1
|
Pourabdollah-Kaleybar V, Pourabdollah-Kaleybar P, Eskandani M, Nazemiyeh H. Toxicity of bioactive compounds from Halocnemum strobilaceum against A549 lung cancer cells. Toxicon 2025; 253:108186. [PMID: 39608464 DOI: 10.1016/j.toxicon.2024.108186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/10/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024]
Abstract
This study investigates the toxicity of bioactive compounds isolated from the halophytic plant Halocnemum strobilaceum against A549 lung cancer cells. Sequential extraction using petroleum ether, chloroform, and methanol yielded various fractions, with the petroleum ether extract demonstrating the highest cytotoxicity. Through bioassay-guided fractionation and isolation techniques, including vacuum liquid chromatography and column chromatography, three compounds were identified: (1) Di(2-ethylhexyl) phthalate, (2) isorhamnetin-3-glucoside, and (3) quercetin-3-glucoside. The cytotoxic effects of these compounds were assessed using the MTT assay, revealing significant toxicity on A549 cells, with quercetin-3-glucoside exhibiting 78% cytotoxicity and isorhamnetin-3-glucoside showing 69% cytotoxicity at a concentration of 100 μg/mL. These findings suggest that the toxic effects of H. strobilaceum may be attributed to the presence of bioactive compounds, such as flavonoids and polyphenols, known for their antioxidant and free-radical scavenging capabilities. This highlights the potential of H. strobilaceum as a source of novel anti-cancer agents, warranting further studies to elucidate the mechanisms of action and explore therapeutic applications.
Collapse
Affiliation(s)
- Vahid Pourabdollah-Kaleybar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parinaz Pourabdollah-Kaleybar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmacognosy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hossein Nazemiyeh
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmacognosy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Dadashi H, Mashinchian M, Karimian-Shaddel A, Mohabbat A, Vandghanooni S, Eskandani M, Jahanban-Esfahlan R. Chitosan nanoparticles loaded with metformin and digoxin synergistically inhibit MCF-7 breast cancer cells through suppression of NOTCH-1 and HIF-1α gene expression. Int J Biol Macromol 2025; 287:138418. [PMID: 39645125 DOI: 10.1016/j.ijbiomac.2024.138418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 11/12/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
This study investigated the potential anticancer efficacy of co-treating the MCF-7 breast cancer cell line with chitosan nanoparticles (Cs NPs) loaded with metformin (Met) and digoxin (Dig). The Cs NPs had a size range of 90.6-148.7 nm and a zeta potential of +11.7 to +11.9 mV, indicating a positive surface charge. Notably, the Cs NPs demonstrated high encapsulation efficiencies, with values of 90.97 ± 5.14 % for Met and 92.12 ± 3.81 % for Dig, indicating effective loading of both drugs. The results revealed that the co-delivery of Met and Dig via Cs NPs significantly enhanced the anticancer efficacy, outperforming the treatment with individual free drugs or their combination, thereby demonstrating the potential benefits of nanoparticle-mediated co-administration. The drugs-loaded Cs NPs induced a marked increase in apoptosis in MCF-7 cells, with a cell death rate of 67.56 %, and significantly reduced mammosphere size by 48.08 %, thereby demonstrating a superior therapeutic efficacy compared to treatment with individual free drugs or their combination. Notably, the drug-loaded Cs NPs exhibited potent anti-migratory and anti-angiogenic effects, significantly inhibiting cell migration and new blood vessel formation, which may contribute to overcoming the inherent resistance of tumors to conventional therapies. Mechanistically, the co-treatment with drugs-loaded Cs NPs was found to downregulate the expression of NOTCH-1 and HIF-1α, two key transcription factors involved in tumor cell survival and adaptation, suggesting that their inhibition is a crucial component of the therapeutic efficacy of this treatment strategy. Collectively, the findings of this study suggest that the co-delivery of Met and Dig via chitosan Cs NPs represents a promising therapeutic strategy for breast cancer, as it effectively targets key pathways involved in tumor growth and progression, and underscores the potential of Cs NPs as a versatile platform for cancer therapy.
Collapse
Affiliation(s)
- Hamed Dadashi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Mashinchian
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Karimian-Shaddel
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aria Mohabbat
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz university of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Bunev AS, Shetnev AA, Shemchuk OS, Kozhukhov PK, Sharonova TV, Tyuryaeva II, Khotin MG, Ageev SV, Kholmurodova DK, Rizaev JA, Semenov KN, Sharoyko VV. Combination of Carbonic Anhydrase Isoform IX Inhibitors and Gefitinib Suppresses on the Invasive Potential of Non-Small Cell Lung Cancer Cells. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:2227-2237. [PMID: 39865035 DOI: 10.1134/s0006297924120113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 01/28/2025]
Abstract
Human carbonic anhydrase IX (CAIX) plays a key role in maintaining pH homeostasis of malignant neoplasms, thus creating a favorable microenvironment for the growth, invasion, and metastasis of tumor cells. Recent studies have established that inhibition of CAIX expressed on the surface of tumor cells significantly increases the efficacy of classical chemotherapeutic agents and makes it possible to suppress the resistance of tumor cells to chemotherapy, as well as to increase their sensitivity to drugs (in particular, to reduce the required dose of cytostatic agents). In this work, we studied the ability of new CAIX inhibitors based on substituted 1,2,4-oxadiazole-containing primary aromatic sulfonamides, to potentiate the cytostatic effect of gefitinib (selective inhibitor of epidermal growth factor receptor tyrosine kinase domain) under hypoxic conditions. We investigated a combined effect of gefitinib and CAIX inhibitors 4-(3-phenyl-1,2,4-oxadiazol-5-yl)thiophene-2-sulfonamide (1), 4-(5-(thiophene-3-yl)-1,2,4-oxadiazol-3-yl)benzenesulfonamide (2), 4-(3-(pyridin-2-yl)-1,2,4-oxadiazol-5-yl)thiophene-2-sulfonamide (3), and 4-(5-methyl-1,2,4-oxadiazol-3-yl)benzenesulfonamide (4) on gefitinib cytotoxicity, cell proliferation, activation of caspases-3/7, and cell cycle control in human lung adenocarcinoma A549 cells. It was found that the combinations of compounds 1 and 2 with gefitinib suppressed the invasive potential of A549 cells. Compound 1 had the greatest effect and can be considered as a promising candidate for further research.
