1
|
Chen Y, Liu X, Zheng JN, Yang LJ, Luo Y, Yao YL, Liu MQ, Xie TT, Lin HF, He YT, Zhou P, Hu B, Tian RJ, Shi ZL. N-linked glycoproteins and host proteases are involved in swine acute diarrhea syndrome coronavirus entry. J Virol 2023; 97:e0091623. [PMID: 37772826 PMCID: PMC10617469 DOI: 10.1128/jvi.00916-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/16/2023] [Indexed: 09/30/2023] Open
Abstract
IMPORTANCE Gaining insight into the cell-entry mechanisms of swine acute diarrhea syndrome coronavirus (SADS-CoV) is critical for investigating potential cross-species infections. Here, we demonstrated that pretreatment of host cells with tunicamycin decreased SADS-CoV attachment efficiency, indicating that N-linked glycosylation of host cells was involved in SADS-CoV entry. Common N-linked sugars Neu5Gc and Neu5Ac did not interact with the SADS-CoV S1 protein, suggesting that these molecules were not involved in SADS-CoV entry. Additionally, various host proteases participated in SADS-CoV entry into diverse cells with different efficiencies. Our findings suggested that SADS-CoV may exploit multiple pathways to enter cells, providing insights into intervention strategies targeting the cell entry of this virus.
Collapse
Affiliation(s)
- Ying Chen
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xi Liu
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiang-Nan Zheng
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen, China
| | - Li-Jun Yang
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen, China
| | - Yun Luo
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yu-Lin Yao
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Mei-Qin Liu
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ting-ting Xie
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hao-Feng Lin
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yan-Tong He
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Peng Zhou
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Ben Hu
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Rui-Jun Tian
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen, China
| | - Zheng-Li Shi
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
2
|
Tuğrul B, Balcan E, Öztel Z, Çöllü F, Gürcü B. Prion protein-dependent regulation of p53-MDM2 crosstalk during endoplasmic reticulum stress and doxorubicin treatments might be essential for cell fate in human breast cancer cell line, MCF-7. Exp Cell Res 2023:113656. [PMID: 37245583 DOI: 10.1016/j.yexcr.2023.113656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/09/2023] [Accepted: 05/21/2023] [Indexed: 05/30/2023]
Abstract
In this study, we investigated the effect of doxorubicin and tunicamycin treatment alone or in combination on MDM-, Cul9-and prion protein (PrP)-mediated subcellular regulation of p53 in the context of apoptosis and autophagy. MTT analysis was performed to determine the cytotoxic effect of the agents. Apoptosis was monitorized by ELISA, flow cytometry and JC-1 assay. Monodansylcadaverine assay was performed for autophagy. Western blotting and immunofluorescence were performed to determine p53, MDM2, CUL9 and PrP levels. Doxorubicin increased p53, MDM2 and CUL9 levels in a dose-dependent manner. Expression of p53 and MDM2 was higher at the 0.25 μM concentration of tunicamycin compared to the control, but it decreased at 0.5 μM and 1 μM concentrations. CUL9 expression was significantly decreased only after treatment of tunicamycin at 0.25 μM. According to its glycosylation status, the upper band of PrP increased only in combination treatment. In combination treatment, p53 expression was higher than control, whereas MDM2 and CUL9 expressions were decreased. Combination treatments may make MCF-7 cells more susceptible to apoptosis rather than autophagy. In conclusion, PrP may be important in determining the fate of cell death through crosstalk between proteins such as p53 and MDM2 under endoplasmic reticulum (ER) stress conditions. Further studies are needed to obtain in-depth information on these potential molecular networks.
Collapse
Affiliation(s)
- Berrin Tuğrul
- Manisa Celal Bayar University, Faculty of Science and Letters, Department of Biology, Molecular Biology Section, 45140, Yunusemre, Manisa, Turkey.
| | - Erdal Balcan
- Manisa Celal Bayar University, Faculty of Science and Letters, Department of Biology, Molecular Biology Section, 45140, Yunusemre, Manisa, Turkey.
| | - Zübeyde Öztel
- Manisa Celal Bayar University, Faculty of Science and Letters, Department of Biology, Molecular Biology Section, 45140, Yunusemre, Manisa, Turkey.
| | - Fatih Çöllü
- Manisa Celal Bayar University, Faculty of Science and Letters, Department of Biology, Zoology Section, 45140, Yunusemre, Manisa, Turkey.
| | - Beyhan Gürcü
- Manisa Celal Bayar University, Faculty of Science and Letters, Department of Biology, Zoology Section, 45140, Yunusemre, Manisa, Turkey.
| |
Collapse
|
3
|
Dawood AA. Determination of binding affinity of tunicamycin with SARS-CoV-2 proteins: Proteinase, protease, nsp2, nsp9, ORF3a, ORF7a, ORF8, ORF9b, envelope and RBD of spike glycoprotein. VACUNAS (ENGLISH EDITION) 2023; 24. [PMCID: PMC9969538 DOI: 10.1016/j.vacune.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Introduction Despite the availability of several COVID-19 vaccines, the incidence of infections remains a serious issue. Tunicamycin (TM), an antibiotic, inhibited tumor growth, reduced coronavirus envelope glycoprotein subunit 2 synthesis, and decreased N-linked glycosylation of coronavirus glycoproteins. Objectives Our study aimed to determine how tunicamycin interacts with certain coronavirus proteins (proteinase, protease, nsp9, ORF7a, ORF3a, ORF9b, ORF8, envelope protein, nsp2, and RBD of spike glycoprotein). Methods: Several types of chemo and bioinformatics tools were used to achieve the aim of the study. As a result, virion's effectiveness may be impaired. Results TM can bind to viral proteins with various degrees of affinity. The proteinase had the highest binding affinity with TM. Proteins (ORF9b, ORF8, nsp9, and RBD) were affected by unfavorable donor or acceptor bonds that impact the degree of docking. ORF7a had the weakest affinities. Conclusions This antibiotic is likely to effect on SARS-CoV-2 in clinical studies.
Collapse
|
4
|
Alshememry AK, Alsaleh NB, Alkhudair N, Alzhrani R, Alshamsan A. Recent nanotechnology advancements to treat multidrug-resistance pancreatic cancer: Pre-clinical and clinical overview. Front Pharmacol 2022; 13:933457. [PMID: 36091785 PMCID: PMC9449524 DOI: 10.3389/fphar.2022.933457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic cancer (PC) remains one of the most lethal and incurable forms of cancer and has a poor prognosis. One of the significant therapeutic challenges in PC is multidrug resistance (MDR), a phenomenon in which cancer cells develop resistance toward administered therapy. Development of novel therapeutic platforms that could overcome MDR in PC is crucial for improving therapeutic outcomes. Nanotechnology is emerging as a promising tool to enhance drug efficacy and minimize off-target responses via passive and/or active targeting mechanisms. Over the past decade, tremendous efforts have been made to utilize nanocarriers capable of targeting PC cells while minimizing off-target effects. In this review article, we first give an overview of PC and the major molecular mechanisms of MDR, and then we discuss recent advancements in the development of nanocarriers used to overcome PC drug resistance. In doing so, we explore the developmental stages of this research in both pre-clinical and clinical settings. Lastly, we discuss current challenges and gaps in the literature as well as potential future directions in the field.
Collapse
Affiliation(s)
- Abdullah K. Alshememry
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nasser B. Alsaleh
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nora Alkhudair
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Rami Alzhrani
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Aws Alshamsan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- *Correspondence: Aws Alshamsan,
| |
Collapse
|
5
|
An Integrated Mass Spectrometry-Based Glycomics-Driven Glycoproteomics Analytical Platform to Functionally Characterize Glycosylation Inhibitors. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123834. [PMID: 35744954 PMCID: PMC9228227 DOI: 10.3390/molecules27123834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/27/2022] [Accepted: 06/11/2022] [Indexed: 12/24/2022]
Abstract
Cancer progression is linked to aberrant protein glycosylation due to the overexpression of several glycosylation enzymes. These enzymes are underexploited as potential anticancer drug targets and the development of rapid-screening methods and identification of glycosylation inhibitors are highly sought. An integrated bioinformatics and mass spectrometry-based glycomics-driven glycoproteomics analysis pipeline was performed to identify an N-glycan inhibitor against lung cancer cells. Combined network pharmacology and in silico screening approaches were used to identify a potential inhibitor, pictilisib, against several glycosylation-related proteins, such as Alpha1-6FucT, GlcNAcT-V, and Alpha2,6-ST-I. A glycomics assay of lung cancer cells treated with pictilisib showed a significant reduction in the fucosylation and sialylation of N-glycans, with an increase in high mannose-type glycans. Proteomics analysis and in vitro assays also showed significant upregulation of the proteins involved in apoptosis and cell adhesion, and the downregulation of proteins involved in cell cycle regulation, mRNA processing, and protein translation. Site-specific glycoproteomics analysis further showed that glycoproteins with reduced fucosylation and sialylation were involved in apoptosis, cell adhesion, DNA damage repair, and chemical response processes. To determine how the alterations in N-glycosylation impact glycoprotein dynamics, modeling of changes in glycan interactions of the ITGA5-ITGB1 (Integrin alpha 5-Integrin beta-1) complex revealed specific glycosites at the interface of these proteins that, when highly fucosylated and sialylated, such as in untreated A549 cells, form greater hydrogen bonding interactions compared to the high mannose-types in pictilisib-treated A549 cells. This study highlights the use of mass spectrometry to identify a potential glycosylation inhibitor and assessment of its impact on cell surface glycoprotein abundance and protein-protein interaction.
