1
|
Xu M, Liu J, Yu J, Wang J, Li H, Zhong T, Hao Y, Li Z, Wang J, Huang X, Wang H, Tian Y, Zhao H, Wei Q, Zhang X. Methyl-β-cyclodextrin Enhances Tumor Cellular Uptake and Accumulation of α-Linolenic Acid-Paclitaxel Conjugate Nanoparticles. Mol Pharm 2024. [PMID: 39495317 DOI: 10.1021/acs.molpharmaceut.4c00190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Improving nanomedicine uptake by tumor cells is key to achieving intracellular drug delivery. In this study, we found that methyl-β-cyclodextrin (MβCD) can significantly promote the intracellular accumulation of nanoparticulated α-linolenic acid-paclitaxel conjugates (ALA-PTX NPs) via enhanced clathrin-mediated endocytosis and limited degradation in lysosomes. Our in vitro results indicated that MβCD not only reduced the plasma membrane cholesterol content and increased plasma membrane fluidity, leading to ALA-PTX NPs being more easily incorporated into the plasma membrane, further enhancing membrane fluidity and making the plasma membrane more susceptible to tensile deformation, forming intracellular vesicles to enhance ALA-PTX NP cellular uptake, but also destroyed lysosomes and then limited ALA-PTX NPs' degradation in lysosomes. In HepG2 tumor-bearing mice, MβCD was also able to enhance the antitumor activity of ALA-PTX NPs in vivo. Moreover, we found that MβCD specifically promoted PUFA-paclitaxel conjugate NP cellular uptake. The cellular uptake of PTX liposome which shares an endocytosis pathway with ALA-PTX NPs could be enhanced by MβCD combined with ALA or ALA-PTX NPs. Therefore, we suggested that MβCD combined with polyunsaturated fatty acid-conjugation would be an effective strategy for improving intracellular delivery of nanoparticulated chemotherapeutic drugs used for combination administration to enhance antitumor efficiency.
Collapse
Affiliation(s)
- Meiqi Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Junwei Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jianming Yu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jingwen Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hui Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ting Zhong
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yanli Hao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhuoyue Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jingru Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xu Huang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hui Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yubo Tian
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Heng Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qingchao Wei
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xuan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
2
|
Meng J, Yasui C, Shida M, Toshima K, Takahashi D. Designed Mannosylerythritol Lipid Analogues Exhibiting Both Selective Cytotoxicity Against Human Skin Cancer Cells and Recovery Effects on Damaged Skin Cells. Chemistry 2024; 30:e202401319. [PMID: 38802321 DOI: 10.1002/chem.202401319] [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: 04/03/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 05/29/2024]
Abstract
Mannosylerythritol lipids (MELs) are a class of amphipathic molecules bearing a hydrophilic 4-O-β-D-mannopyranosyl-D-erythritol skeleton. Here, we designed and synthesized four kinds of MEL analogues R-MEL-A ([2R,3S]-erythritol type), S-mannosylthreitol lipid (MTL)-A ([2S,3S]-threitol type), R-MTL-A ([2R,3R]-threitol type), and α-S-MEL-A ([2S,3R]-erythritol type) using our previously reported boron-mediated aglycon delivery (BMAD) method and a neighboring group assisted glycosylation method. The selective cytotoxicity of the target compounds against cancer cells was evaluated, with R-MTL-A showing the highest selective cytotoxicity against human skin squamous carcinoma HSC-5 cells. Our findings suggest that R-MTL-A induces necrosis-like cell death against HSC-5 cells by decreasing cell membrane fluidity. R-MTL-A also exhibits an efficient recovery effect on damaged skin cells, indicating that R-MTL-A has potential as a lead compound for new cosmeceuticals with both cancer cell-selective toxicity and recovery effects on damaged skin cells.
Collapse
Affiliation(s)
- Jikun Meng
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| | - Chihiro Yasui
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| | - Mai Shida
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| | - Kazunobu Toshima
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| | - Daisuke Takahashi
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, 223-8522, Japan
| |
Collapse
|
3
|
Alam MM, Alsenani NI, Abdelhamid AA, Ahmad A, Baothman OA, Hosawi SA, Altayeb H, Nadeem MS, Ahmad V, Nazreen S, Elhenawy AA. New paracetamol hybrids as anticancer and COX-2 inhibitors: Synthesis, biological evaluation and docking studies. Arch Pharm (Weinheim) 2024; 357:e2300340. [PMID: 37880869 DOI: 10.1002/ardp.202300340] [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] [Revised: 08/31/2023] [Accepted: 09/28/2023] [Indexed: 10/27/2023]
Abstract
Drug repurposing is an emerging field in drug development that has provided many successful drugs. In the current study, paracetamol, a known antipyretic and analgesic agent, was chemically modified to generate paracetamol derivatives as anticancer and anticyclooxygenase-2 (COX-2) agents. Compound 11 bearing a fluoro group was the best cytotoxic candidate with half-maximal inhibitory concentration (IC50 ) values ranging from 1.51 to 6.31 μM and anti-COX-2 activity with IC50 = 0.29 μM, compared to the standard drugs, doxorubicin and celecoxib. The cell cycle and apoptosis studies revealed that compound 11 possesses the ability to induce cell cycle arrest in the S phase and apoptosis in colon Huh-7 cells. These results were strongly supported by docking studies, which showed strong interactions with the amino acids of the COX-2 protein, and in silico pharmacokinetic predictions were found to be favorable for these newly synthesized paracetamol derivatives. It can be concluded that compound 11 could block cell growth and proliferation by inhibiting the COX-2 enzyme in cancer therapy.
Collapse
Affiliation(s)
- Mohammad Mahboob Alam
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| | - Nawaf I Alsenani
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| | - Antar A Abdelhamid
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
- Department of Chemistry, Faculty of Science, Sohag University, Sohag, Egypt
| | - Abrar Ahmad
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Othman A Baothman
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Salman A Hosawi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Hisham Altayeb
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Mohammad Shahid Nadeem
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Varish Ahmad
- Department of Health Information Technology, Faculty of Applied Studies, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Syed Nazreen
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| | - Ahmed A Elhenawy
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, Egypt
| |
Collapse
|
4
|
Mohsin NUA, Aslam S, Ahmad M, Irfan M, Al-Hussain SA, Zaki MEA. Cyclooxygenase-2 (COX-2) as a Target of Anticancer Agents: A Review of Novel Synthesized Scaffolds Having Anticancer and COX-2 Inhibitory Potentialities. Pharmaceuticals (Basel) 2022; 15:ph15121471. [PMID: 36558921 PMCID: PMC9783503 DOI: 10.3390/ph15121471] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer is a serious threat to human beings and is the second-largest cause of death all over the globe. Chemotherapy is one of the most common treatments for cancer; however, drug resistance and severe adverse effects are major problems associated with anticancer therapy. New compounds with multi-target inhibitory properties are targeted to surmount these challenges. Cyclooxygenase-2 (COX-2) is overexpressed in cancers of the pancreas, breast, colorectal, stomach, and lung carcinoma. Therefore, COX-2 is considered a significant target for the synthesis of new anticancer agents. This review discusses the biological activity of recently prepared dual anticancer and COX-2 inhibitory agents. The most important intermolecular interactions with the COX-2 enzyme have also been presented. Analysis of these agents in the active area of the COX-2 enzyme could guide the introduction of new lead compounds with extreme selectivity and minor side effects.
