1
|
Patil VS, Patil CR, Patel HM, Kumar A. Exploring disulfiram mechanisms in renal fibrosis: insights from biological data and computational approaches. Front Pharmacol 2025; 16:1480732. [PMID: 40170735 PMCID: PMC11958968 DOI: 10.3389/fphar.2025.1480732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/03/2025] [Indexed: 04/03/2025] Open
Abstract
Background Disulfiram (DSF) is an anti-alcoholic drug that has been reported to inhibit the epithelial-to-mesenchymal transition and crosslinking during fibrosis, pyroptosis, and inflammatory NF-κB and Nrf-2 signaling pathways. However, there is insufficient evidence to support the mechanisms of DSF in preventing renal fibrosis (RF). Therefore, the current study aimed to elucidate the DSF-modulated targets and pathways in renal fibrosis. Methods The common proteins between DSF and RF were screened for protein-protein interaction, pathway enrichment, cluster, and gene ontology analysis. Molecular docking was executed for core genes using AutoDock Vina through the POAP pipeline. Molecular dynamics (MD) simulation (100 ns) was performed to infer protein-ligand stability, and conformational changes were analyzed by free energy landscape (FEL). Results A total of 78 targets were found to be common between DSF and RF, of which NFKB, PIK3CA/R1, MTOR, PTGS2, and MMP9 were the core genes. PI3K-Akt signaling followed by JAK-STAT, TNF, Ras, ErbB, p53, phospholipase D, mTOR, IL-17, NF-κB, AMPK, VEGF, and MAPK signaling pathways were modulated by DSF in RF. DSF showed a direct binding affinity with active site residues of core genes, and except for DSF with NF-κB, all other complexes, including the standard, were found to be stable during 100 ns MD simulation with minimal protein-ligand root mean squared deviation and residual fluctuations and higher compactness with broad conformational changes. Conclusion DSF protects against renal fibrosis, and this study paves the way for experimental investigation to repurpose DSF for treating RF.
Collapse
Affiliation(s)
- Vishal S. Patil
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Chandragouda R. Patil
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Harun M. Patel
- Department of Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Anoop Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
| |
Collapse
|
2
|
Liang S, Li L, Guo Z, Sun H, Yang Y. Co-expression of CD44v6 and MMP2 predicts lung metastasis and unfavorable prognosis in osteosarcoma. Future Oncol 2024; 20:1799-1806. [PMID: 39011948 PMCID: PMC11485780 DOI: 10.1080/14796694.2024.2370234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024] Open
Abstract
Aim: To evaluate the prognostic significance of CD44 variant v6 (CD44v6) and matrix metalloproteinases 2 (MMP2) expression in patients with surgically resected osteosarcoma.Methods: CD44v6 and MMP2 expression were immunohistochemically detected in 113 primary osteosarcoma patients at our institute between 2001 and 2019.Results: Both CD44v6 and MMP2 were independent predictors for metastasis-free and overall survival. An extended predictive range and improved sensitivity were observed when the combined effects of CD44v6 and MMP2 were considered. Specifically, patients with CD44v6+ and MMP2+ expression were more susceptible to lung metastasis and exhibited the poorest survival rates compared with the other groups.Conclusion: The combination of CD44v6 and MMP2 may serve as a precise prognostic indicator for predicting metastatic progression and survival outcomes in patients with osteosarcoma.
Collapse
Affiliation(s)
- Shoulei Liang
- Department of orthopedic, The Second Hospital of Tangshan, No.21, Jianshe North Road, Tangshan063000, China
| | - Liang Li
- Department of orthopedic, The Second Hospital of Tangshan, No.21, Jianshe North Road, Tangshan063000, China
| | - Zhiliang Guo
- Department of orthopedic, The Second Hospital of Tangshan, No.21, Jianshe North Road, Tangshan063000, China
| | - Haijing Sun
- Department of orthopedic, The Second Hospital of Tangshan, No.21, Jianshe North Road, Tangshan063000, China
| | - Yan Yang
- Department of orthopedic, The Second Hospital of Tangshan, No.21, Jianshe North Road, Tangshan063000, China
| |
Collapse
|
3
|
Zhao P, Tang X, Huang Y. Teaching new tricks to old dogs: A review of drug repositioning of disulfiram for cancer nanomedicine. VIEW 2021. [DOI: 10.1002/viw.20200127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Pengfei Zhao
- School of Chinese Materia Medica Nanjing University of Chinese Medicine Nanjing China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai China
| | - Xueping Tang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai China
- Artemisinin Research Center Guangzhou University of Chinese Medicine Guangzhou China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients Shanghai China
- Zhongshan Institute for Drug Discovery, Institutes of Drug Discovery and Development Chinese Academy of Sciences Zhongshan China
| |
Collapse
|
4
|
Mo J, Zheng T, Lei L, Dai P, Liu J, He H, Shi J, Chen X, Guo T, Yuan B, Ji G. MicroRNA-1253 Suppresses Cell Proliferation Migration and Invasion of Osteosarcoma by Targeting MMP9. Technol Cancer Res Treat 2021; 20:1533033821995278. [PMID: 34036868 PMCID: PMC8161890 DOI: 10.1177/1533033821995278] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Purpose: MicroRNAs play an important role in osteosarcoma (OS) development and progress. Although miR-1253 was considered as a tumor-inhibitor in some cancers, it’s function in the OS is not clear. Methods: In our study, we examined the expression of miR-1253 in OS cells and osteoblast cells using quantitative real-time PCR. The proliferation of OS cells was measured by BrdU assay, and we performed transwell to detect migration and invasion of OS cells. Meanwhile, EMT proteins were tested by western blot. We used Bioinformatics to predict the target genes of miR-1253 and found out Matrix metalloproteinases9 (MMP9) was one of that. The direct combination between miR-1253 and MMP9 was verified by double luciferase reporting experiment. Quantitative real-time PCR and western blot were used to detect the expression of MMP9. Results: We found that the expression level of miR-1253 in OS cells was significantly lower than that in osteoblast cells. Overexpression of miR-1253 could significantly inhibit OS cell proliferation, migration, invasion and EMT. And then, MMP9 was predicted as a downstream target of miR-1253 by Bioinformatics analysis. Further experiments showed that miR-1253 could reduce the protein level of MMP9 by directly binding to the 3’-UTR of MMP9. Afterward, we performed a rescue experiment, in which both MMP9 and miR-1253 were overexpressed. Compared with the groups overexpressed miR-1253 alone, cell proliferation, migration and invasion in co-overexpression groups were improved. Conclusions: In summary, these results suggested that miR-1253 down-regulated in OS cells, and could suppress the proliferation, migration and invasion of OS cells. Its molecular regulatory mechanism was that inhibits the expression of the downstream target gene MMP9 by directly binding, thus affect OS cell functions. Therefore, miR-1253 has the potential to become a biomarker and therapeutic target for OS therapy.
Collapse
Affiliation(s)
- Jianwen Mo
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, People's Republic of China
| | - Tiansheng Zheng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, People's Republic of China
| | - Li Lei
- Department of Dental, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, People's Republic of China
| | - Peng Dai
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, People's Republic of China
| | - Jun Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, People's Republic of China
| | - Huabin He
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, People's Republic of China
| | - Jin Shi
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, People's Republic of China
| | - Xi Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, People's Republic of China
| | - Tianting Guo
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, People's Republic of China
| | - Bin Yuan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, People's Republic of China
| | - Guanglin Ji
- Department of Orthopedic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, People's Republic of China
| |
Collapse
|
5
|
Glycogen synthase kinase 3β promotes osteosarcoma invasion and migration via regulating PTEN and phosphorylation of focal adhesion kinase. Biosci Rep 2021; 41:228616. [PMID: 33969873 PMCID: PMC8314432 DOI: 10.1042/bsr20193514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 12/01/2022] Open
Abstract
Aim: Typical features of human osteosarcoma are highly invasive and migratory capacities. Our study aimed to investigate the roles of glycogen synthase kinase 3β (GSK3β) in human osteosarcoma metastasis. Methods: GSK3β expressions in clinical osteosarcoma tissues with or without metastasis were examined by immunohistochemical staining. The expressions of GSK3β, p-GSK3βSer9, and p-GSK3βTyr216 in human osteoblast cells (hFOB1.19) and human osteosarcoma cells (MG63, SaOS-2, and U2-OS) were detected by Western blotting. The GSK3β activity was measured by non-radio isotopic in vitro kinase assay. Migration and invasion abilities of MG-63 cells treated with small-molecular GSK3β inhibitors were respectively examined by monolayer-based wound-healing assay and transwell assay. The mRNA expressions of GSK3β, matrix metalloproteinase-2 (MMP-2), MMP-9, phosphatase with tensin homology (PTEN), and focal adhesion kinase (FAK) were detected after siRNA transfection for 72 h. Meanwhile, protein expressions of GSK3β, FAK, p-FAKY397, PTEN, MMP-2, and MMP-9 were measured by Western blotting. Results: Clinical osteosarcoma tissues with metastasis showed higher GSK3β expressions. MG63 and U2-OS cells that were easy to occur metastasis showed significantly higher expressions and activities of GSK3β than SaOS-2 cells. Inhibition of GSK3β with small-molecular GSK3β inhibitors in MG63 cells significantly attenuated cell migration and invasion. These effects were associated with reduced expressions of MMP-2 and MMP-9. Moreover, increased PTEN and decreased p-FAKY397 expressions were observed following GSK3β knockdown by siRNA transfection. Conclusion: GSK3β might promote osteosarcoma invasion and migration via pathways associated with PTEN and phosphorylation of FAK.
Collapse
|
6
|
Qian C, Wang J, Qian Y, Hu R, Zou J, Zhu C, Zhu Y, Qi S, Jia X, Wu L, Li W, Chen Z. Tumor-Cell-Surface Adherable Peptide-Drug Conjugate Prodrug Nanoparticles Inhibit Tumor Metastasis and Augment Treatment Efficacy. NANO LETTERS 2020; 20:4153-4161. [PMID: 32462880 DOI: 10.1021/acs.nanolett.0c00152] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cancer metastasis is the main cause of chemotherapeutic failure. Inhibiting the activity of matrix metalloproteinases (MMPs) is a common strategy for reducing metastasis. However, broad-spectrum MMP-inhibitors (MMPI) may cause undesired side effects. Here, we screened a selective MMP2 inhibitor (CCKIGLFRWR) and linked it with doxorubicin (DOX) to produce an amphiphilic peptide-drug conjugate (PDC). Then novel core-shell nanoparticles were self-assembled from PDC core and modified polylysine (MPL) shell. When the particles were passively targeted to the tumor site, the PDC core was exposed for charge switch of the MPL shell, aggregated for its transformation behavior, and specially adhered to the cell membrane. The disulfide bond between the MMPI peptide and DOX was broken via a low concentration of glutathione-mediated reduction in tumor microenvironment. DOX could effectively enter the tumor cells. Meanwhile, the MMPI peptide could selectively inhibit the activity of the MMP2 and effectively inhibit tumor metastasis.
Collapse
Affiliation(s)
- Chen Qian
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jingjing Wang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ying Qian
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Rongfeng Hu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, Anhui 230038, China
| | - Jiayu Zou
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chenqi Zhu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Department of Pharmacy, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, China
| | - Yuan Zhu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shuyang Qi
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaobin Jia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Li Wu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weidong Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhipeng Chen
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
7
|
Xia P, Gu R, Zhang W, Sun YF. lncRNA CEBPA-AS1 Overexpression Inhibits Proliferation and Migration and Stimulates Apoptosis of OS Cells via Notch Signaling. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 19:1470-1481. [PMID: 32160715 PMCID: PMC7056609 DOI: 10.1016/j.omtn.2019.10.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 10/16/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022]
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy derived from primitive bone-forming mesenchymal cells. Long noncoding RNA (lncRNA) expression profiles have been intensively studied for their involvement in OS. Herein, we clarify whether lncRNA CEBPA-AS1 is a regulator of NCOR2 in OS cells. Microarray-based expression analysis identified OS-related differentially expressed lncRNA and predicted microRNAs (miRs) binding to lncRNA and mRNA. lncRNA CEBPA-AS1 and NCOR2 were found to be weakly expressed in OS tissues and cells. Next, functional investigation revealed that lncRNAs CEBPA-AS1 bound to miR-10b-5p to upregulate NCOR2. Following that, gene-targeted knockdown and overexpressed recombinant vectors of lncRNA CEBPA-AS1 and NCOR2 were constructed to explore the effects of lncRNA CEBPA-AS1 and NCOR2 on cell proliferation, differentiation, migration, and apoptosis. Finally, tumor formation was measured in nude mice. lncRNA CEBPA-AS1 overexpression or NCOR2 elevation inhibited cell proliferation and migration, and alkaline phosphatase (ALP) and bone gla protein (BGP) activity, while enhancing apoptosis and tumor formation. Furthermore, NCOR2 was elevated in response to lncRNA CEBPA-AS1 overexpression, thus repressing the Notch signaling pathway. Taken together, lncRNA CEBPA-AS1 overexpression inhibits OS progression through diminishing activation of the Notch signaling pathway via upregulating NCOR2. Therefore, lncRNA CEBPA-AS1 may serve as a molecular target for treating OS.
