1
|
Zhu L, Shen Y, Deng S, Wan Y, Luo J, Su Y, You M, Fan C, Ren K. Controllable mitochondrial aggregation and fusion by a programmable DNA binder. Chem Sci 2023; 14:8084-8094. [PMID: 37538820 PMCID: PMC10395312 DOI: 10.1039/d2sc07095b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 07/05/2023] [Indexed: 08/05/2023] Open
Abstract
DNA nanodevices have been feasibly applied for various chemo-biological applications, but their functions as precise regulators of intracellular organelles are still limited. Here, we report a synthetic DNA binder that can artificially induce mitochondrial aggregation and fusion in living cells. The rationally designed DNA binder consists of a long DNA chain, which is grafted with multiple mitochondria-targeting modules. Our results indicated that the DNA binder-induced in situ self-assembly of mitochondria can be used to successfully repair ROS-stressed neuron cells. Meanwhile, this DNA binder design is highly programmable. Customized molecular switches can be easily implanted to further achieve stimuli-triggered mitochondrial aggregation and fusion inside living cells. We believe this new type of DNA regulator system will become a powerful chemo-biological tool for subcellular manipulation and precision therapy.
Collapse
Affiliation(s)
- Longyi Zhu
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology Nanjing 210094 China
| | - Yiting Shen
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology Nanjing 210094 China
| | - Shengyuan Deng
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology Nanjing 210094 China
| | - Ying Wan
- Intelligent Microsystem Technology and Engineering Center, School of Mechanical Engineering, Nanjing University of Science and Technology Nanjing 210094 China
| | - Jun Luo
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology Nanjing 210094 China
| | - Yan Su
- Intelligent Microsystem Technology and Engineering Center, School of Mechanical Engineering, Nanjing University of Science and Technology Nanjing 210094 China
| | - Mingxu You
- Department of Chemistry, University of Massachusetts Amherst MA 01003 USA
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University Shanghai 200127 China
| | - Kewei Ren
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology Nanjing 210094 China
| |
Collapse
|
2
|
Wang Z, Ma Y, Jiang Y, Zhou F, Wu Y, Jiang H, Wang R, Xu Q, Hua C. Encapsulating quercetin in cyclodextrin metal-organic frameworks improved its solubility and bioavailability. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:3887-3896. [PMID: 34952968 DOI: 10.1002/jsfa.11738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/30/2021] [Accepted: 12/24/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Quercetin (Que) has many pharmacological activities, such as anticancer, antioxidant, cardiovascular protection, antihypertensive and lipid-lowering activities. However, its poor water solubility greatly limits its application in medicine and food. γ-Cyclodextrin metal-organic frameworks (γ-CD-MOFs) are novel porous carriers for loading functional products. In this study, Que was successfully loaded into γ-CD-MOFs, and the new compound (Que-CD-MOFs) was characterised by X-ray diffraction, infrared spectroscopy, thermogravimetric analysis and scanning electron microscopy. RESULTS The apparent solubility of Que-CD-MOFs was enhanced by 100-fold compared with that of pure Que. The free radical scavenging ability of the encapsulated Que was significantly improved. The cytotoxicity of Que-CD-MOFs to HK-2 cells was decreased, and their inhibition on HT-29 tumour cells was maintained, as confirmed by CCK-8 assays. Flow cytometry of HT-29 cells showed that Que-CD-MOFs can inhibit G2 phase cells. Based on molecular modelling, Que molecules were preferentially located inside the cavities of γ-CD pairs in γ-CD-MOFs. CONCLUSION γ-CD-MOFs are promising carriers for bioactive agents in food and pharmaceutical applications. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zhenjiong Wang
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing, People's Republic of China
| | - Yiding Ma
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People's Republic of China
| | - Ying Jiang
- School of life, Nanjing Normal University, Nanjing, People's Republic of China
| | - Feng Zhou
- School of Food Science, Nanjing Xiaozhuang University, Nanjing, People's Republic of China
- Jiangsu Provincial Key Construction Laboratory of Special Biomass by-product Resource Utilization, Nanjing Xiaozhuang University, Nanjing, People's Republic of China
| | - Yulong Wu
- School of Food Science, Nanjing Xiaozhuang University, Nanjing, People's Republic of China
- Jiangsu Provincial Key Construction Laboratory of Special Biomass by-product Resource Utilization, Nanjing Xiaozhuang University, Nanjing, People's Republic of China
| | - Haitao Jiang
- School of Food Science, Nanjing Xiaozhuang University, Nanjing, People's Republic of China
- Jiangsu Provincial Key Construction Laboratory of Special Biomass by-product Resource Utilization, Nanjing Xiaozhuang University, Nanjing, People's Republic of China
| | - Renlei Wang
- Biology Department, Jiangsu Second Normal University, Nanjing, People's Republic of China
| | - Qing Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People's Republic of China
| | - Chun Hua
- School of Food Science, Nanjing Xiaozhuang University, Nanjing, People's Republic of China
- Jiangsu Provincial Key Construction Laboratory of Special Biomass by-product Resource Utilization, Nanjing Xiaozhuang University, Nanjing, People's Republic of China
| |
Collapse
|
3
|
Chen Y, Wang X, Shi H, Zou P. Montelukast Inhibits HCoV-OC43 Infection as a Viral Inactivator. Viruses 2022; 14:v14050861. [PMID: 35632604 PMCID: PMC9143845 DOI: 10.3390/v14050861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023] Open
Abstract
Coronaviruses (CoVs) consist of a large group of RNA viruses causing various diseases in humans and in lots of animals. Human coronavirus (HCoV) OC43, the prototype of beta-coronavirus discovered in the 1960s, has been circulating in humans for long time, and infection with other emerging strains of beta-coronavirus (SARS-CoV, SARS-CoV-2, and MERS-CoV) can lead to severe illness and death. In this study, we found that montelukast, a leukotriene receptor antagonist, potently inhibited the infection of HCoV-OC43 in distinct cells in a dose- and time- dependent manner. Additionally, the results showed that montelukast induced release of HCoV-OC43 genomic RNA by disrupting the integrity of the viral lipid membrane, and irreversibly inhibited viral infection. Considering the similarity among HCoV-OC43, MERS-CoV, and SARS-CoV-2, it suggests that montelukast may be a potential candidate for the treatment of human beta-coronavirus infection.
Collapse
Affiliation(s)
| | | | | | - Peng Zou
- Correspondence: ; Tel.: +86-21-3799-0333 (ext. 5273)
| |
Collapse
|
4
|
Bär SI, Biersack B, Schobert R. 3D cell cultures, as a surrogate for animal models, enhance the diagnostic value of preclinical in vitro investigations by adding information on the tumour microenvironment: a comparative study of new dual-mode HDAC inhibitors. Invest New Drugs 2022; 40:953-961. [PMID: 35796910 PMCID: PMC9395463 DOI: 10.1007/s10637-022-01280-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/01/2022] [Indexed: 12/15/2022]
Abstract
Anchorage-independent 3D-cultures of multicellular tumour spheroids (MCTS) and in vitro microtumours of cancer cells can provide upfront information on the effects of anticancer drug candidates, tantamount to that obtained from animal xenograft studies. Unlike 2D cancer cell cultures, 3D-models take into account the influence of the tumour microenvironment and the location dependence of drug effects and accumulation. We exemplified this by comparison of the effects of two new dual-mode anticancer agents, Troxbam and Troxham, and their monomodal congeners SAHA (suberoylanilide hydroxamic acid) and CA-4 (combretastatin A-4). We assessed the growth of MCTS of HCT116<sup>wt</sup> human colon carcinoma cells exposed to these compounds, as well as the spatial distribution of dead HCT116<sup>wt</sup> cells in these MCTS. Also, fluorescence imaging of live and fixed MCTS was used to assess the type of cellular death induced by test compounds. Furthermore, an innovative perfusion bioreactor system was used to grow microtumours in the presence or absence of test compounds. Both new investigational compounds led to significant reductions of the size of such MCTS and also of corresponding in vitro microtumours by inducing caspase-9 dependent apoptosis and elevated levels of reactive oxygen species. 3D multicellular tumour spheroids are easy to grow and employ for compound tests in the familiar well-plate set-up. Together with 3D microtumours grown at scaffolds in continuously perfused bioreactors they allow to study, early on in the course of drug evaluations, the communication of tumour cells with their microenvironment to an extent hitherto available only in animal experiments.
Collapse
Affiliation(s)
- Sofia I Bär
- Organic Chemistry Laboratory, University of Bayreuth, Universitätsstraße 30, 95447, Bayreuth, Germany.
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, Universitätsstraße 30, 95447, Bayreuth, Germany
| | - Rainer Schobert
- Organic Chemistry Laboratory, University of Bayreuth, Universitätsstraße 30, 95447, Bayreuth, Germany
| |
Collapse
|
5
|
Saravanakumar K, Park S, Sathiyaseelan A, Mariadoss AVA, Park S, Kim SJ, Wang MH. Isolation of Polysaccharides from Trichoderma harzianum with Antioxidant, Anticancer, and Enzyme Inhibition Properties. Antioxidants (Basel) 2021; 10:1372. [PMID: 34573005 PMCID: PMC8471597 DOI: 10.3390/antiox10091372] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 01/15/2023] Open
Abstract
In this work, a total of six polysaccharides were isolated from culture filtrate (EPS1, EPS2) and mycelia (IPS1-IPS4) of Trichoderma harzianum. The HPLC analysis results showed that EPS1, EPS2, IPS1, and IPS2 were composed of mannose, ribose, glucose, galactose, and arabinose. The FT-IR, 1H, and 13C NMR chemical shifts confirmed that the signals in EPS1 mainly consist of (1→4)-linked α-d-glucopyranose. EPS1 and IPS1 showed a smooth and clean surface, while EPS2, IPS2, and IPS3 exhibited a microporous structure. Among polysaccharides, EPS1 displayed higher ABTS+ (47.09 ± 2.25% and DPPH (26.44 ± 0.12%) scavenging activities, as well as higher α-amylase (69.30 ± 1.28%) and α-glucosidase (68.22 ± 0.64%) inhibition activity than the other polysaccharides. EPS1 exhibited high cytotoxicity to MDA-MB293 cells, with an IC50 of 0.437 mg/mL, and this was also confirmed by cell staining and FACS assays. These results report the physicochemical and bioactive properties of polysaccharides from T. harzianum.
Collapse
Affiliation(s)
- Kandasamy Saravanakumar
- Department of Bio Health Convergence, Kangwon National University, Chuncheon 200-701, Korea; (K.S.); (A.S.); (A.V.A.M.); (S.P.)
| | - SeonJu Park
- Chuncheon Center, Korea Basic Science Institute (KBSI), Chuncheon 24341, Korea;
| | - Anbazhagan Sathiyaseelan
- Department of Bio Health Convergence, Kangwon National University, Chuncheon 200-701, Korea; (K.S.); (A.S.); (A.V.A.M.); (S.P.)
| | - Arokia Vijaya Anand Mariadoss
- Department of Bio Health Convergence, Kangwon National University, Chuncheon 200-701, Korea; (K.S.); (A.S.); (A.V.A.M.); (S.P.)
| | - Soyoung Park
- Department of Bio Health Convergence, Kangwon National University, Chuncheon 200-701, Korea; (K.S.); (A.S.); (A.V.A.M.); (S.P.)
| | - Seong-Jung Kim
- Department of Physical Therapy, College of Health and Science, Kangwon National University, Samcheok-si 24949, Korea
| | - Myeong-Hyeon Wang
- Department of Bio Health Convergence, Kangwon National University, Chuncheon 200-701, Korea; (K.S.); (A.S.); (A.V.A.M.); (S.P.)
| |
Collapse
|
6
|
Hudáčová M, Hamuľaková S, Konkoľová E, Jendželovský R, Vargová J, Ševc J, Fedoročko P, Soukup O, Janočková J, Ihnatova V, Kučera T, Bzonek P, Novakova N, Jun D, Junova L, Korábečný J, Kuča K, Kožurková M. Synthesis of New Biscoumarin Derivatives, In Vitro Cholinesterase Inhibition, Molecular Modelling and Antiproliferative Effect in A549 Human Lung Carcinoma Cells. Int J Mol Sci 2021; 22:ijms22083830. [PMID: 33917200 PMCID: PMC8068036 DOI: 10.3390/ijms22083830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 12/29/2022] Open
Abstract
A series of novel C4-C7-tethered biscoumarin derivatives (12a–e) linked through piperazine moiety was designed, synthesized, and evaluated biological/therapeutic potential. Biscoumarin 12d was found to be the most effective inhibitor of both acetylcholinesterase (AChE, IC50 = 6.30 µM) and butyrylcholinesterase (BChE, IC50 = 49 µM). Detailed molecular modelling studies compared the accommodation of ensaculin (well-established coumarin derivative tested in phase I of clinical trials) and 12d in the human recombinant AChE (hAChE) active site. The ability of novel compounds to cross the blood–brain barrier (BBB) was predicted with a positive outcome for compound 12e. The antiproliferative effects of newly synthesized biscoumarin derivatives were tested in vitro on human lung carcinoma cell line (A549) and normal colon fibroblast cell line (CCD-18Co). The effect of derivatives on cell proliferation was evaluated by MTT assay, quantification of cell numbers and viability, colony-forming assay, analysis of cell cycle distribution and mitotic activity. Intracellular localization of used derivatives in A549 cells was confirmed by confocal microscopy. Derivatives 12d and 12e showed significant antiproliferative activity in A549 cancer cells without a significant effect on normal CCD-18Co cells. The inhibition of hAChE/human recombinant BChE (hBChE), the antiproliferative activity on cancer cells, and the ability to cross the BBB suggest the high potential of biscoumarin derivatives. Beside the treatment of cancer, 12e might be applicable against disorders such as schizophrenia, and 12d could serve future development as therapeutic agents in the prevention and/or treatment of Alzheimer’s disease.