Collapse
Affiliation(s)
- Noor ul Amin Mohsin
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Sana Aslam
- Department of Chemistry, Government College Women University, Faisalabad 38000, Pakistan
| | - Matloob Ahmad
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
- Correspondence: (M.A.); (M.E.A.Z.)
| | - Muhammad Irfan
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Sami A. Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Magdi E. A. Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
- Correspondence: (M.A.); (M.E.A.Z.)
| |
Collapse
|
5
|
Szlasa W, Janicka N, Sauer N, Michel O, Nowak B, Saczko J, Kulbacka J. Chemotherapy and Physical Therapeutics Modulate Antigens on Cancer Cells. Front Immunol 2022; 13:889950. [PMID: 35874714 PMCID: PMC9299262 DOI: 10.3389/fimmu.2022.889950] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/06/2022] [Indexed: 12/29/2022] Open
Abstract
Cancer cells possess specific properties, such as multidrug resistance or unlimited proliferation potential, due to the presence of specific proteins on their cell membranes. The release of proliferation-related proteins from the membrane can evoke a loss of adaptive ability in cancer cells and thus enhance the effects of anticancer therapy. The upregulation of cancer-specific membrane antigens results in a better outcome of immunotherapy. Moreover, cytotoxic T-cells may also become more effective when stimulated ex-vivo toward the anticancer response. Therefore, the modulation of membrane proteins may serve as an interesting attempt in anticancer therapy. The presence of membrane antigens relies on various physical factors such as temperature, exposure to radiation, or drugs. Therefore, changing the tumor microenvironment conditions may lead to cancer cells becoming sensitized to subsequent therapy. This paper focuses on the therapeutic approaches modulating membrane antigens and enzymes in anticancer therapy. It aims to analyze the possible methods for modulating the antigens, such as pharmacological treatment, electric field treatment, photodynamic reaction, treatment with magnetic field or X-ray radiation. Besides, an overview of the effects of chemotherapy and immunotherapy on the immunophenotype of cancer cells is presented. Finally, the authors review the clinical trials that involved the modulation of cell immunophenotype in anticancer therapy.
Collapse
Affiliation(s)
- Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Natalia Janicka
- Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Natalia Sauer
- Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Olga Michel
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Bernadetta Nowak
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
6
|
De Mel JU, Gupta S, Harmon S, Stingaciu L, Roth EW, Siebenbuerger M, Bleuel M, Schneider GJ. Acetaminophen Interactions with Phospholipid Vesicles Induced Changes in Morphology and Lipid Dynamics. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:9560-9570. [PMID: 34328747 PMCID: PMC8359007 DOI: 10.1021/acs.langmuir.1c01458] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/12/2021] [Indexed: 05/28/2023]
Abstract
Acetaminophen (APAP) or paracetamol, despite its wide and common use for pain and fever symptoms, shows a variety of side effects, toxic effects, and overdose effects. The most common form of toxic effects of APAP is in the liver where phosphatidylcholine is the major component of the cell membrane with additional associated functionalities. Although this is the case, the effects of APAP on pure phospholipid membranes have been largely ignored. Here, we used 1,2-di-(octadecenoyl)-sn-glycero-3-phosphocholine (DOPC), a commonly found phospholipid in mammalian cell membranes, to synthesize large unilamellar vesicles to investigate how the incorporation of APAP changes the pure lipid vesicle structure, morphology, and fluidity at different concentrations. We used a combination of dynamic light scattering, small-angle neutron and X-ray scattering (SANS, SAXS), and cryo-TEM for structural characterization, and neutron spin-echo (NSE) spectroscopy to investigate the dynamics. We showed that the incorporation of APAP in the lipid bilayer significantly impacts the spherical phospholipid self-assembly in terms of its morphology and influences the lipid content in the bilayer, causing a decrease in bending rigidity. We observe a decrease in the number of lipids per vesicle by almost 28% (0.06 wt % APAP) and 19% (0.12 wt % APAP) compared to the pure DOPC (0 wt % APAP). Our results showed that the incorporation of APAP reduces the membrane rigidity by almost 50% and changes the spherical unilamellar vesicles into much more irregularly shaped vesicles. Although the bilayer structure did not show much change when observed by SAXS, NSE and cryo-TEM results showed the lipid dynamics change with the addition of APAP in the bilayer, which causes the overall decreased membrane rigidity. A strong effect on the lipid tail motion showed that the space explored by the lipid tails increases by a factor of 1.45 (for 0.06 wt % APAP) and 1.75 (for 0.12 wt % APAP) compared to DOPC without the drug.
Collapse
Affiliation(s)
- Judith U. De Mel
- Department
of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | - Sudipta Gupta
- Department
of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | - Sydney Harmon
- Department
of Chemistry, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Laura Stingaciu
- Neutron
Sciences Directorate, Oak Ridge National
Laboratory (ORNL), P.O.B 2008, 1 Bethel Valley Road, Oak Ridge, Tennessee 37831, United States
| | - Eric W. Roth
- Department
of Materials Science and Engineering and NUANCE Center, Northwestern University, 2220 Campus Drive, Evanston, Illinois 60208, United States
| | - Miriam Siebenbuerger
- Center
of Advanced Microstructures and Devices, Louisiana State University, 6980 Jefferson Highway, Baton Rouge, Louisiana 70806, United States
| | - Markus Bleuel
- NIST Center
for Neutron Research, National Institute
of Standards and Technology, Gaithersburg, Maryland 20899-8562, United States
| | - Gerald J. Schneider
- Department
of Chemistry, Louisiana State University, Baton Rouge, Louisiana 70803, United States
- Department
of Physics & Astronomy, Louisiana State
University, Baton
Rouge, Louisiana 70803, United States
| |
Collapse
|
7
|
Oliver TE, Piantavigna S, Andrews PC, Holt SA, Dillon CT. Interactions of Non-steroidal Anti-inflammatory Drugs and Their Bismuth Analogues (BiNSAIDs) with Biological Membrane Mimics at Physiological pH. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:1337-1352. [PMID: 33478220 DOI: 10.1021/acs.langmuir.0c02212] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Previous studies have demonstrated the potential for non-steroidal anti-inflammatory drugs (NSAIDs), in particular aspirin, to be used as chemopreventives for colorectal cancer; however, a range of unwanted gastrointestinal side effects limit their effectiveness. Due to the role of bismuth in the treatment of gastrointestinal disorders, it is hypothesized that bismuth-coordinated NSAIDs (BiNSAIDs) could be used to combat the gastrointestinal side effects of NSAIDs while still maintaining their chemopreventive potential. To further understand the biological activity of these compounds, the present study examined four NSAIDs, namely, tolfenamic acid (tolfH), aspirin (aspH), indomethacin (indoH), and mefenamic acid (mefH) and their analogous homoleptic BiNSAIDs ([Bi(L)3]n), to determine how these compounds interact with biological membrane mimics composed of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) or a mixture of POPC and cholesterol. Electrical impedance spectroscopy studies revealed that each of the NSAIDs and BiNSAIDs influenced membrane conductance, suggesting that temporary pore formation may play a key role in the previously observed cytotoxicity of tolfH and Bi(tolf)3. Quartz crystal microbalance with dissipation monitoring showed that all the compounds were able to interact with membrane mimics composed of solely POPC or POPC/cholesterol. Finally, neutron reflectometry studies showed changes in membrane thickness and composition. The location of the compounds within the bilayer could not be determined with certainty; however, a complex interplay of interactions governs the location of small molecules, such as NSAIDs, within lipid membranes. The charged nature of the parent NSAIDs means that interactions with the polar headgroup region are most likely with larger hydrophobic sections, potentially leading to deeper penetration.