Collapse
Affiliation(s)
- Peng Xia
- Department of Orthopaedics, Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Rui Gu
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China
| | - Wei Zhang
- Department of Orthopaedics, Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Yi-Fu Sun
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun 130033, P.R. China.
| |
Collapse
|
8
|
Lézot F, Corre I, Morice S, Rédini F, Verrecchia F. SHH Signaling Pathway Drives Pediatric Bone Sarcoma Progression. Cells 2020; 9:cells9030536. [PMID: 32110934 PMCID: PMC7140443 DOI: 10.3390/cells9030536] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/19/2020] [Accepted: 02/23/2020] [Indexed: 02/07/2023] Open
Abstract
Primary bone tumors can be divided into two classes, benign and malignant. Among the latter group, osteosarcoma and Ewing sarcoma are the most prevalent malignant primary bone tumors in children and adolescents. Despite intensive efforts to improve treatments, almost 40% of patients succumb to the disease. Specifically, the clinical outcome for metastatic osteosarcoma or Ewing sarcoma remains poor; less than 30% of patients who present metastases will survive 5 years after initial diagnosis. One common and specific point of these bone tumors is their ability to deregulate bone homeostasis and remodeling and divert them to their benefit. Over the past years, considerable interest in the Sonic Hedgehog (SHH) pathway has taken place within the cancer research community. The activation of this SHH cascade can be done through different ways and, schematically, two pathways can be described, the canonical and the non-canonical. This review discusses the current knowledge about the involvement of the SHH signaling pathway in skeletal development, pediatric bone sarcoma progression and the related therapeutic options that may be possible for these tumors.
Collapse
|
9
|
Zhang Y, Weng Q, Han J, Chen J. Alantolactone suppresses human osteosarcoma through the PI3K/AKT signaling pathway. Mol Med Rep 2019; 21:675-684. [PMID: 31974628 PMCID: PMC6947914 DOI: 10.3892/mmr.2019.10882] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/18/2019] [Indexed: 12/24/2022] Open
Abstract
Osteosarcoma is the most common type of malignant bone cancer and results in cancer-related deaths among adolescents. Alantolactone (ALT) demonstrates antitumor properties in various diseases; however, its potential role in osteosarcoma is relatively unclear. The aim of the present study was to evaluate the effect of ALT on osteosarcoma. ALT significantly decreased the viability of U2OS and HOS osteosarcoma cell lines. Cells flow cytometry assay and Hoechst 33258 staining assay revealed that ALT significantly increased the proportion of apoptotic U2OS cells. In addition, wound healing and Transwell invasion assays demonstrated that the invasion and migration of osteosarcoma were markedly reduced upon ALT treatment. It was hypothesized that the antitumor functions of ALT are mediated through inhibition of the PI3K/AKT signaling pathway. In conclusion, the results of the present study confirmed the inhibition of ALT on osteosarcoma cells via downregulation of PI3K/AKT signaling pathways, suggesting ALT as a potential therapeutic candidate for osteosarcoma.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Trauma Orthopedics, Ningbo No. 6 Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Qiuyan Weng
- Department of Neurology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang 315000, P.R. China
| | - Jinming Han
- Department of Spinal Surgery, Ningbo No. 6 Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Jianming Chen
- Department of Trauma Orthopedics, Ningbo No. 6 Hospital, Ningbo, Zhejiang 315000, P.R. China
| |
Collapse
|
10
|
Drug Repurposing as an Antitumor Agent: Disulfiram-Mediated Carbonic Anhydrase 12 and Anion Exchanger 2 Modulation to Inhibit Cancer Cell Migration. Molecules 2019; 24:molecules24183409. [PMID: 31546841 PMCID: PMC6767608 DOI: 10.3390/molecules24183409] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/15/2019] [Accepted: 09/17/2019] [Indexed: 12/18/2022] Open
Abstract
Disulfiram has been used in the treatment of alcoholism and exhibits an anti-tumor effect. However, the intracellular mechanism of anti-tumor activity of Disulfiram remains unclear. In this study, we focused on the modulatory role of Disulfiram via oncogenic factor carbonic anhydrase CA12 and its associated transporter anion exchanger AE2 in lung cancer cell line A549. The surface expression of CA12 and AE2 were decreased by Disulfiram treatment with a time-dependent manner. Disulfiram treatment did not alter the expression of Na+-bicarbonate cotransporters, nor did it affect autophagy regulation. The chloride bicarbonate exchanger activity of A549 cells was reduced by Disulfiram treatment in a time-dependent manner without change in the resting pH level. The expression and activity of AE2 and the expression of CA12 were also reduced by Disulfiram treatment in the breast cancer cell line. An invasion assay and cell migration assay revealed that Disulfiram attenuated the invasion and migration of A549 cells. In conclusion, the attenuation of AE2 and its supportive enzyme CA12, and the inhibitory effect on cell migration by Disulfiram treatment in cancer cells provided the molecular evidence supporting the potential of Disulfiram as an anticancer agent.
Collapse
|
11
|
Bu W, Wang Z, Meng L, Li X, Liu X, Chen Y, Xin Y, Li B, Sun H. Disulfiram inhibits epithelial-mesenchymal transition through TGFβ-ERK-Snail pathway independently of Smad4 to decrease oral squamous cell carcinoma metastasis. Cancer Manag Res 2019; 11:3887-3898. [PMID: 31118804 PMCID: PMC6504671 DOI: 10.2147/cmar.s199912] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 04/07/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose: Smad4 loss is highly related to poor prognosis and decreased patient survival in oral squamous cell carcinoma (OSCC), suggesting that agents that target both Smad4-mutated and Smad4 wild-type cells could treat OSCC more effectively. Disulfiram (Dsf) has anticancer activity through a variety of mechanisms, including inhibition of epithelial–mesenchymal transition (EMT). It remains unclear whether Dsf has the same effect on Smad4-mutated and Smad4 wild-type OSCC or not and what mechanism is involved. Methods: Effect of Dsf on TGFβ1-induced EMT in CAL27 (Smad4 mutation) and SCC25 (Smad4 wild-type) cells were evaluated through analyzing changes in morphology, expression of EMT markers, and migration and invasion of cells. The ERK-pathway inhibitor U0126 was used to confirm TGFβ–ERK–Snail pathway–mediated cell behavior. Dsf’s effects on tumor growth and metastasis in vivo were examined through a subcutaneous xenograft mouse model and an intravenous tumor mouse model. Results: Dsf inhibited TGFβ1-induced EMT through suppression of morphological change, EMT-marker expression, and cell migration and invasion in both CAL27 and SCC25. Phosphorylation of ERK and expression of Snail were blocked by Dsf treatment. Like Dsf, U0126 had a similar effect on EMT of CAL27 and SCC25. Dsf also reduced tumor growth and metastasis in vivo, accompanied by decreased expression of EMT markers in tumors. Conclusion: These results indicated that Dsf inhibited EMT of OSCC in vitro and in vivo independently of Smad4 through suppression of the TGFβ–ERK–Snail pathway, suggesting the broad-spectrum anticancer potential of Dsf for clinical use against OSCC.
Collapse
Affiliation(s)
- Wenhuan Bu
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, 130000 Changchun, People's Republic of China
| | - Zilin Wang
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, 130000 Changchun, People's Republic of China
| | - Lin Meng
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, 130000 Changchun, People's Republic of China
| | - Xing Li
- School and Hospital of Stomatology, China Medical University, 110000 Shenyang, People's Republic of China
| | - Xinchen Liu
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, 130000 Changchun, People's Republic of China
| | - Yumeng Chen
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, 130000 Changchun, People's Republic of China
| | - Ying Xin
- Department of Oral Pathology, Hospital of Stomatology, Xi'an Jiaotong University, Xi'an, 710000, People's Republic of China
| | - Baoquan Li
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Jilin University, 130000 Changchun, People's Republic of China
| | - Hongchen Sun
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, 130000 Changchun, People's Republic of China
| |
Collapse
|
12
|
MicroRNA-Based Diagnosis and Treatment of Metastatic Human Osteosarcoma. Cancers (Basel) 2019; 11:cancers11040553. [PMID: 31003401 PMCID: PMC6521107 DOI: 10.3390/cancers11040553] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/15/2019] [Accepted: 04/16/2019] [Indexed: 12/24/2022] Open
Abstract
Osteosarcoma is a malignant tumor of the bones that commonly occurs in young individuals. The 5-year survival rate of osteosarcoma patients is 60-70%. Metastasis to the lungs leads to death in 30-40% of osteosarcoma patients. Therefore, the development of effective strategies for early detection and treatment of this disease are important to improve the survival of osteosarcoma patients. However, metastatic markers for osteosarcoma and molecules that might be targeted for the treatment of metastatic osteosarcoma have not been identified yet. Therefore, the mechanism of metastasis to the lungs needs to be explored from a novel viewpoint. Recently, the aberrant expression of microRNAs (miRNAs) has been reported to be involved in the carcinogenesis and cancer progression of many cancers. Furthermore, miRNAs in the blood have been reported to show an aberrant expression unique to several cancers. Therefore, miRNAs are gaining attention as potential diagnostic markers for cancers. On the other hand, normalizing the dysregulated expression of miRNAs in cancer cells has been shown to alter the phenotype of cancer cells, and thus treatment strategies targeting miRNAs are also being considered. This review summarizes the abnormality of miRNA expression associated with the metastasis of osteosarcoma and describes the present situation and issues regarding the early diagnosis and development of treatment strategies for metastatic osteosarcoma based on the current understanding of this disease.