Collapse
Affiliation(s)
- Alexander S Bunev
- Medicinal Chemistry Center, Togliatti State University, Togliatti, 445020, Russia
| | - Anton A Shetnev
- Institute of Biophysics of the Future, Dolgoprudny, Moscow Region, 141701, Russia
| | - Olga S Shemchuk
- Pavlov First St. Petersburg State Medical University, St. Petersburg, 197022, Russia
| | - Pavel K Kozhukhov
- Pavlov First St. Petersburg State Medical University, St. Petersburg, 197022, Russia
| | | | - Irina I Tyuryaeva
- St. Petersburg State University, St. Petersburg, 199034, Russia
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
| | - Mikhail G Khotin
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
| | - Sergey V Ageev
- Pavlov First St. Petersburg State Medical University, St. Petersburg, 197022, Russia
- St. Petersburg State University, St. Petersburg, 199034, Russia
| | - Dilafruz K Kholmurodova
- Scientific and Practice Center for Immunology, Allergology and Human Genomics, Samarkand State Medical University, Samarkand, 100400, Uzbekistan
| | - Jasur A Rizaev
- Scientific and Practice Center for Immunology, Allergology and Human Genomics, Samarkand State Medical University, Samarkand, 100400, Uzbekistan
| | - Konstantin N Semenov
- Pavlov First St. Petersburg State Medical University, St. Petersburg, 197022, Russia
- St. Petersburg State University, St. Petersburg, 199034, Russia
- Scientific and Practice Center for Immunology, Allergology and Human Genomics, Samarkand State Medical University, Samarkand, 100400, Uzbekistan
| | - Vladimir V Sharoyko
- Medicinal Chemistry Center, Togliatti State University, Togliatti, 445020, Russia.
- Pavlov First St. Petersburg State Medical University, St. Petersburg, 197022, Russia
- St. Petersburg State University, St. Petersburg, 199034, Russia
- Scientific and Practice Center for Immunology, Allergology and Human Genomics, Samarkand State Medical University, Samarkand, 100400, Uzbekistan
| |
Collapse
|
4
|
Al-Medhtiy MH, Mohammed MT, M Raouf MMH, Al-Qaaneh AM, Jabbar AAJ, Abdullah FO, Mothana RA, Alanzi AR, Hassan RR, Abdulla MA, Saleh MI, Hasson S. A triterpenoid (corosolic acid) ameliorated AOM-mediated aberrant crypt foci in rats: modulation of Bax/PCNA, antioxidant and inflammatory mechanisms. J Mol Histol 2024; 55:765-783. [PMID: 39122895 DOI: 10.1007/s10735-024-10229-x] [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: 06/21/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024]
Abstract
Corosolic acid (CA) is a well-known natural pentacyclic triterpene found in numerous therapeutic plants that can exhibit many bioactivities including anti-inflammatory and anti-tumor actions. The current investigation explores the chemoprotective roles of CA against azoxymethane (AOM)-induced colonic aberrant crypt foci (ACF) in rats. Thirty Sprague Dawley rats were grouped in 5 cages; Group A, normal control rats inoculated subcutaneously (sc) with two doses of normal saline and fed orally on 10% tween 20; Groups B-E received two doses (sc) of azoxymethane in two weeks and treated with either 10% tween 20 (group B) or two intraperitoneal injections of 35 mg/kg 5-fluorouracil each week for one month (group C), while group D and E treated with 30 and 60 mg/kg, respectively, for 2 months. The toxicity results showed lack of any behavioral abnormalities or mortality in rats ingested with up-to 500 mg/kg of CA. The present AOM induction caused a significant initiation of ACF characterized by an increased number, larger in size, and well-matured tissue clusters in cancer controls. AOM inoculation created a bizarrely elongated nucleus, and strained cells, and significantly lowered the submucosal glands in colon tissues of cancer controls compared to 5-FU or CA-treated rats. CA treatment led to significant suppression of ACF incidence, which could be mediated by its modulatory effects on the immunohistochemical proteins (pro-apoptotic (Bax) and reduced PCNA protein expressions in colon tissues). Moreover, CA-treated rats had improved oxidative stress-mediated cytotoxicity indicated by increased endogenous antioxidants (SOD and CAT) and reduced lipid peroxidation indicators (MDA). In addition, CA ingestion (30 and 60 mg/kg) suppressed the inflammatory cascades, indicated by decreased serum TNF-α and IL-6 cytokines and increased anti-inflammatory (IL-10) cytokines consequently preventing further tumor development. CA treatment maintained liver and kidney functions in rats exposed to AOM cytotoxicity. CA could be a viable alternative for the treatment of oxidative-related human disorders including ACF.