Collapse
Affiliation(s)
- Tara E Oliver
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales 2522, Australia
- Molecular Horizons, University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Stefania Piantavigna
- School of Chemistry, Monash University, Clayton, Melbourne, Victoria 3800, Australia
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, New South Wales 2232, Australia
| | - Philip C Andrews
- School of Chemistry, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| | - Stephen A Holt
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation, Locked Bag 2001, Kirrawee DC, New South Wales 2232, Australia
| | - Carolyn T Dillon
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales 2522, Australia
- Molecular Horizons, University of Wollongong, Wollongong, New South Wales 2522, Australia
| |
Collapse
|
8
|
Soltani Rad MN, Behrouz S, Atashbasteh E, Hashemi SS. Butyl methyl imidazolium silica sulfate (BMIm)SS: A novel hybrid nano-catalyst for highly efficient synthesis of new 1,2-diol monoesters of ibuprofen as the novel prodrugs of ibuprofen having potent analgesic property. Bioorg Chem 2020; 107:104570. [PMID: 33373759 DOI: 10.1016/j.bioorg.2020.104570] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/04/2020] [Accepted: 12/16/2020] [Indexed: 11/25/2022]
Abstract
The fabrication, characterization of butyl methyl imidazolium silica sulfate [BMIm]SS as a novel nano hybrid catalyst and its application in synthesis of new ibuprofen (IBP) 1,2-diol mono esters were described. [BMIm]SS catalyzed the reaction of IBP with epoxides to afford the new IBP 1,2-diol mono esters in good to excellent yields. The products were tested in vivo for the analgesic properties on female mice using formalin test. The test results revealed that most compounds, in particular compounds 1h, 1k and 1o displayed potent analgesic activity compare to IBP as a reference drug. No mortality was observed due to the toxicity of the synthesized compounds. The docking analysis was conducted that confirmed the strong binding affinity of active compounds to active site of murine cyclooxygenase-2 (COX-2) enzyme compare to IBP. The in silico pharmacokinetic profile, drug likeness and toxicity predictions were carried out for all compounds which determined that 1h can be suggested as an appropriate future drug candidate.
Collapse
Affiliation(s)
- Mohammad Navid Soltani Rad
- Medicinal Chemistry Research Laboratory, Department of Chemistry, Shiraz University of Technology, Shiraz 71555-313, Iran.
| | - Somayeh Behrouz
- Medicinal Chemistry Research Laboratory, Department of Chemistry, Shiraz University of Technology, Shiraz 71555-313, Iran.
| | - Esmaeil Atashbasteh
- Medicinal Chemistry Research Laboratory, Department of Chemistry, Shiraz University of Technology, Shiraz 71555-313, Iran
| | - Seyedeh-Sara Hashemi
- Burn and Wound Healing Research Center, Division of Food and Nutrition, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
9
|
Shawky AM, Abourehab MA, Abdalla AN, Gouda AM. Optimization of pyrrolizine-based Schiff bases with 4-thiazolidinone motif: Design, synthesis and investigation of cytotoxicity and anti-inflammatory potency. Eur J Med Chem 2020; 185:111780. [DOI: 10.1016/j.ejmech.2019.111780] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/04/2019] [Accepted: 10/09/2019] [Indexed: 12/17/2022]
|
10
|
Hastie R, Brownfoot FC, Cannon P, Nguyen V, Tuohey L, Hannan NJ, Tong S, Kaitu'u-Lino TJ. Sulfasalazine decreases soluble fms-like tyrosine kinase-1 secretion potentially via inhibition of upstream placental epidermal growth factor receptor signalling. Placenta 2019; 87:53-57. [DOI: 10.1016/j.placenta.2019.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/27/2019] [Accepted: 09/09/2019] [Indexed: 10/26/2022]
|
11
|
Karpisheh V, Nikkhoo A, Hojjat-Farsangi M, Namdar A, Azizi G, Ghalamfarsa G, Sabz G, Yousefi M, Yousefi B, Jadidi-Niaragh F. Prostaglandin E2 as a potent therapeutic target for treatment of colon cancer. Prostaglandins Other Lipid Mediat 2019; 144:106338. [PMID: 31100474 DOI: 10.1016/j.prostaglandins.2019.106338] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/30/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023]
Abstract
Although colon cancer is one of the most important triggers of cancer related mortality, a few therapeutic options exist for this disease, including combination chemotherapy, anti-EGFR and anti-angiogenic agents. However, none of these therapeutics are fully effective for complete remission, and this issue needs further investigations, particularly in the patients with advanced disease. It has been shown that colon carcinogenesis process is associated with upregulation of prostaglandin (PG) levels. Moreover, conversion of pre-malignant cells to malignant was also related with increased generation of PGs in susceptible subjects. Among the prostanoids, PGE2 is the most important produced member which generated in high levels by colon tumor cells. Generation of PGE2 by action of cyclooxygenase (COX)-2 can promote growth and development, resistance to apoptosis, proliferation, invasion and metastasis, angiogenesis and drug resistance in colon cancer. Increased levels of PGE2 and COX-2 in colon cancer is reported by various investigators which was associated with disease progression. It is suggested that there is a positive feedback loop between COX-2 and PGE2, in which function of COX-2 induces generation of PGE2, and upregulation of PGE2 increases the expression of COX-2 in colon cancer. Although an existence of this feedback loop is well-documented, its precise mechanism, signaling pathways, and the particular E-type prostanoid (EP) receptor mediating this feedback are elusive. Therefore, it seems that targeting COX-2/PGE2/EP receptors may be supposed as a potent therapeutic strategy for treatment of colon cancer. In this review, we try to clarify the role of PGE2 in cancer progression and its targeting for treatment of colon cancer.
Collapse
Affiliation(s)
- Vahid Karpisheh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Nikkhoo
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden; The Persian Gulf Marine Biotechnology Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Afshin Namdar
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, T6G 2E1 Canada
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Gholamabas Sabz
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mehdi Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
12
|
Wang H, Han R. Zinc as a modulator of membrane stability parameters during prostate cancer. MINERVA BIOTECNOL 2018. [DOI: 10.23736/s1120-4826.18.02427-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
13
|
Chan WY, Lau PM, Yeung KW, Kong SK. The second generation tyrosine kinase inhibitor dasatinib induced eryptosis in human erythrocytes-An in vitro study. Toxicol Lett 2018; 295:10-21. [PMID: 29803841 DOI: 10.1016/j.toxlet.2018.05.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 05/04/2018] [Accepted: 05/23/2018] [Indexed: 12/12/2022]
Abstract
Dasatinib, a new tyrosine kinase inhibitor, is used clinically to kill chronic myelogenous leukemia and acute lymphoblastic leukemia through apoptosis. Obviously, anemia is developed in many patients receiving dasatinib for treatment. Until now, the mechanism for the cytotoxic effects of dasatinib in human erythrocytes is not fully understood. As many tyrosine kinases are found in human erythrocytes, it is therefore logical to hypothesize that dasatinib is able to induce apoptosis (or eryptosis) in human erythrocytes. True to our expectation, dasatinib inhibited tyrosine kinase and induced eryptosis in human erythrocytes with early denature of esterase, cell shrinkage, loss of membrane integrity with inside-out phosphatidylserine, increase in the cytosolic Ca2+ ion concentration ([Ca2+]i), caspase-3 activation and change in cellular redox state. Mechanistically, the rise of [Ca2+]i seems to be a key mediator in the dasatinib-mediated eryptosis because depletion of external Ca2+ could suppress the eryptotic effects. Also, dasatinib was able to reduce membrane fluidity in human RBCs. For the direct action on membrane, dasatinib permeabilized RBC ghosts in a way similar to digitonin. Taken together, we report here for the first time that dasatinib inhibited tyrosine kinase and induced eryptosis in human erythrocytes through Ca2+ loading and membrane permeabilization.