Collapse
Affiliation(s)
- Monika Hudáčová
- Department of Biochemistry, Faculty of Science, Pavol Jozef Šafárik University in Košice, Šrobárova 2, 041 54 Košice, Slovakia; (M.H.); (E.K.); (M.K.)
| | - Slávka Hamuľaková
- Department of Organic Chemistry, Institute of Chemical Sciences, Faculty of Science, Pavol Jozef Šafárik University in Košice, Moyzesova 11, 040 01 Kosice, Slovakia
- Correspondence:
| | - Eva Konkoľová
- Department of Biochemistry, Faculty of Science, Pavol Jozef Šafárik University in Košice, Šrobárova 2, 041 54 Košice, Slovakia; (M.H.); (E.K.); (M.K.)
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Náměstí 542/2, 160 00 Prague 6, Czech Republic
| | - Rastislav Jendželovský
- Department of Cellular Biology, Faculty of Science, Pavol Jozef Šafárik University in Košice, Šrobárova 2, 041 54 Košice, Slovakia; (R.J.); (J.V.); (J.Š.); (P.F.)
| | - Jana Vargová
- Department of Cellular Biology, Faculty of Science, Pavol Jozef Šafárik University in Košice, Šrobárova 2, 041 54 Košice, Slovakia; (R.J.); (J.V.); (J.Š.); (P.F.)
| | - Juraj Ševc
- Department of Cellular Biology, Faculty of Science, Pavol Jozef Šafárik University in Košice, Šrobárova 2, 041 54 Košice, Slovakia; (R.J.); (J.V.); (J.Š.); (P.F.)
| | - Peter Fedoročko
- Department of Cellular Biology, Faculty of Science, Pavol Jozef Šafárik University in Košice, Šrobárova 2, 041 54 Košice, Slovakia; (R.J.); (J.V.); (J.Š.); (P.F.)
| | - Ondrej Soukup
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Trebesska 1575, 500 05 Hradec Kralove, Czech Republic; (O.S.); (V.I.); (T.K.); (P.B.); (D.J.); (L.J.); (J.K.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (J.J.); (N.N.); (K.K.)
| | - Jana Janočková
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (J.J.); (N.N.); (K.K.)
| | - Veronika Ihnatova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Trebesska 1575, 500 05 Hradec Kralove, Czech Republic; (O.S.); (V.I.); (T.K.); (P.B.); (D.J.); (L.J.); (J.K.)
| | - Tomáš Kučera
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Trebesska 1575, 500 05 Hradec Kralove, Czech Republic; (O.S.); (V.I.); (T.K.); (P.B.); (D.J.); (L.J.); (J.K.)
| | - Petr Bzonek
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Trebesska 1575, 500 05 Hradec Kralove, Czech Republic; (O.S.); (V.I.); (T.K.); (P.B.); (D.J.); (L.J.); (J.K.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (J.J.); (N.N.); (K.K.)
| | - Nikola Novakova
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (J.J.); (N.N.); (K.K.)
| | - Daniel Jun
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Trebesska 1575, 500 05 Hradec Kralove, Czech Republic; (O.S.); (V.I.); (T.K.); (P.B.); (D.J.); (L.J.); (J.K.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (J.J.); (N.N.); (K.K.)
| | - Lucie Junova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Trebesska 1575, 500 05 Hradec Kralove, Czech Republic; (O.S.); (V.I.); (T.K.); (P.B.); (D.J.); (L.J.); (J.K.)
| | - Jan Korábečný
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defense, Trebesska 1575, 500 05 Hradec Kralove, Czech Republic; (O.S.); (V.I.); (T.K.); (P.B.); (D.J.); (L.J.); (J.K.)
- National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
| | - Kamil Kuča
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (J.J.); (N.N.); (K.K.)
| | - Mária Kožurková
- Department of Biochemistry, Faculty of Science, Pavol Jozef Šafárik University in Košice, Šrobárova 2, 041 54 Košice, Slovakia; (M.H.); (E.K.); (M.K.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (J.J.); (N.N.); (K.K.)
| |
Collapse
|
7
|
Metabolite Profiling of Methanolic Extract of Gardenia jaminoides by LC-MS/MS and GC-MS and Its Anti-Diabetic, and Anti-Oxidant Activities. Pharmaceuticals (Basel) 2021; 14:ph14020102. [PMID: 33525758 PMCID: PMC7912419 DOI: 10.3390/ph14020102] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/20/2021] [Accepted: 01/25/2021] [Indexed: 12/18/2022] Open
Abstract
In this study, the methanolic extract from seeds of Gardenia jasminoides exhibited strong antioxidant and enzyme inhibition activities with less toxicity to NIH3T3 and HepG2 cells at the concentration of 100 µg/mL. The antioxidant activities (DPPH and ABTS), α-amylase, and α-glucosidase inhibition activities were found higher in methanolic extract (MeOH-E) than H2O extract. Besides, 9.82 ± 0.62 µg and 6.42 ± 0.26 µg of MeOH-E were equivalent to 1 µg ascorbic acid for ABTS and DPPH scavenging, respectively while 9.02 ± 0.25 µg and 6.52 ± 0.15 µg of MeOH-E were equivalent to 1 µg of acarbose for inhibition of α-amylase and α-glucosidase respectively. Moreover, the cell assay revealed that the addition of MeOH-E (12.5 µg/mL) increased about 37% of glucose uptake in insulin resistant (IR) HepG2 as compared to untreated IR HepG2 cells. The LC- MS/MS and GC-MS analysis of MeOH-E revealed a total of 54 compounds including terpenoids, glycosides, fatty acid, phenolic acid derivatives. Among the identified compounds, chlorogenic acid and jasminoside A were found promising for anti-diabetic activity revealed by molecular docking study and these molecules are deserving further purification and molecular analysis.
Collapse
|
8
|
Gonzalez G, Hodoň J, Kazakova A, D'Acunto CW, Kaňovský P, Urban M, Strnad M. Novel pentacyclic triterpenes exhibiting strong neuroprotective activity in SH-SY5Y cells in salsolinol- and glutamate-induced neurodegeneration models. Eur J Med Chem 2021; 213:113168. [PMID: 33508480 DOI: 10.1016/j.ejmech.2021.113168] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Novel triterpene derivatives were prepared and evaluated in salsolinol (SAL)- and glutamate (Glu)-induced models of neurodegeneration in neuron-like SH-SY5Y cells. Among the tested compounds, betulin triazole 4 bearing a tetraacetyl-β-d-glucose substituent showed a highly potent neuroprotective effect. Further studies revealed that removal of tetraacetyl-β-d-glucose part (free triazole derivative 10) resulted in strong neuroprotection in the SAL model at 1 μM, but this derivative suffered from cytotoxicity at higher concentrations. Both compounds modulated oxidative stress and caspase-3,7 activity, but 10 showed a superior effect comparable to the Ac-DEVD-CHO inhibitor. Interestingly, while both 4 and 10 outperformed the positive controls in blocking mitochondrial permeability transition pore opening, only 4 demonstrated potent restoration of the mitochondrial membrane potential (MMP) in the model. Derivatives 4 and 10 also showed neuroprotection in the Glu model, with 10 exhibiting the strongest oxidative stress reducing effect among the tested compounds, while the neuroprotective activity of 4 was probably due recovery of the MMP.
Collapse
Affiliation(s)
- Gabriel Gonzalez
- Laboratory of Growth Regulators, Faculty of Science, Palacký University and the Institute of Experimental Botany of the Czech Academy of Sciences, Šlechtitelů 27, CZ-78371, Olomouc, Czech Republic; Department of Neurology, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacký University Olomouc, CZ-775 20, Olomouc, Czech Republic
| | - Jiří Hodoň
- Department of Organic Chemistry, Faculty of Science, Palacky University, 17. Listopadu 1192/12, 771 46, Olomouc, Czech Republic
| | - Anna Kazakova
- Department of Organic Chemistry, Faculty of Science, Palacky University, 17. Listopadu 1192/12, 771 46, Olomouc, Czech Republic
| | - Cosimo Walter D'Acunto
- Laboratory of Growth Regulators, Faculty of Science, Palacký University and the Institute of Experimental Botany of the Czech Academy of Sciences, Šlechtitelů 27, CZ-78371, Olomouc, Czech Republic
| | - Petr Kaňovský
- Department of Neurology, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacký University Olomouc, CZ-775 20, Olomouc, Czech Republic
| | - Milan Urban
- Department of Medicinal Chemistry, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University in Olomouc, Hnevotinska 5, 779 00, Olomouc, Czech Republic.
| | - Miroslav Strnad
- Laboratory of Growth Regulators, Faculty of Science, Palacký University and the Institute of Experimental Botany of the Czech Academy of Sciences, Šlechtitelů 27, CZ-78371, Olomouc, Czech Republic; Department of Neurology, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacký University Olomouc, CZ-775 20, Olomouc, Czech Republic.
| |
Collapse
|
9
|
Geck RC, Foley JR, Murray Stewart T, Asara JM, Casero RA, Toker A. Inhibition of the polyamine synthesis enzyme ornithine decarboxylase sensitizes triple-negative breast cancer cells to cytotoxic chemotherapy. J Biol Chem 2020; 295:6263-6277. [PMID: 32139506 PMCID: PMC7212655 DOI: 10.1074/jbc.ra119.012376] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/28/2020] [Indexed: 12/19/2022] Open
Abstract
Treatment of patients with triple-negative breast cancer (TNBC) is limited by a lack of effective molecular therapies targeting this disease. Recent studies have identified metabolic alterations in cancer cells that can be targeted to improve responses to standard-of-care chemotherapy regimens. Using MDA-MB-468 and SUM-159PT TNBC cells, along with LC-MS/MS and HPLC metabolomics profiling, we found here that exposure of TNBC cells to the cytotoxic chemotherapy drugs cisplatin and doxorubicin alter arginine and polyamine metabolites. This alteration was because of a reduction in the levels and activity of a rate-limiting polyamine biosynthetic enzyme, ornithine decarboxylase (ODC). Using gene silencing and inhibitor treatments, we determined that the reduction in ODC was mediated by its negative regulator antizyme, targeting ODC to the proteasome for degradation. Treatment with the ODC inhibitor difluoromethylornithine (DFMO) sensitized TNBC cells to chemotherapy, but this was not observed in receptor-positive breast cancer cells. Moreover, TNBC cell lines had greater sensitivity to single-agent DFMO, and ODC levels were elevated in TNBC patient samples. The alterations in polyamine metabolism in response to chemotherapy, as well as DFMO-induced preferential sensitization of TNBC cells to chemotherapy, reported here suggest that ODC may be a targetable metabolic vulnerability in TNBC.
Collapse
Affiliation(s)
- Renee C Geck
- Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215
- Harvard Medical School, Boston, Massachusetts 02115
| | - Jackson R Foley
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | - Tracy Murray Stewart
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | - John M Asara
- Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215
| | - Robert A Casero
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | - Alex Toker
- Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215
- Harvard Medical School, Boston, Massachusetts 02115
- Ludwig Center at Harvard, Boston, Massachusetts 02115
| |
Collapse
|
10
|
Wenzel TJ, Bajwa E, Klegeris A. Cytochrome c can be released into extracellular space and modulate functions of human astrocytes in a toll-like receptor 4-dependent manner. Biochim Biophys Acta Gen Subj 2019; 1863:129400. [PMID: 31344401 DOI: 10.1016/j.bbagen.2019.07.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 06/23/2019] [Accepted: 07/18/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND Chronic activation of glial cells contributes to neurodegenerative diseases. Cytochrome c (CytC) is a soluble mitochondrial protein that can act as a damage-associated molecular pattern (DAMP) when released into the extracellular space from damaged cells. CytC causes immune activation of microglia in a toll-like receptor (TLR) 4-dependent manner. The effects of extracellular CytC on astrocytes are unknown. Astrocytes, which are the most abundant glial cell type in the brain, express TLR 4 and secrete inflammatory mediators; therefore, we hypothesized that extracellular CytC can interact with the TLR 4 of astrocytes inducing their release of inflammatory molecules and cytotoxins. METHOD Experiments were conducted using primary human astrocytes, U118 MG human astrocytic cells, BV-2 murine microglia, and SH-SY5Y human neuronal cells. RESULTS Extracellularly applied CytC increased the secretion of interleukin (IL)-1β, granulocyte-macrophage colony stimulating factor (GM-CSF) and IL-12 p70 by cultured primary human astrocytes. Anti-TLR 4 antibodies blocked the CytC-induced secretion of IL-1β and GM-CSF by astrocytes. Supernatants from CytC-activated astrocytes were toxic to human SH-SY5Y neuronal cells. We also demonstrated CytC release from damaged glial cells by measuring CytC in the supernatants of BV-2 microglia after their exposure to cytotoxic concentrations of staurosporine, amyloid-β peptides (Aβ42) and tumor necrosis factor-α. CONCLUSION CytC can be released into the extracellular space from damaged glial cells causing immune activation of astrocytes in a TLR 4-dependent manner. GENERAL SIGNIFICANCE Astrocyte activation by CytC may contribute to neuroinflammation and neuronal death in neurodegenerative diseases. Astrocyte TLR 4 could be a potential therapeutic target in these diseases.
