidrug resistance (MDR). Thus, MDR is a major drawback in anticancer therapy. In that sense, the leukemic cell line K-562 shows resistance to edelfosine. This resistance is overcome by the use of nanotechnology. The present work describes the rate and mechanism of internalization of free and nanoencapsulated edelfosine. The molecular mechanisms underlying cell death are described in the present paper by characterization of several molecules implied in the apoptosic and autophagic pathways (PARP, LC3IIB, caspases-3, -9 and -7), and their pattern of expression is compared with the cell induction in a sensitive cell line HL-60. Results showed different internalization patterns in both cells. Clathrin and lipid raft mediated endocytosis were observable in edelfosine uptake, whereas these mechanism were not visible in the uptake of lipid nanoparticles, which might suffer phagocytosis and macropinocytosis. Both treatments induced caspase-mediated apoptosis in HL-60 cells, whereas this cell death mechanism was unnoticeable in K-562 cells. Moreover, an important increase in autophagic vesicles was visible in K-562 cells. Thus, this mechanism might be implicated in overcoming K-562 resistance with the treatment by lipid nanoparticles.
Collapse
Affiliation(s)
- María Ángela Aznar
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra , C/Irunlarrea 1, E-31080 Pamplona, Spain
| | | | | |
Collapse
|
8
|
Xu D, Cobb MG, Gavilano L, Witherspoon SM, Williams D, White CD, Taverna P, Bednarski BK, Kim HJ, Baldwin AS, Baines AT. Inhibition of oncogenic Pim-3 kinase modulates transformed growth and chemosensitizes pancreatic cancer cells to gemcitabine. Cancer Biol Ther 2014; 14:492-501. [PMID: 23760491 DOI: 10.4161/cbt.24343] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer with a 5-year survival rate of only 6%. Although the cytosine analog gemcitabine is the drug commonly used to treat PDAC, chemoresistance unfortunately renders the drug ineffective. Thus, strategies that can decrease this resistance will be essential for improving the dismal outcome of patients suffering from this disease. We previously observed that oncogenic Pim-1 kinase was aberrantly expressed in PDAC tissues and cell lines and was responsible for radioresistance. Furthermore, members of the Pim family have been shown to reduce the efficacy of chemotherapeutic drugs in cancer. Therefore, we attempted to evaluate the role of Pim-3 in chemoresistance of PDAC cells. We were able to confirm upregulation of the Pim-3 oncogene in PDAC tissues and cell lines versus normal samples. Biological consequences of inhibiting Pim-3 expression with shRNA-mediated suppression included decreases in anchorage-dependent growth, invasion through Matrigel and chemoresistance to gemcitabine as measured by caspase-3 activity. Additionally, we were able to demonstrate that Pim-1 and Pim-3 play overlapping but non-identical roles as it relates to gemcitabine sensitivity of pancreatic cancer cells. To further support the role of Pim-3 suppression in sensitizing PDAC cells to gemcitabine, we used the pharmacological Pim kinase inhibitor SGI-1776. Treatment of PDAC cells with SGI-1776 resulted in decreased phosphorylation of the proapoptotic protein Bad and cell cycle changes. When SGI-1776 was combined with gemcitabine, there was a greater decrease in cell viability in the PDAC cells versus cells treated with either of the drugs separately. These results suggest combining drug therapies that inhibit Pim kinases, such as Pim-3, with chemotherapeutic agents, to aid in decreasing chemoresistance in pancreatic cancer.
Collapse
Affiliation(s)
- Dapeng Xu
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Cheng Y, Diao D, Zhang H, Guo Q, Wu X, Song Y, Dang C. High glucose-induced resistance to 5-fluorouracil in pancreatic cancer cells alleviated by 2-deoxy-D-glucose. Biomed Rep 2013; 2:188-192. [PMID: 24649094 DOI: 10.3892/br.2013.211] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 10/22/2013] [Indexed: 12/14/2022] Open
Abstract
Abnormal glucose metabolism from hyperglycemia or diabetes aggravates the progression of pancreatic cancer. It is unknown whether high glucose has an impact on the antitumor effect of 5-fluorouracil (5-Fu) and whether targeting aberrant glucose metabolism using 2-deoxy-D-glucose (2-DG) may reverse this effect in high-glucose microenvironments. The cell viability of AsPC-1 and Panc-1 was analyzed by MTT assay following 5-Fu treatment at different glucose concentrations. Altered sensitivity to 5-Fu by 2-DG was also analyzed. LY294002 was used to inhibit PI3K-Akt signaling to determine the mechanism involved. In response to glucose, 5-Fu-induced cell growth inhibition was attenuated in a dose-dependent manner, accompanied with activated p-Akt, while 2-DG enhanced 5-Fu-induced cell growth inhibition. Moreover, blocking the PI3K/Akt pathway by LY294002 effectively eliminated 2-DG-induced apoptosis. In conclusion, high glucose weakens the antitumor effect of 5-Fu via PI3K/Akt signaling. Using 2-DG in combination with 5-Fu significantly increased their therapeutic effectiveness in high-glucose microenvironments.