Collapse
Affiliation(s)
- Morteta H Al-Medhtiy
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, University of Kufa, Kufa, Najaf Region, 540011, Iraq
| | - Mohammed T Mohammed
- Department of Microbiology, Faculty of veterinary medicine, University of Kufa, Kufa, Iraq
| | - Mohammed M Hussein M Raouf
- Department of Biomedical Sciences, College of Applied Science, Cihan University-Erbil, Erbil, Kurdistan Region, 44001, Iraq
| | - Ayman M Al-Qaaneh
- Department of Allied Health Sciences, Al-Balqa Applied University (BAU), Al-Salt, 19117, Jordan
| | - Ahmed A J Jabbar
- Department of Medical Laboratory Technology, Erbil Technical Health and Medical College, Erbil Polytechnic University, Erbil, 44001, Iraq.
| | - Fuad Othman Abdullah
- Department of Chemistry, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Ramzi A Mothana
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| | - Abdullah R Alanzi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| | - Rawaz Rizgar Hassan
- Department of Medical Laboratory Science, College of Science, Knowledge University, Kirkuk Road, Erbil, 44001, Iraq
| | - Mahmood Ameen Abdulla
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Musher Ismail Saleh
- Department of Chemistry, Faculty of Science and Health, Koya University, Koya KOY45, Kurdistan Region, Erbil, 44001, Iraq
| | - Sidgi Hasson
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, L3 3AF, UK
| |
Collapse
|
5
|
Dadashi H, Vandghanooni S, Karamnejad-Faragheh S, Karimian-Shaddel A, Eskandani M, Jahanban-Esfahlan R. A rapid protocol for synthesis of chitosan nanoparticles with ideal physicochemical features. Heliyon 2024; 10:e32228. [PMID: 38961950 PMCID: PMC11219308 DOI: 10.1016/j.heliyon.2024.e32228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 07/05/2024] Open
Abstract
In this research, an innovative protocol is introduced to address crucial deficiencies in the formulation of chitosan nanoparticles (Cs NPs). While NPs show potential in drug delivery systems (DDSs), their application in the clinic is hindered by various drawbacks, such as toxicity, high material costs, and time-consuming and challenging preparation procedures. Within polymer-based NPs, Cs is a plentiful natural substance derived from the deacetylation of chitin, which can be sourced from the shells of shrimp or crab. Cs NPs can be formulated using the ionic gelation technique, which involves the use of a negatively charged agent, such as tripolyphosphate (TPP), as a crosslinking agent. Even though Cs is a cost-effective and biocompatible material, the formulation of Cs NPs with the correct size and surface electrical charge (zeta potential) presents a persistent challenge. In this study, various techniques were employed to analyze the prepared Cs NPs. The size and surface charge of the NPs were evaluated using dynamic light scattering (DLS). Morphological analysis was conducted using field emission-scanning electron microscopy (FE-SEM). The chemical composition and formation of Cs NPs were investigated using Fourier transform infrared (FTIR). The stability analysis was confirmed through X-ray diffraction (XRD) analysis. Lastly, the biocompatibility of the NPs was assessed through cell cytotoxicity evaluation using the MTT assay. Moreover, here, 11 formulations with different parameters such as reaction pH, Cs:TPP ratio, type of Cs/TPP, and ultrasonication procedure were prepared. Formulation 11 was chosen as the optimized formulation based on its high stability of more than three months, biocompatibility, nanosize of 75.6 ± 18.24 nm, and zeta potential of +26.7 mV. To conclude, the method described here is easy and reproducible and can be used for facile preparation of Cs NPs with desirable physicochemical characteristics and engineering ideal platforms for drug delivery purposes.
Collapse
Affiliation(s)
- Hamed Dadashi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz university of Medical Sciences, Tabriz, Iran
| | - Shahrbanoo Karamnejad-Faragheh
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Karimian-Shaddel
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Jin M, Li J, Zheng L, Huang M, Wu Y, Huang Q, Huang G. Corosolic acid delivered by exosomes from Eriobotrya japonica decreased pancreatic cancer cell proliferation and invasion by inducing SAT1-mediated ferroptosis. Int Immunopharmacol 2024; 132:111939. [PMID: 38608471 DOI: 10.1016/j.intimp.2024.111939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND In this study, we investigated whether Exo regulate the proliferation and invasion of PC. METHODS In this study, we isolated the Eriobotrya japonica Exo using Ultra-high speed centrifugal method. Mass spectrum were used for Exo active components analysis. PC (Capan-1 and Bxpc-3) cells proliferation, migration, and apoptosis were detected using CCK8, ethynyldeoxyuridine, transwell, wound healing, and flow cytometry analyses. We also constructed a lung metastatic mouse model and subcutaneous tumor model to illustrate the regulation effect of Exo or active components. Proteomics were used to reveal the regulatory mechanism responsible for the observed effects. RESULTS We isolated Eriobotrya japonica Exo and found that Exo treatment significantly suppressed cell migration and proliferation in both in vivo and in vitro using Capan-1. Mass spectrum for Exo active components analysis found that Exo contains high amounts of corosolic acid (CRA). The further study found that CRA treatment inhibit the proliferation, migration, and increased cell death of both Capan-1 and Bxpc-3 cells in a concentration-dependent manner. In vivo experiments confirmed that CRA inhibited pulmonary metastasis by decreasing the number of metastatic foci. Cell proteomics analysis showed that CRA treatment induced spermidine/spermine N1-acetyltransferase 1 (SAT1)-dependent ferroptosis. Treatment with the ferroptosis suppressor ferrostatin-1 significantly reversed CRA-induced cell apoptosis. CONCLUSION The data suggested that corosolic acid delivered by exosomes from Eriobotrya japonica decreased pancreatic cancer cell proliferation and invasion by inducing SAT1-mediated ferroptosis.