Collapse
|
6
|
Costa Catta-Preta CM, Cézar de Azevedo-Martins A, de Souza W, Motta MCM. Effect of the endoplasmic reticulum stressor tunicamycin in Angomonas deanei heat-shock protein expression and on the association with the endosymbiotic bacterium. Exp Cell Res 2022; 417:113162. [PMID: 35460679 DOI: 10.1016/j.yexcr.2022.113162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/04/2022] [Accepted: 04/16/2022] [Indexed: 01/01/2023]
Abstract
The endoplasmic reticulum (ER) presents unique properties to establishing bacterium symbiosis in eukaryotic cells since it synthesizes and glycosylates essential molecules like proteins and lipids. Tunicamycin (TM) is an antibiotic that inhibits the first step in the N-linked glycosylation in eukaryotes and has been used as an ER stress inducer to activate the Unfolded Protein Response (UPR). Mutualistic symbiosis in trypanosomatids is characterized by structural adaptations and intense metabolic exchanges, thus we investigated the effects of TM in the association between Angomonas deanei and its symbiotic bacterium, through ultrastructural and proteomic approaches. Cells treated with the inhibitor showed a decrease in proliferation, enlargement of the ER and Golgi cisternae and an increased distance between the symbiont and the ER. TM proved to be an important tool to better understand ER stress in trypanosomatids, since changes in protein composition were observed in the host protozoan, especially the expression of the Hsp90 chaperone. Furthermore, data obtained indicates the importance of the ER for the adaptation and maintenance of symbiotic associations between prokaryotes and eukaryotes, considering that this organelle has recognized importance in the biogenesis and division of cell structures.
Collapse
Affiliation(s)
- Carolina Moura Costa Catta-Preta
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21491-590, Rio de Janeiro, RJ, Brazil
| | - Allan Cézar de Azevedo-Martins
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21491-590, Rio de Janeiro, RJ, Brazil
| | - Wanderley de Souza
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21491-590, Rio de Janeiro, RJ, Brazil; Centro Nacional de Biologia Estrutural e Bioimagem, RJ, Brazil
| | - Maria Cristina M Motta
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21491-590, Rio de Janeiro, RJ, Brazil; Centro Nacional de Biologia Estrutural e Bioimagem, RJ, Brazil.
| |
Collapse
|
7
|
Inhibition of N-linked Glycosylation by Tunicamycin May Contribute to The Treatment of SARS-CoV-2. Microb Pathog 2020; 149:104586. [PMID: 33091582 PMCID: PMC7573633 DOI: 10.1016/j.micpath.2020.104586] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 12/14/2022]
Abstract
SARS-CoV-2 remains a medical and economic challenge, due to the lack of a suitable drug or vaccine. The glycans in some proteins play a pivotal role in protein folding, oligomerization, quality control, sorting, and transport so the hindering of N-linked glycosylation of glycoproteins will prevent assembly of the virion. Tunicamycin an anticancer drug inhibit the N- linked glycans. Our study aimed to find out the mechanism action of tunicamycin on the viral glycoproteins. The growth of coronavirus in the presence inhibitor tunicamycin resulted in the production of spikeless, non-infectious virions which were devoid of S protein. We concluded that tunicamycin inhibits E2, S, and M glycoproteins of coronaviruses. Tunicamycin is also diminished glycosylation of PTMs such as HE, and 8 ab of SARS-CoV. Finally, we recommend using this drug to treat the SARS-CoV-2. Tunicamycin an anticancer drug inhibits the N- linked glycans. Nucleocapsid phosphoprotein is one of the most structural protein of the virus. The growth of coronavirus in the presence inhibitor tunicamycin resulted in the production of spikeless. Tunicamycin inhibits E2, S, and M glycoproteins of coronaviruses. Tunicamycin is also diminish glycosylation od PTMs such as HE, and 8 ab of SARS-CoV.
Collapse
|
8
|
Hou S, Zhou S, Chen S, Lu Q. Polyphosphazene-Based Drug Self-Framed Delivery System as a Universal Intelligent Platform for Combination Therapy against Multidrug-Resistant Tumors. ACS APPLIED BIO MATERIALS 2020; 3:2284-2294. [DOI: 10.1021/acsabm.0c00072] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Shenglei Hou
- School of Chemistry and Chemical Engineering, The State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shiliu Zhou
- School of Chemistry and Chemical Engineering, The State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shuangshuang Chen
- School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China
| | - Qinghua Lu
- School of Chemistry and Chemical Engineering, The State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
9
|
Ivanova IG, Park CV, Yemm AI, Kenneth NS. PERK/eIF2α signaling inhibits HIF-induced gene expression during the unfolded protein response via YB1-dependent regulation of HIF1α translation. Nucleic Acids Res 2019. [PMID: 29529249 PMCID: PMC5934640 DOI: 10.1093/nar/gky127] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
HIF1α (hypoxia inducible factor 1α) is the central regulator of the cellular response to low oxygen and its activity is deregulated in multiple human pathologies. Consequently, given the importance of HIF signaling in disease, there is considerable interest in developing strategies to modulate HIF1α activity and down-stream signaling events. In the present study we find that under hypoxic conditions, activation of the PERK branch of the unfolded protein response (UPR) can suppress the levels and activity of HIF1α by preventing efficient HIF1α translation. Activation of PERK inhibits de novo HIF1α protein synthesis by preventing the RNA-binding protein, YB-1, from interacting with the HIF1α mRNA 5′UTR. Our data indicate that activation of the UPR can sensitise tumor cells to hypoxic stress, indicating that chemical activation of the UPR could be a strategy to target hypoxic malignant cancer cells.
Collapse
Affiliation(s)
- Iglika G Ivanova
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Catherine V Park
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Adrian I Yemm
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Niall S Kenneth
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
10
|
Chatterjee BK, Jayaraj A, Kumar V, Blagg B, Davis RE, Jayaram B, Deep S, Chaudhuri TK. Stimulation of heat shock protein 90 chaperone function through binding of a novobiocin analog KU-32. J Biol Chem 2019; 294:6450-6467. [PMID: 30792306 DOI: 10.1074/jbc.ra118.002502] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 02/17/2019] [Indexed: 12/13/2022] Open
Abstract
Heat shock protein 90 (Hsp90) is a eukaryotic chaperone responsible for the folding and functional activation of numerous client proteins, many of which are oncoproteins. Thus, Hsp90 inhibition has been intensely pursued, resulting in the development of many potential Hsp90 inhibitors, not all of which are well-characterized. Hsp90 inhibitors not only abrogate its chaperone functions, but also could help us gain insight into the structure-function relationship of this chaperone. Here, using biochemical and cell-based assays along with isothermal titration calorimetry, we investigate KU-32, a derivative of the Hsp90 inhibitor novobiocin (NB), for its ability to modulate Hsp90 chaperone function. Although NB and KU-32 differ only slightly in structure, we found that upon binding, they induce completely opposite conformational changes in Hsp90. We observed that NB and KU-32 both bind to the C-terminal domain of Hsp90, but surprisingly, KU-32 stimulated the chaperone functions of Hsp90 via allosteric modulation of its N-terminal domain, responsible for the chaperone's ATPase activity. In vitro and in silico studies indicated that upon KU-32 binding, Hsp90 undergoes global structural changes leading to the formation of a "partially closed" intermediate that selectively binds ATP and increases ATPase activity. We also report that KU-32 promotes HeLa cell survival and enhances the refolding of an Hsp90 substrate inside the cell. This discovery explains the effectiveness of KU-32 analogs in the management of neuropathies and may facilitate the design of molecules that promote cell survival by enhancing Hsp90 chaperone function and reducing the load of misfolded proteins in cells.
Collapse
Affiliation(s)
| | - Abhilash Jayaraj
- the Supercomputing Facility for Bioinformatics and Computational Biology, and
| | - Vinay Kumar
- the Department of Chemistry, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi 110016, India and
| | - Brian Blagg
- the Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66045
| | - Rachel E Davis
- the Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas 66045
| | - B Jayaram
- the Supercomputing Facility for Bioinformatics and Computational Biology, and
| | - Shashank Deep
- the Department of Chemistry, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi 110016, India and
| | | |
Collapse
|
11
|
Ahmmed B, Khan MN, Nisar MA, Kampo S, Zheng Q, Li Y, Yan Q. Tunicamycin enhances the suppressive effects of cisplatin on lung cancer growth through PTX3 glycosylation via AKT/NF-κB signaling pathway. Int J Oncol 2018; 54:431-442. [PMID: 30483742 PMCID: PMC6317655 DOI: 10.3892/ijo.2018.4650] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 07/05/2018] [Indexed: 12/22/2022] Open
Abstract
Long pentraxin-3 (PTX3) is an inflammatory molecule related to cancer proliferation, invasion, and metastasis. Many studies have highlighted the significance of glycosylated molecules in immune modulation, inflammation and cancer progression. Moreover, aberrant glycosylation of cancer cells is linked to chemoresistance. This study aimed to develop effective therapeutic strategies for deglycosylation of PTX3 (dePTX3) in order to enhance chemosensitivity to cisplatin (Cis) in lung cancer treatment. The A549 and SPCA1 cells were used to determine the role of PTX3 glycosylation in lung cancer growth. Our results revealed that PTX3 was higher in both human lung cancer tissues and serum in comparison with control. Furthermore, we found that deglycosylated PTX3 (dePTX3) by tunicamycin (TM), which is N-glycan precursor biosynthesis blocker, and PNGase F significantly reduced the survival and migration of lung cancer cells. To further confirm this, we also generated glycosylation-site mutant of PTX3 (mPTX3) to characterize the loss of glyco-function. dePTX3 and TM enhanced the suppressive effects of Cis on lung cancer cell growth, migration and invasion compared to individual treatment. Treatment with a combination of TM and Cis significantly inactivated AKT/NF-κB signaling pathway and induced apoptosis. In conclusion, these findings suggest that PTX3 is an important mediator of lung cancer progression, and dePTX3 by TM enhances the anticancer effects of Cis. The deglycosylation in chemotherapy may represent a potential novel therapeutic strategy against lung cancer.