Collapse
Affiliation(s)
- Wai Yin Chan
- Programme of Biochemistry, School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Pui Man Lau
- Programme of Biochemistry, School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Ka Wing Yeung
- Programme of Biochemistry, School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Siu Kai Kong
- Programme of Biochemistry, School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong.
| |
Collapse
|
14
|
Pyrrolizine-5-carboxamides: Exploring the impact of various substituents on anti-inflammatory and anticancer activities. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2018; 68:251-273. [PMID: 31259695 DOI: 10.2478/acph-2018-0026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/18/2018] [Indexed: 01/05/2023]
Abstract
Towards optimization of the pyrrolizine-5-carboxamide scaffold, a novel series of six derivatives (4a-c and 5a-c) was prepared and evaluated for their anti-inflammatory, analgesic and anticancer activities. The (EZ)-7-cyano-6-((4-hydroxybenzylidene)amino)-N-(p-tolyl)-2,3-dihydro-1H-pyrrolizine-5-carboxamide (4b) and (EZ)-6-((4-chlorobenzylidene)-amino)-7-cyano-N-(p-tolyl)-2,3-dihydro-1H-pyrrolizine-5-carboxamide (5b) bearing the electron donating methyl group showed the highest anti-inflammatory activity while (EZ)-6-((4-chlorobenzylidene)amino)-7-cyano-N-phenyl-2,3-dihydro-1H-pyrrolizine-5-carboxamide (5a) was the most active analgesic agent. Cytotoxicity of the new compounds was evaluated against the MCF-7, A2780 and HT29 cancer cell lines using the MTT assay. Compounds 4b and 5b displayed high anticancer activity with IC50 in the range of 0.30-0.92 μmol L-1 against the three cell lines, while compound (EZ)-N-(4-chlorophenyl)-7-cyano-6-((4-hydroxybenzylidene)-amino)-2,3-dihydro-1H-pyrrolizine-5-carboxamide (4c) was the most active against MCF-7 cells (IC50 = 0.08 μmol L-1). Both the anti-inflammatory and anticancer activities of the new compounds were dependent on the type of substituent on the phenyl rings. Substituents with opposite electronic effects on the two phenyl rings are preferable for high cytotoxicity against the MCF-7 and A2780 cells. COX inhibition was suggested as the molecular mechanism of the anti-inflammatory activity of the new compounds while no clear relationship could be observed between COX inhibition and anticancer activity. Compound 5b, the most active against the three cell lines, induced dose-dependent early apoptosis with 0.1-0.2 % necrosis in MCF-7 cells. New compounds showed promising drug-likeness scores while the docking study revealed high binding affinity to COX-2. Taken together, this study highlighted the significant impact of the substituents on the anti-inflammatory and anticancer activity of pyrrolizine-5-carboxamides, which could help in further optimization to discover good leads for the treatment of cancer and inflammation.
Collapse
|
15
|
Jung SH, Lee M, Park HA, Lee HC, Kang D, Hwang HJ, Park C, Yu DM, Jung YR, Hong MN, Kim YN, Park HJ, Ko YG, Lee JS. Integrin α6β4-Src-AKT signaling induces cellular senescence by counteracting apoptosis in irradiated tumor cells and tissues. Cell Death Differ 2018; 26:245-259. [PMID: 29786073 DOI: 10.1038/s41418-018-0114-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 03/07/2018] [Accepted: 03/22/2018] [Indexed: 02/06/2023] Open
Abstract
Cellular senescence refers to an irreversible growth arrest that is triggered by various intrinsic and extrinsic stresses. Many recent studies have demonstrated that cellular senescence plays a crucial role in the regression of tumors exposed to ionizing radiation (IR), but the underlying mechanism remains unknown. Here we show that the activation of integrin β4 is essential for IR-induced cellular senescence. IR treatment results in the phosphorylation of integrin β4 at tyrosine residue 1510, leading to activation of the integrin α6β4-Src-AKT signaling pathway. We further reveal that the IR-induced phosphorylation of integrin β4 is regulated by the cholesterol content and membrane fluidity. We also find that IR-induced p53-caspase signaling is independent of integrin α6β4-Src-AKT signaling. Finally, we show that siRNA- or inhibitor-mediated blockade of integrin α6β4-Src-AKT signaling switches the post-irradiation fate from senescence to apoptosis, under p53 activated condition, in both cancer cells and tumor tissues of xenograft mice. On the basis of our finding that, integrin α6β4 is specifically activated and acts primarily to induce premature senescence in irradiated cancer cells, we propose that this integrin may be a valuable target and biomarker for radiotherapy.
Collapse
Affiliation(s)
- Seung Hee Jung
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea
| | - Minyoung Lee
- Radiation Non-clinical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Hyun A Park
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea
| | - Hyung Chul Lee
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea
| | - Donghee Kang
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea
| | - Hyun Jung Hwang
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea.,Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea
| | - Chanho Park
- Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea
| | - Dong-Min Yu
- Division of Life Sciences, Korea University, Seoul, Korea
| | - Yu Ri Jung
- Radiation Non-clinical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Mi-Na Hong
- Radiation Non-clinical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Yong-Nyun Kim
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang, Korea
| | - Heon Joo Park
- Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea.,Department of Microbiology, Inha University College of Medicine, Incheon, Korea
| | - Young-Gyu Ko
- Division of Life Sciences, Korea University, Seoul, Korea
| | - Jae-Seon Lee
- Department of Molecular Medicine, Inha University College of Medicine, Incheon, Korea. .,Hypoxia-related Disease Research Center, Inha University College of Medicine, Incheon, Korea.
| |
Collapse
|
16
|
Angelucci C, D'Alessio A, Iacopino F, Proietti G, Di Leone A, Masetti R, Sica G. Pivotal role of human stearoyl-CoA desaturases (SCD1 and 5) in breast cancer progression: oleic acid-based effect of SCD1 on cell migration and a novel pro-cell survival role for SCD5. Oncotarget 2018; 9:24364-24380. [PMID: 29849946 PMCID: PMC5966257 DOI: 10.18632/oncotarget.25273] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 04/05/2018] [Indexed: 01/01/2023] Open
Abstract
The influence of cell membrane fluidity on cancer progression has been established in different solid tumors. We previously reported that “cancer-associated fibroblasts” (CAFs) induced epithelial-mesenchymal transition and increased cell membrane fluidity and migration in poorly (MCF-7) and highly invasive (MDA-MB-231) breast cancer cells. We also found that the membrane fluidity regulating enzyme stearoyl-CoA desaturase 1 (SCD1) was upregulated in tumor cells co-cultured with CAFs and established its essential role for both intrinsic and CAF-driven tumor cell motility. Here, we further explored the mechanisms involved in the SCD1-based modulation of breast cancer cell migration and investigated the role of the other human SCD isoform, SCD5. We showed that the addition of oleic acid, the main SCD1 product, nullified the inhibitory effects produced on MCF-7 and MDA-MB-231 cell migration by SCD1 depletion (pharmacological or siRNA-based). Conversely, SCD5 seemed not involved in the regulation of cancer cell motility. Interestingly, a clear induction of necrosis was observed as a result of the depletion of SCD5 in MCF-7 cells, where the expression of SCD5 was found to be upregulated by CAFs. The necrotic effect was rescued by a 48-h treatment of cells with oleic acid. These results provide further insights in understanding the role of SCD1 in both intrinsic and CAF-stimulated mammary tumor cell migration, unveiling the metabolic basis of this desaturase-triggered effect. Moreover, our data suggest the ability of CAFs to promote the maintenance of tumor cell survival by the induction of SCD5 levels.