Collapse
Affiliation(s)
- Mingming Jin
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jingjing Li
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Liying Zheng
- Postgraduate Training base at Shanghai Gongli Hospital, Ningxia Medical University, Shanghai 200135, China; Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, China
| | - Mi Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Yue Wu
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Qingqing Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China; Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
7
|
Anticancer properties of curcumin-treated Lactobacillus plantarum against the HT-29 colorectal adenocarcinoma cells. Sci Rep 2023; 13:2860. [PMID: 36801895 PMCID: PMC9938284 DOI: 10.1038/s41598-023-29462-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 02/06/2023] [Indexed: 02/19/2023] Open
Abstract
Probiotic bacteria with functions of importance to the health and well-being of the host exhibit various medicinal properties including anti-proliferative properties against cancer cells. There are observations demonstrating probiotic bacteria and their metabolomics can be different in various populations with different eating habits. Here, Lactobacillus plantarum was treated with curcumin (the major compound of turmeric), and its resistance to the curcumin was determined. After then the cell-free supernatants of untreated bacteria (CFS) and bacteria treated with curcumin (cur-CFS) were isolated and their anti-proliferative properties against HT-29 colon cancer cells were compared. The ability of L. plantarum treated with curcumin to combat a variety of pathogenic bacterial species and its ability to survive in acidic conditions were evidence that the probiotic properties of the bacterium were unaffected by the curcumin treatment. L. plantarum treated with curcumin and intact L. plantarum were both able to live in acidic conditions, according to the results of the resistance to low pH test. The MTT result showed that CFS and cur-CFS dose-dependently decreased the growth of HT29 cells with a half-maximal inhibitory concentration of 181.7 and 116.3 µL/mL at 48 h, respectively. Morphological alteration of DAPI-stained cells also exhibited significant fragmentation in the chromatin within the nucleus of cur-CFS-treated cells compared to CFS-treated HT29 cells. Moreover, flow cytometry analyses of apoptosis and cell cycle confirmed DAPI staining and MTT assay results and stipulated the increased occurrence of programmed cell death (apoptosis) in cur-CFS-treated cells (~ 57.65%) compared to CFS-treated cells (~ 47%). These results were more confirmed with qPCR and exhibited the upregulation of Caspase 9-3 and BAX genes, and downregulation of the BCL-2 gene in cur-CFS- and CFS-treated cells. In conclusion, turmeric spice and curcumin may affect the metabolomics of probiotics in intestinal flora which could subsequently influence their anticancer properties.
Collapse
|
8
|
Zare S, Pirhadi S, El Seedi HR, Jassbi AR. Anti-COVID-19 and antidiabetic activities of new oleanane and ursane-type triterpenoids from Salvia grossheimii: an in-silico approach. J Recept Signal Transduct Res 2022; 42:540-548. [PMID: 35544359 DOI: 10.1080/10799893.2022.2072891] [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: 12/15/2022]
Abstract
Salvia grossheimii is a perennial herb with antidiabetic and cytotoxic constituents. In continuation of our study on S. grosshiemii to identify the bioactive phytochemicals, we have reported the characterization of seven undescribed triterpenoids. The aerial parts of the plant were extracted in dichloromethane and its constituents were isolated using chromatography techniques. The structures of compounds were identified using 1D, 2D NMR, and ESI-MS spectral data. Seven new oleanane- and ursane-type triterpenoids (1-7) were identified in S. grossheimii. The structures of 1-7 were characterized as; 2α-hydroxy-3β-acetoxy-olean-9(11),12-diene (1), 2α-acetoxy-3β-hydroxy-olean-9(11),12-diene (2), 3β-acetoxy-olean-18-ene,2α,11α-diol (3), 2α-hydroxy-3β-acetoxy-urs-9(11),12-diene (4), 2α-acetoxy-3β-hydroxy-urs-9(11),12-diene (5), 2α,3β-diacetoxy-urs-12-ene-11α,20β-diol (6), 2α,3β-diacetoxy-urs-9(11),12-diene-20β-ol (7). Triterpenoids (2, 5, and 7) were intramolecular transesterification or dehydration products of their corresponding isomers or allylic alcohol in the C rings, respectively, produced in-situ during NMR spectroscopy. Virtual screening of 1-7 was performed with molecular docking analysis to identify the potential SARS-CoV-2 and α-glucosidase inhibitors using the smina molecular docking algorithm. The best binding energy values (kcal/mol) against COVID-19 main protease Mpro were calculated for 6 (-8.77) and 7 (-8.68), and the higher binding affinities toward human α-glucosidase were obtained for 2 (-9.39) and 6 (-8.63). This study suggests S. grossheimii as a rich source of bioactive triterpenoids and introduces new natural compounds. Considering the high binding energy values of 2, 6, and 7, these structures could be candidates for anti-COVID-19 and antidiabetic drug development in the future.