Collapse
Affiliation(s)
- Bulbul Ahmmed
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Muhammad Noman Khan
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Muhammad Azhar Nisar
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Sylvanus Kampo
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Qin Zheng
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yulin Li
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Qiu Yan
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
12
|
Gurunathan S, Kang MH, Qasim M, Kim JH. Nanoparticle-Mediated Combination Therapy: Two-in-One Approach for Cancer. Int J Mol Sci 2018; 19:E3264. [PMID: 30347840 PMCID: PMC6214025 DOI: 10.3390/ijms19103264] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/16/2018] [Accepted: 10/16/2018] [Indexed: 02/07/2023] Open
Abstract
Cancer represents a group of heterogeneous diseases characterized by uncontrolledgrowth and spread of abnormal cells, ultimately leading to death. Nanomedicine plays a significantrole in the development of nanodrugs, nanodevices, drug delivery systems and nanocarriers. Someof the major issues in the treatment of cancer are multidrug resistance (MDR), narrow therapeuticwindow and undesired side effects of available anticancer drugs and the limitations of anticancerdrugs. Several nanosystems being utilized for detection, diagnosis and treatment such as theranosticcarriers, liposomes, carbon nanotubes, quantum dots, polymeric micelles, dendrimers and metallicnanoparticles. However, nonbiodegradable nanoparticles causes high tissue accumulation andleads to toxicity. MDR is considered a major impediment to cancer treatment due to metastatictumors that develop resistance to chemotherapy. MDR contributes to the failure of chemotherapiesin various cancers, including breast, ovarian, lung, gastrointestinal and hematological malignancies.Moreover, the therapeutic efficiency of anticancer drugs or nanoparticles (NPs) used alone is lessthan that of the combination of NPs and anticancer drugs. Combination therapy has long beenadopted as the standard first-line treatment of several malignancies to improve the clinical outcome.Combination therapy with anticancer drugs has been shown to generally induce synergistic drugactions and deter the onset of drug resistance. Therefore, this review is designed to report andanalyze the recent progress made to address combination therapy using NPs and anticancer drugs.We first provide a comprehensive overview of the angiogenesis and of the different types of NPscurrently used in treatments of cancer; those emphasized in this review are liposomes, polymericNPs, polymeric micelles (PMs), dendrimers, carbon NPs, nanodiamond (ND), fullerenes, carbonnanotubes (CNTs), graphene oxide (GO), GO nanocomposites and metallic NPs used forcombination therapy with various anticancer agents. Nanotechnology has provided the convenienttools for combination therapy. However, for clinical translation, we need continued improvementsin the field of nanotechnology.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Min-Hee Kang
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Muhammad Qasim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
13
|
Zhang D, Liu Q, Visvanathan R, Tuchband MR, Sheetah GH, Fairbanks BD, Clark NA, Smalyukh II, Bowman CN. A supramolecular hydrogel prepared from a thymine-containing artificial nucleolipid: study of assembly and lyotropic mesophases. SOFT MATTER 2018; 14:7045-7051. [PMID: 30112539 DOI: 10.1039/c8sm01383g] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
An artificial nucleolipid containing thymine, a triazole-ring, and phosphatidylcholine (TTPC) moieties was prepared by copper catalyzed azide alkyne cycloaddition (CuAAC) under aqueous conditions. The resulting TTPC molecules assembled in situ into a fibrous aggregation. The study of the TTPC fiber assembly using XRD and NMR spectroscopy revealed that the formation of fibers was driven by the unique combination of the lipid and nucleobase moieties in the structure of TTPC. At a critical TTPC concentration, entanglement of the fibers resulted in the formation of a supramolecular hydrogel. Investigation of the lyotropic mesophases in the TTPC supramolecular hydrogel showed the presence of multiple phases including two liquid crystal phases (i.e., nematic and lamellar), which have a certain degree of structural order and are promising templates for constructing functional biomaterials.
Collapse
Affiliation(s)
- Dawei Zhang
- Department of Chemical and Biological Engineering, University of Colorado Boulder, UCB 596, Colorado 80309, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Combination therapy of simvastatin and 5, 6-dimethylxanthenone-4-acetic acid synergistically suppresses the aggressiveness of B16.F10 melanoma cells. PLoS One 2018; 13:e0202827. [PMID: 30138430 PMCID: PMC6107259 DOI: 10.1371/journal.pone.0202827] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 08/09/2018] [Indexed: 12/15/2022] Open
Abstract
The major drawback of current anti-angiogenic therapies is drug resistance, mainly caused by overexpression of the transcription factor, hypoxia-inducible factor 1α (HIF-1α) as a result of treatment-induced hypoxia, which stimulates cancer cells to develop aggressive and immunosuppressive phenotypes. Moreover, the cancer cell resistance to anti-angiogenic therapies is deeply mediated by the communication between tumor cells and tumor-associated macrophages (TAMs)-the most important microenvironmental cells for the coordination of all supportive processes in tumor development. Thus, simultaneous targeting of TAMs and cancer cells could improve the outcome of the anti-angiogenic therapies. Since our previous studies proved that simvastatin (SIM) exerts strong antiproliferative actions on B16.F10 murine melanoma cells via reduction of TAMs-mediated oxidative stress and inhibition of intratumor production of HIF-1α, we investigated whether the antitumor efficacy of the anti-angiogenic agent-5,6-dimethylxanthenone-4-acetic acid (DMXAA) could be improved by its co-administration with the lipophilic statin. Our results provide confirmatory evidence for the ability of the combined treatment to suppress the aggressive phenotype of the B16.F10 melanoma cells co-cultured with TAMs under hypoxia-mimicking conditions in vitro. Thus, proliferation and migration capacity of the melanoma cells were strongly decelerated after the co-administration of SIM and DMXAA. Moreover, our data suggested that the anti-oxidant action of the combined treatment, as a result of melanogenesis stimulation, might be the principal cause for the simultaneous suppression of key molecules involved in melanoma cell aggressiveness, present in melanoma cells (HIF-1α) as well as in TAMs (arginase-1). Finally, the concomitant suppression of these proteins might have contributed to a very strong inhibition of the angiogenic capacity of the cell co-culture microenvironment.
Collapse
|
15
|
Tekin E, Beppler C, White C, Mao Z, Savage VM, Yeh PJ. Enhanced identification of synergistic and antagonistic emergent interactions among three or more drugs. J R Soc Interface 2017; 13:rsif.2016.0332. [PMID: 27278366 DOI: 10.1098/rsif.2016.0332] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 05/17/2016] [Indexed: 02/02/2023] Open
Abstract
Interactions among drugs play a critical role in the killing efficacy of multi-drug treatments. Recent advances in theory and experiment for three-drug interactions enable the search for emergent interactions-ones not predictable from pairwise interactions. Previous work has shown it is easier to detect synergies and antagonisms among pairwise interactions when a rescaling method is applied to the interaction metric. However, no study has carefully examined whether new types of normalization might be needed for emergence. Here, we propose several rescaling methods for enhancing the classification of the higher order drug interactions based on our conceptual framework. To choose the rescaling that best separates synergism, antagonism and additivity, we conducted bacterial growth experiments in the presence of single, pairwise and triple-drug combinations among 14 antibiotics. We found one of our rescaling methods is far better at distinguishing synergistic and antagonistic emergent interactions than any of the other methods. Using our new method, we find around 50% of emergent interactions are additive, much less than previous reports of greater than 90% additivity. We conclude that higher order emergent interactions are much more common than previously believed, and we argue these findings for drugs suggest that appropriate rescaling is crucial to infer higher order interactions.
Collapse
Affiliation(s)
- Elif Tekin
- Department of Biomathematics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Casey Beppler
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA 90095, USA
| | - Cynthia White
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA 90095, USA
| | - Zhiyuan Mao
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA 90095, USA
| | - Van M Savage
- Department of Biomathematics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA 90095, USA Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Pamela J Yeh
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
16
|
Peiris D, Spector AF, Lomax-Browne H, Azimi T, Ramesh B, Loizidou M, Welch H, Dwek MV. Cellular glycosylation affects Herceptin binding and sensitivity of breast cancer cells to doxorubicin and growth factors. Sci Rep 2017; 7:43006. [PMID: 28223691 PMCID: PMC5320443 DOI: 10.1038/srep43006] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 01/12/2017] [Indexed: 01/07/2023] Open
Abstract
Alterations in protein glycosylation are a key feature of oncogenesis and have been shown to affect cancer cell behaviour perturbing cell adhesion, favouring cell migration and metastasis. This study investigated the effect of N-linked glycosylation on the binding of Herceptin to HER2 protein in breast cancer and on the sensitivity of cancer cells to the chemotherapeutic agent doxorubicin (DXR) and growth factors (EGF and IGF-1). The interaction between Herceptin and recombinant HER2 protein and cancer cell surfaces (on-rate/off-rate) was assessed using a quartz crystal microbalance biosensor revealing an increase in the accessibility of HER2 to Herceptin following deglycosylation of cell membrane proteins (deglycosylated cells Bmax: 6.83 Hz; glycosylated cells Bmax: 7.35 Hz). The sensitivity of cells to DXR and to growth factors was evaluated using an MTT assay. Maintenance of SKBR-3 cells in tunicamycin (an inhibitor of N-linked glycosylation) resulted in an increase in sensitivity to DXR (0.1 μM DXR P < 0.001) and a decrease in sensitivity to IGF-1 alone and to IGF-1 supplemented with EGF (P < 0.001). This report illustrates the importance of N-linked glycosylation in modulating the response of cancer cells to chemotherapeutic and biological treatments and highlights the potential of glycosylation inhibitors as future combination treatments for breast cancer.