Collapse
Affiliation(s)
- Tyler J Wenzel
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Ekta Bajwa
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada.
| |
Collapse
|
11
|
Katebi B, Mahdavimehr M, Meratan AA, Ghasemi A, Nemat-Gorgani M. Protective effects of silibinin on insulin amyloid fibrillation, cytotoxicity and mitochondrial membrane damage. Arch Biochem Biophys 2018; 659:22-32. [PMID: 30266624 DOI: 10.1016/j.abb.2018.09.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 09/12/2018] [Accepted: 09/25/2018] [Indexed: 02/02/2023]
Abstract
A growing body of evidence suggests that secretion and assembly of insulin to amyloid fibrils reduce its efficacy in treating type II diabetes and may lead to dysfunctioning of several organs. The research presented here explores the effects of silibinin on the in vitro amyloid fibrillation and cytotoxicity of bovine insulin fibrils on SH-SY5Y human neuroblastoma cells. Interaction of the resulting structures with rat brain mitochondria was also investigated. Using a range of methods for amyloid detection we showed that insulin fibrillation was significantly inhibited by silibinin in a dose-dependent fashion. Moreover, we found that silibinin was very effective in attenuating insulin fibril-induced neuronal toxicity characterized by decrease of cell viability, the release of lactate dehydrogenase, intracellular reactive oxygen species enhancement, morphological alterations, and apoptotic cell death induction. While insulin fibrillation products showed the capacity to damage mitochondria, the resultant structures produced in the presence of silibinin were totally ineffective. Together, results demonstrate the capacity of insulin fibrils to cause SH-SY5Y cell death by inducing necrosis/apoptosis changes and suggest how silibinin may afford protection. It is concluded that elucidation of such protection may provide important insights into the development of preventive and therapeutic agents for amyloid-related diseases.
Collapse
Affiliation(s)
- Bentolhoda Katebi
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 45137-66731, Iran
| | - Mohsen Mahdavimehr
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 45137-66731, Iran
| | - Ali Akbar Meratan
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, 45137-66731, Iran.
| | - Atiyeh Ghasemi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | |
Collapse
|
12
|
Lien EC, Ghisolfi L, Geck RC, Asara JM, Toker A. Oncogenic PI3K promotes methionine dependency in breast cancer cells through the cystine-glutamate antiporter xCT. Sci Signal 2017; 10:10/510/eaao6604. [PMID: 29259101 DOI: 10.1126/scisignal.aao6604] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The precursor homocysteine is metabolized either through the methionine cycle to produce methionine or through the transsulfuration pathway to synthesize cysteine. Alternatively, cysteine can be obtained through uptake of its oxidized form, cystine. Many cancer cells exhibit methionine dependency such that their proliferation is impaired in growth media in which methionine is replaced by homocysteine. We showed that oncogenic PIK3CA and decreased expression of SLC7A11, a gene that encodes a cystine transporter also known as xCT, correlated with increased methionine dependency in breast cancer cells. Oncogenic PIK3CA was sufficient to confer methionine dependency to mammary epithelial cells, partly by decreasing cystine uptake through the transcriptional and posttranslational inhibition of xCT. Manipulation of xCT activity altered the proliferation of breast cancer cells in methionine-deficient, homocysteine-containing media, suggesting that it functionally contributed to methionine dependency. We propose that concurrent with decreased cystine uptake through xCT, PIK3CA mutant cells use homocysteine through the transsulfuration pathway to synthesize cysteine. Consequently, less homocysteine is available to produce methionine, contributing to methionine dependency. These results indicate that oncogenic PIK3CA alters methionine and cysteine utilization, partly by inhibiting xCT to contribute to the methionine dependency phenotype in breast cancer cells.
Collapse
Affiliation(s)
- Evan C Lien
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Laura Ghisolfi
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Renee C Geck
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - John M Asara
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston 02215 MA, USA.,Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Alex Toker
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA. .,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston 02215 MA, USA.,Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.,Ludwig Center at Harvard, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
13
|
Szatmary P, Liu T, Abrams ST, Voronina S, Wen L, Chvanov M, Huang W, Wang G, Criddle DN, Tepikin AV, Toh CH, Sutton R. Systemic histone release disrupts plasmalemma and contributes to necrosis in acute pancreatitis. Pancreatology 2017; 17:884-892. [PMID: 29102149 DOI: 10.1016/j.pan.2017.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 10/01/2017] [Accepted: 10/06/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND Clinical and experimental acute pancreatitis feature histone release within the pancreas from innate immune cells and acinar cell necrosis. In this study, we aimed to detail the source of circulating histones and assess their role in the pathogenesis of acute pancreatitis. METHODS Circulating nucleosomes were measured in patient plasma, taken within 24 and 48 h of onset of acute pancreatitis and correlated with clinical outcomes. Using caerulein hyperstimulation, circulating histones were measured in portal, systemic venous and systemic arterial circulation in mice, and the effects of systemic administration of histones in this model were assessed. The sites of actions of circulating histones were assessed by administration of FITC-labelled histones. The effects of histones on isolated pancreatic acinar cells were further assessed by measuring acinar cell death and calcium permeability in vitro. RESULTS Cell-free histones were confirmed to be abundant in human acute pancreatitis and found to derive from pancreatitis-associated liver injury in a rodent model of the disease. Fluorescein isothianate-labelled histones administered systemically targeted the pancreas and exacerbated injury in experimental acute pancreatitis. Histones induce charge- and concentration-dependent plasmalemma leakage and necrosis in isolated pancreatic acinar cells, independent of extracellular calcium. CONCLUSION We conclude that histones released systemically in acute pancreatitis concentrate within the inflamed pancreas and exacerbate injury. Circulating histones may provide meaningful biomarkers and targets for therapy in clinical acute pancreatitis.
Collapse
Affiliation(s)
- Peter Szatmary
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, University of Liverpool, Liverpool, L69 3GA, UK; Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, L69 3BX, UK
| | - Tingting Liu
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, University of Liverpool, Liverpool, L69 3GA, UK; Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK; Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Simon T Abrams
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK
| | - Svetlana Voronina
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, L69 3BX, UK
| | - Li Wen
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, University of Liverpool, Liverpool, L69 3GA, UK
| | - Michael Chvanov
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, L69 3BX, UK
| | - Wei Huang
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, University of Liverpool, Liverpool, L69 3GA, UK; Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Guozheng Wang
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK
| | - David N Criddle
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, L69 3BX, UK
| | - Alexey V Tepikin
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, L69 3BX, UK
| | - Cheng-Hock Toh
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, UK; Roald Dahl Haemostasis and Thrombosis Centre, Royal Liverpool University Hospital, Liverpool, L7 8XP, UK.
| | - Robert Sutton
- NIHR Liverpool Pancreas Biomedical Research Unit, Royal Liverpool University Hospital, University of Liverpool, Liverpool, L69 3GA, UK
| |
Collapse
|
14
|
Endo S, Nakata K, Ohuchida K, Takesue S, Nakayama H, Abe T, Koikawa K, Okumura T, Sada M, Horioka K, Zheng B, Mizuuchi Y, Iwamoto C, Murata M, Moriyama T, Miyasaka Y, Ohtsuka T, Mizumoto K, Oda Y, Hashizume M, Nakamura M. Autophagy Is Required for Activation of Pancreatic Stellate Cells, Associated With Pancreatic Cancer Progression and Promotes Growth of Pancreatic Tumors in Mice. Gastroenterology 2017; 152:1492-1506.e24. [PMID: 28126348 DOI: 10.1053/j.gastro.2017.01.010] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 01/04/2017] [Accepted: 01/11/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Pancreatic stellate cells (PSCs) change from a quiescent to activated state in the tumor environment and secrete extracellular matrix (ECM) molecules and cytokines to increase the aggressiveness of tumors. However, it is not clear how PSCs are activated to produce these factors, or whether this process can be inhibited. PSCs have morphologic and functional similarities to hepatic stellate cells, which undergo autophagy to promote fibrosis and tumor growth. We investigated whether autophagy activates PSCs, which promotes development of the tumor stroma and growth of pancreatic tumors in mice. METHODS We used immunofluorescence microscopy and immunohistochemistry to analyze pancreatic tumor specimens from 133 patients who underwent pancreatectomy in Japan from 2000 to 2009. PSCs were cultured from pancreatic tumor tissues or tissues of patients with chronic pancreatitis; these were analyzed by immunofluorescence microscopy, immunoblots, quantitative reverse transcription polymerase chain reaction, and in assays for invasiveness, proliferation, and lipid droplets. Autophagy was inhibited in PSCs by administration of chloroquine or transfection with small interfering RNAs. Proteins were knocked down in immortalized PSCs by expression of small hairpin RNAs. Cells were transplanted into pancreatic tails of nude mice, and tumor growth and metastasis were quantified. RESULTS Based on immunohistochemical analyses, autophagy was significantly associated with tumor T category (P = .018), histologic grade (P = .001), lymph node metastases (P < .001), stage (P = .009), perilymphatic invasion (P = .001), and perivascular invasion (P = .003). Autophagy of PSCs was associated with shorter survival times of patients with pancreatic cancer. PSC expression of microtubule-associated protein 1 light chain 3, a marker of autophagosomes, was associated with poor outcomes (shorter survival time, disease recurrence) for patients with pancreatic cancer (relative risk of shorter survival time, 1.56). Immunoblots showed that PSCs from pancreatic tumor samples expressed higher levels of markers of autophagy than PSCs from chronic pancreatitis samples. Inhibitors of autophagy increased the number of lipid droplets of PSCs, indicating a quiescent state of PSCs, and reduced their production of ECM molecules and interleukin 6, as well as their proliferation and invasiveness in culture. PSCs exposed to autophagy inhibitors formed smaller tumors in nude mice (P = .001) and fewer liver metastases (P = .018) with less peritoneal dissemination (P = .018) compared to PSCs not exposed to autophagy inhibitors. CONCLUSIONS Autophagic PSCs produce ECM molecules and interleukin 6 and are associated with shorter survival times and disease recurrence in patients with pancreatic cancer. Inhibitors of PSC autophagy might reduce pancreatic tumor invasiveness by altering the tumor stroma.
Collapse
Affiliation(s)
- Sho Endo
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohei Nakata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Shin Takesue
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiromichi Nakayama
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiya Abe
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuhiro Koikawa
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Okumura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Sada
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohei Horioka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Biao Zheng
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yusuke Mizuuchi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Anatomical Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Chika Iwamoto
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaharu Murata
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taiki Moriyama
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Miyasaka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takao Ohtsuka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuhiro Mizumoto
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomical Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Makoto Hashizume
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
15
|
Brown KK, Spinelli JB, Asara JM, Toker A. Adaptive Reprogramming of De Novo Pyrimidine Synthesis Is a Metabolic Vulnerability in Triple-Negative Breast Cancer. Cancer Discov 2017; 7:391-399. [PMID: 28255083 DOI: 10.1158/2159-8290.cd-16-0611] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 11/22/2016] [Accepted: 01/17/2017] [Indexed: 02/06/2023]
Abstract
Chemotherapy resistance is a major barrier to the treatment of triple-negative breast cancer (TNBC), and strategies to circumvent resistance are required. Using in vitro and in vivo metabolic profiling of TNBC cells, we show that an increase in the abundance of pyrimidine nucleotides occurs in response to chemotherapy exposure. Mechanistically, elevation of pyrimidine nucleotides induced by chemotherapy is dependent on increased activity of the de novo pyrimidine synthesis pathway. Pharmacologic inhibition of de novo pyrimidine synthesis sensitizes TNBC cells to genotoxic chemotherapy agents by exacerbating DNA damage. Moreover, combined treatment with doxorubicin and leflunomide, a clinically approved inhibitor of the de novo pyrimidine synthesis pathway, induces regression of TNBC xenografts. Thus, the increase in pyrimidine nucleotide levels observed following chemotherapy exposure represents a metabolic vulnerability that can be exploited to enhance the efficacy of chemotherapy for the treatment of TNBC.Significance: The prognosis for patients with TNBC with residual disease after chemotherapy is poor. We find that chemotherapy agents induce adaptive reprogramming of de novo pyrimidine synthesis and show that this response can be exploited pharmacologically, using clinically approved inhibitors of de novo pyrimidine synthesis, to sensitize TNBC cells to chemotherapy. Cancer Discov; 7(4); 391-9. ©2017 AACR.See related article by Mathur et al., p. 380This article is highlighted in the In This Issue feature, p. 339.