Collapse
Affiliation(s)
- Cristiana Angelucci
- Istituto di Istologia e Embriologia, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Alessio D'Alessio
- Istituto di Istologia e Embriologia, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Fortunata Iacopino
- Istituto di Istologia e Embriologia, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Gabriella Proietti
- Istituto di Istologia e Embriologia, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Alba Di Leone
- Unità Operativa di Chirurgia Senologica, Università Cattolica Del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli, Roma, Italia
| | - Riccardo Masetti
- Unità Operativa di Chirurgia Senologica, Università Cattolica Del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli, Roma, Italia
| | - Gigliola Sica
- Istituto di Istologia e Embriologia, Università Cattolica del Sacro Cuore, Roma, Italia
| |
Collapse
|
17
|
Gouda AM, Abdelazeem AH, Omar HA, Abdalla AN, Abourehab MAS, Ali HI. Pyrrolizines: Design, synthesis, anticancer evaluation and investigation of the potential mechanism of action. Bioorg Med Chem 2017; 25:5637-5651. [PMID: 28916158 DOI: 10.1016/j.bmc.2017.08.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/18/2017] [Accepted: 08/22/2017] [Indexed: 02/08/2023]
Abstract
A novel set of pyrrolizine-5-carboxamides has been synthesized and evaluated for their anticancer potential against human breast MCF-7, lung carcinoma A549 and hepatoma Hep3B cancer cell lines. Compound 10c was the most active against MCF-7 with IC50 value of 4.72µM, while compound 12b was the most active against A549 and Hep3B cell lines. Moreover, kinases/COXs inhibition and apoptosis induction were suggested as potential molecular mechanisms for the anticancer activity of the novel pyrrolizines based on their structural features. The new compounds significantly inhibited COX-1 and COX-2 with IC50 values in the ranges of 5.78-11.96µM and 0.1-0.78µM, respectively with high COX-2 selectivity over COX-1. Interestingly, the most potent compound in MTT assay, compound 12b, exhibited high inhibitory activity against COX-2 with selectivity index (COX-1/COX-2)>100. Meanwhile, compound 12b displayed weak to moderate inhibition of six kinases with inhibition% (7-20%) compared to imatinib (inhibition%=1-38%). The results of cell cycle analysis, annexin V PI/FITC apoptosis assay and caspase-3/7 assay revealed that compound 12b has the ability to induce apoptosis. The docking results of compound 12b into the active sites of COXs, ALK1 and Aurora kinases indicated that it fits nicely inside their active sites. Overall, the current study highlighted the significant anticancer activity of the newly synthesized pyrrolizines with a potential multi-targeted mechanism which could serve as a base for future studies and further structural optimization into potential anticancer agents.
Collapse
Affiliation(s)
- Ahmed M Gouda
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; Department of Medicinal Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - Ahmed H Abdelazeem
- Department of Medicinal Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Hany A Omar
- Sharjah Institute for Medical Research and College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; Department of Pharmacology, Medicinal and Aromatic Plants Research Institute, National Center for Research, Khartoum 2404, Sudan
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Hamed I Ali
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Egypt; Rangel College of Pharmacy, Health Science Center, Texas A&M University, Kingsville, TX 78363, United States.
| |
Collapse
|
18
|
Hsu HH, Lin YM, Shen CY, Shibu MA, Li SY, Chang SH, Lin CC, Chen RJ, Viswanadha VP, Shih HN, Huang CY. Prostaglandin E2-Induced COX-2 Expressions via EP2 and EP4 Signaling Pathways in Human LoVo Colon Cancer Cells. Int J Mol Sci 2017; 18:E1132. [PMID: 28587064 PMCID: PMC5485956 DOI: 10.3390/ijms18061132] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/12/2017] [Accepted: 05/15/2017] [Indexed: 12/11/2022] Open
Abstract
Metastasis is the most dangerous risk faced by patients with hereditary non-polyposis colon cancer (HNPCC). The expression of matrix metalloproteinases (MMPs) has been observed in several types of human cancers and regulates the efficacy of many therapies. Here, we show that treatment with various concentrations of prostaglandin E2 (PGE2; 0, 1, 5 or 10 μM) promotes the migration ability of the human LoVo colon cancer cell line. As demonstrated by mRNA and protein expression analyses, EP2 and EP4 are the major PGE2 receptors expressed on the LoVo cell membrane. The Phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/Akt cell survival pathway was upregulated by EP2 and EP4 activation. Following the activation of the PI3K/Akt pathway, β-catenin translocated into the nucleus and triggered COX2 transcription via LEF-1 and TCF-4 and its subsequent translation. COX2 expression correlated with the elevation in the migration ability of LoVo cells. The experimental evidence shows a possible mechanism by which PGE2 induces cancer cell migration and further suggests PGE2 to be a potential therapeutic target in colon cancer metastasis. On inhibition of PGE2, in order to determine the downstream pathway, the levels of PI3K/Akt pathway were suppressed and the β-catenin expression was also modulated. Inhibition of EP2 and EP4 shows that PGE2 induces protein expression of COX-2 through EP2 and EP4 receptors in LoVo colon cancer cells.
Collapse
Affiliation(s)
- Hsi-Hsien Hsu
- Division of Colorectal Surgery, Mackay Memorial Hospital, Freshwater 25160, Taiwan.
- Mackay Medicine, Nursing and Management College, Taipei 10449, Taiwan.
| | - Yueh-Min Lin
- Department of pathology, Changhua Christian Hospital, Changhua 500, Taiwan.
- Medical Technology, Jen-The Junior College of Medicine, Nursing and Management, Miaoli 35664, Taiwan.
| | - Chia-Yao Shen
- Department of Nursing, Mei Ho University, Pingguang Road, Pingtung 912, Taiwan.
| | - Marthandam Asokan Shibu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| | - Shin-Yi Li
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| | - Sheng-Huang Chang
- Tsao-Tun Psychiatric Center, Department of Health, Executive Yuan, Taipei 10058, Taiwan.
| | - Chien-Chung Lin
- Orthopaedic Department, Armed Forces General Hospital, Taichung 404, Taiwan.
| | - Ray-Jade Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | | | - Hui-Nung Shih
- Division of Colorectal Surgery, Mackay Memorial Hospital, Freshwater 25160, Taiwan.
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung 40402, Taiwan.
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan.
| |
Collapse
|
19
|
Rani I, Sharma B, Kumar S, Kaur S, Agnihotri N. Apoptosis mediated chemosensitization of tumor cells to 5-fluorouracil on supplementation of fish oil in experimental colon carcinoma. Tumour Biol 2017; 39:1010428317695019. [PMID: 28349837 DOI: 10.1177/1010428317695019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
5-Fluorouracil has been considered as a cornerstone therapy for colorectal cancer; however, it suffers from low therapeutic response rate and severe side effects. Therefore, there is an urgent need to increase the clinical efficacy of 5-fluorouracil. Recently, fish oil rich in n-3 polyunsaturated fatty acids has been reported to chemosensitize tumor cells to anti-cancer drugs. This study is designed to understand the underlying mechanisms of synergistic effect of fish oil and 5-fluorouracil by evaluation of tumor cell-associated markers such as apoptosis and DNA damage. The colon cancer was developed by administration of N,N-dimethylhydrazine dihydrochloride and dextran sulfate sodium salt. Further these animals were treated with 5-fluorouracil, fish oil, or a combination of both. In carcinogen-treated animals, a decrease in DNA damage and apoptotic index was observed. There was also a decrease in the expression of Fas, FasL, caspase 8, and Bax, and an increase in Bcl-2. In contrast, administration of 5-fluorouracil and fish oil as an adjuvant increased both DNA damage and apoptotic index by activation of both extrinsic and intrinsic apoptotic pathways as compared to the other groups. The increased pro-apoptotic effect by synergism of 5-fluorouracil and fish oil may be attributed to the incorporation of n-3 polyunsaturated fatty acids in membrane, which alters membrane fluidity in cancer cells. In conclusion, this study highlights that the induction of apoptotic pathway by fish oil may increase the susceptibility of tumors to chemotherapeutic regimens.