Collapse
Affiliation(s)
- Diluka Peiris
- Attana AB, Bjornnasvagen 21, SE-11419, Stockholm, Sweden
| | - Alexander F Spector
- Division of Surgery and Interventional Science, UCL Medical School Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Hannah Lomax-Browne
- Department of Biomedical Sciences, Faculty of Science and Technology, University of Westminster, 115 New Cavendish St, W1W 6UW, UK
| | - Tayebeh Azimi
- Department of Biomedical Sciences, Faculty of Science and Technology, University of Westminster, 115 New Cavendish St, W1W 6UW, UK
| | - Bala Ramesh
- Division of Surgery and Interventional Science, UCL Medical School Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Marilena Loizidou
- Division of Surgery and Interventional Science, UCL Medical School Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Hazel Welch
- Division of Surgery and Interventional Science, UCL Medical School Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Miriam V Dwek
- Department of Biomedical Sciences, Faculty of Science and Technology, University of Westminster, 115 New Cavendish St, W1W 6UW, UK
| |
Collapse
|
17
|
Riccardi C, Musumeci D, Irace C, Paduano L, Montesarchio D. RuIIIComplexes for Anticancer Therapy: The Importance of Being Nucleolipidic. European J Org Chem 2016. [DOI: 10.1002/ejoc.201600943] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences; University of Napoli Federico II; Via Cintia 21 80126 Napoli Italy
| | - Domenica Musumeci
- Department of Chemical Sciences; University of Napoli Federico II; Via Cintia 21 80126 Napoli Italy
| | - Carlo Irace
- Department of Pharmacy; University of Napoli Federico II; Via D. Montesano 49 80131 Napoli Italy
| | - Luigi Paduano
- Department of Chemical Sciences; University of Napoli Federico II; Via Cintia 21 80126 Napoli Italy
| | - Daniela Montesarchio
- Department of Chemical Sciences; University of Napoli Federico II; Via Cintia 21 80126 Napoli Italy
| |
Collapse
|
18
|
Combination Therapy using Co-encapsulated Resveratrol and Paclitaxel in Liposomes for Drug Resistance Reversal in Breast Cancer Cells in vivo. Sci Rep 2016; 6:22390. [PMID: 26947928 PMCID: PMC4780086 DOI: 10.1038/srep22390] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 02/11/2016] [Indexed: 12/19/2022] Open
Abstract
Multidrug resistance (MDR) is a major impediment to cancer treatment. A promising strategy for treating MDR is the joint delivery of combined anticancer agents to tumor cells in a single nanocarrier. Here, for the first time, Resveratrol (Res) was co-encapsulated with paclitaxel (PTX) in a PEGylated liposome to construct a carrier-delivered form of combination therapy for drug-resistant tumors. The composite liposome had an average diameter of 50 nm with encapsulated efficiencies of above 50%. The studies demonstrated that the composite liposome could generate potent cytotoxicity against the drug-resistant MCF-7/Adr tumor cells in vitro and enhance the bioavailability and the tumor-retention of the drugs in vivo. Moreover, systemic therapy with the composite liposome effectively inhibited drug-resistant tumor in mice (p < 0.01), without any notable increase in the toxicity. These results suggested that the co-delivery of Res and a cytotoxic agent in a nanocarrier may potentially improve the treatment of drug-resistant tumors.
Collapse
|
19
|
Liu X, Zou J, Su J, Lu Y, Zhang J, Li L, Yin F. Downregulation of transient receptor potential cation channel, subfamily C, member 1 contributes to drug resistance and high histological grade in ovarian cancer. Int J Oncol 2015; 48:243-52. [PMID: 26647723 DOI: 10.3892/ijo.2015.3254] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 10/15/2015] [Indexed: 11/05/2022] Open
Abstract
Transient receptor potential cation channel, subfamily C, member 1 (TRPC1) participates in many physiological functions but has also been implicated in cancer development. However, little is known about the role of TRPC1 in ovarian cancer (OC), including the drug resistance of these tumors. In the present study, a significant and consistent downregulation of TRPC1 in drug-resistant OC tissues/cells was determined using real-time quantitative polymerase chain reaction assays and the microarrays deposited in Oncomine and Gene Expression Omnibus (GEO) profiles. Protein/gene-protein/gene and protein-chemical interactions indicated that TRPC1 interacts with 14 proteins/genes and 6 chemicals, all of which are involved in the regulation of drug resistance in OC. Biological process annotation of TRPC1, OC, and drug resistance indicated a role for TRPC1 in drug-resistance-related functions in OC, mainly via the cell cycle, gene expression and cell growth and cell death. Analysis of mRNA-microRNA interactions showed that 8 out of 11 major pathways enriched from 38 predominant microRNAs targeting TRPC1 were involved in the regulation of drug resistance in OC, and 8 out of these top 10 microRNAs were implicated in the drug resistance in ovarian and other cancers. In a clinical analysis using data obtained from The Cancer Genome Atlas project (TCGA) cohort on 341 OC patients, TRPC1 expression was found to differ significantly between grade 2 and grade 3 tumors, with low-level expression correlating with higher tumor grade. This is the first report to show a potential association between the downregulation of TRPC1 and both drug resistance and high histological tumor grade in OC. Our results provide the basis for further investigations of the drug-resistance-related functions of TRPC1 in OC and other forms of cancer.
Collapse
Affiliation(s)
- Xia Liu
- Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jing Zou
- Medical Scientific Research Centre, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jie Su
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, Guangxi 530021, P.R. China
| | - Yi Lu
- Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jian Zhang
- Center for Translational Medicine, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Li Li
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, Guangxi 530021, P.R. China
| | - Fuqiang Yin
- Medical Scientific Research Centre, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
20
|
Atil B, Berger-Sieczkowski E, Bardy J, Werner M, Hohenegger M. In vitro and in vivo downregulation of the ATP binding cassette transporter B1 by the HMG-CoA reductase inhibitor simvastatin. Naunyn Schmiedebergs Arch Pharmacol 2015; 389:17-32. [PMID: 26319048 PMCID: PMC4700083 DOI: 10.1007/s00210-015-1169-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 08/18/2015] [Indexed: 12/20/2022]
Abstract
Extrusion of chemotherapeutics by ATP-binding cassette (ABC) transporters like ABCB1 (P-glycoprotein) represents a crucial mechanism of multidrug resistance in cancer therapy. We have previously shown that the 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor simvastatin directly inhibits ABCB1, alters the glycosylation of the transporter, and enhances the intracellular accumulation of doxorubicin with subsequent anti-cancer action. Here, we show that simvastatin reduces endogenous dolichol levels and ABCB1 in human neuroblastoma SH-SY5Y cells. Coapplication with dolichol prevents the downregulation of the ABCB1 transporter. Importantly, dolichol also attenuated simvastatin-induced apoptosis, unmasking involvement of unfolded protein response. Direct monitoring of the fluorescent fusion protein YFP-ABCB1 further confirms concentration-dependent reduction of ABCB1 in HEK293 cells by simvastatin. In simvastatin-treated murine xenografts, ABCB1 was also reduced in the liver and rhabdomyosarcoma but did not reach significance in neuroblastoma. Nevertheless, the in vivo anti-cancer effects of simvastatin are corroborated by increased apoptosis in tumor tissues. These findings provide experimental evidence for usage of simvastatin in novel chemotherapeutic regimens and link dolichol depletion to simvastatin-induced anti-cancer activity.