Collapse
Affiliation(s)
- Kristin K Brown
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts. .,Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.,Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Jessica B Spinelli
- Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts
| | - John M Asara
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Alex Toker
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts. .,Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.,Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
16
|
Le Borgne F, Ravaut G, Bernard A, Demarquoy J. L-carnitine protects C2C12 cells against mitochondrial superoxide overproduction and cell death. World J Biol Chem 2017; 8:86-94. [PMID: 28289521 PMCID: PMC5329717 DOI: 10.4331/wjbc.v8.i1.86] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/29/2016] [Accepted: 12/14/2016] [Indexed: 02/05/2023] Open
Abstract
AIM To identify and characterize the protective effect that L-carnitine exerted against an oxidative stress in C2C12 cells.
METHODS Myoblastic C2C12 cells were treated with menadione, a vitamin K analog that engenders oxidative stress, and the protective effect of L-carnitine (a nutrient involved in fatty acid metabolism and the control of the oxidative process), was assessed by monitoring various parameters related to the oxidative stress, autophagy and cell death.
RESULTS Associated with its physiological function, a muscle cell metabolism is highly dependent on oxygen and may produce reactive oxygen species (ROS), especially under pathological conditions. High levels of ROS are known to induce injuries in cell structure as they interact at many levels in cell function. In C2C12 cells, a treatment with menadione induced a loss of transmembrane mitochondrial potential, an increase in mitochondrial production of ROS; it also induces autophagy and was able to provoke cell death. Pre-treatment of the cells with L-carnitine reduced ROS production, diminished autophagy and protected C2C12 cells against menadione-induced deleterious effects.
CONCLUSION In conclusion, L-carnitine limits the oxidative stress in these cells and prevents cell death.
Collapse
|
17
|
Bondarenko OM, Heinlaan M, Sihtmäe M, Ivask A, Kurvet I, Joonas E, Jemec A, Mannerström M, Heinonen T, Rekulapelly R, Singh S, Zou J, Pyykkö I, Drobne D, Kahru A. Multilaboratory evaluation of 15 bioassays for (eco)toxicity screening and hazard ranking of engineered nanomaterials: FP7 project NANOVALID. Nanotoxicology 2016; 10:1229-42. [PMID: 27259032 PMCID: PMC5030619 DOI: 10.1080/17435390.2016.1196251] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Within EU FP7 project NANOVALID, the (eco)toxicity of 7 well-characterized engineered nanomaterials (NMs) was evaluated by 15 bioassays in 4 laboratories. The highest tested nominal concentration of NMs was 100 mg/l. The panel of the bioassays yielded the following toxicity order: Ag > ZnO > CuO > TiO2 > MWCNTs > SiO2 > Au. Ag, ZnO and CuO proved very toxic in the majority of assays, assumingly due to dissolution. The latter was supported by the parallel analysis of the toxicity of respective soluble metal salts. The most sensitive tests/species were Daphnia magna (towards Ag NMs, 24-h EC50 = 0.003 mg Ag/l), algae Raphidocelis subcapitata (ZnO and CuO, 72-h EC50 = 0.14 mg Zn/l and 0.7 mg Cu/l, respectively) and murine fibroblasts BALB/3T3 (CuO, 48-h EC50 = 0.7 mg Cu/l). MWCNTs showed toxicity only towards rat alveolar macrophages (EC50 = 15.3 mg/l) assumingly due to high aspect ratio and TiO2 towards R. subcapitata (EC50 = 6.8 mg Ti/l) due to agglomeration of TiO2 and entrapment of algal cells. Finally, we constructed a decision tree to select the bioassays for hazard ranking of NMs. For NM testing, we recommend a multitrophic suite of 4 in vitro (eco)toxicity assays: 48-h D. magna immobilization (OECD202), 72-h R. subcapitata growth inhibition (OECD201), 30-min Vibrio fischeri bioluminescence inhibition (ISO2010) and 48-h murine fibroblast BALB/3T3 neutral red uptake in vitro (OECD129) representing crustaceans, algae, bacteria and mammalian cells, respectively. Notably, our results showed that these assays, standardized for toxicity evaluation of “regular” chemicals, proved efficient also for shortlisting of hazardous NMs. Additional assays are recommended for immunotoxicity evaluation of high aspect ratio NMs (such as MWCNTs).
Collapse
Affiliation(s)
- Olesja M Bondarenko
- a Laboratory of Environmental Toxicology , National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Margit Heinlaan
- a Laboratory of Environmental Toxicology , National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Mariliis Sihtmäe
- a Laboratory of Environmental Toxicology , National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Angela Ivask
- a Laboratory of Environmental Toxicology , National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Imbi Kurvet
- a Laboratory of Environmental Toxicology , National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Elise Joonas
- a Laboratory of Environmental Toxicology , National Institute of Chemical Physics and Biophysics , Tallinn , Estonia .,b Faculty of Science and Technology , Institute of Ecology and Earth Sciences, Tartu University , Tartu , Estonia
| | - Anita Jemec
- c Biotechnical Faculty , University of Ljubljana , Ljubljana , Slovenia
| | - Marika Mannerström
- d The Finnish Centre for Alternative Methods (FICAM) , School of Medicine, University of Tampere , Tampere , Finland
| | - Tuula Heinonen
- d The Finnish Centre for Alternative Methods (FICAM) , School of Medicine, University of Tampere , Tampere , Finland
| | - Rohit Rekulapelly
- e The Centre for Cellular & Molecular Biology, Habsiguda , Hyderabad, Telangana , India , and
| | - Shashi Singh
- e The Centre for Cellular & Molecular Biology, Habsiguda , Hyderabad, Telangana , India , and
| | - Jing Zou
- f Hearing and Balance Research Unit , Field of Oto-Laryngology, School of Medicine, University of Tampere , Tampere , Finland
| | - Ilmari Pyykkö
- f Hearing and Balance Research Unit , Field of Oto-Laryngology, School of Medicine, University of Tampere , Tampere , Finland
| | - Damjana Drobne
- c Biotechnical Faculty , University of Ljubljana , Ljubljana , Slovenia
| | - Anne Kahru
- a Laboratory of Environmental Toxicology , National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| |
Collapse
|
18
|
Kongseng S, Yoovathaworn K, Wongprasert K, Chunhabundit R, Sukwong P, Pissuwan D. Cytotoxic and inflammatory responses of TiO2 nanoparticles on human peripheral blood mononuclear cells. J Appl Toxicol 2016; 36:1364-73. [PMID: 27225715 DOI: 10.1002/jat.3342] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 04/01/2016] [Accepted: 04/01/2016] [Indexed: 12/27/2022]
Abstract
Titanium dioxide nanoparticles (TiO2 -NPs) have been widely used in many applications. Owing to their nanoscale size, interactions between cells and NPs have been expansively investigated. With the health concerns raised regarding the adverse effects of these interactions, closer examination of whether TiO2 -NPs can induce toxicity towards human cells is greatly needed. Therefore, in this study, we investigated the cytotoxicity of TiO2 -NPs towards human blood cells (peripheral blood mononuclear cells [PBMCs]) in serum-free medium, for which there is little information regarding the cytotoxic effects of TiO2 -NPs. Our results provide evidence that PBMCs treated with TiO2 -NPs (at concentrations ≥25 μg ml(-1) ) for 24 h significantly reduced cell viability and significantly increased production of toxic mediators such as reactive oxygen species and inflammatory response cytokines such as interleukin-6 and tumor necrosis factor-α (P < 0.05). Cell apoptosis induction also occurred at these concentrations. Significant expressions of cyclooxygenase-2 and interleukin-1β were also observed in PBMCs treated with TiO2 -NPs at concentrations ≥125 μg ml(-1) . Our data presented here clearly indicate that the concentration of TiO2 -NPs (at size ~26.4 ± 1.2 nm) applied to human blood cells has a strong impact on cytotoxic induction. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Supunsa Kongseng
- Toxicology Graduate Program, Multidisciplinary Unit, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Krongtong Yoovathaworn
- Toxicology Graduate Program, Multidisciplinary Unit, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Rodjana Chunhabundit
- Graduate Program in Nutrition, Faculty of Medicine at Ramathibodi Hospital, Mahidol University, Bangkok
| | - Patinya Sukwong
- Toxicology Graduate Program, Multidisciplinary Unit, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Dakrong Pissuwan
- Toxicology Graduate Program, Multidisciplinary Unit, Faculty of Science, Mahidol University, Bangkok, Thailand.,Center of Excellence on Environmental Health and Toxicology, Faculty of Science, Mahidol University, Bangkok.,Materials Science and Engineering Graduate Program, Multidisciplinary Unit, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
19
|
Glutathione biosynthesis is a metabolic vulnerability in PI(3)K/Akt-driven breast cancer. Nat Cell Biol 2016; 18:572-8. [PMID: 27088857 PMCID: PMC4848114 DOI: 10.1038/ncb3341] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 03/11/2016] [Indexed: 02/06/2023]
Abstract
Cancer cells often select for mutations that enhance signalling through pathways that promote anabolic metabolism. Although the PI(3)K/Akt signalling pathway, which is frequently dysregulated in breast cancer, is a well-established regulator of central glucose metabolism and aerobic glycolysis, its regulation of other metabolic processes required for tumour growth is not well defined. Here we report that in mammary epithelial cells, oncogenic PI(3)K/Akt stimulates glutathione (GSH) biosynthesis by stabilizing and activating NRF2 to upregulate the GSH biosynthetic genes. Increased NRF2 stability is dependent on the Akt-mediated accumulation of p21(Cip1/WAF1) and GSK-3β inhibition. Consistently, in human breast tumours, upregulation of NRF2 targets is associated with PI(3)K pathway mutation status and oncogenic Akt activation. Elevated GSH biosynthesis is required for PI(3)K/Akt-driven resistance to oxidative stress, initiation of tumour spheroids, and anchorage-independent growth. Furthermore, inhibition of GSH biosynthesis with buthionine sulfoximine synergizes with cisplatin to selectively induce tumour regression in PI(3)K pathway mutant breast cancer cells, both in vitro and in vivo. Our findings provide insight into GSH biosynthesis as a metabolic vulnerability associated with PI(3)K pathway mutant breast cancers.
Collapse
|
20
|
Chhajed SS, Sonawane SS, Upasani CD, Kshirsagar SJ, Gupta PP. Design, synthesis and molecular modeling studies of few chalcone analogues of benzimidazole for epidermal growth factor receptor inhibitor in search of useful anticancer agent. Comput Biol Chem 2016; 61:138-44. [DOI: 10.1016/j.compbiolchem.2016.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/01/2016] [Indexed: 11/26/2022]
|
21
|
CASES ANAINES, OHTSUKA TAKAO, KIMURA HIDEYO, ZHENG BIAO, SHINDO KOJI, ODA YOSHINAO, MIZUMOTO KAZUHIRO, NAKAMURA MASAFUMI, TANAKA MASAO. Significance of expression of glucagon-like peptide 1 receptor in pancreatic cancer. Oncol Rep 2015; 34:1717-25. [DOI: 10.3892/or.2015.4138] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 06/29/2015] [Indexed: 11/05/2022] Open
|
22
|
Yu C, Wang M, Chen M, Huang Y, Jiang J. Upregulation of microRNA‑138‑5p inhibits pancreatic cancer cell migration and increases chemotherapy sensitivity. Mol Med Rep 2015; 12:5135-40. [PMID: 26135834 DOI: 10.3892/mmr.2015.4031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 03/26/2015] [Indexed: 11/06/2022] Open
Abstract
The present study investigated the role of microRNA (miR)‑138‑5p in regulating carcinoma migration and sensitivity to chemotherapy in pancreatic cancer. Reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) was used to assess the expression levels of miR‑138‑5p in pancreatic cancer cell lines and primary carcinoma tissues from human patients. A lentiviral vector, containing miR‑138‑5p mimics (lv‑miR‑138‑m) or miR‑138‑5p inhibitor (lv‑miR‑138‑i), was used to either upregulate or downregulate the expression levels of miR‑138‑5p in PANC‑1 cells, respectively. The effects of miR‑138‑3p regulation on pancreatic cancer cell migration and sensitivity to chemotherapy were examined. The predicted targeting of miR‑138‑5p on vimentin (VIM) was assessed by western blotting in PANC‑1 cells. VIM was subsequently downregulated using small interfering (si)RNA to determine its effect on miR‑138‑5p‑modulated pancreatic cancer cell development. The expression levels of miR‑138‑5p were downregulated in pancreatic cancer cell lines and primary carcinoma tissues. In PANC‑1 cells, lentivirus-mediated upregulation of miR‑138‑5p inhibited cancer cell migration and increased cell chemosensitivity to 5‑fluorouracil (5‑FU). By contrast, downregulation of miR‑138‑5p promoted cancer cell migration and decreased cell chemosensitivity to 5‑FU. A luciferase assay revealed that VIM was a direct target of miR‑138‑5p. Western blotting demonstrated that VIM was downregulated upon the upregulation of miR‑138‑5p in PANC‑1 cells. siRNA‑mediated downregulation of VIM inhibited pancreatic cancer cell migration in the control and miR‑138‑5p downregulated PANC‑1 cells. The present study demonstrated that miR‑138‑5p is important in regulating pancreatic cancer development, possibly through targeting VIM.