Collapse
|
13
|
Trucco MM, Meyer CF, Thornton KA, Shah P, Chen AR, Wilky BA, Carrera-Haro MA, Boyer LC, Ferreira MF, Shafique U, Powell JD, Loeb DM. A phase II study of temsirolimus and liposomal doxorubicin for patients with recurrent and refractory bone and soft tissue sarcomas. Clin Sarcoma Res 2018; 8:21. [PMID: 30410720 PMCID: PMC6217787 DOI: 10.1186/s13569-018-0107-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/12/2018] [Indexed: 01/16/2023] Open
Abstract
Background Relapsed and refractory sarcomas continue to have poor survival rates. The cancer stem cell (CSC) theory provides a tractable explanation for the observation that recurrences occur despite dramatic responses to upfront chemotherapy. Preclinical studies demonstrated that inhibition of the mechanistic target of rapamycin (mTOR) sensitizes the CSC population to chemotherapy. Methods Here we present the results of the Phase II portion of a Phase I/II clinical trial that aimed to overcome the chemoresistance of sarcoma CSC by combining the mTOR inhibitor temsirolimus (20 mg/m2 weekly) with the chemotherapeutic agent liposomal doxorubicin (30 mg/m2 monthly). Results Fifteen patients with relapsed/refractory sarcoma were evaluable at this recommended Phase 2 dose level. The median progression free survival was 315 days (range 27–799). Response rate, defined as stable disease or better for 60 days, was 53%. Nine of the patients had been previously treated with doxorubicin. Therapy was well tolerated. In a small number of patients, pre- and post- treatment tumor biopsies were available for assessment of ALDH expression as a marker of CSCs and showed a correlation between response and decreased ALDH expression. We also found a correlation between biopsy-proven inhibition of mTOR and response. Conclusions Our study adds to the literature supporting the addition of mTOR inhibition to chemotherapy agents for the treatment of sarcomas, and proposes that a mechanism by which mTOR inhibition enhances the efficacy of chemotherapy may be through sensitizing the chemoresistant CSC population. Further study, ideally with pre- and post-therapy assessment of ALDH expression in tumor cells, is warranted. Trial registration The trial was registered on clinicaltrials.gov (NCT00949325) on 30 July 2009. http://www.editorialmanager.com/csrj/default.aspx
Collapse
Affiliation(s)
- Matteo M Trucco
- 1Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD USA.,3Present Address: Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL USA
| | - Christian F Meyer
- 2Division of Medical Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD USA
| | - Katherine A Thornton
- 2Division of Medical Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD USA.,4Present Address: Dana Farber Cancer Institute, Boston, MA USA
| | - Preeti Shah
- 1Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD USA.,Present Address: Akan Biosciences, Gaithersburg, MD USA
| | - Allen R Chen
- 1Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD USA
| | - Breelyn A Wilky
- 2Division of Medical Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD USA.,3Present Address: Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL USA
| | - Maria A Carrera-Haro
- 1Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD USA.,6Present Address: Columbia University College of Physicians and Surgeons, New York, NY USA
| | - Lillian C Boyer
- 1Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD USA
| | - Margaret F Ferreira
- 1Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD USA
| | - Umber Shafique
- 1Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD USA
| | - Jonathan D Powell
- 2Division of Medical Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD USA
| | - David M Loeb
- 1Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD USA.,7Department of Pediatrics, Albert Einstein College of Medicine, Children's Hospital at Montefiore, 3411 Wayne Ave., Room 910, Bronx, NY 10467 USA
| |
Collapse
|
14
|
Chalcone Derivatives 4'-Amino-1-Naphthyl-Chalcone (D14) and 4'-Amino-4-Methyl-1-Naphthyl-Chalcone (D15) Suppress Migration and Invasion of Osteosarcoma Cells Mediated by p53 Regulating EMT-Related Genes. Int J Mol Sci 2018; 19:ijms19092838. [PMID: 30235848 PMCID: PMC6163733 DOI: 10.3390/ijms19092838] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/04/2018] [Accepted: 09/04/2018] [Indexed: 12/11/2022] Open
Abstract
Osteosarcoma (OS) is a primary malignant bone tumor that mainly affects children, adolescents, and young adults. The inhibition of metastasis is a main strategy of OS therapy since the development of metastatic disease due to drug resistance remains the most important cause of death from this cancer. Considering the severe side effects of current OS chemotherapy, the identification of anti-metastatic drugs with reduced toxicity is of great interest. Chalcones are polyphenols with a basic structure consisting of an α-, β-unsaturated carbonyl system linking two aryl rings. These compounds exhibit anticancer activity against a variety of tumor cell lines through multiple mechanisms, including the regulation of the tumor-suppressor protein p53 and its target genes. An important process regulated by p53 is epithelial-mesenchymal transition (EMT), which facilitates tumor metastasis by conferring migratory and invasive properties to cancer cells. The activation of p53 can revert EMT and reduce migration and invasion. This study aimed to examine the inhibitory effects of two 4′-aminochalcones on the migration/invasion of the U2OS (p53+/+) and SAOS-2 (p53−/−) OS cell lines as well as the underlying molecular mechanisms. Cell viability was examined by MTT assay. Transwell assays were used to evaluate the migratory and invasive ability of the cells. The two 4′-aminochalcones showed low capacity to inhibit the viability of OS cells independent of p53 status, but preferentially suppressed the migration of U2OS cells and of a SAOS-2 cell line expressing p53. Invasion was strongly inhibited by both chalcones independent of p53 status. RT-PCR, zymography, and Western blot were used to study the expression of matrix metalloproteinases and EMT markers after treatment with the chalcones. The results indicated that the 4′-aminochalcone-induced antimigratory and anti-invasive effects are potentially associated with the inhibition of extracellular matrix (ECM) enzymatic degradation in OS cells and with the modulation of EMT genes. These effects probably result from the induced increase of p53 protein expression by the two chalcones. In conclusion, chalcones D14 and D15 have potential anti-metastatic activity mediated by p53 that can be exploited for OS treatment.
Collapse
|
15
|
Butcher K, Kannappan V, Kilari RS, Morris MR, McConville C, Armesilla AL, Wang W. Investigation of the key chemical structures involved in the anticancer activity of disulfiram in A549 non-small cell lung cancer cell line. BMC Cancer 2018; 18:753. [PMID: 30031402 PMCID: PMC6054747 DOI: 10.1186/s12885-018-4617-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/20/2018] [Indexed: 12/11/2022] Open
Abstract
Background Disulfiram (DS), an antialcoholism medicine, demonstrated strong anticancer activity in the laboratory but did not show promising results in clinical trials. The anticancer activity of DS is copper dependent. The reaction of DS and copper generates reactive oxygen species (ROS). After oral administration in the clinic, DS is enriched and quickly metabolised in the liver. The associated change of chemical structure may make the metabolites of DS lose its copper-chelating ability and disable their anticancer activity. The anticancer chemical structure of DS is still largely unknown. Elucidation of the relationship between the key chemical structure of DS and its anticancer activity will enable us to modify DS and speed its translation into cancer therapeutics. Methods The cytotoxicity, extracellular ROS activity, apoptotic effect of DS, DDC and their analogues on cancer cells and cancer stem cells were examined in vitro by MTT assay, western blot, extracellular ROS assay and sphere-reforming assay. Results Intact thiol groups are essential for the in vitro cytotoxicity of DS. S-methylated diethyldithiocarbamate (S-Me-DDC), one of the major metabolites of DS in liver, completely lost its in vitro anticancer activity. In vitro cytotoxicity of DS was also abolished when its thiuram structure was destroyed. In contrast, modification of the ethyl groups in DS had no significant influence on its anticancer activity. Conclusions The thiol groups and thiuram structure are indispensable for the anticancer activity of DS. The liver enrichment and metabolism may be the major obstruction for application of DS in cancer treatment. A delivery system to protect the thiol groups and development of novel soluble copper-DDC compound may pave the path for translation of DS into cancer therapeutics. Electronic supplementary material The online version of this article (10.1186/s12885-018-4617-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kate Butcher
- Faculty of Science & Engineering, University of Wolverhampton, Wolverhampton, WV1 1LY, UK
| | - Vinodh Kannappan
- Faculty of Science & Engineering, University of Wolverhampton, Wolverhampton, WV1 1LY, UK
| | | | - Mark R Morris
- Faculty of Science & Engineering, University of Wolverhampton, Wolverhampton, WV1 1LY, UK
| | | | - Angel L Armesilla
- Faculty of Science & Engineering, University of Wolverhampton, Wolverhampton, WV1 1LY, UK
| | - Weiguang Wang
- Faculty of Science & Engineering, University of Wolverhampton, Wolverhampton, WV1 1LY, UK.
| |
Collapse
|
16
|
Chiappetta C, Puggioni C, Carletti R, Petrozza V, Della Rocca C, Di Crisfofano C. The nuclear-cytoplasmic trafficking of a chromatin-modifying and remodelling protein (KMT2C), in osteosarcoma. Oncotarget 2018; 9:30624-30634. [PMID: 30093974 PMCID: PMC6078128 DOI: 10.18632/oncotarget.25755] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/25/2018] [Indexed: 02/05/2023] Open
Abstract
Osteosarcoma is the most common paediatric primary non-hematopoietic bone tumor; the survival is related to the response to chemotherapy and development of metastases. KMT2C is a chromatin-modifying and remodelling protein and its expression has never been studied in osteosarcoma. The aim of this study was to understand the role of KMT2C in the osteosarcoma carcinogenesis and metastatic progression to identify a new molecular target and to provide new therapeutic approach. We performed the immunohistochemical and gene expression analysis of KMT2C in 32 samples of patients with diagnosis of osteosarcoma with known clinic-pathological data and we analysed the expression of genes involved in the metastatic pathway in four osteosarcoma cell lines by blocking the KMT2C expression using siRNA. We found a nuclear-cytoplamic trafficking of KMT2C and the cytoplasmic localization was higher than the nuclear localization (p < 0.0001). Moreover, the percentage of cells with cytoplasmic positivity increased from low grade primary tissue to metastatic tissues. The cytoplasmic localization of KMT2C could lead to a change in its function supporting osteosarcoma carcinogenesis and progression. Our hypothesis is that KMT2C could affect the enhancer activity of genes influencing the invasive properties and metastatic potential of osteosarcoma.
Collapse
Affiliation(s)
- Caterina Chiappetta
- UOC of Pathology, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Latina, Italy
| | - Chiara Puggioni
- UOC of Pathology, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Latina, Italy
| | - Raffaella Carletti
- UOC of Pathology, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Latina, Italy
| | - Vincenzo Petrozza
- UOC of Pathology, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Latina, Italy
| | - Carlo Della Rocca
- UOC of Pathology, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Latina, Italy
| | - Claudio Di Crisfofano
- UOC of Pathology, Department of Medical-Surgical Sciences and Bio-Technologies, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
17
|
Crasto JA, Fourman MS, Morales-Restrepo A, Mahjoub A, Mandell JB, Ramnath K, Tebbets JC, Watters RJ, Weiss KR. Disulfiram reduces metastatic osteosarcoma tumor burden in an immunocompetent Balb/c or-thotopic mouse model. Oncotarget 2018; 9:30163-30172. [PMID: 30046395 PMCID: PMC6059028 DOI: 10.18632/oncotarget.25733] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/14/2018] [Indexed: 12/16/2022] Open
Abstract
Introduction The overall survival rate of patients with osteosarcoma (OS) and pulmonary metastases has remained stagnant at 15–30% for several decades. Disulfiram (DSF) is an FDA-approved aldehyde dehydrogenase inhibitor that reduces the metastatic phenotype of OS cells in vitro. Here we evaluate its in vivo efficacy, as compared to doxorubicin chemotherapy, in a previously-validated orthotopic model of metastatic OS. Results All treatment groups displayed a significantly reduced quantitative OS metastatic burden compared with controls. The metastatic burden of Lo DSF-treated animals was equivalent to the DXR group. Ninety-five percent of control animals displayed evidence of metastatic disease, which was significantly greater than all treatment groups. Discussion Disulfiram treatment resulted in a reduced burden of OS metastatic disease compared with controls. This was statistically-equivalent to doxorubicin. No additive effect was observed between these two therapies. Materials and Methods One-hundred twenty immunocompetent Balb/c mice received proximal tibia paraphyseal injections of 5 × 105 K7M2 murine OS cells. Therapy began three weeks after injection: saline (control), low-dose disulfiram (Lo DSF), high-dose disulfiram (Hi DSF), doxorubicin (DXR), Lo DSF + DXR, and Hi DSF + DXR. Transfemoral amputations were performed at 4 weeks. Quantitative metastatic tumor burden was measured using near-infrared indocyanine green (ICG) angiography.
Collapse
Affiliation(s)
- Jared Anthony Crasto
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mitchell Stephen Fourman
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alejandro Morales-Restrepo
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adel Mahjoub
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan Brendan Mandell
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kavita Ramnath
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Carnegie Mellon University, Pittsburgh, PA, USA
| | - Jessica C Tebbets
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rebecca J Watters
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, PA, USA
| | - Kurt Richard Weiss
- Musculoskeletal Oncology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Departments of Anatomic Pathology and General Surgical Oncology, University of Pittsburgh, PA, USA
| |
Collapse
|
18
|
Huang Z, Li J, Du S, Tang Y, Huang L, Xiao L, Tong P. FKBP14 overexpression contributes to osteosarcoma carcinogenesis and indicates poor survival outcome. Oncotarget 2018; 7:39872-39884. [PMID: 27223089 PMCID: PMC5129977 DOI: 10.18632/oncotarget.9524] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 04/16/2016] [Indexed: 12/21/2022] Open
Abstract
The FK506-binding protein 14 (FKBP14) is a subfamily of immunophilins, has been implicated in various biochemical processes. However, its effects on the primary malignant bone tumor, osteosarcoma, are unclear. Here, we reported that FKBP14 may be an oncogene as it overexpressed in osteosarcoma tissues and cell lines, and FKBP14 expression was correlated with metastases, recurrence, tumor maximum diameter and poor survival time. FKBP14 was associated with the biological pathways including cell cycle, apoptosis and metastasis. Furthermore, we detected FKBP14 knockdown induced cell cycle arrest, apoptosis, invasion and adhesion in vitro. FKBP14 knockdown decreased the protein levels of PCNA, CDK1 and CCNB1 that promotes cell cycle, increased Bax, caspase-3 and caspase-7 protein involved in promoting cell apoptosis, and increased KIF4A expression as well as decreased SMC4 and TMEM33 proteins that contribute to cell invasion and adhesion. In addition, FKBP14 knockdown also caused a significant inhibition in tumor growth in vivo. Then, we found that the protein RhoA was identified as a binding partner of FKBP14. Taken together, FKBP14 may act as an oncogene in osteosarcoma via suppressing apoptosis and promoting invasion and adhesion in osteosarcoma carcinogenesis. FKBP14 may be a prognostic factor and potential target for osteosarcoma treatment.