Collapse
Affiliation(s)
- Yao Cheng
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Dongmei Diao
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Hao Zhang
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Qi Guo
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Xuandi Wu
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Yongchun Song
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Chengxue Dang
- Department of Surgical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
10
|
Lee JY, Song SY, Park JY. Notch pathway activation is associated with pancreatic cancer treatment failure. Pancreatology 2013; 14:48-53. [PMID: 24555978 DOI: 10.1016/j.pan.2013.11.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/23/2013] [Accepted: 11/19/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pancreatic cancer is resistant to conventional treatment. The aim of the study was to confirm the hypothesis that changes in cancer stem cells (CSCs) and developmental pathway after treatment was responsible for treatment failure in pancreatic cancer. METHODS After recovery from a gemcitabine treatment, the percentage of pancreatic cancer CSCs and Notch pathway in BxPC3 and HPAC pancreatic cancer cell lines were analyzed by FACS (CD24 and CD44) and western blot (Notch1, Hes1, β-catenin, and pAKT). The effect of DAPT, a gamma-secretase inhibitor, was similarly investigated. The association between immunohistochemical expression of Hes1 and survival was analyzed. RESULTS The percentage of CD24(+)CD44(+) cells was higher in gemcitabine-treated BxPC3 and HPAC cells than at pre-treatment. CD24(+)CD44(+) cells sorted from the gemcitabine-treated cell lines showed higher migration and invasion ability than CD24(-)CD44(-) or CD24(-)CD44(+) cells from the same cell lines. Western blot analysis showed an increased expression of Notch1 and Hes1 in gemcitabine-treated cell lines. The overall survival of pancreatic cancer patients with strong expression of Hes1 was shorter than that in patients with no or weak expression (11.1 vs. 21.6 months, P = 0.036). Treatment with DAPT reversed the increase in Hes1, β-catenin, and pAKT expression and the proportion of CD24(+)CD44(+) cells in gemcitabine-treated cell lines. The treatment also decreased migration and invasion ability. CONCLUSION Our data suggested that an increase in CSCs and activation of the Notch pathway might contribute to the failure of treatment in pancreatic cancer. Notch pathway can be a potential target to overcome treatment failure.
Collapse
Affiliation(s)
- Jin Young Lee
- Division of Gastroenterology, Department of Internal Medicine, Institute of Gastroenterology, College of Medicine Yonsei University, South Korea
| | - Si Young Song
- Division of Gastroenterology, Department of Internal Medicine, Institute of Gastroenterology, College of Medicine Yonsei University, South Korea; Brain Korea 21 Project for Medical Science, Seoul, South Korea
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal Medicine, Institute of Gastroenterology, College of Medicine Yonsei University, South Korea.