Collapse
Affiliation(s)
- Chao Yu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou 550004, P.R. China
| | - Min Wang
- Department of Biliary‑Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Meiyuan Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou 550004, P.R. China
| | - Yang Huang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou 550004, P.R. China
| | - Jianxin Jiang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
23
|
Yu C, Wang M, Li Z, Xiao J, Peng F, Guo X, Deng Y, Jiang J, Sun C. MicroRNA-138-5p regulates pancreatic cancer cell growth through targeting FOXC1. Cell Oncol (Dordr) 2015; 38:173-81. [PMID: 25875420 PMCID: PMC4445488 DOI: 10.1007/s13402-014-0200-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2014] [Indexed: 12/17/2022] Open
Abstract
PURPOSE The prognosis of pancreatic cancer ranks among the worst of all cancer types, which is primarily due to the fact that during the past decades little progress has been made in its diagnosis and treatment. Here, we set out to investigate the role of microRNA 138 (miR-138-5p) in the regulation of pancreatic cancer cell growth and to assess its role as putative therapeutic target. METHODS qRT-PCR was used to examine the expression of miR-138-5p in 8 pancreatic cancer cell lines and 18 primary human pancreatic cancer samples. A lentivirual vector containing miR-138-5p mimics (lv-miR-138-5p) was used to exogenously over-express miR-138-5p in the pancreatic cancer cells lines Capan-2 and PANC-1. The effect of this over-expression on cell proliferation was examined using an in vitro propidium iodide fluorescence assay. Capan-2 cells exogenously over-expressing miR-138-5p were transplanted into nude mice to examine its in vivo effect on tumor growth. A predicted target of miR-138-5p (FOXC1) was first validated using a luciferase assay and, subsequently, down-regulated by siRNA to assess its effect on pancreatic cancer cell growth. RESULTS We found that miR-138-5p was markedly down-regulated in both pancreatic cancer cell lines and primary human pancreatic cancer samples, compared to a human pancreas ductal epithelial (HPDE) cell line and normal pancreatic tissues, respectively (P < 0.05). In addition, we found that in the pancreatic cancer cells lines Capan-2 and PANC-1 lentiviral transfection of miR-138-5p mimicked up-regulation of the endogenous expression of miR-138-5p and, concomitantly, inhibited cancer cell proliferation (P < 0.05). The exogenous over-expression of miR-138-5p also led to a significant inhibition of tumor formation in vivo. Using a luciferase assay, we found that miR-138-5p directly targets FOXC1. In conformity with this notion, we found that FOXC1 was down-regulated upon miR-138-5p over-expression in pancreatic cancer cells. Finally, we found that silencing of FOXC1 by siRNA had an inhibitory effect on pancreatic cancer cell growth. CONCLUSIONS Our data indicate that miR-138-5p may play an important role in regulating pancreatic cancer cell growth, possibly through targeting FOXC1. Over-expression of miR-138-5p may serve as a novel approach for the treatment of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Chao Yu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou Province 550004 China
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Hazhong University of Science and Technology, Wuhan, Hubei Province 430074 China
| | - Zhipeng Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou Province 550004 China
| | - Jie Xiao
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou Province 550004 China
| | - Feng Peng
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Hazhong University of Science and Technology, Wuhan, Hubei Province 430074 China
| | - Xingjun Guo
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Hazhong University of Science and Technology, Wuhan, Hubei Province 430074 China
| | - Yazhu Deng
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou Province 550004 China
| | - Jianxin Jiang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou Province 550004 China
| | - Chengyi Sun
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou Province 550004 China
| |
Collapse
|
24
|
Abbad S, Wang C, Waddad AY, Lv H, Zhou J. Preparation, in vitro and in vivo evaluation of polymeric nanoparticles based on hyaluronic acid-poly(butyl cyanoacrylate) and D-alpha-tocopheryl polyethylene glycol 1000 succinate for tumor-targeted delivery of morin hydrate. Int J Nanomedicine 2015; 10:305-20. [PMID: 25609946 PMCID: PMC4293365 DOI: 10.2147/ijn.s73971] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Herein, we describe the preparation of a targeted cellular delivery system for morin hydrate (MH), based on a low-molecular-weight hyaluronic acid-poly(butyl cyanoacrylate) (HA-PBCA) block copolymer. In order to enhance the therapeutic effect of MH, D-alpha-tocopheryl polyethylene glycol 1000 succinate (TPGS) was mixed with HA-PBCA during the preparation process. The MH-loaded HA-PBCA “plain” nanoparticle (MH-PNs) and HA-PBCA/TPGS “mixed” nanoparticles (MH-MNs) were concomitantly characterized in terms of loading efficiency, particle size, zeta potential, critical aggregation concentration, and morphology. The obtained MH-PNs and MH-MNs exhibited a spherical morphology with a negative zeta potential and a particle size less than 200 nm, favorable for drug targeting. Remarkably, the addition of TPGS resulted in about 1.6-fold increase in drug-loading. The in vitro cell viability experiment revealed that MH-MNs enhanced the cytotoxicity of MH in A549 cells compared with MH solution and MH-PNs. Furthermore, blank MNs containing TPGS exhibited selective cytotoxic effects against cancer cells without diminishing the viability of normal cells. In addition, the cellular uptake study indicated that MNs resulted in 2.28-fold higher cellular uptake than that of PNs, in A549 cells. The CD44 receptor competitive inhibition and the internalization pathway studies suggested that the internalization mechanism of the nanoparticles was mediated mainly by the CD44 receptors through a clathrin-dependent endocytic pathway. More importantly, MH-MNs exhibited a higher in vivo antitumor potency and induced more tumor cell apoptosis than did MH-PNs, following intravenous administration to S180 tumor-bearing mice. Overall, the results imply that the developed nanoparticles are promising vehicles for the targeted delivery of lipophilic anticancer drugs.
Collapse
Affiliation(s)
- Sarra Abbad
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People's Republic of China ; Department of Pharmacy, Abou Bekr Belkaid University, Tlemcen, Algeria
| | - Cheng Wang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Ayman Yahia Waddad
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Huixia Lv
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Jianping Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
25
|
Nagayoshi K, Ueki T, Tashiro K, Mizuuchi Y, Manabe T, Araki H, Oda Y, Kuhara S, Tanaka M. Galanin plays an important role in cancer invasiveness and is associated with poor prognosis in stage II colorectal cancer. Oncol Rep 2014; 33:539-46. [PMID: 25504183 PMCID: PMC4306273 DOI: 10.3892/or.2014.3660] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/20/2014] [Indexed: 12/13/2022] Open
Abstract
Reliable predictors of tumor recurrence for patients with stage II colorectal cancer (CRC) are needed to select patients who should receive adjuvant chemotherapy. Although galanin (GAL) is expressed in several malignant tumors and is associated with cell proliferation and tumor growth, the prognostic value of GAL expression in CRC is poorly understood. We compared GAL expression between 56 patients with stage II and III CRC who developed tumor recurrences and 56 patients who did not. The clinical and prognostic significance of GAL expression was examined using our data and independent public datasets. We also analyzed the influence of GAL expression on the proliferation and invasive activity of CRC cells. Higher expression of GAL was associated with tumor recurrence among the CRC patients (P<0.001). Stage II CRC patients who presented with high expression levels of GAL had significantly poorer prognosis than those with low expression levels of GAL [5-year overall survival: hazard ratio (HR), 7.31; 95% confidence interval (CI), 2.38–24.04; P<0.001; 5-year recurrence-free survival: HR, 3.99; 95% CI, 1.61–9.44; P=0.004], but there was no association between GAL expression and survival in stage III CRC patients. These findings were supported by analysis of two public datasets. Functionally, siRNA-mediated silencing of GAL resulted in a significant decrease in the proliferative and invasive activities of CRC cells. In conclusion, high expression of GAL is associated with poor prognosis of stage II CRC patients and GAL expression may be related to the aggressive behavior of CRC.
Collapse
Affiliation(s)
- Kinuko Nagayoshi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Higashi‑ku, Fukuoka 812-8582, Japan
| | - Takashi Ueki
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Higashi‑ku, Fukuoka 812-8582, Japan
| | - Kosuke Tashiro
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| | - Yusuke Mizuuchi
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Tatsuya Manabe
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Higashi‑ku, Fukuoka 812-8582, Japan
| | - Hiromitsu Araki
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | - Satoru Kuhara
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Higashi-ku, Fukuoka 812-8581, Japan
| | - Masao Tanaka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Higashi‑ku, Fukuoka 812-8582, Japan
| |
Collapse
|
26
|
Nakata K, Ohuchida K, Mizumoto K, Aishima S, Oda Y, Nagai E, Tanaka M. Micro RNA-373 is Down-regulated in Pancreatic Cancer and Inhibits Cancer Cell Invasion. Ann Surg Oncol 2014; 21:564-574. [DOI: 10.1245/s10434-014-3676-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
27
|
Liu A, Shao C, Jin G, Liu R, Hao J, Shao Z, Liu Q, Hu X. Downregulation of CPE regulates cell proliferation and chemosensitivity in pancreatic cancer. Tumour Biol 2014; 35:12459-65. [PMID: 25374060 DOI: 10.1007/s13277-014-2564-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 08/26/2014] [Indexed: 01/03/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most common cancers worldwide and a leading cause of cancer-related death. Discovering novel targets is a key for its therapy. Carboxypeptidase E (CPE), a subtype of the pro-protein convertases, has been shown to be upregulated in many types of cancer, yet its function in PC remains elusive. The expressions of CPE in PC cell lines and cancer patients were investigated by Western blot and qRT-PCR. In PC cell line BX-pc-3, CPE was downregulated and its effect on cancer cell proliferation, migration, cisplatin chemosensitivity, and in vivo tumor growth was analyzed by Western blot, proliferation assay, invasion assay, and in vivo transplantation, respectively. The expression of nuclear factor-kappaB (NF-κB), a possible downstream target of CPE was examined by Western blot upon CPE regulation in PC cells, and the effects of inhibiting NF-κB on PC cell invasion and proliferation were examined. CPE was significantly upregulated in PC cell lines and tumor tissues. Proliferation and invasion assays indicated that downregulation of CPE inhibited cancer cell growth and migration and increased chemosensitivity to cisplatin. Inoculation of small interfering RNA (siRNA) transfected BX-pc-3 cells into null mice demonstrated that downregulation of CPE prevented tumor growth in vivo. NF-κB was directly regulated by CPE in pancreatic cancer, and siRNA-mediated inhibition of NF-κB exerted similar anti-tumor effect as downregulating CPE. Taken together, our results demonstrate that CPE plays an important role in pancreatic cancer. Inhibition of CPE may serve as a potential target for PC therapeutics.
Collapse
Affiliation(s)
- Anan Liu
- Department of Pancreatic Surgery, Changhai Hospital of Second Military Medical University, Shanghai, 200433, China
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Li Z, Li X, Yu C, Wang M, Peng F, Xiao J, Tian R, Jiang J, Sun C. MicroRNA-100 regulates pancreatic cancer cells growth and sensitivity to chemotherapy through targeting FGFR3. Tumour Biol 2014; 35:11751-9. [PMID: 25344675 DOI: 10.1007/s13277-014-2271-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 06/20/2014] [Indexed: 12/30/2022] Open
Abstract
We intended to investigate the role of microRNA 100 (miR-100) in regulating pancreatic cancer cells' growth in vitro and tumor development in vivo. QTR-PCR was used to examine the expression of miR-100 in pancreatic cancer cell lines and tumor cells from human patients. Lentivirual vector containing miR-100 mimics (lv-miR-100) was used to overexpress miR-100 in MIA PaCa-2 and FCPAC-1 cells. The effects of overexpressing miR-100 on pancreatic cancer cell proliferation and chemosensitivity to cisplatin were examined by cell proliferation essay in vitro. MIA PaCa-2 cells with endogenously overexpressed miR-100 were transplanted into null mice to examine tumor growth in vivo. The predicted target of miR-100, fibroblast growth factor receptor 3 (FGFR3), was downregulated by siRNA to examine its effect on pancreatic cancer cells. We found miR-100 was markedly underexpressed in both pancreatic cancer cell lines and tumor cells from patients. In cancer cells, transfection of lv-miR-100 was able to upregulate endogenous expression of miR-100, inhibited cancer cell proliferation, and increased sensitivities to cisplatin. Overexpressing miR-100 led to significant inhibition on tumor formation in vivo. Luciferase essay showed FGFR3 was direct target of miR-100. FGFR3 was significantly downregulated by overexpressing miR-100 in pancreatic cancer cells and knocking down FGFR3 by siRNA exerted similar effect as miR-100. Our study demonstrated that miR-100 played an important role in pancreatic cancer development, possibly through targeting FGFR3. It may become a new therapeutic target for gene therapy in patients suffered from pancreatic cancer.