Collapse
Affiliation(s)
- Zhongming Huang
- Department of Orthopaedic Surgery, Affiliated Jiangnan Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China.,Department of Orthopaedic Surgery, Xiaoshan Chinese Medical Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China.,Zhejiang Chinese Medical University, Hangzhou 310053, China.,Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou 310053, China
| | - Junhua Li
- Department of Orthopaedic Surgery, Affiliated Jiangnan Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China.,Department of Orthopaedic Surgery, Xiaoshan Chinese Medical Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Shaohua Du
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310053, China
| | - Yanghua Tang
- Department of Orthopaedic Surgery, Affiliated Jiangnan Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China.,Department of Orthopaedic Surgery, Xiaoshan Chinese Medical Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Ligang Huang
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Luwei Xiao
- Zhejiang Chinese Medical University, Hangzhou 310053, China.,Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou 310053, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Peijian Tong
- Zhejiang Chinese Medical University, Hangzhou 310053, China.,Institute of Orthopaedics and Traumatology of Zhejiang Province, Hangzhou 310053, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
19
|
Li Y, Wang LH, Zhang HT, Wang YT, Liu S, Zhou WL, Yuan XZ, Li TY, Wu CF, Yang JY. Disulfiram combined with copper inhibits metastasis and epithelial-mesenchymal transition in hepatocellular carcinoma through the NF-κB and TGF-β pathways. J Cell Mol Med 2017; 22:439-451. [PMID: 29148232 PMCID: PMC5742719 DOI: 10.1111/jcmm.13334] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 06/25/2017] [Indexed: 01/01/2023] Open
Abstract
Late‐stage hepatocellular carcinoma (HCC) usually has a low survival rate because of the high risk of metastases and the lack of an effective cure. Disulfiram (DSF) has copper (Cu)‐dependent anticancer properties in vitro and in vivo. The present work aims to explore the anti‐metastasis effects and molecular mechanisms of DSF/Cu on HCC cells both in vitro and in vivo. The results showed that DSF inhibited the proliferation, migration and invasion of HCC cells. Cu improved the anti‐metastatic activity of DSF, while Cu alone had no effect. Furthermore, DSF/Cu inhibited both NF‐κB and TGF‐β signalling, including the nuclear translocation of NF‐κB subunits and the expression of Smad4, leading to down‐regulation of Snail and Slug, which contributed to phenotype epithelial–mesenchymal transition (EMT). Finally, DSF/Cu inhibited the lung metastasis of Hep3B cells not only in a subcutaneous tumour model but also in an orthotopic liver metastasis assay. These results indicated that DSF/Cu suppressed the metastasis and EMT of hepatic carcinoma through NF‐κB and TGF‐β signalling. Our study indicates the potential of DSF/Cu for therapeutic use.
Collapse
Affiliation(s)
- Yi Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, China
| | - Li-Hui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, China
| | - Hao-Tian Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, China
| | - Ya-Ting Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Shuai Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, China
| | - Wen-Long Zhou
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiang-Zhong Yuan
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Tian-Yang Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Chun-Fu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, China
| | - Jing-Yu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China.,Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
20
|
Li Y, Du W, Han J, Ge J. LAMP3 promotes the invasion of osteosarcoma cells via SPP1 signaling. Mol Med Rep 2017; 16:5947-5953. [PMID: 28849219 PMCID: PMC5865773 DOI: 10.3892/mmr.2017.7349] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 06/09/2017] [Indexed: 01/27/2023] Open
Abstract
Osteosarcoma is the most common type of primary bone cancer in children and young adults. The prognosis of osteosarcoma is very poor when it is diagnosed with metastasis. Lysosomal‑associated membrane protein 3 (LAMP3) is a tumor‑specific protein induced by hypoxia, which stimulates invasion and metastasis of various cancer cells via hypoxia‑inducible factor (HIF). A previous study from our group has reported that expression of LAMP3 is significantly increased in lung metastatic osteosarcoma compared with primary osteosarcoma using microarray analysis, suggesting that LAMP3 may be involved in metastatic osteosarcoma. The present study therefore aimed to investigate the role of LAMP3 in osteosarcoma metastasis. Knockdown of LAMP3 decreased the invasion of two osteosarcoma cell lines in vitro. Furthermore, knockdown of LAMP3 increased the expression of secreted phosphoprotein 1 (SPP1), cadherin 1, and keratin 19, while it decreased the expression of matrix metallopeptidase 2, collagen type III α 1, twist family bHLH transcription factor 1 and cadherin 2. Concurrent knockdown of SPP1 and LAMP3 attenuated the changes in gene expression profile induced by LAMP3 knockdown alone. Gene ontology and KEGG analysis demonstrated that SPP1 was involved in cell adhesion, focal adhesion, and extracellular matrix‑receptor interaction. In conclusion, the present results suggest that LAMP3 may be involved in the invasion and metastasis of osteosarcoma via regulating signaling downstream of SPP1. Thus, LAMP3/SPP1 signaling may serve as a potential target in the future to prevent osteosarcoma metastasis.
Collapse
Affiliation(s)
- Yu Li
- Department of Bone Trauma, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Wei Du
- Department of Spine Branch, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Jian Han
- Department of Bone Oncology, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Junbo Ge
- Department of Bone Trauma, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
21
|
Huang S, Jin A. ZIC2 promotes viability and invasion of human osteosarcoma cells by suppressing SHIP2 expression and activating PI3K/AKT pathways. J Cell Biochem 2017; 119:2248-2257. [PMID: 28857346 DOI: 10.1002/jcb.26387] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/24/2017] [Indexed: 12/20/2022]
Abstract
Osteosarcoma is a malignant tumor of the skeletal system. The zinc finger transcription factor ZIC2 has been reported to be highly expressed in human cancers. The present study evaluated the effects of ZIC2 and the possible underlying mechanisms in the human osteosarcoma cells. The expression levels of ZIC2 in human fetal osteoblastic cell line (hFOB1.19), osteosarcoma cell lines (U-2OS, SaoS2, and MG63), normal bone tissue, and osteosarcoma tumor were analyzed by Western blot, and real-time quantitative RT-PCR (qRT-PCR). Osteosarcoma cells with either overexpressed ZIC2 or suppressed ZIC2 were analyzed to determine cell viability, colony formation, and cell invasion. The expressions of SHIP2 and PI3K/AKT signal pathway-related proteins were analyzed by Western blot and qRT-PCR. We first showed that ZIC2 is highly expressed in osteosarcoma cells and tissues. Then we demonstrated that overexpression of ZIC2 promoted viability, migration, and invasion of osteosarcoma cells, whereas suppression of ZIC2 showed opposite effects. Furthermore, SHIP2 expression was negatively regulated by ZIC2. Importantly, ZIC2 overexpression activated the PI3K/AKT signal pathway; however, overexpressed SHIP2 inhibited these effects. Lastly, we showed that activation of the PI3K/AKT signal pathway is essential for the effects of ZIC2 on osteosarcoma cells, as the effects of ZIC2 on the osteosarcoma cells were reversed by a PI3K/AKT inhibitor. Overall, ZIC2 is highly expressed in osteosarcoma cells and tissues, and its overexpression promotes viability, invasion of osteosarcoma cells via SHIP2 suppression, and PI3K/AKT activation. Thus, ZIC2 can be considered as a novel drug target for osteosarcoma management.
Collapse
Affiliation(s)
- Shuaihao Huang
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Anmin Jin
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
22
|
Sun W, Ma X, Shen J, Yin F, Wang C, Cai Z. Bioinformatics analysis of differentially expressed pathways related to the metastatic characteristics of osteosarcoma. Int J Mol Med 2016; 38:466-74. [PMID: 27353415 PMCID: PMC4935462 DOI: 10.3892/ijmm.2016.2657] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 06/03/2016] [Indexed: 12/17/2022] Open
Abstract
In this study, gene expression data of osteosarcoma (OSA) were analyzed to identify metastasis-related biological pathways. Four gene expression data sets (GSE21257, GSE9508, GSE49003 and GSE66673) were downloaded from Gene Expression Omnibus (GEO). An analysis of differentially expressed genes (DEGs) was performed using the Significance Analysis of Microarray (SAM) method. Gene expression levels were converted into scores of pathways by the Functional Analysis of Individual Microarray Expression (FAIME) algorithm and the differentially expressed pathways (DEPs) were then disclosed by a t-test. The distinguishing and prediction ability of the DEPs for metastatic and non-metastatic OSA was further confirmed using the principal component analysis (PCA) method and 3 gene expression data sets (GSE9508, GSE49003 and GSE66673) based on the support vector machines (SVM) model. A total of 616 downregulated and 681 upregulated genes were identified in the data set, GSE21257. The DEGs could not be used to distinguish metastatic OSA from non-metastatic OSA, as shown by PCA. Thus, an analysis of DEPs was further performed, resulting in 14 DEPs, such as NRAS signaling, Toll-like receptor (TLR) signaling, matrix metalloproteinase (MMP) regulation of cytokines and tumor necrosis factor receptor-associated factor (TRAF)-mediated interferon regulatory factor 7 (IRF7) activation. Cluster analysis indicated that these pathways could be used to distinguish between metastatic OSA from non-metastatic OSA. The prediction accuracy was 91, 66.7 and 87.5% for the data sets, GSE9508, GSE49003 and GSE66673, respectively. The results of PCA further validated that the DEPs could be used to distinguish metastatic OSA from non-metastatic OSA. On the whole, several DEPs were identified in metastatic OSA compared with non-metastatic OSA. Further studies on these pathways and relevant genes may help to enhance our understanding of the molecular mechanisms underlying metastasis and may thus aid in the development of novel therapies.
Collapse
Affiliation(s)
- Wei Sun
- Department of Orthopedics, Shanghai General Hospital, Nanjing Medical University, Shanghai 200072, P.R. China
| | - Xiaojun Ma
- Department of Orthopedics, Shanghai General Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai 200080, P.R. China
| | - Jiakang Shen
- Department of Orthopedics, Shanghai General Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai 200080, P.R. China
| | - Fei Yin
- Department of Orthopedics, Shanghai General Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai 200080, P.R. China
| | | | - Zhengdong Cai
- Department of Orthopedics, Shanghai General Hospital, Nanjing Medical University, Shanghai 200072, P.R. China
| |
Collapse
|
23
|
Kunz P, Sähr H, Lehner B, Fischer C, Seebach E, Fellenberg J. Elevated ratio of MMP2/MMP9 activity is associated with poor response to chemotherapy in osteosarcoma. BMC Cancer 2016; 16:223. [PMID: 26979530 PMCID: PMC4793522 DOI: 10.1186/s12885-016-2266-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 03/10/2016] [Indexed: 11/15/2022] Open
Abstract
Background Matrix metalloproteinases (MMPs) are crucially involved in the regulation of multiple stages of cancer progression. Elevated MMP levels have been associated with the development of metastases and poor prognosis in several types of cancer. However, the role of MMPs in osteosarcoma and their prognostic value is still unclear. Available data are conflicting, most likely due to different technical approaches. We hypothesized that in contrast to total mRNA or protein levels frequently analyzed in previous studies the enzymatic activities of MMPs and their inhibitors the tissue inhibitors of matrix metalloproteinases (TIMPs) are closer related to their biological functions. We therefore aimed to evaluate the reliability of different zymography techniques for the quantification of MMP and TIMP activities in osteosarcoma biopsies in order to investigate their distribution, possible regulation and prognostic value. Methods All analyses were done using cryo-conserved osteosarcoma pretreatment biopsies (n = 18). Gene and protein expression of MMPs and TIMPs were analyzed by RT-qPCR and western blot analysis, respectively. Overall MMP activity was analyzed by in situ zymography, individual MMP activities were analyzed by gelatin zymography. Reverse zymography was used to detect and quantify TIMP activities. Results Strong overall MMP activities could be detected in osteosarcoma pretreatment biopsies with MMP2 and MMP9 as predominant active MMPs. In contrast to total RNA or protein expression MMP2 and MMP9 activities showed significant quantitative differences between good and poor responders. While MMP9 activity was high in the good responder group and significantly decreased in the poor responder group, MMP2 activity showed a reverse distribution. Likewise, significant differences were detected concerning the activity of TIMPs resulting in a negative correlation of TIMP1 activity with MMP2 activity (p = 0.044) and negative correlations of TIMP2 and TIMP3 with MMP9 activity (p = 0.007 and p = 0.006). Conclusion In contrast to mRNA or protein levels MMP and TIMP activities showed significant differences between the analyzed good and poor responder groups. A shift from MMP9 to predominant MMP2 activity is associated with poor response to chemotherapy suggesting that the ratio of MMP2/MMP9 activity might be a valuable and easily accessible marker to predict the response to chemotherapy in osteosarcoma.