Collapse
Affiliation(s)
- Bihter Atil
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Währingerstraße 13A, 1090, Vienna, Austria
| | | | - Johanna Bardy
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Währingerstraße 13A, 1090, Vienna, Austria.,Department of Internal Medicine, Hanuschkrankenhaus, Heinrich-Collin-Strasse 30, 1140, Vienna, Austria
| | - Martin Werner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Währingerstraße 13A, 1090, Vienna, Austria.,Department of Internal Medicine, Hanuschkrankenhaus, Heinrich-Collin-Strasse 30, 1140, Vienna, Austria
| | - Martin Hohenegger
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Währingerstraße 13A, 1090, Vienna, Austria.
| |
Collapse
|
21
|
Wojtowicz K, Januchowski R, Nowicki M, Zabel M. Inhibition of protein glycosylation reverses the MDR phenotype of cancer cell lines. Biomed Pharmacother 2015; 74:49-56. [PMID: 26349962 DOI: 10.1016/j.biopha.2015.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 07/09/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Multidrug resistance proteins are one of the most important factors that cause chemotherapy resistance, which in turn reduces therapeutic efficacy and survival for cancer patients. Tunicamycin is one of the most well-known inhibitors of N-glycosylation and is considered a powerful adjunct that can increase the effectiveness of many drugs. Tunicamycin blocks the first step of P-gp (glycoprotein P) and BCRP (breast cancer resistance protein) N-glycosylation, which is a very important modification for the activity and cellular localisation of these proteins. METHODS The effects of tunicamycin on ovarian and colorectal cancer cells were examined in multiple cell lines. The primary ovarian cancer cell line W1 and the established ovarian cancer cell line A2780 were compared against their drug-resistant derivatives W1TR/W1PR (TR: topotecan resistant; PR: paclitaxel resistant) and A2780T1 (topotecan resistant), respectively. We also compared the colorectal cancer cell line LoVo against its doxorubicin-resistant derivative LoVo/Dx. Cell viability was determined by the MTT assay. The glycopeptides were subjected to deglycosylation using the endoglycosidase PNGase F. A2780T1, LoVo/Dx and W1PR cells were treated with the protein degradation inhibitors MG132 and BMA. Protein expression was detected by western blot and immunocytochemistry. RESULTS In this study, we showed via the MTT assay that tunicamycin significantly decreased the viability of cancer cell lines that were co-treated with a chemotherapeutic drug. Western blot analysis showed that, in LoVo/Dx and W1PR cells, tunicamycin treatment resulted in the expression of a 70kDa P-gp protein instead of the mature 170kDa P-gp. Treatment with MG132 or BMA fully suppressed the effect of tunicamycin in the case of W1PR cells only. In tunicamycin-treated W1TR cells, the size of the BCRP protein did not differ from that of its native unglycosylated form. In tunicamycin-treated A2780T1 cells, BCRP expression was completely inhibited, but pre-treatment with MG132 or BMA suppressed the effect of tunicamycin. Immunocytochemistry analysis indicated that tunicamycin only affected the translocation of P-gp but not that of BCRP. After treatment, we observed higher P-gp expression in the cytoplasm than at the cell membrane. CONCLUSIONS Our results indicated that tunicamycin may enhance the effect of chemotherapy by interfering with the localisation and function of transporter proteins that are responsible for multidrug resistance.
Collapse
Affiliation(s)
- Karolina Wojtowicz
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland.
| | - Radosław Januchowski
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland.
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland.
| | - Maciej Zabel
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; Department of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland.
| |
Collapse
|
22
|
Ng PL, Rajab NF, Then SM, Mohd Yusof YA, Wan Ngah WZ, Pin KY, Looi ML. Piper betle leaf extract enhances the cytotoxicity effect of 5-fluorouracil in inhibiting the growth of HT29 and HCT116 colon cancer cells. J Zhejiang Univ Sci B 2015; 15:692-700. [PMID: 25091987 DOI: 10.1631/jzus.b1300303] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The combination effect of Piper betle (PB) and 5-fluorouracil (5-FU) in enhancing the cytotoxic potential of 5-FU in inhibiting the growth of colon cancer cells was investigated. METHODS HT29 and HCT116 cells were subjected to 5-FU or PB treatment. 5-FU and PB were then combined and their effects on both cell lines were observed after 24 h of treatment. PB-5-FU interaction was elucidated by isobologram analysis. Apoptosis features of the treated cells were revealed by annexin V/PI stain. High-performance liquid chromatography (HPLC) was performed to exclude any possible chemical interaction between the compounds. RESULTS In the presence of PB extract, the cytotoxicity of 5-FU was observed at a lower dose (IC50 12.5 µmol/L) and a shorter time (24 h) in both cell lines. Both cell lines treated with 5-FU or PB alone induced a greater apoptosis effect compared with the combination treatment. Isobologram analysis indicated that PB and 5-FU interacted synergistically and antagonistically in inhibiting the growth of HT29 and HCT116 cells, respectively. CONCLUSIONS In the presence of PB, a lower dosage of 5-FU is required to achieve the maximum drug effect in inhibiting the growth of HT29 cells. However, PB did not significantly reduce 5-FU dosage in HCT116 cells. Our result showed that this interaction may not solely contribute to the apoptosis pathway.
Collapse
Affiliation(s)
- Pek Leng Ng
- Department of Biomedical Sciences, Faculty of Allied Health, Universiti Kebangsaan Malaysia (UKM), Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia; UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, 56000 Kuala Lumpur, Malaysia; School of Biomedical Science, University of Nottingham Malaysia Campus, 43500 Semenyih, Selangor, Malaysia; Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia; Forest Research Institute Malaysia (FRIM), 52109 Kuala Lumpur, Kepong, Malaysia; School of Biosciences, Taylor's University, Lakeside Campus, 47500 Subang Jaya, Selangor, Malaysia
| | | | | | | | | | | | | |
Collapse
|
23
|
Goodall ML, Wang T, Martin KR, Kortus MG, Kauffman AL, Trent JM, Gately S, MacKeigan JP. Development of potent autophagy inhibitors that sensitize oncogenic BRAF V600E mutant melanoma tumor cells to vemurafenib. Autophagy 2015; 10:1120-36. [PMID: 24879157 PMCID: PMC4091172 DOI: 10.4161/auto.28594] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a dynamic cell survival mechanism by which a double-membrane vesicle, or autophagosome, sequesters portions of the cytosol for delivery to the lysosome for recycling. This process can be inhibited using the antimalarial agent chloroquine (CQ), which impairs lysosomal function and prevents autophagosome turnover. Despite its activity, CQ is a relatively inadequate inhibitor that requires high concentrations to disrupt autophagy, highlighting the need for improved small molecules. To address this, we screened a panel of antimalarial agents for autophagy inhibition and chemically synthesized a novel series of acridine and tetrahydroacridine derivatives. Structure-activity relationship studies of the acridine ring led to the discovery of VATG-027 as a potent autophagy inhibitor with a high cytotoxicity profile. In contrast, the tetrahydroacridine VATG-032 showed remarkably little cytotoxicity while still maintaining autophagy inhibition activity, suggesting that both compounds act as autophagy inhibitors with differential effects on cell viability. Further, knockdown of autophagy-related genes showed no effect on cell viability, demonstrating that the ability to inhibit autophagy is separate from the compound cytotoxicity profiles. Next, we determined that both inhibitors function through lysosomal deacidification mechanisms and ultimately disrupt autophagosome turnover. To evaluate the genetic context in which these lysosomotropic inhibitors may be effective, they were tested in patient-derived melanoma cell lines driven by oncogenic BRAF (v-raf murine sarcoma viral oncogene homolog B). We discovered that both inhibitors sensitized melanoma cells to the BRAF V600E inhibitor vemurafenib. Overall, these autophagy inhibitors provide a means to effectively block autophagy and have the potential to sensitize mutant BRAF melanomas to first-line therapies.
Collapse
Affiliation(s)
- Megan L Goodall
- Laboratory of Systems Biology; Van Andel Research Institute; Grand Rapids, MI USA; Genetics Graduate Program; Michigan State University; East Lansing, MI USA
| | - Tong Wang
- Translational Drug Development (TD2); Scottsdale, AZ USA
| | - Katie R Martin
- Laboratory of Systems Biology; Van Andel Research Institute; Grand Rapids, MI USA
| | - Matthew G Kortus
- Laboratory of Systems Biology; Van Andel Research Institute; Grand Rapids, MI USA
| | - Audra L Kauffman
- Laboratory of Systems Biology; Van Andel Research Institute; Grand Rapids, MI USA
| | | | - Stephen Gately
- Translational Drug Development (TD2); Scottsdale, AZ USA
| | - Jeffrey P MacKeigan
- Laboratory of Systems Biology; Van Andel Research Institute; Grand Rapids, MI USA
| |
Collapse
|
24
|
Riganti C, Kopecka J, Panada E, Barak S, Rubinstein M. The role of C/EBP-β LIP in multidrug resistance. J Natl Cancer Inst 2015; 107:djv046. [PMID: 25766403 DOI: 10.1093/jnci/djv046] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Chemotherapy triggers endoplasmic reticulum (ER) stress, which in turn regulates levels of the active (LAP) and the natural dominant-negative (LIP) forms of the transcription factor C/EBP-β. LAP upregulates and LIP downregulates the multidrug resistance (MDR) protein P-glycoprotein (Pgp), but it is not known how critical is their role in establishing MDR. METHODS Cell viability was quantitated by crystal violet staining and measuring absorbance at 540nm. Expression of various proteins was determined by immunoblotting. mRNA levels were determined by quantitative reverse transcriptase polymerase chain reaction (RT-PCR). LIP and LAP were overexpressed using expression plasmids followed by selection with blasticidin. Tumor cells expressing doxycycline-inducible LIP were orthotopically implanted in mice (n = 15 mice per group), and tumor size was measured daily by caliper. Tumor sections were stained with hematoxylin and eosin and immunostained for Pgp, proliferation, and ER stress markers. RESULTS MDR cells do not express basal, chemotherapy-triggered, or ER stress-triggered LIP and fail to activate the CHOP-caspase-3 death-triggering axis upon ER stress or chemotherapy challenge. Overexpression of LIP reversed the MDR phenotype in vitro and in tumors implanted in mice. LIP was undetectable in MDR cells, probably due to its ubiquitination, which was 3.56-fold higher, resulting in lysosomal and proteasomal degradation of LIP. CONCLUSIONS Spontaneous and drug-selected MDR cells lack LIP, which is eliminated by ubiquitin-mediated degradation. Loss of LIP drives MDR not only by increasing Pgp expression but also by a two-fold attenuation of ER stress-triggered cell death.