Collapse
Affiliation(s)
- Zhipeng Li
- Department of Biliary-Hepatic Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou Province, 50004, China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Li Z, Li X, Yu C, Wang M, Peng F, Xiao J, Tian R, Jiang J, Sun C. MicroRNA-100 regulates pancreatic cancer cells growth and sensitivity to chemotherapy through targeting FGFR3. Tumour Biol 2014. [PMID: 25344675 DOI: 10.1007/s13277- 014-2271-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
We intended to investigate the role of microRNA 100 (miR-100) in regulating pancreatic cancer cells' growth in vitro and tumor development in vivo. QTR-PCR was used to examine the expression of miR-100 in pancreatic cancer cell lines and tumor cells from human patients. Lentivirual vector containing miR-100 mimics (lv-miR-100) was used to overexpress miR-100 in MIA PaCa-2 and FCPAC-1 cells. The effects of overexpressing miR-100 on pancreatic cancer cell proliferation and chemosensitivity to cisplatin were examined by cell proliferation essay in vitro. MIA PaCa-2 cells with endogenously overexpressed miR-100 were transplanted into null mice to examine tumor growth in vivo. The predicted target of miR-100, fibroblast growth factor receptor 3 (FGFR3), was downregulated by siRNA to examine its effect on pancreatic cancer cells. We found miR-100 was markedly underexpressed in both pancreatic cancer cell lines and tumor cells from patients. In cancer cells, transfection of lv-miR-100 was able to upregulate endogenous expression of miR-100, inhibited cancer cell proliferation, and increased sensitivities to cisplatin. Overexpressing miR-100 led to significant inhibition on tumor formation in vivo. Luciferase essay showed FGFR3 was direct target of miR-100. FGFR3 was significantly downregulated by overexpressing miR-100 in pancreatic cancer cells and knocking down FGFR3 by siRNA exerted similar effect as miR-100. Our study demonstrated that miR-100 played an important role in pancreatic cancer development, possibly through targeting FGFR3. It may become a new therapeutic target for gene therapy in patients suffered from pancreatic cancer.
Collapse
Affiliation(s)
- Zhipeng Li
- Department of Biliary-Hepatic Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou Province, 50004, China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Fujiwara K, Ohuchida K, Sada M, Horioka K, Ulrich CD, Shindo K, Ohtsuka T, Takahata S, Mizumoto K, Oda Y, Tanaka M. CD166/ALCAM expression is characteristic of tumorigenicity and invasive and migratory activities of pancreatic cancer cells. PLoS One 2014; 9:e107247. [PMID: 25221999 PMCID: PMC4164537 DOI: 10.1371/journal.pone.0107247] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 08/08/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND CD166, also known as activated leukocyte cell adhesion molecule (ALCAM), is expressed by various cells in several tissues including cancer. However, the role of CD166 in malignant tumors is controversial, especially in pancreatic cancer. This study aimed to clarify the role and significance of CD166 expression in pancreatic cancer. METHODS We performed immunohistochemistry and flow cytometry to analyze the expression of CD166 in surgical pancreatic tissues and pancreatic cancer cell lines. The differences between isolated CD166+ and CD166- pancreatic cancer cells were analyzed by invasion and migration assays, and in mouse xenograft models. We also performed quantitative RT-PCR and microarray analyses to evaluate the expression levels of CD166 and related genes in cultured cells. RESULTS Immunohistochemistry revealed high expression of CD166 in pancreatic cancer tissues (12.2%; 12/98) compared with that in normal pancreas controls (0%; 0/17) (p = 0.0435). Flow cytometry indicated that CD166 was expressed in 33.8-70.2% of cells in surgical pancreatic tissues and 0-99.5% of pancreatic cancer cell lines. Invasion and migration assays demonstrated that CD166- pancreatic cancer cells showed stronger invasive and migratory activities than those of CD166+ cancer cells (p<0.05). On the other hand, CD166+ Panc-1 cells showed a significantly stronger colony formation activity than that of CD166- Panc-1 cells (p<0.05). In vivo analysis revealed that CD166+ cells elicited significantly greater tumor growth than that of CD166- cells (p<0.05) in both subcutaneous and orthotopic mouse tumor models. mRNA expression of the epithelial-mesenchymal transition activator Zeb1 was over-expressed in CD166- cells (p<0.001). Microarray analysis showed that TSPAN8 and BST2 were over-expressed in CD166+ cells, while BMP7 and Col6A1 were over-expressed in CD166- cells. CONCLUSIONS CD166+ pancreatic cancer cells are strongly tumorigenic, while CD166- pancreatic cancer cells exhibit comparatively stronger invasive and migratory activities. These findings suggest that CD166 expression is related to different functions in pancreatic cancer cells.
Collapse
Affiliation(s)
- Kenji Fujiwara
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Sada
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohei Horioka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Charles D. Ulrich
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koji Shindo
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Research Fellow of the Japan Society for the Promotion of Science, Tokyo, Japan
| | - Takao Ohtsuka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shunichi Takahata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuhiro Mizumoto
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Kyushu University Hospital Cancer Center, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masao Tanaka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
31
|
Synthesis, in vitro aerobic and hypoxic cytotoxicity and radiosensitizing activity of novel metronidazole tethered 5-fluorouracil. ACTA ACUST UNITED AC 2013; 21:76. [PMID: 24359860 PMCID: PMC3914007 DOI: 10.1186/2008-2231-21-76] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 09/13/2013] [Indexed: 11/10/2022]
Abstract
Background and the purpose of the study Several 2, 4-dinitrophenyl and 2,4-dinitrophenylamine tethered 5-FU (5-fluorouracil) compared to their components have shown minimal or no cytotoxicity to HT-29 cell line under aerobic conditions but high cytotoxicity and radiosensitizing effects under hypoxic conditions. In the present study the cytotoxicity and radiation potentiation of three novel analogues of these compounds by replacing 2,4-dinitrophenyl moiety with 2-methyl-5-nitroimidazole, a known radiosensitizer and cytotoxic agent was investigated. Methods Tethered compounds 7–9 were prepared by the reaction of 1-(t-butoxycarbonyl)-5-fluorouracil 6 with metronidazole esters 2–4 followed by removal of the t-butoxycarbonyl protecting group. Cytotoxicity of compounds in HT-29 cells with or without radiation were determined by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), propidium iodide (PI)-digitonin and clonogenic assays. Results Tethered compounds 7–9 induced time-and concentration–dependent cytotoxicity under hypoxia but had no significant effect under aerobic conditions. These compounds also showed selective and concentration- dependent radiosensitization effects under hypoxic conditions. Conclusion Tethered compounds 7–9 compared to 5-FU 5 showed minimal cytotoxicities under aerobic and selective radiosensitizing activities under hypoxic conditions. Also effects of these compounds were higher than those of metronidazole 1 which is a known cytotoxin and radiosensitizer under hypoxic conditions.
Collapse
|
32
|
Abstract
OBJECTIVES CD105 expression correlates with prognosis for several cancers. However, its significance in pancreatic cancer is unclear. METHODS We analyzed CD105 expression in resected pancreatic cancer tissue and pancreatic cancer cell lines, compared the properties of CD105(+) and CD105(-) cells using quantitative RT-PCR and migration assays, and evaluated the relationship between CD105(+) cells and pancreatic stellate cells (PSCs). RESULTS Immunohistochemistry showed that the frequency of CD105 expression was higher in pancreatic cancer than that in normal tissue(8% vs 0%, respectively). In flow cytometry, CD105 was expressed in pancreatic cancer cells, whereas weak CD105 expression was detected in normal pancreatic ductal epithelial cells. Quantitative RT-PCR showed that E-cadherin mRNA expression was suppressed and vimentin mRNA was overexpressed in CD105(+) cells (P < 0.05). Migration of CD105(+) cancer cells was strongly enhanced (more than that of CD105(+) cells) in coculture with PSCs (P < 0.05). CD105 expression did not correlate to clinicopathologic characteristics or the Kaplan-Meier survival analysis. CONCLUSIONS Suppression of an epithelial marker and over expression of a mesenchymal marker suggest that epithelial-mesenchymal transition is induced in CD105(+) pancreatic cancer cells. CD105(+) pancreatic cancer cell migration is strongly enhanced by PSCs, suggesting that these cells play a role in the pancreatic cancer microenvironment.
Collapse
|
33
|
Khalaj A, Abdi K, Ostad SN, Khoshayand MR, Lamei N, Nedaie HA. Synthesis,In VitroCytotoxicity and Radiosensitizing Activity of Novel 3-[(2,4-Dinitrophenylamino)Alkyl] Derivatives of 5-Fluorouracil. Chem Biol Drug Des 2013; 83:183-90. [DOI: 10.1111/cbdd.12211] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 08/10/2013] [Accepted: 08/12/2013] [Indexed: 11/30/2022]
Affiliation(s)
- Ali Khalaj
- Department of Medicinal Chemistry; Faculty of Pharmacy; Tehran University of Medical Sciences; Tehran Iran
- Drug Design & Development Research Center; Tehran University of Medical Sciences; Tehran Iran
| | - Khosrou Abdi
- Department of Medicinal Chemistry; Faculty of Pharmacy; Tehran University of Medical Sciences; Tehran Iran
- Department of Radiopharmacy; Faculty of Pharmacy; Tehran University of Medical Sciences; Tehran Iran
| | - Seyed Nasser Ostad
- Department of Toxicology and Pharmacology; Faculty of Pharmacy; Tehran University of Medical Sciences; Tehran Iran
| | - Mohammad Reza Khoshayand
- Department of Drug and Food Control; Faculty of Pharmacy; Tehran University of Medical Sciences; Tehran Iran
| | - Navid Lamei
- Drug Design & Development Research Center; Tehran University of Medical Sciences; Tehran Iran
| | - Hasan Ali Nedaie
- Department of Radiotherapy; Oncology Cancer Institute; Tehran University of Medical Sciences; Tehran Iran
| |
Collapse
|
34
|
Kozono S, Ohuchida K, Ohtsuka T, Cui L, Eguchi D, Fujiwara K, Zhao M, Mizumoto K, Tanaka M. S100A4 mRNA expression level is a predictor of radioresistance of pancreatic cancer cells. Oncol Rep 2013; 30:1601-8. [PMID: 23900547 DOI: 10.3892/or.2013.2636] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 04/23/2013] [Indexed: 11/06/2022] Open
Abstract
Improving poor outcomes in patients with pancreatic cancer requires a greater understanding of the biological mechanisms contributing to radioresistance. We, therefore, sought to identify genes involved in the radioresistance of pancreatic cancer cells. Two pancreatic cancer cell lines, CFPAC-1 and Capan-1, were repeatedly exposed to radiation, establishing two radioresistant cell lines. Gene expression profiling using cDNA microarrays was performed to identify genes responsible for radioresistance. The levels of expression of mRNAs encoded by selected genes and their correlation with radiation dose resulting in 50% survival rate were analyzed in pancreatic cancer cell lines. The radiation dose resulting in a 50% survival rate was significantly higher in irradiated (IR) compared to parental CFPAC-1 cells (8.31 ± 0.85 Gy vs. 2.14 ± 0.04 Gy, P<0.0001), but was lower in IR compared with parental Capan-1 cells (2.66 ± 0.24 Gy vs. 2.25 ± 0.03 Gy, P=0.04). cDNA microarray analysis identified 4 genes, including S100 calcium binding protein A4 (S100A4), overexpressed and 23 genes underexpressed in the IR compared with the parental cell lines. The levels of S100A4 mRNA expression were correlated with radiation dose resulting in a 50% survival rate (Pearson's test, R2=0.81, P=0.0025). S100A4 mRNA expression may predict radioresistance of pancreatic cancer cells and may play an important role in the poor response of pancreatic cancer cells to radiation therapy.