Collapse
Affiliation(s)
- Pierre Kunz
- Clinic for Orthopedics and Trauma Surgery/Spinal Cord Injury Center; Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, University Hospital Heidelberg, Heidelberg, Germany
| | - Heiner Sähr
- Clinic for Orthopedics and Trauma Surgery/Spinal Cord Injury Center; Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, University Hospital Heidelberg, Heidelberg, Germany
| | - Burkhard Lehner
- Clinic for Orthopedics and Trauma Surgery/Spinal Cord Injury Center; Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, University Hospital Heidelberg, Heidelberg, Germany
| | - Christian Fischer
- Clinic for Orthopedics and Trauma Surgery/Spinal Cord Injury Center; Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, University Hospital Heidelberg, Heidelberg, Germany
| | - Elisabeth Seebach
- Clinic for Orthopedics and Trauma Surgery/Spinal Cord Injury Center; Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, University Hospital Heidelberg, Heidelberg, Germany
| | - Jörg Fellenberg
- Clinic for Orthopedics and Trauma Surgery/Spinal Cord Injury Center; Center for Orthopedics, Trauma Surgery and Spinal Cord Injury, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
24
|
Sharma V, Verma V, Lal N, Yadav SK, Sarkar S, Mandalapu D, Porwal K, Rawat T, Maikhuri JP, Rajender S, Sharma VL, Gupta G. Disulfiram and its novel derivative sensitize prostate cancer cells to the growth regulatory mechanisms of the cell by re-expressing the epigenetically repressed tumor suppressor-estrogen receptor β. Mol Carcinog 2015; 55:1843-1857. [PMID: 26599461 DOI: 10.1002/mc.22433] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 10/20/2015] [Accepted: 11/03/2015] [Indexed: 11/09/2022]
Abstract
Estrogen Receptor-β (ER-β), a tumor-suppressor in prostate cancer, is epigenetically repressed by hypermethylation of its promoter. DNA-methyltransferases (DNMTs), which catalyze the transfer of methyl-groups to CpG islands of gene promoters, are overactive in cancers and can be inhibited by DNMT-inhibitors to re-express the tumor suppressors. The FDA-approved nucleoside DNMT-inhibitors like 5-Azacytidine and 5-Aza-deoxycytidine carry notable concerns due to their off-target toxicity, therefore non-nucleoside DNMT inhibitors are desirable for prolonged epigenetic therapy. Disulfiram (DSF), an antabuse drug, inhibits DNMT and prevents proliferation of cells in prostate and other cancers, plausibly through the re-expression of tumor suppressors like ER-β. To increase the DNMT-inhibitory activity of DSF, its chemical scaffold was optimized and compound-339 was discovered as a doubly potent DSF-derivative with similar off-target toxicity. It potently and selectively inhibited cell proliferation of prostate cancer (PC3/DU145) cells in comparison to normal (non-cancer) cells by promoting cell-cycle arrest and apoptosis, accompanied with inhibition of total DNMT activity, and re-expression of ER-β (mRNA/protein). Bisulfite-sequencing of ER-β promoter revealed that compound-339 demethylated CpG sites more efficaciously than DSF, restoring near-normal methylation status of ER-β promoter. Compound-339 docked on to the MTase domain of DNMT1 with half the energy of DSF. In xenograft mice-model, the tumor volume regressed by 24% and 50% after treatment with DSF and compound-339, respectively, with increase in ER-β expression. Apparently both compounds inhibit prostate cancer cell proliferation by re-expressing the epigenetically repressed tumor-suppressor ER-β through inhibition of DNMT activity. Compound-339 presents a new lead for further study as an anti-prostate cancer agent. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Vikas Sharma
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Vikas Verma
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Nand Lal
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, India
| | - Santosh K Yadav
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Saumya Sarkar
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Dhanaraju Mandalapu
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, India
| | - Konica Porwal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Tara Rawat
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, India
| | - J P Maikhuri
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Singh Rajender
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - V L Sharma
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, India
| | - Gopal Gupta
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India.
| |
Collapse
|
25
|
Zhang L, Tian B, Li Y, Lei T, Meng J, Yang L, Zhang Y, Chen F, Zhang H, Xu H, Zhang Y, Tang X. A Copper-Mediated Disulfiram-Loaded pH-Triggered PEG-Shedding TAT Peptide-Modified Lipid Nanocapsules for Use in Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2015; 7:25147-25161. [PMID: 26501354 DOI: 10.1021/acsami.5b06488] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Disulfiram, which exhibits marked tumor inhibition mediated by copper, was encapsulated in lipid nanocapsules modified with TAT peptide (TATp) and pH-triggered sheddable PEG to target cancer cells on the basis of tumor environmental specificity. PEG-shedding lipid nanocapsules (S-LNCs) were fabricated from LNCs by decorating short PEG chains with TATp (HS-PEG(1k)-TATp) to form TATp-LNCs and then covered by pH-sensitive graft copolymers of long PEG chains (PGA-g-PEG(2k)). The DSF-S-LNCs had sizes in the range of 60-90 nm and were stable in the presence of 50% plasma. DSF-S-LNCs exhibited higher intracellular uptake and antitumor activity at pH 6.5 than at pH 7.4. The preincubation of Cu showed that the DSF cytotoxicity was based on the accumulation of Cu in Hep G2 cells. Pharmacokinetic studies showed the markedly improved pharmacokinetic profiles of DSF-S-LNCs (AUC= 3921.391 μg/L·h, t(1/2z) = 1.294 h) compared with free DSF (AUC = 907.724 μg/L·h, t(1/2z) = 0.252 h). The in vivo distribution of S-LNCs was investigated using Cy5.5 as a fluorescent probe. In tumor-bearing mice, the delivery efficiency of S-LNCs was found to be 496.5% higher than that of free Cy5.5 and 74.5% higher than that of LNCs in tumors. In conclusion, DSF-S-LNCs increased both the stability and tumor internalization and further increased the cytotoxicity because of the higher copper content.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University , Shenyang, Liaoning, PR China
| | - Bin Tian
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University , Shenyang, Liaoning, PR China
| | - Yi Li
- Department of Pharmacology, Shenyang Pharmaceutical University , Shenyang, Liaoning, PR China
| | - Tian Lei
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University , Shenyang, Liaoning, PR China
| | - Jia Meng
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University , Shenyang, Liaoning, PR China
| | - Liu Yang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University , Shenyang, Liaoning, PR China
| | - Yan Zhang
- Normal College, Shenyang University , Shenyang, Liaoning, PR China
| | - Fen Chen
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University , Shenyang, Liaoning, PR China
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine , Shenyang, Liaoning, PR China
| | - Haotian Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University , Shenyang, Liaoning, PR China
| | - Hui Xu
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University , Shenyang, Liaoning, PR China
| | - Yu Zhang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University , Shenyang, Liaoning, PR China
| | - Xing Tang
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University , Shenyang, Liaoning, PR China
| |
Collapse
|
26
|
Rodriguez-Torres M, Allan AL. Aldehyde dehydrogenase as a marker and functional mediator of metastasis in solid tumors. Clin Exp Metastasis 2015; 33:97-113. [PMID: 26445849 PMCID: PMC4740561 DOI: 10.1007/s10585-015-9755-9] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 10/01/2015] [Indexed: 12/16/2022]
Abstract
There is accumulating evidence indicating that aldehyde dehydrogenase (ALDH) activity selects for cancer cells with increased aggressiveness, capacity for sustained proliferation, and plasticity in primary tumors. However, emerging data also suggests an important mechanistic role for the ALDH family of isoenzymes in the metastatic activity of tumor cells. Recent studies indicate that ALDH correlates with either increased or decreased metastatic capacity in a cellular context-dependent manner. Importantly, it appears that different ALDH isoforms support increased metastatic capacity in different tumor types. This review assesses the potential of ALDH as biological marker and mechanistic mediator of metastasis in solid tumors. In many malignancies, most notably in breast cancer, ALDH activity and expression appears to be a promising marker and potential therapeutic target for treating metastasis in the clinical setting.
Collapse
Affiliation(s)
- Mauricio Rodriguez-Torres
- London Regional Cancer Program, London Health Sciences Centre, London, ON, Canada.,Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Alison L Allan
- London Regional Cancer Program, London Health Sciences Centre, London, ON, Canada. .,Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada. .,Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada. .,Lawson Health Research Institute, London, ON, Canada. .,London Regional Cancer Program, Room A4-132, 790 Commissioners Road East, London, ON, N6A 4L6, Canada.
| |
Collapse
|
27
|
Tawari PE, Wang Z, Najlah M, Tsang CW, Kannappan V, Liu P, McConville C, He B, Armesilla AL, Wang W. The cytotoxic mechanisms of disulfiram and copper(ii) in cancer cells. Toxicol Res (Camb) 2015; 4:1439-1442. [PMID: 27708770 PMCID: PMC5027600 DOI: 10.1039/c5tx00210a] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 08/30/2015] [Indexed: 12/31/2022] Open
Abstract
The anticancer activity of disulfiram (DS) is copper(ii) (Cu)-dependent.
The anticancer activity of disulfiram (DS) is copper(ii) (Cu)-dependent. This study investigated the anticancer mechanisms of DS/Cu using in vitro cytotoxicity and metabolic kinetic analysis. Our study indicates that DS/Cu targets cancer cells by the combination of two types of actions: (1) instant killing executed by DS/Cu reaction generated reactive oxygen species; (2) delayed cytotoxicity introduced by the end product, DDC-Cu. Nanoencapsulation of DS might shed light on repositioning of DS into cancer treatment.
Collapse
Affiliation(s)
- Patricia Erebi Tawari
- Research Institute in Healthcare Science , Faculty of Science & Engineering , University of Wolverhampton , Wolverhampton WV1 1LY , UK . ; ; Tel: +44 (0)1902 322756
| | - Zhipeng Wang
- Research Institute in Healthcare Science , Faculty of Science & Engineering , University of Wolverhampton , Wolverhampton WV1 1LY , UK . ; ; Tel: +44 (0)1902 322756
| | - Mohammad Najlah
- Medicine & Healthcare Science , Faculty of Medical Science , Anglia Ruskin University , UK
| | | | - Vinodh Kannappan
- Research Institute in Healthcare Science , Faculty of Science & Engineering , University of Wolverhampton , Wolverhampton WV1 1LY , UK . ; ; Tel: +44 (0)1902 322756
| | - Peng Liu
- Research Institute in Healthcare Science , Faculty of Science & Engineering , University of Wolverhampton , Wolverhampton WV1 1LY , UK . ; ; Tel: +44 (0)1902 322756
| | - Christopher McConville
- School of Pharmacy , Faculty of Science & Engineering , University of Wolverhampton , Wolverhampton WV1 1LY , UK
| | - Bin He
- National Engineering Res Centre for Biomaterials , Sichuan University , China
| | - Angel L Armesilla
- Research Institute in Healthcare Science , Faculty of Science & Engineering , University of Wolverhampton , Wolverhampton WV1 1LY , UK . ; ; Tel: +44 (0)1902 322756
| | - Weiguang Wang
- Research Institute in Healthcare Science , Faculty of Science & Engineering , University of Wolverhampton , Wolverhampton WV1 1LY , UK . ; ; Tel: +44 (0)1902 322756
| |
Collapse
|
28
|
Rivera-Valentin RK, Zhu L, Hughes DPM. Bone Sarcomas in Pediatrics: Progress in Our Understanding of Tumor Biology and Implications for Therapy. Paediatr Drugs 2015; 17:257-71. [PMID: 26002157 PMCID: PMC4516866 DOI: 10.1007/s40272-015-0134-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The pediatric bone sarcomas osteosarcoma and Ewing sarcoma represent a tremendous challenge for the clinician. Though less common than acute lymphoblastic leukemia or brain tumors, these aggressive cancers account for a disproportionate amount of the cancer morbidity and mortality in children, and have seen few advances in survival in the past decade, despite many large, complicated, and expensive trials of various chemotherapy combinations. To improve the outcomes of children with bone sarcomas, a better understanding of the biology of these cancers is needed, together with informed use of targeted therapies that exploit the unique biology of each disease. Here we summarize the current state of knowledge regarding the contribution of receptor tyrosine kinases, intracellular signaling pathways, bone biology and physiology, the immune system, and the tumor microenvironment in promoting and maintaining the malignant phenotype. These observations are coupled with a review of the therapies that target each of these mechanisms, focusing on recent or ongoing clinical trials if such information is available. It is our hope that, by better understanding the biology of osteosarcoma and Ewing sarcoma, rational combination therapies can be designed and systematically tested, leading to improved outcomes for a group of children who desperately need them.