Collapse
Affiliation(s)
- Somayeh Zare
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Pirhadi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hesham R El Seedi
- Department of Pharmaceutical Biosciences, Pharmacognosy Group, BMC, Uppsala University, Uppsala, Sweden.,International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, China.,Jiangsu Education Department, International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing, Jiangsu University, Zhenjiang, China
| | - Amir Reza Jassbi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
9
|
Jafari S, Bakhshaei A, Eskandani M, Molavi O. Silibinin-Loaded Nanostructured Lipid Carriers for Growth Inhibition of Cisplatin-Resistant Ovarian Cancer Cells. Assay Drug Dev Technol 2022; 20:339-348. [DOI: 10.1089/adt.2022.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Sevda Jafari
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atabak Bakhshaei
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology (RCPN), Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
10
|
Li L, Ding L, Zhang X, Wen D, Zhang M, Liu W, Wang H, Wang B, Yan L, Guo L, Diao H. A nitroreductase-responsive near-infrared phototheranostic probe for in vivo imaging of tiny tumor and photodynamic therapy. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 267:120579. [PMID: 34776373 DOI: 10.1016/j.saa.2021.120579] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/28/2021] [Accepted: 10/31/2021] [Indexed: 06/13/2023]
Abstract
The hypoxia-activated and nitroreductase-responsive phototheranostic probe has been developed by incorporating a nitro group into a hemicyanine fluorophore. The probe displays extremely sensitive and selective near-infrared fluorescence enhancement to nitroreductase with the detection limit of 2.10 ng/mL. The detection mechanism relies on the nitroreductase-catalyzed reduction of the nitro group to an amino group, along with the generation of the fluorophore. The availability of the probe in fluorescence imaging and photodynamic therapy was demonstrated at cellular level and in vivo. The probe can image endogenous nitroreductase and the hypoxia status of living cells. The probe also exhibits significant phototoxicity to hypoxia tumor cells under the 660 nm laser irradiation. More importantly, the probe has been successfully utilized in imaging tiny tumor (about 6 mm3) and tumor photodynamic therapy in vivo. The proposed probe integrates accurate near-infrared fluorescence imaging and photodynamic therapy into the same molecule, which probably become a promising agent in the early diagnosis and therapy of tumors.
Collapse
Affiliation(s)
- Lihong Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, PR China; College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China.
| | - Lei Ding
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Xueyun Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Danning Wen
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Min Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Wen Liu
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China.
| | - Haojiang Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Bin Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Lili Yan
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Lixia Guo
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Haipeng Diao
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, PR China; College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China.
| |
Collapse
|
11
|
Vandghanooni S, Rasoulian F, Eskandani M, Akbari Nakhjavani S, Eskandani M. Acriflavine-loaded solid lipid nanoparticles: preparation, physicochemical characterization, and anti-proliferative properties. Pharm Dev Technol 2021; 26:934-942. [PMID: 34338582 DOI: 10.1080/10837450.2021.1963276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Acriflavine (ACF) is an antiseptic compound with the potential antitumor activity which is used for the fluorescent staining of RNA due to its dominant fluorescent emission at ∼515 nm. Here, solid lipid nanoparticles (SLNs) containing ACF (ACF-SLNs) were prepared and their physicochemical properties, potential geno/cytotoxicity, as well as the fluorescent properties were investigated. FITC-annexin V/PI staining and cell cycle assays were carried out to find the type of cellular death caused. Particle size analysis and SEM images revealed that spherical ACF-SLNs had a homogeneous dispersion with a mean diameter of 106 ± 5.7 nm. Drug loading (DL) of 31.25 ± 4.21 mg/mL and high encapsulation efficiency (EE%) (89.75 ± 5.44) were found. ACF-SLNs physically were relatively stable in terms of dispersion, size, and EE. The uptake study demonstrated the potential use of fluorescent ACF-SLNs in bio-distribution studies. MTT assay showed that plain ACF could induce growth inhibition of A549 cells with IC50 of 8.5, 6, and 4.5 μMol after 24, 48, and 72 hours, respectively, while ACF-SLNs had stable cytotoxic effects after 48 hours. ACF-SLNs induced remarkable apoptosis and even necrosis after 48 h. Conclusively, ACF-SLNs with acceptable physicochemical features showed increased bioimpacts after 48 h compared to plain ACF.
Collapse
Affiliation(s)
- Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Forough Rasoulian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Eskandani
- Department of Animal Science, Faculty of Agricultural Sciences, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Sattar Akbari Nakhjavani
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
12
|
Zheng F, Chen P, Li H, Aschner M. Drp-1-Dependent Mitochondrial Fragmentation Contributes to Cobalt Chloride-Induced Toxicity in Caenorhabditis elegans. Toxicol Sci 2021; 177:158-167. [PMID: 32617571 DOI: 10.1093/toxsci/kfaa105] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Excess cobalt may lead to metallosis, characterized by sensorineural hearing loss, visual, and cognitive impairment, and peripheral neuropathy. In the present study, we sought to address the molecular mechanisms of cobalt-induced neurotoxicity, using Caenorhabditis elegans as an experimental model. Exposure to cobalt chloride for 2 h significantly decreased the survival rate and lifespan in nematodes. Cobalt chloride exposure led to increased oxidative stress and upregulation of glutathione S-transferase 4. Consistently, its upstream regulator skn-1, a mammalian homolog of the nuclear factor erythroid 2-related factor 2, was activated. Among the mRNAs examined by quantitative real-time polymerase chain reactions, apoptotic activator egl-1, proapoptotic gene ced-9, autophagic (bec-1 and lgg-1), and mitochondrial fission regulator drp-1 were significantly upregulated upon cobalt exposure, concomitant with mitochondrial fragmentation, as determined by confocal microscopy. Moreover, drp-1 inhibition suppressed the cobalt chloride-induced reactive oxygen species generation, growth defects, and reduced mitochondrial fragmentation. Our novel findings suggest that the acute toxicity of cobalt is mediated by mitochondrial fragmentation and drp-1 upregulation.