Collapse
Affiliation(s)
- Isha Rani
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Bhoomika Sharma
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Sandeep Kumar
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Satinder Kaur
- Department of Biochemistry, Panjab University, Chandigarh, India
| | | |
Collapse
|
20
|
Mitra D, Basu A, Das B, Jena AK, De A, Das M, Bhattacharya S, Samanta A. Gum odina: an emerging gut modulating approach in colorectal cancer prevention. RSC Adv 2017. [DOI: 10.1039/c7ra04077f] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This study is focused on Gum Odina (GO), a reported prebiotic in our earlier work, and its impact on colorectal cancer (CRC).
Collapse
Affiliation(s)
- Debmalya Mitra
- Division of Microbiology
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata
- India
| | - Abhishek Basu
- Department of Cancer Chemoprevention
- Chittaranjan National Cancer Institute
- Kolkata
- India
| | - Bhaskar Das
- Division of Microbiology
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata
- India
| | - Aditya Kr. Jena
- Division of Microbiology
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata
- India
| | - Arnab De
- Division of Microbiology
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata
- India
| | - Mousumi Das
- Division of Microbiology
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata
- India
| | - Sudin Bhattacharya
- Department of Cancer Chemoprevention
- Chittaranjan National Cancer Institute
- Kolkata
- India
| | - Amalesh Samanta
- Division of Microbiology
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata
- India
| |
Collapse
|
21
|
Zhu X, Li N, Wang Y, Ding L, Chen H, Yu Y, Shi X. Protective effects of quercetin on UVB irradiation-induced cytotoxicity through ROS clearance in keratinocyte cells. Oncol Rep 2016; 37:209-218. [PMID: 27840962 DOI: 10.3892/or.2016.5217] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 10/13/2016] [Indexed: 11/06/2022] Open
|
22
|
Manrique-Moreno M, Heinbockel L, Suwalsky M, Garidel P, Brandenburg K. Biophysical study of the non-steroidal anti-inflammatory drugs (NSAID) ibuprofen, naproxen and diclofenac with phosphatidylserine bilayer membranes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1858:2123-2131. [DOI: 10.1016/j.bbamem.2016.06.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 05/19/2016] [Accepted: 06/10/2016] [Indexed: 11/27/2022]
|
23
|
Wu X, Cao H, Zhao L, Song J, She Y, Feng Y. Metabolomic analysis of glycerophospholipid signatures of inflammation treated with non-steroidal anti-inflammatory drugs-induced-RAW264.7 cells using 1H NMR and U-HPLC/Q-TOF-MS. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1028:199-215. [DOI: 10.1016/j.jchromb.2016.06.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/17/2016] [Accepted: 06/18/2016] [Indexed: 01/29/2023]
|
24
|
Newcomb C, Sur S, Lee SS, Yu JM, Zhou Y, Snead ML, Stupp SI. Supramolecular Nanofibers Enhance Growth Factor Signaling by Increasing Lipid Raft Mobility. NANO LETTERS 2016; 16:3042-3050. [PMID: 27070195 PMCID: PMC4948975 DOI: 10.1021/acs.nanolett.6b00054] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/23/2016] [Indexed: 05/30/2023]
Abstract
The nanostructures of self-assembling biomaterials have been previously designed to tune the release of growth factors in order to optimize biological repair and regeneration. We report here on the discovery that weakly cohesive peptide nanostructures in terms of intermolecular hydrogen bonding, when combined with low concentrations of osteogenic growth factor, enhance both BMP-2 and Wnt mediated signaling in myoblasts and bone marrow stromal cells, respectively. Conversely, analogous nanostructures with enhanced levels of internal hydrogen bonding and cohesion lead to an overall reduction in BMP-2 signaling. We propose that the mechanism for enhanced growth factor signaling by the nanostructures is related to their ability to increase diffusion within membrane lipid rafts. The phenomenon reported here could lead to new nanomedicine strategies to mediate growth factor signaling for translational targets.
Collapse
Affiliation(s)
- Christina
J. Newcomb
- Department of Materials Science and Engineering Northwestern University, Evanston, Illinois 60208, United States
| | - Shantanu Sur
- Department of Materials Science and Engineering Northwestern University, Evanston, Illinois 60208, United States
| | - Sungsoo S. Lee
- Department of Materials Science and Engineering Northwestern University, Evanston, Illinois 60208, United States
| | - Jeong Min Yu
- Simpson
Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois 60611, United States
| | - Yan Zhou
- Center for Craniofacial Molecular Biology,
Herman Ostrow School of Dentistry of USC, The University of Southern California, Los Angeles, California 90033, United States
| | - Malcolm L. Snead
- Center for Craniofacial Molecular Biology,
Herman Ostrow School of Dentistry of USC, The University of Southern California, Los Angeles, California 90033, United States
| | - Samuel I. Stupp
- Department of Materials Science and Engineering Northwestern University, Evanston, Illinois 60208, United States
- Simpson
Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois 60611, United States
- Department
of Biomedical Engineering, Northwestern
University, Evanston, Illinois 60208, United
States
- Department of Chemistry, Northwestern
University, Evanston, Illinois 60208, United
States
- Department of Medicine, Northwestern
University, Chicago, Illinois 60611, United
States
| |
Collapse
|
25
|
Bakirel T, Alkan FÜ, Üstüner O, Çinar S, Yildirim F, Erten G, Bakirel U. Synergistic growth inhibitory effect of deracoxib with doxorubicin against a canine mammary tumor cell line, CMT-U27. J Vet Med Sci 2016; 78:657-68. [PMID: 26822118 PMCID: PMC4873858 DOI: 10.1292/jvms.15-0387] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cyclooxygenase (COX) inhibitors have been shown to exert anti-angiogenic and anti-tumor
activities on many types of malignant tumors. These anticancer properties make it
worthwhile to examine the possible benefit of combining COX inhibitors with other
anti-cancer agents. In the present study, we evaluated the potential of deracoxib (DER) in
potentiating antitumor activity of doxorubicin (DOX) in canine mammary carcinoma cells
(CMT-U27). DER (50–250 µM) enhanced the antiproliferative activity of DOX
by reducing the IC50 (approximately 3- to 3.5 fold). Interaction analysis of
the data showed that combinations of DOX at 0.9 µM with DER (100–250
µM) produced synergism in the CMT-U27 cell line, with a ratio index
ranging from 1.98 to 2.33. In additional studies identifying the mechanism of observed
synergistic effect, we found that DER strongly potentiated DOX-caused
G0/G1 arrest in cell cycle progression. Also, DER (100–250
µM) augmented apoptosis induction with approximately 1.35- and 1.37-
fold increases in apoptotic response caused by DOX in the cells. DER enhanced the
antiproliferative effect of DOX in conjunction with induction of apoptosis by modulation
of Bcl-2 expression and changes in the cell cycle of the CMT-U27 cell line. Although the
exact molecular mechanism of the alterations in the cell cycle and apoptosis observed with
DER and DOX combinations require further investigations, the results suggest that the
synergistic effect of DOX and DER combinations in CMT therapy may be achieved at
relatively lower doses of DOX with lesser side effects. Therefore, combining DER with DOX
may prove beneficial in the clinical treatment of canine mammary cancer.