Collapse
Affiliation(s)
- Rocio K. Rivera-Valentin
- Department of Pediatrics-Research, The Children’s Cancer Hospital at MD Anderson Cancer Center, Unit 853, MOD 1.021d, 1515 Holcombe Blvd, Houston, TX 77030 USA
| | - Limin Zhu
- Department of Pediatrics-Research, The Children’s Cancer Hospital at MD Anderson Cancer Center, Unit 853, MOD 1.021d, 1515 Holcombe Blvd, Houston, TX 77030 USA
| | - Dennis P. M. Hughes
- Department of Pediatrics-Research, The Children’s Cancer Hospital at MD Anderson Cancer Center, Unit 853, MOD 1.021d, 1515 Holcombe Blvd, Houston, TX 77030 USA
| |
Collapse
|
29
|
Alfranca A, Martinez-Cruzado L, Tornin J, Abarrategi A, Amaral T, de Alava E, Menendez P, Garcia-Castro J, Rodriguez R. Bone microenvironment signals in osteosarcoma development. Cell Mol Life Sci 2015; 72:3097-113. [PMID: 25935149 PMCID: PMC11113487 DOI: 10.1007/s00018-015-1918-y] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 04/24/2015] [Accepted: 04/27/2015] [Indexed: 02/06/2023]
Abstract
The bone is a complex connective tissue composed of many different cell types such as osteoblasts, osteoclasts, chondrocytes, mesenchymal stem/progenitor cells, hematopoietic cells and endothelial cells, among others. The interaction between them is finely balanced through the processes of bone formation and bone remodeling, which regulates the production and biological activity of many soluble factors and extracellular matrix components needed to maintain the bone homeostasis in terms of cell proliferation, differentiation and apoptosis. Osteosarcoma (OS) emerges in this complex environment as a result of poorly defined oncogenic events arising in osteogenic lineage precursors. Increasing evidence supports that similar to normal development, the bone microenvironment (BME) underlies OS initiation and progression. Here, we recapitulate the physiological processes that regulate bone homeostasis and review the current knowledge about how OS cells and BME communicate and interact, describing how these interactions affect OS cell growth, metastasis, cancer stem cell fate and therapy outcome.
Collapse
Affiliation(s)
- Arantzazu Alfranca
- Unidad de Biotecnología Celular, Área de Genética Humana, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Lucia Martinez-Cruzado
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| | - Juan Tornin
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| | - Ander Abarrategi
- Unidad de Biotecnología Celular, Área de Genética Humana, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, UK
| | - Teresa Amaral
- Molecular Pathology Program, Institute of Biomedical Research of Salamanca-Centro de Investigación del Cáncer, Centro de Investigación del Cáncer (IBSAL-CIC), Salamanca, Spain
- Department of Pathology and Biobank, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS), CSIC-Universidad de Sevilla, Seville, Spain
| | - Enrique de Alava
- Molecular Pathology Program, Institute of Biomedical Research of Salamanca-Centro de Investigación del Cáncer, Centro de Investigación del Cáncer (IBSAL-CIC), Salamanca, Spain
- Department of Pathology and Biobank, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS), CSIC-Universidad de Sevilla, Seville, Spain
| | - Pablo Menendez
- Cell Therapy Program, School of Medicine, Josep Carreras Leukemia Research Institute, University of Barcelona, Barcelona, Spain
- Instituciò Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| | - Javier Garcia-Castro
- Unidad de Biotecnología Celular, Área de Genética Humana, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Rene Rodriguez
- Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, 33006 Oviedo, Spain
| |
Collapse
|
30
|
Wang B, Su Y, Yang Q, Lv D, Zhang W, Tang K, Wang H, Zhang R, Liu Y. Overexpression of Long Non-Coding RNA HOTAIR Promotes Tumor Growth and Metastasis in Human Osteosarcoma. Mol Cells 2015; 38:432-40. [PMID: 25728753 PMCID: PMC4443285 DOI: 10.14348/molcells.2015.2327] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 01/05/2015] [Accepted: 01/26/2015] [Indexed: 01/13/2023] Open
Abstract
Human osteosarcoma usually presented a high tendency to metastatic spread and caused poor outcomes, however, the underlying mechanism was still largely unknown. In the present study, using a series of in vitro experiments and an animal model, we investigated the roles of HOX antisense intergenic RNA (HOTAIR) during the proliferation and invasion of osteosarcoma. According with our results, HOTAIR was commonly overexpressed in osteosarcoma, which significantly correlated with advanced tumor stage, highly histological grade and poor prognosis. In vitro and in vivo experiments demonstrated that knockdown of HOTAIR could notably suppress cellular proliferation, inhibit invasion and decrease the secretion of MMP2 and MMP9 in osteosarcoma. Collectively, our results suggested that HOTAIR might be a potent therapeutic target for osteosarcoma.
Collapse
Affiliation(s)
- Bo Wang
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| | - Yun Su
- Department of Orthopeadics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning Province, 116001,
China
| | - Qun Yang
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| | - Decheng Lv
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| | - Weiguo Zhang
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| | - Kai Tang
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| | - Hong Wang
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| | - Rui Zhang
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| | - Yang Liu
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| |
Collapse
|
31
|
Pantziarka P, Sukhatme V, Bouche G, Meheus L, Sukhatme VP. Repurposing Drugs in Oncology (ReDO)-itraconazole as an anti-cancer agent. Ecancermedicalscience 2015; 9:521. [PMID: 25932045 PMCID: PMC4406527 DOI: 10.3332/ecancer.2015.521] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Indexed: 12/12/2022] Open
Abstract
Itraconazole, a common triazole anti-fungal drug in widespread clinical use, has evidence of clinical activity that is of interest in oncology. There is evidence that at the clinically relevant doses, itraconazole has potent anti-angiogenic activity, and that it can inhibit the Hedgehog signalling pathway and may also induce autophagic growth arrest. The evidence for these anticancer effects, in vitro, in vivo, and clinical are summarised, and the putative mechanisms of their action outlined. Clinical trials have shown that patients with prostate, lung, and basal cell carcinoma have benefited from treatment with itraconazole, and there are additional reports of activity in leukaemia, ovarian, breast, and pancreatic cancers. Given the evidence presented, a case is made that itraconazole warrants further clinical investigation as an anti- cancer agent. Additionally, based on the properties summarised previously, it is proposed that itraconazole may synergise with a range of other drugs to enhance the anti-cancer effect, and some of these possible combinations are presented in the supplementary materials accompanying this paper.
Collapse
Affiliation(s)
- Pan Pantziarka
- Anticancer Fund, 1853 Strombeek-Bever, Belgium ; The George Pantziarka TP53 Trust, London, KT1 2JP, UK
| | | | | | | | - Vikas P Sukhatme
- GlobalCures, Inc; Newton MA 02459, USA ; Beth Israel Deaconess Medical Centre and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
32
|
Hairy/enhancer-of-split related with YRPW motif protein 1 promotes osteosarcoma metastasis via matrix metallopeptidase 9 expression. Br J Cancer 2015; 112:1232-40. [PMID: 25742474 PMCID: PMC4385965 DOI: 10.1038/bjc.2015.84] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/23/2015] [Accepted: 02/04/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Activation of the Notch pathway has been reported in various types of cancers. However, the role of the hairy/enhancer-of-split related with YRPW motif protein 1 (HEY1) in osteosarcoma is unknown. We examined the function of HEY1 in osteosarcoma. METHODS Expression of HEY1 was studied in human osteosarcoma. The effects of HEY1 in osteosarcoma were evaluated in vitro and in a xenograft model. Moreover, we examined the function of matrix metallopeptidase 9 (MMP9) as a downstream effector of HEY1. RESULTS HEY1 was upregulated in human osteosarcoma. Knockdown of HEY1 inhibited the invasion of osteosarcoma cell lines. In contrast, the forced expression of HEY1 increased the invasion of mesenchymal stem cell. In addition, lung metastases were significantly inhibited by the knockdown of HEY1. We found that MMP9 was a downstream effector of HEY1 that promotes the invasion of osteosarcoma cells. Knockdown of HEY1 decreased the expression of MMP9. Addition of MMP9 rescued the invasion of osteosarcoma cells that had been rendered less invasive by knockdown of HEY1 expression. CONCLUSIONS Our findings suggested that HEY1 augmented the metastasis of osteosarcoma via upregulation of MMP9 expression. Therefore, inhibition of HEY1 may be a novel therapeutic strategy for preventing osteosarcoma metastasis.
Collapse
|
33
|
Feng Y, Sassi S, Shen JK, Yang X, Gao Y, Osaka E, Zhang J, Yang S, Yang C, Mankin HJ, Hornicek FJ, Duan Z. Targeting CDK11 in osteosarcoma cells using the CRISPR-Cas9 system. J Orthop Res 2015; 33:199-207. [PMID: 25348612 PMCID: PMC4304907 DOI: 10.1002/jor.22745] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 09/02/2014] [Indexed: 02/04/2023]
Abstract
Osteosarcoma is the most common type primary malignant tumor of bone. Patients with regional osteosarcoma are routinely treated with surgery and chemotherapy. In addition, many patients with metastatic or recurrent osteosarcoma show poor prognosis with current chemotherapy agents. Therefore, it is important to improve the general condition and the overall survival rate of patients with osteosarcoma by identifying novel therapeutic strategies. Recent studies have revealed that CDK11 is essential in osteosarcoma cell growth and survival by inhibiting CDK11 mRNA expression with RNAi. Here, we apply the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas9 system, a robust and highly efficient novel genome editing tool, to determine the effect of targeting endogenous CDK11 gene at the DNA level in osteosarcoma cell lines. We show that CDK11 can be efficiently silenced by CRISPR-Cas9. Inhibition of CDK11 is associated with decreased cell proliferation and viability, and induces cell death in osteosarcoma cell lines KHOS and U-2OS. Furthermore, the migration and invasion activities are also markedly reduced by CDK11 knockout. These results demonstrate that CRISPR-Cas9 system is a useful tool for the modification of endogenous CDK11 gene expression, and CRISPR-Cas9 targeted CDK11 knockout may be a promising therapeutic regimen for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Yong Feng
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, Massachusetts 02114,Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jie Fang Avenue, Wuhan, China, 430022
| | - Slim Sassi
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Jacson K Shen
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, Massachusetts 02114
| | - Xiaoqian Yang
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, Massachusetts 02114
| | - Yan Gao
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, Massachusetts 02114
| | - Eiji Osaka
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, Massachusetts 02114
| | - Jianming Zhang
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School Boston, MA, 02114
| | - Shuhua Yang
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jie Fang Avenue, Wuhan, China, 430022
| | - Cao Yang
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jie Fang Avenue, Wuhan, China, 430022
| | - Henry J. Mankin
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, Massachusetts 02114
| | - Francis J Hornicek
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, Massachusetts 02114
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Jackson 1115, Boston, Massachusetts 02114
| |
Collapse
|
34
|
Dastjerdi MN, Babazadeh Z, Salehi M, Hashemibeni B, Kazemi M. Comparison of the anti-cancer effect of Disulfiram and 5-Aza-CdR on pancreatic cancer cell line PANC-1. Adv Biomed Res 2014; 3:156. [PMID: 25221759 PMCID: PMC4162084 DOI: 10.4103/2277-9175.137866] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 02/02/2014] [Indexed: 01/31/2023] Open
Abstract
Background: Pancreatic cancer has poor prognosis by surgical and chemotherapy when it is diagnosed, so other anti-cancerous assistant therapeutic drugs are suggested e.g. epigenetic reversal of tumor-suppressor genes on promoter hypermethylation. 5-Aza-CdR is a nucleoside analog of DNMTi but it has long-term cytotoxicity effects. This study compares the anticancer effect of 5-Aza-CdR and Disulfiram potencies on PANC-1 cell line and up-regulation of p21. Materials and Methods: PANC-1 cell line was cultured in DMEM high glucose and treated by 5-Aza-CdR with 5 and 10 μM concentration for four days and 13 μM DSF (Diulfiram) for 24 hours. MS-PCR and RT-PCR were carried out to detect the methylation pattern and estimate the mRNA expression of RASSF1A and p21 in PANC-1. Result: MS-PCR demonstrated partial unmethylation after treatment with 5-Aza-CdR while there was no unmethylated band after DSF treatment. RT-PCR showed significant differences between re-expression of RASSF1A before and after treatment with 10 μM 5-Aza-CdR (P < 0.01) but not after treatment with 13 μM DSF (P > 0.05). The significant correlation was observed between RASSF1A re-expression and p21 up-regulation before and after treatment with 10 μM 5-Aza-CdR (P < 0.01) but not after treatment with 13 μM DSF (P > 0.05), while p21 up-regulation was significantly higher after DSF treatment (P < 0.01). Conclusion: Our findings indicated that 5-Aza-CdR induces the re-expression of RASSF1A and p21 up-regulation in PANC-1. DSF showed no epigenetic reversion while it affected p21 up-regulation.