Collapse
Affiliation(s)
- Chiara Riganti
- Department of Oncology, University of Torino, Italy (CR, JK, EP); Department of Molecular Genetics, the Weizmann Institute of Science, Rehovot, Israel (SB, MR)
| | - Joanna Kopecka
- Department of Oncology, University of Torino, Italy (CR, JK, EP); Department of Molecular Genetics, the Weizmann Institute of Science, Rehovot, Israel (SB, MR)
| | - Elisa Panada
- Department of Oncology, University of Torino, Italy (CR, JK, EP); Department of Molecular Genetics, the Weizmann Institute of Science, Rehovot, Israel (SB, MR)
| | - Sara Barak
- Department of Oncology, University of Torino, Italy (CR, JK, EP); Department of Molecular Genetics, the Weizmann Institute of Science, Rehovot, Israel (SB, MR)
| | - Menachem Rubinstein
- Department of Oncology, University of Torino, Italy (CR, JK, EP); Department of Molecular Genetics, the Weizmann Institute of Science, Rehovot, Israel (SB, MR).
| |
Collapse
|
25
|
Yang H, Liu YH, Xu L, Liu LH. Efficacy of permanent iodine-125 seed implants and gemcitabine chemotherapy in patients with platinum- resistant recurrent ovarian carcinoma. Asian Pac J Cancer Prev 2014; 15:9009-13. [PMID: 25374244 DOI: 10.7314/apjcp.2014.15.20.9009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The aim of this study was to explore the efficacy and adverse reactions of CT-guided radioactive 125I-seed implantation treatment combined with chemotherapy for platinum-resistant recurrent ovarian carcinoma. MATERIALS AND METHODS From September 2010 to December 2012, 23 patients with platinum-resistant recurrent ovarian carcinoma were enrolled. All the patients refused, could not bear, or were not suitable for surgery. They all had no more than 3 lesions, which were detected and could also be measured by CT. All were clarified as single-lesion or multiple-lesion groups. A total of 41 lesions underwent implantation of from 8 to 106 125I seeds (median=43). Multi-plane implanting was adopted and 125I-seeds of (0.4-0.7)mCi were placed at intervals of (0.5-1.0) cm. After implantation treatment, all patients underwent 4 cycles of chemotherapy with gemcitabine 800 mg/m2 (days 1, 8 and 15). RESULTS The outcome was evaluated with CT 3 weeks and every 3 months after implantation treatment. After 6 months, the volume of 32 out of 41 lesions (78.0%) was reduced at least 30%, within which 9 lesions completely disappeared(22.0%). Complete response was observed in 7 cases (30.4%), with a partial response in 4 cases (17.4%),4 cases stable(17.4%)and 8 cases showing progression (34.8%). The total clinical remission rate was 47.8% (11/23). The clinical remission rate was 77.8% (7/9) in the single-lesion group and 28.6% (4/14) in the multiple-lesion group with a significant difference between the two(P=0.036). The common side effects observed were mild gastrointestinal reactions. CONCLUSIONS 125I-seed implantation combined with chemotherapy applies an effective way in the treatment of platinum-resistant recurrent ovarian epithelial carcinoma with the advantages of high local control rates, good short-term effects, little trauma and less side effects.
Collapse
Affiliation(s)
- Hui Yang
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, The Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan, China E-mail : ;
| | | | | | | |
Collapse
|
26
|
Varelas X, Bouchie MP, Kukuruzinska MA. Protein N-glycosylation in oral cancer: dysregulated cellular networks among DPAGT1, E-cadherin adhesion and canonical Wnt signaling. Glycobiology 2014; 24:579-91. [PMID: 24742667 PMCID: PMC4038253 DOI: 10.1093/glycob/cwu031] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 03/17/2014] [Accepted: 04/11/2014] [Indexed: 12/17/2022] Open
Abstract
N-Linked glycosylation (N-glycosylation) of proteins has long been associated with oncogenesis, but not until recently have the molecular mechanisms underlying this relationship begun to be unraveled. Here, we review studies describing how dysregulation of the N-glycosylation-regulating gene, DPAGT1, drives oral cancer. DPAGT1 encodes the first and rate-limiting enzyme in the assembly of the lipid-linked oligosaccharide precursor in the endoplasmic reticulum and thus mediates N-glycosylation of many cancer-related proteins. DPAGT1 controls N-glycosylation of E-cadherin, the major epithelial cell-cell adhesion receptor and a tumor suppressor, thereby affecting intercellular adhesion and cytoskeletal dynamics. DPAGT1 also regulates and is regulated by Wnt/β-catenin signaling, impacting the balance between proliferation and adhesion in homeostatic tissues. Thus, aberrant induction of DPAGT1 promotes a positive feedback network with Wnt/β-catenin that represses E-cadherin-based adhesion and drives tumorigenic phenotypes. Further, modification of receptor tyrosine kinases (RTKs) with N-glycans is known to control their surface presentation via the galectin lattice, and thus increased DPAGT1 expression likely contributes to abnormal activation of RTKs in oral cancer. Collectively, these studies suggest that dysregulation of the DPAGT1/Wnt/E-cadherin network underlies the etiology and pathogenesis of oral cancer.
Collapse
Affiliation(s)
- Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Meghan P Bouchie
- Department of Molecular and Cell Biology, Boston University School of Dental Medicine, Boston, MA, USA
| | - Maria A Kukuruzinska
- Department of Molecular and Cell Biology, Boston University School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
27
|
Hou H, Sun H, Lu P, Ge C, Zhang L, Li H, Zhao F, Tian H, Zhang L, Chen T, Yao M, Li J. Tunicamycin Potentiates Cisplatin Anticancer Efficacy through the DPAGT1/Akt/ABCG2 Pathway in Mouse Xenograft Models of Human Hepatocellular Carcinoma. Mol Cancer Ther 2013; 12:2874-84. [DOI: 10.1158/1535-7163.mct-13-0201] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
28
|
Banerjee A, Johnson KT, Banerjee IA, Banerjee DK. Nanoformulation enhances anti-angiogenic efficacy of tunicamycin. Transl Cancer Res 2013; 2:240-255. [PMID: 33209651 PMCID: PMC7671603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Nanoparticles (<100 nm) evades the immune system's clearing mechanisms long enough to reach the targeted disease tissue efficiently. We have, therefore, hypothesized that nano-formulated Tunicamycin would have a better efficacy and consequently it will be a better candidate for treating solid tumor including breast cancer in the clinic. Tunicamycin, a potent inhibitor of asparagine-linked (N-linked) protein glycosylation has been found earlier (I) inhibits angiogenesis in vitro by arresting cells in G1; (II) in vivo angiogenesis in Matrigel™ implant in nude mice; and (III) prevents the progression of a double- and a triple-negative breast tumor in athymic nude mice by inducing "ER stress" in tumor microvasculature. Tunicamycin could work alone or in combination with radiation/radiotherapy. To evaluate nano-formulated Tunicamycin, we have synthesized Tunicamycin encapsulated in peptide nanotubes, nanotubes bound to gold nanoparticles (Au NPs) conjugated with Tunicamycin, Tunicamycin conjugated with nanotubes, Au NPs bound to tubes and conjugated with Tunicamycin, and Au NPs conjugated with Tunicamycin. Functionalization of the nanoparticles was characterized by transmission electron microscopy (TEM), Fourier Transformed Infrared (FTIR) Spectroscopy, dynamic light scattering, atomic force microscopy (AFM), and absorbance spectroscopy. The 3-(4,5-methylthiazol-2-yl)-2,5-dipheyl-tetrazolium bromide (MTT) assay indicated that nanoparticles (1 μg/mL) inhibited capillary endothelial cells proliferation, i.e., angiogenesis ~50% within one hour of treatment whereas the native Tunicamycin had no effect. The nano-formulated Tunicamycin blocked the cell cycle progression by inhibiting either both cyclin D1 and CDK4, or cyclin D1, or the CDK4 expression as well as the expression of phospho Rb (serine-229/threonine-252). Phosphorylation of p53 at serine-392 was down-regulated but not the total p53. Increased expression of GRP-78/Bip identified "ER stress". Upregulated expression (1.6-5.5 fold) of phopsho-PERK and significant reduction of mannosylphospho dolichol synthase (DPMS) expression supported induction of unfolded protein response (upr) signaling. Down regulated expression of caspase-9 and caspase-3 proposes a non-canonical pathway of cell death during "ER stress" induced by nano-formulated Tunicamycin.