Collapse
Affiliation(s)
- Tülay Bakirel
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Istanbul University, Istanbul, 34320, Turkey
| | | | | | | | | | | | | |
Collapse
|
26
|
Che XH, Chen CL, Ye XL, Weng GB, Guo XZ, Yu WY, Tao J, Chen YC, Chen X. Dual inhibition of COX-2/5-LOX blocks colon cancer proliferation, migration and invasion in vitro. Oncol Rep 2015; 35:1680-8. [PMID: 26707712 DOI: 10.3892/or.2015.4506] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 11/06/2015] [Indexed: 11/06/2022] Open
Abstract
Inflammation is emerging as a new hallmark of cancer. Arachidonic acid (AA) metabolism, the family of cyclooxygenases (COXs) and lipoxygenase (LOX) play important roles in AA-related inflammatory cascades. In 94 colorectal cancer samples collected from the Han population, the immunohistochemical results indicated that 68% of the patients with colorectal cancer had a co-expression of both COX-2 and 5-LOX, while both displayed low expression in the matched normal tissues. In cell lines, three colorectal cancer cell lines exhibited high expression of COX-2 and 5-LOX. During stable silencing of the expression of COX-2 or 5-LOX in LoVo cancer cells, we found that downregulation of either COX-2 or 5-LOX significantly diminished the growth, migration and invasion of the colon cancer cells and specifically, downregulation of COX-2 could elicit upregulation of 5-LOX protein and vice versa. The above results suggested that the simultaneous blocking of COX-2 and 5-LOX activity may bring more potential benefits in managing the progression of colon cancer. Therefore, we sought to explore the effectiveness of a dual COX-2/5-LOX inhibitor darbufelone on the proliferation, migration, invasion and apoptosis of colon cancer cells, as well as the underlying mechanism of action. The results indicated that darbufelone significantly decreased the proliferative and invasive abilities of the colon cancer cells, in a dose-dependent manner. During the study of the related mechanisms, we found an upregulation of p27 and downregulation of cyclin D1 as well as CDK4 after darbufelone treatment, which indicated that darbufelone could arrest the cell cycle of LoVo cells at the G0/G1 phase. Furthermore, the activation of caspase-3 and -9, upregulation of Bax and downregulation of Bcl-2 demonstrated the occurrence of apoptosis by darbufelone. Finally, darbufelone also prevented the migration and invasion of LoVo cells, which may be ascribed to the upregulation of E-cadherin and ZO-1. In summary, our data suggest that the inhibition of both COX-2/5-LOX may be an effective therapeutic approach for colon cancer management, particularly for those patients with high expression of COX-2/5-LOX.
Collapse
Affiliation(s)
- Xiao-Hang Che
- Department of Drugs and Pharmacology, Ningbo Institute of Medical Sciences, Ningbo, Zhejiang 315020, P.R. China
| | - Chun-Lin Chen
- College of Chemistry and Bio-Engineering, Yichun University, Yichun, Jiangxi 336000, P.R. China
| | - Xiao-Lei Ye
- Department of Drugs and Pharmacology, Ningbo Institute of Medical Sciences, Ningbo, Zhejiang 315020, P.R. China
| | - Guo-Bin Weng
- Department of Urology, Ningbo Urinary Kidney Disease Hospital, Ningbo, Zhejiang 315100, P.R. China
| | - Xian-Zhi Guo
- Department of Medical Oncology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, P.R. China
| | - Wen-Ying Yu
- Ningbo Clinical and Pathology Diagnostic Center, Ningbo, Zhejiang 315021, P.R. China
| | - Jin Tao
- Department of Drugs and Pharmacology, Ningbo Institute of Medical Sciences, Ningbo, Zhejiang 315020, P.R. China
| | - Yi-Chen Chen
- Department of Drugs and Pharmacology, Ningbo Institute of Medical Sciences, Ningbo, Zhejiang 315020, P.R. China
| | - Xiaodong Chen
- Department of Drugs and Pharmacology, Ningbo Institute of Medical Sciences, Ningbo, Zhejiang 315020, P.R. China
| |
Collapse
|
27
|
Membrane Interactions of Phytochemicals as Their Molecular Mechanism Applicable to the Discovery of Drug Leads from Plants. Molecules 2015; 20:18923-66. [PMID: 26501254 PMCID: PMC6332185 DOI: 10.3390/molecules201018923] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 10/11/2015] [Accepted: 10/14/2015] [Indexed: 02/02/2023] Open
Abstract
In addition to interacting with functional proteins such as receptors, ion channels, and enzymes, a variety of drugs mechanistically act on membrane lipids to change the physicochemical properties of biomembranes as reported for anesthetic, adrenergic, cholinergic, non-steroidal anti-inflammatory, analgesic, antitumor, antiplatelet, antimicrobial, and antioxidant drugs. As well as these membrane-acting drugs, bioactive plant components, phytochemicals, with amphiphilic or hydrophobic structures, are presumed to interact with biological membranes and biomimetic membranes prepared with phospholipids and cholesterol, resulting in the modification of membrane fluidity, microviscosity, order, elasticity, and permeability with the potencies being consistent with their pharmacological effects. A novel mechanistic point of view of phytochemicals would lead to a better understanding of their bioactivities, an insight into their medicinal benefits, and a strategic implication for discovering drug leads from plants. This article reviews the membrane interactions of different classes of phytochemicals by highlighting their induced changes in membrane property. The phytochemicals to be reviewed include membrane-interactive flavonoids, terpenoids, stilbenoids, capsaicinoids, phloroglucinols, naphthodianthrones, organosulfur compounds, alkaloids, anthraquinonoids, ginsenosides, pentacyclic triterpene acids, and curcuminoids. The membrane interaction’s applicability to the discovery of phytochemical drug leads is also discussed while referring to previous screening and isolating studies.
Collapse
|
28
|
Dong Y, Wu G. Azadircta indica as a modulator of membrane stability parameters and surface changes during 1,2 dimethylhydrazine-induced colorectal carcinogenesis. ACTA ACUST UNITED AC 2015; 63:204-9. [PMID: 26271242 DOI: 10.1016/j.patbio.2015.06.003] [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: 04/04/2015] [Accepted: 06/25/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE The aim of the present study was to study the modulatory potential of Azadirta indica on colonic surface abnormalities and membrane fluidity changes following 1,2 dimethylhydrazine-induced [DMH] colon carcinogenesis. MATERIALS AND METHODS Brush border membranes [BBM] were isolated from the colon of rats and the viscosity as well as fluidity parameters were assessed by using the membrane extrinsic fluorophore pyrene. RESULTS DMH treatment resulted in a significant increase in lipid peroxidation [LPO]. Reduced glutathione levels [GSH] and the activities of glutathione reductase [GR], glutathione transferase [GST], superoxide dismutase [SOD], catalase [CAT] and glutathione peroxidase [GPx] were found to be significantly decreased following DMH treatment. On the other hand, supplementation with AI, DMH-treated rats resulted in a significant decrease in the levels of lipid peroxidation but caused a significant increase in the levels of GSH as well in the activities of GR, GST, SOD, CAT and GPx. The results further demonstrated a marked decrease in membrane microviscosity following DMH treatment. On the other hand, a significant increase was observed in the excimer/monomer ratio and fluidity parameter of DMH-treated rats when compared to normal control rats. However, the alterations in membrane microviscosity and the fluidity parameters were significantly restored following A. indica treatment. Further, histological as well as colon surface alterations were also observed following DMH treatment, which however were greatly prevented upon AI co-administration. CONCLUSIONS The study, therefore, concludes that A. indica proves to be useful in modulating the colonic surface abnormalities and membrane stability following DMH-induced colon carcinogenesis.