Collapse
Affiliation(s)
- Mehdi Nikbakht Dastjerdi
- Department of Anatomical Sciences and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Babazadeh
- Department of Anatomical Sciences and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mansour Salehi
- Department of Anatomical Sciences and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Batool Hashemibeni
- Department of Anatomical Sciences and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Kazemi
- Department of Anatomical Sciences and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
35
|
Fouriki A, Dobson J. Oscillating magnet array-based nanomagnetic gene transfection of human mesenchymal stem cells. Nanomedicine (Lond) 2014; 9:989-97. [DOI: 10.2217/nnm.13.74] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Aim: In this work, the potential of nanomagnetic transfection of primary human mesenchymal stem cells (hMSCs) and the effects of a novel nonviral oscillating magnet array system in enhancing transfection efficiency were investigated. Materials & methods: Green fluorescent protein plasmids coupled to magnetic nanoparticles (MNPs) were introduced onto hMSCs in culture. Magnetic fields generated by arrays of neodymium iron boron magnets positioned below the culture plates direct the MNP/DNA complexes into contact with the cells. The magnet arrays were oscillated, promoting more efficient endocytosis via mechanical stimulation. Green fluorescent protein expression, cell viability and stem cell surface markers were assayed. Results: MNP/DNA complexes were delivered into hMSCs, and the oscillating magnet array system appears to improve transfection efficiency as well as cell viability. The expression of hMSC-specific cell surface markers was unaffected. Conclusion: Nonviral transfection using MNPs and oscillating magnet arrays offers a more efficient and ‘cell-friendly’ method of transfecting hMSCs than other nonviral techniques, while preserving their stem cell characteristics. Original submitted 8 March 2012; Revised submitted 12 February 2013
Collapse
Affiliation(s)
- Angeliki Fouriki
- Institute for Science & Technology in Medicine, Keele University, Thornburrow Drive, Hartshill, Stoke-on-Trent, ST4 7QB, UK
| | - Jon Dobson
- Institute for Science & Technology in Medicine, Keele University, Thornburrow Drive, Hartshill, Stoke-on-Trent, ST4 7QB, UK
- J Crayton Pruitt Family Department of Biomedical Engineering, Department of Materials Science & Engineering, & the Institute for Cell Engineering & Regenerative Medicine University of Florida, PO Box 116131, Gainesville, FL 32611, USA
| |
Collapse
|
36
|
Duan X, Xiao J, Yin Q, Zhang Z, Yu H, Mao S, Li Y. Multi-targeted inhibition of tumor growth and lung metastasis by redox-sensitive shell crosslinked micelles loading disulfiram. NANOTECHNOLOGY 2014; 25:125102. [PMID: 24576956 DOI: 10.1088/0957-4484/25/12/125102] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Metastasis, the main cause of cancer related deaths, remains the greatest challenge in cancer treatment. Disulfiram (DSF), which has multi-targeted anti-tumor activity, was encapsulated into redox-sensitive shell crosslinked micelles to achieve intracellular targeted delivery and finally inhibit tumor growth and metastasis. The crosslinked micelles demonstrated good stability in circulation and specifically released DSF under a reductive environment that mimicked the intracellular conditions of tumor cells. As a result, the DSF-loaded redox-sensitive shell crosslinked micelles (DCMs) dramatically inhibited cell proliferation, induced cell apoptosis and suppressed cell invasion, as well as impairing tube formation of HMEC-1 cells. In addition, the DCMs could accumulate in tumor tissue and stay there for a long time, thereby causing significant inhibition of 4T1 tumor growth and marked prevention in lung metastasis of 4T1 tumors. These results suggested that DCMs could be a promising delivery system in inhibiting the growth and metastasis of breast cancer.
Collapse
Affiliation(s)
- Xiaopin Duan
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China. School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
37
|
Berezin V, Walmod PS, Filippov M, Dityatev A. Targeting of ECM molecules and their metabolizing enzymes and receptors for the treatment of CNS diseases. PROGRESS IN BRAIN RESEARCH 2014; 214:353-88. [DOI: 10.1016/b978-0-444-63486-3.00015-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
38
|
Poudel B, Kim DOK, Ki HH, Kwon YB, Lee YM, Kim DK. Downregulation of ERK signaling impairs U2OS osteosarcoma cell migration in collagen matrix by suppressing MMP9 production. Oncol Lett 2013; 7:215-218. [PMID: 24348851 PMCID: PMC3861606 DOI: 10.3892/ol.2013.1655] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 10/25/2013] [Indexed: 01/16/2023] Open
Abstract
The present study investigated the role of extracellular signal-regulated kinase (ERK) activation in the migratory phenotype of human U2OS osteosarcoma (OS) cells in a collagen matrix. The activation of ERK was inhibited by PD98059, a specific inhibitor of ERK kinase. Additionally, no significant differences were observed in the adhesion and proliferation of the cells with or without PD98059 treatment in collagen-coated dishes. The migratory capacity of the U2OS cells was then examined in non-coated and collagen-coated dishes, and the results depicted that collagen I enhanced the migration of the U2OS cells, the effect of which was significantly blocked by the treatment of the cells with PD98059. Furthermore, enhanced gene and protein expression of matrix metalloproteinase 9 (MMP9), but not MMP2, was observed to be involved in the enhanced migratory phenotype of the U20S cells in the collagen-coated plates. This effect was partially abolished by the treatment of the cells in the collagen-coated dishes with ERK inhibitor. Collectively, the data demonstrate that ERK signaling is important for the migration of U2OS cells through the extracellular matrix (ECM), which is comprised mostly of collagen, by enhancing MMP9 production. These results may contribute to the regulation of MMP9 production in metastatic OS.
Collapse
Affiliation(s)
- Barun Poudel
- Department of Immunology and Institute of Medical Sciences, Chonbuk National University Medical School, Jeonju, Jeonbuk 561-756, Republic of Korea
| | - DO-Kuk Kim
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan 570-749, Republic of Korea
| | - Hyeon-Hui Ki
- Department of Immunology and Institute of Medical Sciences, Chonbuk National University Medical School, Jeonju, Jeonbuk 561-756, Republic of Korea
| | - Young-Bae Kwon
- Department of Pharmacology, Chonbuk National University Medical School, Jeonju 561-756, Republic of Korea
| | - Young-Mi Lee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan 570-749, Republic of Korea
| | - Dae-Ki Kim
- Department of Immunology and Institute of Medical Sciences, Chonbuk National University Medical School, Jeonju, Jeonbuk 561-756, Republic of Korea
| |
Collapse
|
39
|
Reversing the intractable nature of pancreatic cancer by selectively targeting ALDH-high, therapy-resistant cancer cells. PLoS One 2013; 8:e78130. [PMID: 24194908 PMCID: PMC3806801 DOI: 10.1371/journal.pone.0078130] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/17/2013] [Indexed: 12/19/2022] Open
Abstract
Human pancreatic ductal adenocarcinoma (PDAC) is a cancer with a dismal prognosis. The efficacy of PDAC anticancer therapies is often short-lived; however, there is little information on how this disease entity so frequently gains resistance to treatment. We adopted the concept of cancer stem cells (CSCs) to explain the mechanism of resistance and evaluated the efficacy of a candidate anticancer drug to target these therapy-resistant CSCs. We identified a subpopulation of cells in PDAC with CSC features that were enriched for aldehyde dehydrogenase (ALDH), a marker expressed in certain stem/progenitor cells. These cells were also highly resistant to, and were further enriched by, treatment with gemcitabine. Similarly, surgical specimens from PDAC patients showed that those who had undergone preoperative chemo-radiation therapy more frequently displayed cancers with ALDH strongly positive subpopulations compared with untreated patients. Importantly, these ALDH-high cancer cells were sensitive to disulfiram, an ALDH inhibitor, when tested in vitro. Furthermore, in vivo xenograft studies showed that the effect of disulfiram was additive to that of low-dose gemcitabine when applied in combination. In conclusion, human PDAC-derived cells that express high levels of ALDH show CSC features and have a key role in the development of resistance to anticancer therapies. Disulfiram can be used to suppress this therapy-resistant subpopulation.
Collapse
|
40
|
Zhu L, McManus MM, Hughes DPM. Understanding the Biology of Bone Sarcoma from Early Initiating Events through Late Events in Metastasis and Disease Progression. Front Oncol 2013; 3:230. [PMID: 24062983 PMCID: PMC3775316 DOI: 10.3389/fonc.2013.00230] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 08/21/2013] [Indexed: 12/27/2022] Open
Abstract
The two most common primary bone malignancies, osteosarcoma (OS), and Ewing sarcoma (ES), are both aggressive, highly metastatic cancers that most often strike teens, though both can be found in younger children and adults. Despite distinct origins and pathogenesis, both diseases share several mechanisms of progression and metastasis, including neovascularization, invasion, anoikis resistance, chemoresistance, and evasion of the immune response. Some of these processes are well-studies in more common carcinoma models, and the observation from adult diseases may be readily applied to pediatric bone sarcomas. Neovascularization, which includes angiogenesis and vasculogenesis, is a clear example of a process that is likely to be similar between carcinomas and sarcomas, since the responding cells are the same in each case. Chemoresistance mechanisms also may be similar between other cancers and the bone sarcomas. Since OS and ES are mesenchymal in origin, the process of epithelial-to-mesenchymal transition is largely absent in bone sarcomas, necessitating different approaches to study progression and metastasis in these diseases. One process that is less well-studied in bone sarcomas is dormancy, which allows micrometastatic disease to remain viable but not growing in distant sites – typically the lungs – for months or years before renewing growth to become overt metastatic disease. By understanding the basic biology of these processes, novel therapeutic strategies may be developed that could improve survival in children with OS or ES.
Collapse
Affiliation(s)
- Limin Zhu
- Department of Pediatrics - Research, UT MD Anderson Cancer Center , Houston, TX , USA
| | | | | |
Collapse
|
41
|
Pharmacodynamic study of disulfiram in men with non-metastatic recurrent prostate cancer. Prostate Cancer Prostatic Dis 2013; 16:357-61. [PMID: 23958896 PMCID: PMC3830644 DOI: 10.1038/pcan.2013.28] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 07/01/2013] [Accepted: 07/21/2013] [Indexed: 01/15/2023]
Abstract
BACKGROUND Preclinical drug screens identified disulfiram as a potent in vitro inhibitor of prostate cancer (PCa) cell growth. Although many mechanisms for its anticancer activity have been proposed, tumor suppressor gene re-expression through promoter demethylation emerged as one of the more plausible. METHODS We conducted an open-label, dose escalation trial of disulfiram in men with non-metastatic recurrent PCa after local therapy. Dose escalation occurred if a demethylating 'response' (that is, 10% decrease in peripheral blood mononuclear cell (PBMC) global 5-methyl cytosine (5(me)C) content) was observed in <3 patients in cohort 1. Cohorts 1 and 2 received disulfiram 250 mg and 500 mg daily, respectively. The primary end point was the proportion of subjects with a demethylation response. Secondary end points included the rate of PSA progression at 6 months, changes in PSA doubling time and safety/tolerability. RESULTS Changes in global 5(me)C content were observed in two of nine patients (22.2%) in cohort 1 and 3 of 10 (30.0%) in cohort 2. Only five subjects were on trial for 6 months, all were in cohort 1 and all had PSA progression by 6 months. No changes in PSA kinetics were observed in either cohort. Disulfiram was poorly tolerated with six patients experiencing grade 3 adverse events (three per cohort). Three of the responders displayed pretreatment instability in their 5(me)C content. CONCLUSIONS A minority of patients had transient global PBMC demethylation changes. Instability in 5(me)C may limit the reproducibility of these findings, limiting our ability to confirm our hypothesis. Given the toxicities and no clinical benefits, further development of disulfiram should not be pursued in this population.
Collapse
|
42
|
Lv Z, Yang D, Li J, Hu M, Luo M, Zhan X, Song P, Liu C, Bai H, Li B, Yang Y, Chen Y, Shi Q, Weng Y. Bone morphogenetic protein 9 overexpression reduces osteosarcoma cell migration and invasion. Mol Cells 2013; 36:119-26. [PMID: 23807047 PMCID: PMC3887952 DOI: 10.1007/s10059-013-0043-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/08/2013] [Accepted: 05/24/2013] [Indexed: 01/24/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is known to promote tumor migration and invasion. Bone morphogenetic proteins (BMPs) are members of the TGF-β family expressed in a variety of human carcinoma cell lines. The role of bone morphogenetic protein 9 (BMP9), the most powerful osteogenic factor, in osteosarcoma (OS) progression has not been fully clarified. The expression of BMP9 and its receptors in OS cell lines was analyzed by RT-PCR. We found that BMP9 and its receptors were expressed in OS cell lines. We further investigated the influence of BMP9 on the biological behaviors of OS cells. BMP9 overexpression in the OS cell lines 143B and MG63 inhibited in vitro cell migration and invasion. We further investigated the expression of a panel of cancer-related genes and found that BMP9 overexpression increased the phosphorylation of Smad1/5/8 proteins, increased the expression of ID1, and reduced the expression and activity of matrix metalloproteinase 9 (MMP9) in OS cells. BMP9 silencing induced the opposite effects. We also found that BMP9 may not affect the chemokine (C-X-C motif) ligand 12 (CXCL12)/C-X-C chemokine receptor type 4 (CXCR4) axis to regulate the invasiveness and metastatic capacity of OS cells. Interestingly, CXCR4 was expressed in both 143B and MG63 cells, while CXCL12 was only detected in MG63 cells. Taken together, we hypothesize that BMP9 inhibits the migration and invasiveness of OS cells through a Smad-dependent pathway by downregulating the expression and activity of MMP9.