Collapse
Affiliation(s)
- Fuli Zheng
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China.,Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Pan Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Huangyuan Li
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
13
|
Vandghanooni S, Farajzadeh Vahid Z, Nakhlband A, Bahadori MB, Eskandani M. Sclareol Inhibits Hypoxia-Inducible Factor-1α Accumulation and Induces Apoptosis in Hypoxic Cancer Cells. Adv Pharm Bull 2021; 12:593-602. [PMID: 35935045 PMCID: PMC9348540 DOI: 10.34172/apb.2022.062] [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: 04/14/2021] [Revised: 05/27/2021] [Accepted: 07/02/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose: The hypoxia in solid tumors is associated with the resistance to chemo/radiotherapy. Hypoxia-inducible factor-1 (HIF-1) plays a key role in cell remodeling to hypoxia. Therefore, the inhibition of HIF-1 accumulation is considered a hopeful strategy for the treatment of cancer. Here, we aimed to evaluate the geno- and cytotoxicity properties of sclareol, a natural bicyclic diterpene alcohol, on A549 cells in CoCl2-induced hypoxia.
Methods: The cytotoxicity and apoptosis-inducing properties of sclareol on the A549 cell were evaluated using MTT assay and Annexin V/PI staining, respectively in hypoxia. DAPI staining, DNA ladder, and comet assay were used to evaluate the genotoxicity. Further, the qPCR technique was employed to assess the expression of HIF-1α, HIF-1β, and downstream target genes (GluT1, and Eno1). Finally, the level of HIF-1α protein was evaluated through Western blotting in sclareol-treated cells in hypoxia.
Results: The inhibitory concentration (IC50) of sclareol against A549 cells was 8 μg/mL at 48 hours in hypoxia. The genotoxicity of sclareol was confirmed in the cells treated with sclareol in hypoxia. Sclareol induced ~46% apoptosis and also necrosis in the hypoxic condition. The qPCR analyses showed an enhanced suppression of HIF-1α, HIF-1β, GluT1, and Eno1 due to the sclareol treatment in the hypoxia. Moreover, protein quantification analysis showed dose-dependently degradation of HIF-1α in hypoxia upon treatment with sclareol.
Conclusion: The results obtained here indicate that sclareol possesses dose-dependent cytotoxicity effects against A549 cells in hypoxia through inhibition of HIF-1α protein accumulation, increasing cell sensitivity to intracellular oxygen levels, and disruption of cell adaptation to hypoxia.
Collapse
Affiliation(s)
- Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ailar Nakhlband
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mir Babak Bahadori
- Medicinal Plants Research Center, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Zhao J, Zhou H, An Y, Shen K, Yu L. Biological effects of corosolic acid as an anti-inflammatory, anti-metabolic syndrome and anti-neoplasic natural compound. Oncol Lett 2021; 21:84. [PMID: 33363621 PMCID: PMC7723172 DOI: 10.3892/ol.2020.12345] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 09/16/2020] [Indexed: 12/23/2022] Open
Abstract
Accumulating evidence has indicated that corosolic acid exerts anti-diabetic, anti-obesity, anti-inflammatory, anti-hyperlipidemic and anti-viral effects. More importantly, corosolic acid has recently attracted much attention due to its anticancer properties and innocuous effects on normal cells. Furthermore, the increasing proportion of obese and/or diabetic populations has led to an epidemic of non-alcoholic fatty liver disease (NAFLD), which frequently progresses to hepatocellular carcinoma (HCC). Evidence has indicated that NAFLD is closely associated with the development of HCC and comprises a high risk factor. The present review summarizes the anticancer effects of corosolic acid in vitro and in vivo, and its related molecular mechanisms. It also describes the inhibitory effects of corosolic acid on the progression of NAFLD and its associated molecular mechanisms, providing guidance for future research on corosolic acid in NAFLD-related HCC prevention and treatment. To the best of our knowledge, a review of corosolic acid as an anticancer agent has not yet been reported. Due to its multitargeted activity in cancer cells, corosolic acid exerts anticancer effects when administered alone, and acts synergistically when administered with chemotherapeutic drugs, even in drug-resistant cells. In addition, as a novel tool to treat metabolic syndromes, corosolic acid uses the same mechanism in its action against cancer as that used in the progression of NAFLD-related HCC. Therefore, corosolic acid has been suggested as an agent for the prevention and treatment of NAFLD-related HCC.