Collapse
Affiliation(s)
- Y Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China; Department of Oncology, Xiangyang Hospital Affiliated to Hubei University of Medicine, Xiangyang, Hubei 441000, P.R. China
| | - G Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China.
| |
Collapse
|
29
|
Liggett JL, Zhang X, Eling TE, Baek SJ. Anti-tumor activity of non-steroidal anti-inflammatory drugs: cyclooxygenase-independent targets. Cancer Lett 2014; 346:217-24. [PMID: 24486220 DOI: 10.1016/j.canlet.2014.01.021] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/21/2014] [Accepted: 01/22/2014] [Indexed: 12/27/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are used extensively for analgesic and antipyretic treatments. In addition, NSAIDs reduce the risk and mortality to several cancers. Their mechanisms in anti-tumorigenesis are not fully understood, but both cyclooxygenase (COX)-dependent and -independent pathways play a role. We and others have been interested in elucidating molecular targets of NSAID-induced apoptosis. In this review, we summarize updated literature regarding cellular and molecular targets modulated by NSAIDs. Among those NSAIDs, sulindac sulfide and tolfenamic acid are emphasized in this review because these two drugs have been well investigated for their anti-tumorigenic activity in many different types of cancer.
Collapse
Affiliation(s)
- Jason L Liggett
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996 USA
| | - Xiaobo Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Thomas E Eling
- Laboratory of Molecular Carcinogenesis, National Institutes of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Seung Joon Baek
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996 USA.
| |
Collapse
|
30
|
Pereira-Leite C, Nunes C, Reis S. Interaction of nonsteroidal anti-inflammatory drugs with membranes: in vitro assessment and relevance for their biological actions. Prog Lipid Res 2013; 52:571-84. [PMID: 23981364 DOI: 10.1016/j.plipres.2013.08.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 08/01/2013] [Accepted: 08/16/2013] [Indexed: 12/12/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are among the most commonly used drugs in the world due to their anti-inflammatory, analgesic and antipyretic properties. Nevertheless, the consumption of these drugs is still associated with the occurrence of a wide spectrum of adverse effects. Regarding the major role of membranes in cellular events, the hypothesis that the biological actions of NSAIDs may be related to their effect at the membrane level has triggered the in vitro assessment of NSAIDs-membrane interactions. The use of membrane mimetic models, cell cultures, a wide range of experimental techniques and molecular dynamics simulations has been providing significant information about drugs partition and location within membranes and also about their effect on diverse membrane properties. These studies have indeed been providing evidences that the effect of NSAIDs at membrane level may be an additional mechanism of action and toxicity of NSAIDs. In fact, the pharmacokinetic properties of NSAIDs are closely related to the ability of these drugs to interact and overcome biological membranes. Moreover, the therapeutic actions of NSAIDs may also result from the indirect inhibition of cyclooxygenase due to the disturbing effect of NSAIDs on membrane properties. Furthermore, increasing evidences suggest that the disordering effects of these drugs on membranes may be in the basis of the NSAIDs-induced toxicity in diverse organ systems. Overall, the study of NSAIDs-membrane interactions has proved to be not only important for the better understanding of their pharmacological actions, but also for the rational development of new approaches to overcome NSAIDs adverse effects.
Collapse
Affiliation(s)
- Catarina Pereira-Leite
- REQUIMTE, Laboratório de Química Aplicada, Departamento de Ciências Químicas, Faculdade de Farmácia da Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | | | | |
Collapse
|
31
|
McArt DG, Dunne PD, Blayney JK, Salto-Tellez M, Van Schaeybroeck S, Hamilton PW, Zhang SD. Connectivity Mapping for Candidate Therapeutics Identification Using Next Generation Sequencing RNA-Seq Data. PLoS One 2013; 8:e66902. [PMID: 23840550 PMCID: PMC3694114 DOI: 10.1371/journal.pone.0066902] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 05/14/2013] [Indexed: 12/29/2022] Open
Abstract
The advent of next generation sequencing technologies (NGS) has expanded the area of genomic research, offering high coverage and increased sensitivity over older microarray platforms. Although the current cost of next generation sequencing is still exceeding that of microarray approaches, the rapid advances in NGS will likely make it the platform of choice for future research in differential gene expression. Connectivity mapping is a procedure for examining the connections among diseases, genes and drugs by differential gene expression initially based on microarray technology, with which a large collection of compound-induced reference gene expression profiles have been accumulated. In this work, we aim to test the feasibility of incorporating NGS RNA-Seq data into the current connectivity mapping framework by utilizing the microarray based reference profiles and the construction of a differentially expressed gene signature from a NGS dataset. This would allow for the establishment of connections between the NGS gene signature and those microarray reference profiles, alleviating the associated incurring cost of re-creating drug profiles with NGS technology. We examined the connectivity mapping approach on a publicly available NGS dataset with androgen stimulation of LNCaP cells in order to extract candidate compounds that could inhibit the proliferative phenotype of LNCaP cells and to elucidate their potential in a laboratory setting. In addition, we also analyzed an independent microarray dataset of similar experimental settings. We found a high level of concordance between the top compounds identified using the gene signatures from the two datasets. The nicotine derivative cotinine was returned as the top candidate among the overlapping compounds with potential to suppress this proliferative phenotype. Subsequent lab experiments validated this connectivity mapping hit, showing that cotinine inhibits cell proliferation in an androgen dependent manner. Thus the results in this study suggest a promising prospect of integrating NGS data with connectivity mapping.
Collapse
Affiliation(s)
- Darragh G. McArt
- Centre for Cancer Research and Cell Biology(CCRCB), Queen's University Belfast, Belfast, United Kingdom
| | - Philip D. Dunne
- Centre for Cancer Research and Cell Biology(CCRCB), Queen's University Belfast, Belfast, United Kingdom
| | - Jaine K. Blayney
- Centre for Cancer Research and Cell Biology(CCRCB), Queen's University Belfast, Belfast, United Kingdom
| | - Manuel Salto-Tellez
- Centre for Cancer Research and Cell Biology(CCRCB), Queen's University Belfast, Belfast, United Kingdom
| | - Sandra Van Schaeybroeck
- Centre for Cancer Research and Cell Biology(CCRCB), Queen's University Belfast, Belfast, United Kingdom
| | - Peter W. Hamilton
- Centre for Cancer Research and Cell Biology(CCRCB), Queen's University Belfast, Belfast, United Kingdom
| | - Shu-Dong Zhang
- Centre for Cancer Research and Cell Biology(CCRCB), Queen's University Belfast, Belfast, United Kingdom
- * E-mail:
| |
Collapse
|
32
|
Pereira-Leite C, Carneiro C, Soares JX, Afonso C, Nunes C, Lúcio M, Reis S. Biophysical characterization of the drug–membrane interactions: The case of propranolol and acebutolol. Eur J Pharm Biopharm 2013; 84:183-91. [DOI: 10.1016/j.ejpb.2012.12.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 12/11/2012] [Indexed: 11/16/2022]
|
33
|
Tekpli X, Holme JA, Sergent O, Lagadic-Gossmann D. Role for membrane remodeling in cell death: Implication for health and disease. Toxicology 2013; 304:141-57. [DOI: 10.1016/j.tox.2012.12.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 11/29/2012] [Accepted: 12/20/2012] [Indexed: 12/31/2022]
|
34
|
Selenium as a modulator of membrane stability parameters and surface changes during the initiation phase of 1,2-dimethylhydrazine induced colorectal carcinogenesis. Mol Cell Biochem 2012; 369:119-26. [PMID: 22752389 DOI: 10.1007/s11010-012-1374-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 06/20/2012] [Indexed: 01/02/2023]
|