Collapse
Affiliation(s)
- Zilan Lv
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Dandan Yang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Jie Li
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Min Hu
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Min Luo
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Xiaoqin Zhan
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Peipei Song
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Chen Liu
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Huili Bai
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Baolin Li
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Yang Yang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Yingying Chen
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Qiong Shi
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Yaguang Weng
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| |
Collapse
|
43
|
Oreščanin-Dusić Z, Milovanović S, Blagojević D, Nikolić-Kokić A, Radojičić R, Spasojević I, Spasić M. Diethyldithiocarbamate potentiates the effects of protamine sulphate in the isolated rat uterus. Redox Rep 2013; 14:48-54. [DOI: 10.1179/135100009x392476] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
44
|
Mu X, Isaac C, Greco N, Huard J, Weiss K. Notch Signaling is Associated with ALDH Activity and an Aggressive Metastatic Phenotype in Murine Osteosarcoma Cells. Front Oncol 2013; 3:143. [PMID: 23805413 PMCID: PMC3678113 DOI: 10.3389/fonc.2013.00143] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 05/20/2013] [Indexed: 12/13/2022] Open
Abstract
Osteosarcoma (OS) is the most common primary malignancy of bone, and pulmonary metastatic disease accounts for nearly all mortality. However, little is known about the biochemical signaling alterations that drive the progression of metastatic disease. Two murine OS cell populations, K7M2 and K12, are clonally related but differ significantly in their metastatic phenotypes and therefore represent excellent tools for studying metastatic OS molecular biology. K7M2 cells are highly metastatic, whereas K12 cells display limited metastatic potential. Here we report that the expression of Notch genes (Notch1, 2, 4) are up-regulated, including downstream targets Hes1 and Stat3, in the highly metastatic K7M2 cells compared to the less metastatic K12 cells, indicating that the Notch signaling pathway is more active in K7M2 cells. We have previously described that K7M2 cells exhibit higher levels of aldehyde dehydrogenase (ALDH) activity. Here we report that K7M2 cell ALDH activity is reduced with Notch inhibition, suggesting that ALDH activity may be regulated in part by the Notch pathway. Notch signaling is also associated with increased resistance to oxidative stress, migration, invasion, and VEGF expression in vitro. However, Notch inhibition did not significantly alter K7M2 cell proliferation. In conclusion, we provide evidence that Notch signaling is associated with ALDH activity and increased metastatic behavior in OS cells. Both Notch and ALDH are putative molecular targets for the treatment and prevention of OS metastasis.
Collapse
Affiliation(s)
- Xiaodong Mu
- Stem Cell Research Laboratory, University of Pittsburgh Medical Center , Pittsburgh, PA , USA
| | | | | | | | | |
Collapse
|
45
|
Kast RE, Boockvar JA, Brüning A, Cappello F, Chang WW, Cvek B, Dou QP, Duenas-Gonzalez A, Efferth T, Focosi D, Ghaffari SH, Karpel-Massler G, Ketola K, Khoshnevisan A, Keizman D, Magné N, Marosi C, McDonald K, Muñoz M, Paranjpe A, Pourgholami MH, Sardi I, Sella A, Srivenugopal KS, Tuccori M, Wang W, Wirtz CR, Halatsch ME. A conceptually new treatment approach for relapsed glioblastoma: coordinated undermining of survival paths with nine repurposed drugs (CUSP9) by the International Initiative for Accelerated Improvement of Glioblastoma Care. Oncotarget 2013; 4:502-30. [PMID: 23594434 PMCID: PMC3720600 DOI: 10.18632/oncotarget.969] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 04/11/2013] [Indexed: 02/06/2023] Open
Abstract
To improve prognosis in recurrent glioblastoma we developed a treatment protocol based on a combination of drugs not traditionally thought of as cytotoxic chemotherapy agents but that have a robust history of being well-tolerated and are already marketed and used for other non-cancer indications. Focus was on adding drugs which met these criteria: a) were pharmacologically well characterized, b) had low likelihood of adding to patient side effect burden, c) had evidence for interfering with a recognized, well-characterized growth promoting element of glioblastoma, and d) were coordinated, as an ensemble had reasonable likelihood of concerted activity against key biological features of glioblastoma growth. We found nine drugs meeting these criteria and propose adding them to continuous low dose temozolomide, a currently accepted treatment for relapsed glioblastoma, in patients with recurrent disease after primary treatment with the Stupp Protocol. The nine adjuvant drug regimen, Coordinated Undermining of Survival Paths, CUSP9, then are aprepitant, artesunate, auranofin, captopril, copper gluconate, disulfiram, ketoconazole, nelfinavir, sertraline, to be added to continuous low dose temozolomide. We discuss each drug in turn and the specific rationale for use- how each drug is expected to retard glioblastoma growth and undermine glioblastoma's compensatory mechanisms engaged during temozolomide treatment. The risks of pharmacological interactions and why we believe this drug mix will increase both quality of life and overall survival are reviewed.
Collapse
Affiliation(s)
| | | | | | | | - Wen-Wei Chang
- Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Boris Cvek
- Palacky University, Olomouc, Czech Republic
| | | | - Alfonso Duenas-Gonzalez
- Instituto de Investigaciones Biomedicas UNAM, Instituto Nacional de Cancerología, Mexico City, Mexico
| | | | | | | | | | - Kirsi Ketola
- University of British Columbia, Vancouver, Canada
| | | | - Daniel Keizman
- Oncology Department, Meir Medical Center, Tel Aviv University, Israel
| | - Nicolas Magné
- Institut de Cancérologie Lucien Neuwirth, Saint-Priest en Jarez, France
| | | | | | - Miguel Muñoz
- Virgen del Rocío University Hospital, Sevilla, Spain
| | - Ameya Paranjpe
- Texas Tech University Health Sciences Center, Amarillo, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Effects of baicalein on apoptosis, cell cycle arrest, migration and invasion of osteosarcoma cells. Food Chem Toxicol 2013; 53:325-33. [DOI: 10.1016/j.fct.2012.12.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 11/28/2012] [Accepted: 12/12/2012] [Indexed: 11/20/2022]
|
47
|
Rapamycin Inhibits ALDH Activity, Resistance to Oxidative Stress, and Metastatic Potential in Murine Osteosarcoma Cells. Sarcoma 2013; 2013:480713. [PMID: 23476113 PMCID: PMC3586506 DOI: 10.1155/2013/480713] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 12/04/2012] [Accepted: 12/22/2012] [Indexed: 01/08/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary malignancy of bone. Mortality is determined by the presence of metastatic disease, but little is known regarding the biochemical events that drive metastases. Two murine OS cell lines, K7M2 and K12, are related but differ significantly in their metastatic potentials: K7M2 is highly metastatic whereas K12 displays much less metastatic potential. Using this experimental system, the mammalian target of rapamycin (mTOR) pathway has been implicated in OS metastasis. We also discovered that aldehyde dehydrogenase (ALDH, a stem cell marker) activity is higher in K7M2 cells than K12 cells. Rapamycin treatment reduces the expression and enzymatic activity of ALDH in K7M2 cells. ALDH inhibition renders these cells more susceptible to apoptotic death when exposed to oxidative stress. Furthermore, rapamycin treatment reduces bone morphogenetic protein-2 (BMP2) and vascular endothelial growth factor (VEGF) gene expression and inhibits K7M2 proliferation, migration, and invasion in vitro. Inhibition of ALDH with disulfiram correlated with decreased mTOR expression and activity. In conclusion, we provide evidence for interaction between mTOR activity, ALDH activity, and metastatic potential in murine OS cells. Our work suggests that mTOR and ALDH are therapeutic targets for the treatment and prevention of OS metastasis.
Collapse
|
48
|
Nanomagnetic Gene Transfection for Non-Viral Gene Delivery in NIH 3T3 Mouse Embryonic Fibroblasts. MATERIALS 2013; 6:255-264. [PMID: 28809306 PMCID: PMC5452119 DOI: 10.3390/ma6010255] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 01/08/2013] [Accepted: 01/11/2013] [Indexed: 11/17/2022]
Abstract
The objective of this work was to examine the potential of oscillating nanomagnetic gene transfection systems (magnefect-nano™) for improving the transfection efficiency of NIH3T3 mouse embryonic fibroblasts (MEFs) in comparison to other non-viral transfection techniques-static magnetofection™ and the cationic lipid agent, Lipofectamine 2000™. Magnetic nanoparticles (MNPs) associated with the plasmid coding for green fluorescent protein (GFP) were used to transfect NIH3T3 cells. The magnefect-nano system was evaluated for transfection efficiency, and any potential associated effects on cell viability were investigated. MNPs associated with the plasmid coding for GFP were efficiently delivered into NIH3T3 cells, and the magnefect-nano system significantly enhanced overall transfection efficiency in comparison to lipid-mediated gene delivery. MNP dosage used in this work was not found to affect the cell viability and/or morphology of the cells. Non-viral transfection using MNPs and the magnefect-nano system can be used to transfect NIH3T3 cells and direct reporter gene delivery, highlighting the wide potential of nanomagnetic gene transfection in gene therapy.
Collapse
|
49
|
Srikoon P, Kariya R, Kudo E, Goto H, Vaeteewoottacharn K, Taura M, Wongkham S, Okada S. Diethyldithiocarbamate suppresses an NF-kappaB dependent metastatic pathway in cholangiocarcinoma cells. Asian Pac J Cancer Prev 2013; 14:4441-4446. [PMID: 23992017 DOI: 10.7314/apjcp.2013.14.7.4441] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a tumor of biliary ducts, which has a high mortality rate and dismal prognosis. Constitutively activation of the transcription factor nuclear factor kappa-B (NF-κB) has been previously demonstrated in CCA. It is therefore a potential target for CCA treatment. Effects of diethyldithiocarbamate (DDTC) on NF-κB-dependent apoptosis induction in cancer have been reported; however, anti-metastasis has never been addressed. Therefore, here the focus was on DDTC effects on CCA migration and adhesion. Anti-proliferation, anti-migration and anti-adhesion activities were determined in CCA cell lines, along with p65 protein levels and function. NF-κB target gene expression was determined by quantitative RT-PCR. DDTC inhibited CCA cell proliferation. Suppression of migration and adhesion were observed prior to anti-CCA proliferation. These effects were related to decreased p65, reduction in NF-κB DNA binding, and impaired activity. Moreover, suppression of ICAM-1 expression supported NF-kB-dependent anti-metastatic effects of DDTC. Taken together, DDTC suppression of CCA migration and adhesion through inhibition of NF-κB signaling pathway is suggested from the current study. This might be a promising treatment choice against CCA metastasis.
Collapse
Affiliation(s)
- Pattaravadee Srikoon
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Li Y, Zhang J, Ma D, Zhang L, Si M, Yin H, Li J. Curcumin inhibits proliferation and invasion of osteosarcoma cells through inactivation of Notch-1 signaling. FEBS J 2012; 279:2247-59. [PMID: 22521131 DOI: 10.1111/j.1742-4658.2012.08607.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The Notch signaling pathway plays critical roles in human cancers, including osteosarcoma, suggesting that the discovery of specific agents targeting Notch would be extremely valuable for osteosarcoma. Curcumin, a naturally occurring phenolic compound found in curcuma longa, has been shown to inhibit proliferation and induce apoptosis of osteosarcoma cells in vitro and tumor growth in xenotransplant or orthotransplant models. However, the precise molecular mechanisms by which curcumin exerts its antitumor activity remain unclear. Here we used multiple molecular approaches, such as the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, the invasion assay, gene transfection, real-time RT-PCR, western blot and gelatin zymography, to investigate whether the downregulation of Notch-1 contributes to curcumin-induced inhibition of proliferation and invasion in osteosarcoma cells. The results showed that curcumin caused marked inhibition of osteosarcoma cell growth and G2/M phase cell cycle arrest. This was associated with concomitant attenuation of Notch-1 and downregulation of its downstream genes, such as matrix metalloproteinases, resulting in the inhibition of osteosarcoma cell invasion through Matrigel. We also found that specific downregulation of Notch-1 via small-interfering RNA prior to curcumin treatment resulted in enhanced inhibition of cell growth and invasion. These results suggest that antitumor activity of curcumin is mediated through a novel mechanism involving inactivation of the Notch-1 signaling pathway. Our data provide the first evidence that the downregulation of Notch-1 by curcumin may be an effective approach for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Yonggang Li
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan, China
| | | | | | | | | | | | | |
Collapse
|