Collapse
Affiliation(s)
- Jinwei Zhao
- Key Laboratory for Zoonosis Research, Department of Hepatopancreatobiliary Surgery, Institute of Zoonosis, The Second Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine Jilin University, Changchun, Jilin 130062, P.R. China
| | - Hong Zhou
- Key Laboratory for Zoonosis Research, Department of Hepatopancreatobiliary Surgery, Institute of Zoonosis, The Second Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine Jilin University, Changchun, Jilin 130062, P.R. China
| | - Yanan An
- Key Laboratory for Zoonosis Research, Department of Hepatopancreatobiliary Surgery, Institute of Zoonosis, The Second Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine Jilin University, Changchun, Jilin 130062, P.R. China
| | - Keshu Shen
- Department of Hepatobiliary Medicine of Jilin Hepatobiliary Hospital, Changchun, Jilin 130062, P.R. China
| | - Lu Yu
- Key Laboratory for Zoonosis Research, Department of Hepatopancreatobiliary Surgery, Institute of Zoonosis, The Second Hospital of Jilin University, Ministry of Education, College of Veterinary Medicine Jilin University, Changchun, Jilin 130062, P.R. China
| |
Collapse
|
15
|
Zare S, Mirkhani H, Firuzi O, Moheimanian N, Asadollahi M, Pirhadi S, Chandran JN, Schneider B, Jassbi AR. Antidiabetic and cytotoxic polyhydroxylated oleanane and ursane type triterpenoids from Salvia grossheimii. Bioorg Chem 2020; 104:104297. [DOI: 10.1016/j.bioorg.2020.104297] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 09/11/2020] [Accepted: 09/16/2020] [Indexed: 12/22/2022]
|
16
|
Liu T, Gao Q, Yang B, Yin C, Chang J, Qian H, Xing G, Wang S, Li F, Zhang Y, Chen D, Cai J, Shi H, Aschner M, Appiah-Kubi K, He D, Lu R. Differential susceptibility of PC12 and BRL cells and the regulatory role of HIF-1α signaling pathway in response to acute methylmercury exposure under normoxia. Toxicol Lett 2020; 331:82-91. [PMID: 32461003 PMCID: PMC7366344 DOI: 10.1016/j.toxlet.2020.05.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 12/24/2022]
Abstract
Hypoxia-inducible factor 1 (HIF-1) is a critical nuclear transcription factor for adaptation to hypoxia; its regulatable subunit, HIF-1α, is a cytoprotective regulatory factor. We examined the effects of methylmercury (MeHg) in rat adrenal pheochromocytoma (PC12) cells and the rat hepatocyte cell line BRL. MeHg treatment led to time- and concentration-dependent toxicity in both lines with statistically significant cytotoxic effects at 5 μM and 10 μM in PC12 and BRL, respectively, at 0.5 h. HIF-1α protein levels were significantly decreased at 2.5 (PC12) and 5 (BRL) μM MeHg. Furthermore, MeHg reduced the protein levels of HIF-1α and its target genes (glucose transporter-1, vascular endothelial growth factor-A and erythropoietin). Overexpression of HIF-1α significantly attenuated MeHg-induced toxicity in both cell types. Notably, cobalt chloride, a pharmacological inducer of HIF-1α, significantly attenuated MeHg-induced toxicity in BRL but not PC12. In both cell lines, an inhibitor of prolyl hydroxylase, 3, 4-dihydroxybenzoic acid, and the proteasome inhibitor carbobenzoxy-L-leucyl-L-leucyl-L-leucinal(MG132), antagonized MeHg toxicity, while 2-methoxyestradiol, a HIF-1α inhibitor, significantly increased it. These data establish that: (a) neuron-like PC12 cells are more sensitive to MeHg than non-neuronal BRL cells; (b) HIF-1α plays a similar role in MeHg-induced toxicity in both cell lines; and (c) upregulation of HIF-1α offers general cytoprotection against MeHg toxicity in PC12 and BRL cell lines.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Qianqian Gao
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Bobo Yang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Changsheng Yin
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jie Chang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Hai Qian
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Guangwei Xing
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Suhua Wang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Fang Li
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yubin Zhang
- Department of Occupational Health and Toxicology, School of Public Health, Fudan University, Shanghai 200032, China
| | - Da Chen
- School of Environment, Jinan University, Guangzhou, Guangdong 510632, China
| | - Jiyang Cai
- Department of Physiology, College of Medicine, University of Oklahoma Health Science Center, Lindsay, Oklahoma City, OK 73104, USA
| | - Haifeng Shi
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kwaku Appiah-Kubi
- Department of Applied Biology, C. K. Tedam University of Technology and Applied Sciences, Navrongo, UK-0215-5321, Ghana
| | - Dawei He
- Center for Experimental Research, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215130, China
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Center for Experimental Research, Kunshan Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215130, China.
| |
Collapse
|
17
|
Apaza T L, Antognoni F, Potente G, Rumbero Sánchez Á. Triterpenoids isolated from Jatropha macrantha (Müll. Arg.) inhibit the NF-κB and HIF-1α pathways in tumour cells. Nat Prod Res 2020; 35:5843-5847. [PMID: 32691666 DOI: 10.1080/14786419.2020.1795851] [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: 10/23/2022]
Abstract
Activity-guided fractionations of Jatropha macrantha Müll. Arg. led to the isolation of pomolic acid (1) and euscaphic acid (2). The potential for inhibition against NF-κB and HIF-1α production of these two compounds was tested in different tumour cell lines. Compounds 1 and 2 showed an inhibitory activity of HIF-1α in the SK-MEL-28 (IC50=3.01 ± 0.02 μM and 3.78 ± 0.02 μM), A549 (IC50=9.97 ± 0.01 μM and 10.25 ± 0.01 μM) and U-373 MG (IC50=6.34 ± 0.02 μM and 8.85 ± 0.02 μM) cell lines. In addition, compounds 1 and 2 showed an inhibitory activity on NF-κB in SK-MEL-28 (IC50=1.05 ± 0.02 μM and 2.71 ± 0.01 μM), A549 (IC50=3.63 ± 0.01 μM and 3.73 ± 0.02 μM) and U-373 MG (IC50=2.55 ± 0.02 μM and 3.39 ± 0.01 μM) cell lines. This is the first report that isolates these compounds from J. macrantha and tests their antitumor potential.