Collapse
Affiliation(s)
- Aditi Banerjee
- Department of Biochemistry, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, USA
| | - Karen T. Johnson
- Department of Chemistry, Fordham University, Bronx, NY 10458, USA
| | | | - Dipak K. Banerjee
- Department of Biochemistry, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, USA
- Institute of Functional Nanomaterials, University of Puerto Rico, Rio Piedras Campus, San Juan, PR 00931-1907, USA
| |
Collapse
|
29
|
de-Freitas-Junior JCM, Bastos LG, Freire-Neto CA, Rocher BD, Abdelhay ESFW, Morgado-Díaz JA. N-glycan biosynthesis inhibitors induce in vitro anticancer activity in colorectal cancer cells. J Cell Biochem 2012; 113:2957-66. [DOI: 10.1002/jcb.24173] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
30
|
Hiss D. Optimizing molecular-targeted therapies in ovarian cancer: the renewed surge of interest in ovarian cancer biomarkers and cell signaling pathways. JOURNAL OF ONCOLOGY 2012; 2012:737981. [PMID: 22481932 PMCID: PMC3306947 DOI: 10.1155/2012/737981] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 11/24/2011] [Indexed: 12/18/2022]
Abstract
The hallmarks of ovarian cancer encompass the development of resistance, disease recurrence and poor prognosis. Ovarian cancer cells express gene signatures which pose significant challenges for cancer drug development, therapeutics, prevention and management. Despite enhancements in contemporary tumor debulking surgery, tentative combination regimens and abdominal radiation which can achieve beneficial response rates, the majority of ovarian cancer patients not only experience adverse effects, but also eventually relapse. Therefore, additional therapeutic possibilities need to be explored to minimize adverse events and prolong progression-free and overall response rates in ovarian cancer patients. Currently, a revival in cancer drug discovery is devoted to identifying diagnostic and prognostic ovarian cancer biomarkers. However, the sensitivity and reliability of such biomarkers may be complicated by mutations in the BRCA1 or BRCA2 genes, diverse genetic risk factors, unidentified initiation and progression elements, molecular tumor heterogeneity and disease staging. There is thus a dire need to expand existing ovarian cancer therapies with broad-spectrum and individualized molecular targeted approaches. The aim of this review is to profile recent developments in our understanding of the interrelationships among selected ovarian tumor biomarkers, heterogeneous expression signatures and related molecular signal transduction pathways, and their translation into more efficacious targeted treatment rationales.
Collapse
Affiliation(s)
- Donavon Hiss
- Molecular Oncology Research Laboratory, Department of Medical BioSciences, University of the Western Cape, Bellville 7535, South Africa
| |
Collapse
|
31
|
Šereš M, Cholujová D, Bubenčíkova T, Breier A, Sulová Z. Tunicamycin depresses P-glycoprotein glycosylation without an effect on its membrane localization and drug efflux activity in L1210 cells. Int J Mol Sci 2011; 12:7772-84. [PMID: 22174631 PMCID: PMC3233437 DOI: 10.3390/ijms12117772] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2011] [Revised: 10/20/2011] [Accepted: 11/03/2011] [Indexed: 01/13/2023] Open
Abstract
P-glycoprotein (P-gp), also known as ABCB1, is a member of the ABC transporter family of proteins. P-gp is an ATP-dependent drug efflux pump that is localized to the plasma membrane of mammalian cells and confers multidrug resistance in neoplastic cells. P-gp is a 140-kDa polypeptide that is glycosylated to a final molecular weight of 170 kDa. Our experimental model used two variants of L1210 cells in which overexpression of P-gp was achieved: either by adaptation of parental cells (S) to vincristine (R) or by transfection with the human gene encoding P-gp (T). R and T cells were found to differ from S cells in transglycosylation reactions in our recent studies. The effects of tunicamycin on glycosylation, drug efflux activity and cellular localization of P-gp in R and T cells were examined in the present study. Treatment with tunicamycin caused less concentration-dependent cellular damage to R and T cells compared with S cells. Tunicamycin inhibited P-gp N-glycosylation in both of the P-gp-positive cells. However, tunicamycin treatment did not alter either the P-gp cellular localization to the plasma membrane or the P-gp transport activity. The present paper brings evidence that independently on the mode of P-gp expression (selection with drugs or transfection with a gene encoding P-gp) in L1210 cells, tunicamycin induces inhibition of N-glycosylation of this protein, without altering its function as plasma membrane drug efflux pump.
Collapse
Affiliation(s)
- Mário Šereš
- Institute of Molecular Physiology and Genetics, Centre of Excellence of the Slovak Research and Development Agency “BIOMEMBRANES2008”, Slovak Academy of Sciences, Vlárska 5, Bratislava 83334, Slovakia; E-Mails: (M.Š.); (T.B.)
| | - Dana Cholujová
- Cancer Research Institute, Slovak Academy of Sciences, Vlárska 7, Bratislava 83391, Slovakia; E-Mail:
| | - Tatiana Bubenčíkova
- Institute of Molecular Physiology and Genetics, Centre of Excellence of the Slovak Research and Development Agency “BIOMEMBRANES2008”, Slovak Academy of Sciences, Vlárska 5, Bratislava 83334, Slovakia; E-Mails: (M.Š.); (T.B.)
| | - Albert Breier
- Institute of Molecular Physiology and Genetics, Centre of Excellence of the Slovak Research and Development Agency “BIOMEMBRANES2008”, Slovak Academy of Sciences, Vlárska 5, Bratislava 83334, Slovakia; E-Mails: (M.Š.); (T.B.)
- Authors to whom correspondence should be addressed; E-Mails: (A.B.); (Z.S.); Tel.: +421-903-472-606 (A.B.); +421-903-246-360 (Z.S.); Fax: +421-2-5477-3666 (A.B.); +421-2-5477-3666 (Z.S.)
| | - Zdenka Sulová
- Institute of Molecular Physiology and Genetics, Centre of Excellence of the Slovak Research and Development Agency “BIOMEMBRANES2008”, Slovak Academy of Sciences, Vlárska 5, Bratislava 83334, Slovakia; E-Mails: (M.Š.); (T.B.)
- Authors to whom correspondence should be addressed; E-Mails: (A.B.); (Z.S.); Tel.: +421-903-472-606 (A.B.); +421-903-246-360 (Z.S.); Fax: +421-2-5477-3666 (A.B.); +421-2-5477-3666 (Z.S.)
| |
Collapse
|
32
|
Simeone L, Mangiapia G, Irace C, Di Pascale A, Colonna A, Ortona O, De Napoli L, Montesarchio D, Paduano L. Nucleolipid nanovectors as molecular carriers for potential applications in drug delivery. MOLECULAR BIOSYSTEMS 2011; 7:3075-86. [PMID: 21897988 DOI: 10.1039/c1mb05143a] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Novel thymidine- or uridine-based nucleolipids, containing one hydrophilic oligo(ethylene glycol) chain and one or two oleic acid residues (called ToThy, HoThy and DoHu), have been synthesized with the aim to develop bio-compatible nanocarriers for drug delivery and/or produce pro-drugs. Microstructural characterization of their aggregates has been determined in pure water and in pseudo-physiological conditions through DLS and SANS experiments. In all cases stable vesicles, with mean hydrodynamic radii ranging between 120 nm and 250 nm have been revealed. Biological validation of the nucleolipidic nanocarriers was ensured by evaluation of their toxicological profiles, performed by administration of the nanoaggregates to a panel of different cell lines. ToThy exhibited a weak cytotoxicity and, at high concentration, some ability to interfere with cell viability and/or proliferation. In contrast, DoHu and HoThy exhibited no toxicological relevance, behaving similarly to POPC-based liposomes, widely used for systemic drug delivery. Taken together, these results show nucleolipid-based nanocarriers as finely tunable, multi-functional self-assembling materials of interest for the in vivo transport of biomolecules or drugs.
Collapse
Affiliation(s)
- Luca Simeone
- Dipartimento di Chimica Organica e Biochimica, Università degli Studi di Napoli Federico II, Complesso Universitario di Monte S. Angelo, via Cinthia, 80126 Naples, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Hadley KC, Borrelli MJ, Lepock JR, McLaurin J, Croul SE, Guha A, Chakrabartty A. Multiphoton ANS fluorescence microscopy as an in vivo sensor for protein misfolding stress. Cell Stress Chaperones 2011; 16:549-61. [PMID: 21484286 PMCID: PMC3156256 DOI: 10.1007/s12192-011-0266-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 03/23/2011] [Accepted: 03/24/2011] [Indexed: 11/30/2022] Open
Abstract
The inability of cells to maintain protein folding homeostasis is implicated in the development of neurodegenerative diseases, malignant transformation, and aging. We find that multiphoton fluorescence imaging of 1-anilinonaphthalene-8-sulfonate (ANS) can be used to assess cellular responses to protein misfolding stresses. ANS is relatively nontoxic and enters live cells and cells or tissues fixed in formalin. In an animal model of Alzheimer's disease, ANS fluorescence imaging of brain tissue sections reveals the binding of ANS to fibrillar deposits of amyloid peptide (Aβ) in amyloid plaques and in cerebrovascular amyloid. ANS imaging also highlights non-amyloid deposits of glial fibrillary acidic protein in brain tumors. Cultured cells under normal growth conditions possess a number of ANS-binding structures. High levels of ANS fluorescence are associated with the endoplasmic reticulum (ER), Golgi, and lysosomes-regions of protein folding and degradation. Nuclei are virtually devoid of ANS binding sites. Additional ANS binding is triggered by hyperthermia, thermal lesioning, proteasome inhibition, and induction of ER stress. We also use multiphoton imaging of ANS binding to follow the in vivo recovery of cells from protein-damaging insults over time. We find that ANS fluorescence tracks with the binding of the molecular chaperone Hsp70 in compartments where Hsp70 is present. ANS highlights the sensitivity of specific cellular targets, including the nucleus and particularly the nucleolus, to thermal stress and proteasome inhibition. Multiphoton imaging of ANS binding should be a useful probe for monitoring protein misfolding stress in cells.