Collapse
Affiliation(s)
- Shingo Kozono
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kono H, Nakamura M, Ohtsuka T, Nagayoshi Y, Mori Y, Takahata S, Aishima S, Tanaka M. High expression of microRNA-155 is associated with the aggressive malignant behavior of gallbladder carcinoma. Oncol Rep 2013; 30:17-24. [PMID: 23660842 PMCID: PMC3729208 DOI: 10.3892/or.2013.2443] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 03/12/2013] [Indexed: 12/11/2022] Open
Abstract
The prognosis of gallbladder cancer (GBC) remains poor despite recent advances in diagnostics and therapeutic strategies. Although the role of microRNAs (miRs) in GBC have not been well documented, miR-155 is known to be associated with inflammation-associated carcinogenesis in various types of cancers. The aim of this study was to investigate the clinical significance of miR-155 expression and the biological functions of miR-155 in GBC. The expression levels of miR-155 in surgically resected GBCs and gallbladders with pancreaticobiliary maljunction (PBM) were assessed by quantitative reverse transcription-polymerase chain reaction. The relationship between the expression levels of miR-155 and clinicopathological features of GBCs was analyzed. Human GBC cell lines were transfected with miR-155 inhibitors or mimics, and the effects on proliferation and invasion were assessed. miR-155 was significantly overexpressed in GBCs when compared with that in gallbladders with PBM (P=0.007) and normal gallbladders (P=0.04). The high expression level of miR-155 in GBCs was significantly associated with the presence of lymph node metastasis (P=0.01) and a poor prognosis (P=0.02). In vitro assays showed that aberrant expression of miR-155 significantly enhanced GBC cell proliferation and invasion. In conclusion, high miR-155 expression correlates with the aggressive behavior of GBCs, and miR-155 may become a prognostic marker and therapeutic target for GBC.
Collapse
Affiliation(s)
- Hiroshi Kono
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | |
Collapse
|
36
|
MAHAWITHITWONG PRAWEJ, OHUCHIDA KENOKI, IKENAGA NAOKI, FUJITA HAYATO, ZHAO MING, KOZONO SHINGO, SHINDO KOJI, OHTSUKA TAKAO, AISHIMA SHINICHI, MIZUMOTO KAZUHIRO, TANAKA MASAO. Kindlin-1 expression is involved in migration and invasion of pancreatic cancer. Int J Oncol 2013; 42:1360-6. [DOI: 10.3892/ijo.2013.1838] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 05/30/2012] [Indexed: 11/06/2022] Open
|
37
|
Kozono S, Ohuchida K, Eguchi D, Ikenaga N, Fujiwara K, Cui L, Mizumoto K, Tanaka M. Pirfenidone inhibits pancreatic cancer desmoplasia by regulating stellate cells. Cancer Res 2013; 73:2345-56. [PMID: 23348422 DOI: 10.1158/0008-5472.can-12-3180] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pancreatic stellate cells (PSC), which are implicated in desmoplasia in pancreatic cancer, enhance the malignancy of cancer cells and confer resistance to established treatments. We investigated whether the antifibrotic agent pirfenidone can suppress desmoplasia and exert antitumor effects against pancreatic cancer. Primary PSCs were established from pancreatic cancer tissue obtained during surgery. In vitro, pirfenidone inhibited the proliferation, invasiveness, and migration of PSCs in a dose-dependent manner. Although supernatants of untreated PSCs increased the proliferation, invasiveness, and migration of pancreatic cancer cells (PCC), supernatants of pirfenidone-treated PSCs decreased these effects. Exposure to PCC supernatant increased the production of platelet-derived growth factor-A, hepatic growth factor, collagen type I, fibronectin, and periostin in PSCs, which was significantly reduced by pirfenidone. Mice were subcutaneously implanted with PCCs (SUIT-2 cells) and PSCs into the right flank and PCCs alone into the left flank. Oral administration of pirfenidone to these mice significantly reduced tumor growth of co-implanted PCCs and PSCs, but not of PCCs alone. Pirfenidone also decreased the proliferation of PSCs and the deposition of collagen type I and periostin in tumors. In mice with orthotopic tumors consisting of PCCs co-implanted with PSCs, pirfenidone suppressed tumor growth, reduced the number of peritoneal disseminated nodules, and reduced the incidence of liver metastasis. Pirfenidone in combination with gemcitabine more effectively suppressed orthotopic tumor growth compared with pirfenidone or gemcitabine alone. In conclusion, our findings indicate that pirfenidone is a promising antitumor agent for pancreatic cancer, owing to its suppression of desmoplasia through regulating PSCs.
Collapse
Affiliation(s)
- Shingo Kozono
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abdi K, Khalaj A, Ostad SN, Khoshayand MR. Cytotoxicity and radiosensitising activity of synthesized dinitrophenyl derivatives of 5-fluorouracil. ACTA ACUST UNITED AC 2012; 20:3. [PMID: 23226112 PMCID: PMC3514535 DOI: 10.1186/1560-8115-20-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 07/19/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND THE PURPOSE OF THE STUDY Dual functional agents in which nitroaromatic or nitroheterocyclic compounds are attached through a linker unit to mustards and aziridines have shown higher cytotoxicities than the corresponding counterparts to both aerobic and hypoxic cells and enhanced radiosensitizing activity. In the present investigation cytotoxicity and radiosensitizing activity of 2,4-dinitrobenzyl, 2,4-dinitrobenzoyl, and 2,4-dinitrophenacetyl derivatives of 5-fluorouracil which was assumed to release cytotoxic active quinone methidide and 5-fluorouracil under hypoxic conditions on HT-29 cell line under both aerobic and hypoxic conditions was investigated. METHODS 5-fluorouracil derivative X-XIII were prepared by the reaction of the corresponding di-nitro substituted benzyl, benzoyl and phenacetyl halides with 5-fluorouracil protected at N-1 with di-t-butoxydicarbonate (BOC) in dimethyl formamide (DMF) in the presence of the potassium carbonate followed by hydrolysis of the blocking group by potassium carbonate in methanol. Cytotoxicity of fluorouracil VIII and tested compounds X-XIII against HT-29 cell line under both aerobic and hypoxic conditions after 48 hrs incubation were measured by determination of the percent of the survival cells using 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and percent of the dead cells using propidium iodide(PI)-digitonine assay and results were used to calculate the corresponding IC(50) values. Radiosensitization experiments were carried out by irradiation of the incubations with a (60)Co source and clonogenic assay was performed to determine the cell viabilities following treatment with the tested compounds and/or radiation. Sensitization Enhancement Ratio (SER) of each tested compound was obtained from the radiation survival curves in the absence and presence of each sensitizer for 37% survival respectively. RESULTS AND MAJOR CONCLUSION Findings of the present study showed that alkylation or acylation of 5-fluorouracil result in compounds which have little or no cytotoxicity and radiosensitizing activity under aerobic conditions, but have high cytotoxicity and radiosensitizing effects under hypoxic conditions. Furthermore radiosensitizing activities of compounds under hypoxic conditions increased by increase in their concentrations and SER of the tested 5-FU derivatives at concentrations higher than 50 μmol were equal or higher than 1.6 which is the minimum effective SER of a radiosensitizer in an in vitro assay.
Collapse
Affiliation(s)
- Khosrou Abdi
- Department of Medicinal Chemistry, Tehran, Iran ; Department of Nuclear Pharmacy, Tehran, Iran
| | | | | | | |
Collapse
|
39
|
Folate Conjugated Chitosan Grafted Thiazole Orange Derivative with High Targeting for Early Breast Cancer Cells Diagnosis. J Fluoresc 2012; 22:1555-61. [DOI: 10.1007/s10895-012-1094-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 06/20/2012] [Indexed: 10/28/2022]
|
40
|
Ohuchida K, Mizumoto K, Lin C, Yamaguchi H, Ohtsuka T, Sato N, Toma H, Nakamura M, Nagai E, Hashizume M, Tanaka M. MicroRNA-10a is overexpressed in human pancreatic cancer and involved in its invasiveness partially via suppression of the HOXA1 gene. Ann Surg Oncol 2012; 19:2394-402. [PMID: 22407312 DOI: 10.1245/s10434-012-2252-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Indexed: 12/15/2022]
Abstract
BACKGROUND There is increasing evidence that microRNAs are differentially expressed in many types of cancers. Despite progress in analyses of microRNAs in several types of cancers, the functional contributions of microRNAs to pancreatic cancer remain unclear. METHODS In the present study, the expression levels of specific microRNAs identified by microarray analyses were examined in a panel of 15 pancreatic cancer cell lines. We then investigated the functional roles of these microRNAs in the proliferation and invasion of pancreatic cancer cells. RESULTS Based on the microarray data, we found frequent and marked overexpression of miR-10a, miR-92, and miR-17-5p in pancreatic cancer cell lines. Microdissection analyses revealed that miR-10a was overexpressed in pancreatic cancer cells isolated from a subset of primary tumors (12 of 20, 60%) compared with precursor lesions and normal ducts (P<.01). In vitro experiments revealed that miR-10a inhibitors decreased the invasiveness of pancreatic cancer cells (P<.01), but had no effect on their proliferation. Inhibition of HOXA1, a target of miR-10a, promoted the invasiveness of pancreatic cancer cells (P<.01). CONCLUSIONS The present data suggest that miR-10a is overexpressed in a subset of pancreatic cancers and is involved in the invasive potential of pancreatic cancer cells partially via suppression of HOXA1.
Collapse
Affiliation(s)
- Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Zhao M, Tominaga Y, Ohuchida K, Mizumoto K, Cui L, Kozono S, Fujita H, Maeyama R, Toma H, Tanaka M. Significance of combination therapy of zoledronic acid and gemcitabine on pancreatic cancer. Cancer Sci 2012; 103:58-66. [PMID: 21954965 PMCID: PMC11164147 DOI: 10.1111/j.1349-7006.2011.02113.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
In the present study, we examined the cytotoxic effects of combination therapy with zoledronic acid (ZOL) and gemcitabine (GEM) on pancreatic cancer cells in vitro and in vivo. Four human pancreatic cancer cell lines were treated with ZOL, GEM or a combination of both, and the effects of the respective drug regimens on cell proliferation, invasion and matrix metalloproteinase (MMP) expression were examined. A pancreatic cancer cell line was also intrasplenically or orthotopically implanted into athymic mice and the effects of these drugs on tumor metastasis and growth in vivo were evaluated by histological and immunohistochemical analyses. Combination treatment with low doses of ZOL and GEM efficiently inhibited the proliferation (P < 0.001) and invasion (P < 0.001) of pancreatic cancer cells in vitro. Western blotting assay revealed that MMP-2 and MMP-9 expression levels were decreased after ZOL treatment. In vivo, combined treatment significantly inhibited tumor growth (P < 0.05) and the development of liver metastasis (P < 0.05). These data revealed that ZOL and GEM, when used in combination, have significant antitumor, anti-metastatic and anti-angiogenic effects on pancreatic cancer cells. The present study is the first to report the significance of the combination treatment of ZOL and GEM in pancreatic cancer using an in vivo model. These data are promising for the future application of this drug regimen in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Sakai H, Ohuchida K, Mizumoto K, Cui L, Nakata K, Toma H, Nagai E, Tanaka M. Inhibition of p600 expression suppresses both invasiveness and anoikis resistance of gastric cancer. Ann Surg Oncol 2011; 18:2057-65. [PMID: 21347795 PMCID: PMC3115059 DOI: 10.1245/s10434-010-1523-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Indexed: 12/22/2022]
Abstract
BACKGROUND Advanced gastric cancers often metastasize to distant organs and the peritoneum, leading to a poor prognosis. Both invasiveness and resistance to anchorage-independent cell death (anoikis) are important factors in the process of metastasis. p600 (600-kDa protein), recently identified from a cervical cancer cell line, plays a role in both anoikis resistance and cell migration. In this study, we examined whether p600 is involved in the progression of gastric cancer. METHODS We used both normal gastric mucosal cells and cancer cells laser-microdissected from 42 gastric cancers and their normal counterparts, and compared their p600 mRNA expression levels with quantitative reverse transcriptase-polymerase chain reaction. We inhibited p600 expression in two gastric cancer cell lines with siRNA and examined its effect on the invasiveness and anoikis resistance both in vitro and in vivo. RESULTS Expression of p600 mRNA was significantly higher in gastric cancer cells than in normal mucosal cells (P = 0.027). The invasion assay revealed that invasiveness was significantly reduced by inhibition of p600 (P < 0.01). In vitro experiments revealed that cell viability and colony-formation capacity under anchorage-independent conditions were significantly reduced by inhibition of p600 (P < 0.05). In vivo experiments also showed that the establishment of intraperitoneal disseminated tumors was significantly suppressed by transient inhibition of p600 (P < 0.001). CONCLUSIONS Our results strongly suggest that p600 is involved in gastric cancer progression, and has a potential to be a new molecular target for gastric cancer therapy.