Collapse
Affiliation(s)
- Luis Apaza T
- Department of Organic Chemistry, Faculty of Sciences, University Autónoma of Madrid, Madrid, Spain.,Department of Pharmacology, Pharmacognosy and Botany, Faculty of Pharmacy, University Complutense of Madrid, Madrid, Spain
| | - Fabiana Antognoni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Giulia Potente
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Ángel Rumbero Sánchez
- Department of Organic Chemistry, Faculty of Sciences, University Autónoma of Madrid, Madrid, Spain
| |
Collapse
|
18
|
Jahanban-Esfahlan R, Derakhshankhah H, Haghshenas B, Massoumi B, Abbasian M, Jaymand M. A bio-inspired magnetic natural hydrogel containing gelatin and alginate as a drug delivery system for cancer chemotherapy. Int J Biol Macromol 2020; 156:438-445. [PMID: 32298719 DOI: 10.1016/j.ijbiomac.2020.04.074] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/09/2020] [Accepted: 04/11/2020] [Indexed: 12/11/2022]
Abstract
This study aimed to design and development of a magnetic natural hydrogel based on alginate (Alg), gelatin (Gel), and Fe3O4 magnetic nanoparticles (MNPs) as an efficient and "smart" drug delivery system (DDS) for cancer therapy. First, Alg was partially oxidized (OAlg), and then the Alg-Gel chemical hydrogel was synthesized through "Shift-Base" condensation reaction. Afterward, Fe3O4 NPs were incorporated into the hydrogel through in situ chemical co-precipitation approach. The scanning electron microscopy (SEM) image exhibited that the fabricated Alg-Gel hydrogel has porous microstructure without microphase separation. Transmission electron microscopy (TEM) revealed the well-defined formation of Fe3O4 NPs throughout the Alg-Gel hydrogel with spherical shapes in the size range of 25 ± 10 nm. Saturation magnetization (δs) value of the Alg-Gel/Fe3O4 was obtained to be 31 emu g-1 that represent proper magnetic property for "smart" drug delivery purposes. The obtained Alg-Gel/Fe3O4 was loaded with doxorubicin hydrochloride (Dox), and its drug loading and encapsulation efficiencies as well as its anticancer activity was investigated against Hela cells. The formulated Alg-Gel/Fe3O4-Dox exhibited pH-dependent drug release behavior due to presence of carboxylic acid groups in the DDS. According to the results, the Alg-Gel/Fe3O4 magnetic hydrogel can be considered as an efficient and "smart" DDS for cancer therapy and diagnosis.
Collapse
Affiliation(s)
- Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Derakhshankhah
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Babak Haghshenas
- Regenerative Medicine Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | | | - Mehdi Jaymand
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
19
|
Vandghanooni S, Eskandani M, Barar J, Omidi Y. Antisense LNA-loaded nanoparticles of star-shaped glucose-core PCL-PEG copolymer for enhanced inhibition of oncomiR-214 and nucleolin-mediated therapy of cisplatin-resistant ovarian cancer cells. Int J Pharm 2020; 573:118729. [PMID: 31705975 DOI: 10.1016/j.ijpharm.2019.118729] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/17/2019] [Accepted: 09/21/2019] [Indexed: 02/07/2023]
Abstract
We aimed to inhibit overexpressed oncomiR-214 in cisplatin (CIS)-resistant ovarian cancer (OC) and perform targeted therapy of sensitized cells using a novel polymeric drug delivery system (DDS). A system of nanoparticles (NPs) of star-shaped glucose-core polycaprolactone-polyethylene glycol (Glu-PCL-PEG) block copolymer containing cisplatin (CIS-PCL NPs) and locked nucleic acid (LNA) anti-miR-214 (LNA-PCL NPs) were prepared and anti-nucleolin aptamer was conjugated to the surface of prepared NPs to prepare Ap-CIS-PCL NPs and Ap-LNA-PCL NPs, respectively. The cancer-targeting ability of the NPs was confirmed and the CIS-resistant A2780 (A2780 R) cells were transfected with Ap-LNA-PCL NPs to inhibit oncomiR-214 and sensitize the cells to CIS. Next, the miR-214-inhibited cells were exposed to the Ap-CIS-NPs and the deracination efficiency of targeted DDS was evaluated. The oncomiR-214 in A2780 R cells were harnessed by Ap-LNA-PCL NPs, and nucleolin-mediated endocytosis of targeted polymeric DDSs containing CIS into miR-214-inhibited A2780 R cells caused enhanced apoptosis, which was further confirmed by apoptosis detection and evaluation of downstream genes expression. Targeted inhibition of miR-214 using the developed NPs containing LNA can decrease drug-resistant properties of cancer cells and may enhance the efficiency of targeted DDSs.
Collapse
Affiliation(s)
- Somayeh Vandghanooni
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran,; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran,; Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
20
|
Zamanlu M, Eskandani M, Barar J, Jaymand M, Pakchin PS, Farhoudi M. Enhanced thrombolysis using tissue plasminogen activator (tPA)-loaded PEGylated PLGA nanoparticles for ischemic stroke. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101165] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|