Collapse
Affiliation(s)
- Kevin C. Hadley
- Department of Medical Biophysics, University of Toronto. Ontario Cancer Institute, 101 College Street, Toronto, ON M5G 1L7 Canada
| | - Michael J. Borrelli
- Department of Radiology, University of Arkansas for Medical Sciences, 4301 W. Markham St., Little Rock, AR 72205 USA
| | - James R. Lepock
- Department of Medical Biophysics, University of Toronto. Ontario Cancer Institute, 101 College Street, Toronto, ON M5G 1L7 Canada
| | - JoAnne McLaurin
- Department of Laboratory Medicine and Pathobiology, Centre for Research in Neurodegenerative Diseases, University of Toronto, 6 Queen’s Park Cres. W., Toronto, ON M5S 3H2 Canada
| | - Sidney E. Croul
- Department of Laboratory Medicine and Pathobiology, University of Toronto, UHN Path 11E426 Toronto General Hospital, 200 Elizabeth St., Toronto, ON M5G 2C4 Canada
| | - Abhijit Guha
- Arthur and Sonia Labatt Brain Tumour Centre, Hospital for Sick Children’s Research Institute, Toronto, ON M5G 1X8 Canada
| | - Avijit Chakrabartty
- Campbell Family Institute for Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, ON Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON Canada
- Department of Biochemistry, University of Toronto, Toronto, ON Canada
- Toronto Medical Discovery Tower 4-307, MaRS Center 101 College Street, Toronto, ON M5G 1L7 Canada
| |
Collapse
|
34
|
Yap YHY, Say YH. Resistance against apoptosis by the cellular prion protein is dependent on its glycosylation status in oral HSC-2 and colon LS 174T cancer cells. Cancer Lett 2011; 306:111-9. [DOI: 10.1016/j.canlet.2011.02.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 02/23/2011] [Accepted: 02/24/2011] [Indexed: 11/25/2022]
|
35
|
A haploid genetic screen identifies the major facilitator domain containing 2A (MFSD2A) transporter as a key mediator in the response to tunicamycin. Proc Natl Acad Sci U S A 2011; 108:11756-65. [PMID: 21677192 DOI: 10.1073/pnas.1018098108] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tunicamycin (TM) inhibits eukaryotic asparagine-linked glycosylation, protein palmitoylation, ganglioside production, proteoglycan synthesis, 3-hydroxy-3-methylglutaryl coenzyme-A reductase activity, and cell wall biosynthesis in bacteria. Treatment of cells with TM elicits endoplasmic reticulum stress and activates the unfolded protein response. Although widely used in laboratory settings for many years, it is unknown how TM enters cells. Here, we identify in an unbiased genetic screen a transporter of the major facilitator superfamily, major facilitator domain containing 2A (MFSD2A), as a critical mediator of TM toxicity. Cells without MFSD2A are TM-resistant, whereas MFSD2A-overexpressing cells are hypersensitive. Hypersensitivity is associated with increased cellular TM uptake concomitant with an enhanced endoplasmic reticulum stress response. Furthermore, MFSD2A mutant analysis reveals an important function of the C terminus for correct intracellular localization and protein stability, and it identifies transmembrane helical amino acid residues essential for mediating TM sensitivity. Overall, our data uncover a critical role for MFSD2A by acting as a putative TM transporter at the plasma membrane.
Collapse
|
36
|
Labbé R, Caveney S, Donly C. Genetic analysis of the xenobiotic resistance-associated ABC gene subfamilies of the Lepidoptera. INSECT MOLECULAR BIOLOGY 2011; 20:243-256. [PMID: 21199020 DOI: 10.1111/j.1365-2583.2010.01064.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Some ATP-binding cassette (ABC) transporters of subfamilies B, C and G confer resistance to xenobiotics including insecticides. We identified genes of these subfamilies expressed by the lepidopterans Trichoplusia ni and Bombyx mori. The B. mori genome includes eight, six and 13 ABC-B, -C and -G genes, respectively, which encode P-glycoprotein, multidrug resistance protein, MRP, and breast cancer resistance protein, BCRP, homologues. Among the ABC-C and -G subfamilies, gene duplication contributes to protein diversity. We have identified three ABC-B and two ABC-C T. ni genes. Analyses of the T. ni MRP (TrnMRP) revealed unique features, including the potential for TrnMRP4 hyperglycosylation and the alternative splicing of TrnMRP1. Taken together, these attributes of moth multidrug resistance-associated ABCs may confer distinct functional capacities to xenobiotic efflux.
Collapse
Affiliation(s)
- R Labbé
- Department of Biology, University of Western Ontario, London, ON, Canada
| | | | | |
Collapse
|
37
|
Hiss DC, Gabriels GA. Implications of endoplasmic reticulum stress, the unfolded protein response and apoptosis for molecular cancer therapy. Part II: targeting cell cycle events, caspases, NF-κB and the proteasome. Expert Opin Drug Discov 2009; 4:907-21. [PMID: 23480539 DOI: 10.1517/17460440903055032] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Endoplasmic reticulum stress (ERS), the unfolded protein response (UPR) and apoptosis signal transduction pathways are fundamental to normal cellular homeostasis and survival, but are exploited by cancer cells to promote the cancer phenotype. OBJECTIVE Collateral activation of ERS and UPR role players impact on cell growth, cell cycle arrest or apoptosis, genomic stability, tumour initiation and progression, tumour aggressiveness and drug resistance. An understanding of these processes affords promising prospects for specific cancer drug targeting of the ERS, UPR and apoptotic pathways. METHOD This review (Part II of II) brings forward the latest developments relevant to the molecular connections among cell cycle regulators, caspases, NF-κB, and the proteasome with ERS and UPR signalling cascades, their functions in apoptosis induction, apoptosis resistance and oncogenesis, and how these relationships can be exploited for targeted cancer therapy. CONCLUSION Overall, ERS, the UPR and apoptosis signalling cascades (the molecular therapeutic targets) and the development of drugs that attack these targets signify a success story in cancer drug discovery, but a more reductionist approach is necessary to determine the precise molecular switches that turn on antiapoptotic and pro-apoptotic programmes.
Collapse
Affiliation(s)
- Donavon C Hiss
- Head, Molecular Oncology Research Programme, University of the Western Cape, Department of Medical BioSciences, Bellville, 7535, South Africa +27 21 959 2334 ; +27 959 1563 ;
| | | |
Collapse
|
38
|
Gissot A, Camplo M, Grinstaff MW, Barthélémy P. Nucleoside, nucleotide and oligonucleotide based amphiphiles: a successful marriage of nucleic acids with lipids. Org Biomol Chem 2008; 6:1324-33. [PMID: 18385837 DOI: 10.1039/b719280k] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Amphiphilic molecules based on nucleosides, nucleotides and oligonucleotides are finding more and more biotechnological applications. This Perspective highlights their synthesis, supramolecular organization as well as their applications in the field of biotechnology.
Collapse
Affiliation(s)
- Arnaud Gissot
- Université Victor Segalen, Bordeaux, F-33076, France
| | | | | | | |
Collapse
|
39
|
Hamaguchi J, Nakagawa H, Takahashi M, Kudo T, Kamiyama N, Sun B, Oshima T, Sato Y, Deguchi K, Todo S, Nishimura SI. Swainsonine reduces 5-fluorouracil tolerance in the multistage resistance of colorectal cancer cell lines. Mol Cancer 2007; 6:58. [PMID: 17883871 PMCID: PMC2071919 DOI: 10.1186/1476-4598-6-58] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Accepted: 09/21/2007] [Indexed: 01/11/2023] Open
Abstract
Background Drug resistance is a major problem in cancer chemotherapy. Acquisition of chemo-resistance not only reduces the effectiveness of drugs, but also promotes side effects and markedly reduces the patient's quality of life. However, a number of resistance mechanisms have been reported and are thought to be the reason for the difficulties in solving drug-resistance problems. Result To investigate the mechanisms of drug resistance, a set of cell lines with different levels of sensitivity and possessing different mechanisms of resistance to 5-fluorouracil (5-FU) was established from a colorectal cancer cell line. The expression of thymidylate synthase, orotic acid phosphoribosyltransferase and dihydropyrimidine dehydrogenase, which are well known to be related to drug resistance, differed among these cell lines, indicating that these cell lines acquired different resistance mechanisms. However, swainsonine, an inhibitor of N-glycan biosynthesis, reduced 5-FU-tolerance in all resistant cells, whereas the sensitivity of the parental cells was unchanged. Further analysis of the N-glycan profiles of all cell lines showed partial inhibition of biosynthesis and no cytotoxicity at the swainsonine dosage tested. Conclusion These observations suggest that N-linked oligosaccharides affect 5-FU resistance more widely than do drug-resistance related enzymes in colorectal cancer cells, and that the N-glycan could be a universal target for chemotherapy. Further, swainsonine may enhance the performance of chemotherapy by reducing tolerance.
Collapse
Affiliation(s)
- Jun Hamaguchi
- Department of General Surgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Hiroaki Nakagawa
- Graduate School of Advanced Life Science, Hokkaido University, Sapporo 001-0021, 001-0021, Japan
| | - Masato Takahashi
- Department of General Surgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Takeaki Kudo
- Department of General Surgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Naoya Kamiyama
- Department of Sensory Physiology, Asahikawa Medical College, Asahikawa 078-8510, Japan
| | - Bailong Sun
- Department of General Surgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Takahiro Oshima
- Department of General Surgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Yuji Sato
- Department of General Surgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Kisaburo Deguchi
- Graduate School of Advanced Life Science, Hokkaido University, Sapporo 001-0021, 001-0021, Japan
| | - Satoru Todo
- Department of General Surgery, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Shin-Ichiro Nishimura
- Graduate School of Advanced Life Science, Hokkaido University, Sapporo 001-0021, 001-0021, Japan
| |
Collapse
|