Collapse
Affiliation(s)
- Hiroshi Sakai
- Department of Surgery and Oncology, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Gene expression levels as predictive markers of outcome in pancreatic cancer after gemcitabine-based adjuvant chemotherapy. Neoplasia 2011; 12:807-17. [PMID: 20927319 DOI: 10.1593/neo.10458] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Revised: 06/09/2010] [Accepted: 06/15/2010] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND AIMS The standard palliative chemotherapy for pancreatic ductal adenocarcinoma (PDAC) is gemcitabine-based chemotherapy; however, PDAC still presents a major therapeutic challenge. The aims of this study were to investigate the expression pattern of genes involved in gemcitabine sensitivity in resected PDAC tissues and to determine correlations of gene expression with treatment outcome. MATERIALS AND METHODS We obtained formalin-fixed paraffin-embedded (FFPE) tissue samples from 70 patients with PDAC. Of the 70 patients, 40 received gemcitabine-based adjuvant chemotherapy (AC). We measured hENT1, dCK, CDA, RRM1, and RRM2 messenger RNA (mRNA) levels by quantitative real-time reverse transcription-polymerase chain reaction and determined the combined score (GEM score), based on the expression levels of hENT1, dCK, RRM1, and RRM2, to investigate the association with survival time. By determining the expression levels of these genes in endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) cytologic specimens, we investigated the feasibility of individualized chemotherapy. RESULTS High dCK (P = .0067), low RRM2 (P = .003), and high GEM score (P = .0003) groups had a significantly longer disease-free survival in the gemcitabine-treated group. A low GEM score (<2) was an independent predictive marker for poor outcome to gemcitabine-based AC as shown by multivariate analysis (P = .0081). Altered expression levels of these genes were distinguishable in microdissected neoplastic cells from EUS-FNA cytologic specimens. CONCLUSIONS Quantitative analyses of hENT1, dCK, RRM1, and RRM2 mRNA levels using FFPE tissue samples and microdissected neoplastic cells from EUS-FNA cytologic specimens may be useful in predicting the gemcitabine sensitivity of patients with PDAC.
Collapse
|
44
|
Gene expression levels as predictive markers of outcome in pancreatic cancer after gemcitabine-based adjuvant chemotherapy. Neoplasia 2010. [PMID: 20927319 DOI: 10.1593/neo.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND AND AIMS The standard palliative chemotherapy for pancreatic ductal adenocarcinoma (PDAC) is gemcitabine-based chemotherapy; however, PDAC still presents a major therapeutic challenge. The aims of this study were to investigate the expression pattern of genes involved in gemcitabine sensitivity in resected PDAC tissues and to determine correlations of gene expression with treatment outcome. MATERIALS AND METHODS We obtained formalin-fixed paraffin-embedded (FFPE) tissue samples from 70 patients with PDAC. Of the 70 patients, 40 received gemcitabine-based adjuvant chemotherapy (AC). We measured hENT1, dCK, CDA, RRM1, and RRM2 messenger RNA (mRNA) levels by quantitative real-time reverse transcription-polymerase chain reaction and determined the combined score (GEM score), based on the expression levels of hENT1, dCK, RRM1, and RRM2, to investigate the association with survival time. By determining the expression levels of these genes in endoscopic ultrasound-guided fine needle aspiration (EUS-FNA) cytologic specimens, we investigated the feasibility of individualized chemotherapy. RESULTS High dCK (P = .0067), low RRM2 (P = .003), and high GEM score (P = .0003) groups had a significantly longer disease-free survival in the gemcitabine-treated group. A low GEM score (<2) was an independent predictive marker for poor outcome to gemcitabine-based AC as shown by multivariate analysis (P = .0081). Altered expression levels of these genes were distinguishable in microdissected neoplastic cells from EUS-FNA cytologic specimens. CONCLUSIONS Quantitative analyses of hENT1, dCK, RRM1, and RRM2 mRNA levels using FFPE tissue samples and microdissected neoplastic cells from EUS-FNA cytologic specimens may be useful in predicting the gemcitabine sensitivity of patients with PDAC.
Collapse
|
45
|
Moriyama T, Ohuchida K, Mizumoto K, Cui L, Ikenaga N, Sato N, Tanaka M. Enhanced cell migration and invasion of CD133+ pancreatic cancer cells cocultured with pancreatic stromal cells. Cancer 2010; 116:3357-68. [PMID: 20564084 DOI: 10.1002/cncr.25121] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Recently, cancer stem cells have been reported as a new therapeutic target in pancreatic cancer as well as other cancers, but the specific role of these cells is unknown. METHODS The authors investigated the functional roles of CD133+ cells, 1 of the putative cancer stem cell candidates in pancreatic cancer. CD133 expression was assessed in human pancreatic cancer and cancer cell lines by quantitative real-time reverse transcriptase polymerase chain reaction and flow cytometry. Next, they compared the ability of CD133+ and CD133- cells to proliferate, migrate, and invade using 2 pancreatic cancer cell lines. In particular, they evaluated the relationship between CD133+ cells and primary pancreatic stromal cells. RESULTS CD133 was expressed in primary human pancreatic cancer tissues and some cancer cell lines, whereas there was little expression in primary normal pancreatic epithelial cells and primary pancreatic stromal cells. CD133+ cells, isolated by flow cytometry, showed increased cell proliferation under anchorage-independent conditions (P<.01), and enhanced migration and invasion, particularly when cocultured with primary pancreatic stromal cells (P<.001). Chemokine-related receptor-4 (CXCR4), markedly overexpressed in CD133+ cells, may be responsible for the increased invasive ability of the cells cocultured with pancreatic stromal cells, which express stromal derived factor-1, the ligand to CXCR4. CONCLUSIONS These data suggest that CD133+ cells exhibit more aggressive behavior, such as increased cell proliferation, migration, and invasion, especially in the presence of pancreatic stromal cells. The targeting therapy for the interaction between CD133+ cancer cells and stromal cells may be a new approach for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Taiki Moriyama
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Kyushu University Hospital Cancer Center, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
46
|
Yu J, Ohuchida K, Mizumoto K, Sato N, Kayashima T, Fujita H, Nakata K, Tanaka M. MicroRNA, hsa-miR-200c, is an independent prognostic factor in pancreatic cancer and its upregulation inhibits pancreatic cancer invasion but increases cell proliferation. Mol Cancer 2010; 9:169. [PMID: 20579395 PMCID: PMC2909980 DOI: 10.1186/1476-4598-9-169] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Accepted: 06/28/2010] [Indexed: 01/05/2023] Open
Abstract
Background Recently, the microRNA-200 family was reported to affect cancer biology by regulating epithelial to mesenchymal transition (EMT). Especially, the expression of miR-200c has been shown to be associated with upregulating the expression of E-cadherin, a gene known to be involved in pancreatic cancer behavior. However, the significance of miR-200c in pancreatic cancer is unknown. Methods In the present study, we investigated the relationship between E-cadherin and miR-200c expression in a panel of 14 pancreatic cancer cell lines and in macro-dissected formalin-fixed paraffin-embedded (FFPE) tissue samples obtained from 99 patients who underwent pancreatectomy for pancreatic cancer. We also investigated the effects of miR-200c on the proliferation and invasion of pancreatic cancer cells. Results We found that patients with high levels of miR-200c expression had significantly better survival rates than those with low levels of miR-200c expression. We also found a remarkably strong correlation between the levels of miR-200c and E-cadherin expression. Conclusions These data indicate that miR-200c may play a role in the pancreatic cancer biology and may be a novel marker for the prognosis of pancreatic cancer.
Collapse
Affiliation(s)
- Jun Yu
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Onimaru M, Ohuchida K, Egami T, Mizumoto K, Nagai E, Cui L, Toma H, Matsumoto K, Hashizume M, Tanaka M. Gemcitabine synergistically enhances the effect of adenovirus gene therapy through activation of the CMV promoter in pancreatic cancer cells. Cancer Gene Ther 2010; 17:541-9. [DOI: 10.1038/cgt.2010.9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
48
|
Onimaru M, Ohuchida K, Mizumoto K, Nagai E, Cui L, Toma H, Takayama K, Matsumoto K, Hashizume M, Tanaka M. hTERT-promoter-dependent oncolytic adenovirus enhances the transduction and therapeutic efficacy of replication-defective adenovirus vectors in pancreatic cancer cells. Cancer Sci 2010; 101:735-42. [PMID: 20059477 PMCID: PMC11159899 DOI: 10.1111/j.1349-7006.2009.01445.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Adenovirus-mediated gene therapy shows promise for cancer therapy, but transgene expression of replication-defective adenovirus may be low and transient in clinical settings. Recent reports have shown that the use of a conditionally replication-competent adenovirus (CRAd) enhanced the gene transduction of a replication-defective adenovirus vector. The control of tumor-stromal interactions has also been determined to be important in cancer therapy. In this study, we investigated the effect of the human telomerase reverse transcriptase (hTERT)-CRAd, Ad5/3hTERTE1, which possesses the tumor-specific hTERT promoter with the chimeric fiber 5/3, on the transgene expression and therapeutic efficacy of a replication-defective adenovirus vector expressing NK4 under the control of the CMV promoter, Ad-NK4. In addition, we established a new strategy to target both cancer cells and cancer-stromal interactions. Human pancreatic cancer cells were infected with Ad-NK4 and either Ad5/3hTERTE1 (CRAd-combination group) or Ad5/3hTERTLuc (control-combination group). In the CRAd-combination group, Ad-NK4-delivered transgene expression was increased, leading to an enhanced inhibitory effect on the invasion of cancer cells. In in vivo experiments, NK4 expression within tumors and its inhibitory effect on tumor growth, angiogenesis, and metastasis were enhanced in the CRAd-combination group. These results suggest that hTERT-CRAd enhances the transgene expression and therapeutic efficacies of Ad-NK4, possibly through the in-trans replication of Ad-NK4 induced by adenovirus E1 derived from co-infected hTERT-CRAd. This approach may be a promising combination therapy against advanced pancreatic cancer.
Collapse
Affiliation(s)
- Manabu Onimaru
- Department of Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Onimaru M, Ohuchida K, Nagai E, Mizumoto K, Egami T, Cui L, Sato N, Uchino J, Takayama K, Hashizume M, Tanaka M. Combination with low-dose gemcitabine and hTERT-promoter-dependent conditionally replicative adenovirus enhances cytotoxicity through their crosstalk mechanisms in pancreatic cancer. Cancer Lett 2010; 294:178-86. [PMID: 20163915 DOI: 10.1016/j.canlet.2010.01.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Revised: 01/26/2010] [Accepted: 01/26/2010] [Indexed: 12/31/2022]
Abstract
To overcome the limited clinical efficacy of conditionally replicative adenoviruses (CRAds), we investigated the effects of combination therapy with gemcitabine (GEM) and the hTERT-promoter-dependent CRAd (hTERT-CRAd), Ad5/3hTERTE1. This combination therapy exhibited enhanced cytotoxic effects on pancreatic cancer both in vitro and in vivo. Furthermore, we revealed that this enhancement effect was due to the multiple bidirectional interactions between hTERT-CRAd and GEM. The GEM-sensitizing effect of E1 expression derived from hTERT-CRAd, and the enhancement effect by GEM on hTERT promoter activity which led to the increase of adenovirus E1 and viral infectivity. This combination therapy may be a promising therapeutic approach for pancreatic cancer.
Collapse
Affiliation(s)
- Manabu Onimaru
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Fujita H, Ohuchida K, Mizumoto K, Egami T, Miyoshi K, Moriyama T, Cui L, Yu J, Zhao M, Manabe T, Tanaka M. Tumor-stromal interactions with direct cell contacts enhance proliferation of human pancreatic carcinoma cells. Cancer Sci 2009; 100:2309-17. [PMID: 19735487 PMCID: PMC11159841 DOI: 10.1111/j.1349-7006.2009.01317.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is often characterized by an abundant desmoplastic stroma that is partially induced by activated pancreatic stellate cells (PSCs). Indirect co-culture has often been used to investigate the effects of cancer-stromal interactions on the proliferation of cancer cells, but the effects of cell-cell adhesion and juxtacrine signaling between cancer and stromal cells cannot be evaluated using this method. This study aimed to establish a simplified direct co-culture system that could be used to quantify populations of cancer cells in co-culture with PSCs, and to evaluate the effects of direct cell contact on the proliferation of cancer cells. We established three green fluorescent protein (GFP)-expressing pancreatic cancer cell lines and were able to quantify them with high reliability and reproducibility, even when co-cultured directly with PSCs, using a color plate reader. We assessed the differential effects of direct and indirect co-culture with PSCs on the proliferation of cancer cells, and found that the proliferation of GFP-expressing pancreatic cancer cell lines was dramatically enhanced by direct co-culture with PSCs, compared with the indirect co-culture system. We also found that direct co-culture of cancer cells and PSCs activated the Notch signaling pathway in both cell types. Direct cell contact between cancer cells and PSCs plays an important role in the control of cancer cell proliferation, and is essential to the understanding of tumor-stromal interactions.
Collapse
Affiliation(s)
- Hayato Fujita
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|