| |
Collapse
|
11
|
Quint K, Tonigold M, Di Fazio P, Montalbano R, Lingelbach S, Rückert F, Alinger B, Ocker M, Neureiter D. Pancreatic cancer cells surviving gemcitabine treatment express markers of stem cell differentiation and epithelial-mesenchymal transition. Int J Oncol 2012; 41:2093-2102. [PMID: 23026911 DOI: 10.3892/ijo.2012.1648] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 09/03/2012] [Indexed: 02/07/2023] Open
Abstract
Objective response rates to standard chemotherapeutic regimens remain low in pancreatic cancer. Subpopulations of cells have been identified in various solid tumors which express stem cell-associated markers and are associated with increased resistance against radiochemotherapy. We investigated the expression of stem cell genes and markers of epithelial-mesenchymal transition in pancreatic cancer cells that survived high concentrations of gemcitabine treatment. Capan-1 and Panc-1 cells were continuously incubated with 1 and 10 µM gemcitabine. Surviving cells were collected after 1, 3 and 6 days. Expression of PDX-1, SHH, CD24, CD44, CD133, EpCAM, CBX7, OCT4, SNAIL, SLUG, TWIST, Ki-67, E-cadherin, β-catenin and vimentin were quantified by qPCR or immunocytochemistry. Migration was assessed by wound‑healing assay. SHH was knocked down using RNA interference. Five primary pancreatic cancer cell lines were used to validate the qPCR results. All investigated genes were upregulated after 6 days of gemcitabine incubation. Highest relative expression levels were observed for OCT4 (13.4-fold), CD24 (47.3-fold) and EpCAM (15.9-fold) in Capan-1 and PDX-1 (13.3‑fold), SHH (24.1-fold), CD44 (17.4-fold), CD133 (20.2-fold) and SLUG (15.2-fold) in Panc-1 cells. Distinct upregulation patterns were observed in the primary cells. Migration was increased in Panc-1 cells and changes in the expression of E-cadherin and β-catenin were typical of epithelial-mesenchymal transition in both cell lines. SHH knockdown reduced IC(50) from 30.1 to 27.6 nM in Capan-1 while it strongly inhibited proli-feration in Panc-1 cells. Cells surviving high-dose gemcitabine treatment express increased levels of stem cell genes, show characteristics associated with epithelial-mesenchymal transition and retain their proliferative capacity.
Collapse
Affiliation(s)
- Karl Quint
- Institute for Surgical Research, Philipps University of Marburg, D-35043 Marburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Abstract
Emerging evidence proposes that most cancers originate from a rare subpopulation of cells, called cancer stem cells (CSCs), which possess characteristics including differentiation, self-renewal, and tumorigenicity. Currently, available therapeutic agents cannot effectively eliminate CSCs. Therefore, the development of a nontoxic, natural treatment that can either overcome chemoresistance or promote the elimination of CSCs is highly desirable. The current study examined whether mulberry leaf (ML) ethanolic extract can effectively eliminate neuroblastoma stem cell-like population. Our data demonstrated that 10-40 μg/ml of ML extract significantly enhanced differentiation by elongating neurites and reducing clonogenicity and sphere formation as shown by the decreased expression of stem cell markers and increased expression of differentiation markers. The knock-down of delta-like 1 homologue by siRNA enhanced the significant inhibitory effects of 40 μg/ml of ML extract on colony formation. Furthermore, phosphorylation of the extracellular signal-regulated kinase (ERK) was increased by 20 or 40 μg/ml of ML extract and the MEK/ERK inhibitors completely blocked differentiation induced by the extract. Taken together, these findings provide experimental evidence that ML may have chemopreventive effects on neuroblastoma cells by inhibiting CSCs characteristics as well as regulating CSCs pathways, which may provide a therapeutic option for controlling the growth of neuroblastoma cells.
Collapse
Affiliation(s)
- Seolhyun Park
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Korea
| | | | | |
Collapse
|
13
|
Liu QH, Zhao CY, Zhang J, Chen Y, Gao L, Ni CY, Zhu MH. Role of heat shock protein 27 in gemcitabine-resistant human pancreatic cancer: comparative proteomic analyses. Mol Med Rep 2012; 6:767-73. [PMID: 22858734 DOI: 10.3892/mmr.2012.1013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 07/19/2012] [Indexed: 11/06/2022] Open
Abstract
The most notable obstacle hindering the effective treatment of human pancreatic cancer is intrinsic chemoresistance. In order to identify the candidate protein(s) responsible for the intrinsic chemoresistance, the protein expression profiling of human pancreatic adenocarcinoma cell line Capan-1 and its distinct surviving cells following primary treatment with gemcitabine (GEM) were compared by two-dimensional electrophoresis (2-DE) combined with liquid chromatography-mass spectrometry (LC-MS) or mass spectrometry (MS). In total, nine proteins were identified, and heat shock protein B1 (HSP27), one of the differentially expressed proteins, was selected for further validation. Furthermore, the results of western blotting and immunohistochemical staining indicated that HSP27 may be significant in pancreatic intrinsic chemoresistance to GEM. The findings of this study provide a platform for further elucidation of the underlying mechanisms of pancreatic cancer intrinsic chemoresistance and demonstrate that HSP27 may be a valid target for anticancer drug development.
Collapse
Affiliation(s)
- Qing-Hua Liu
- Department of Pathology, Changhai Hospital, Secondary Military Medical University, Shanghai, PR China
| | | | | | | | | | | | | |
Collapse
|