1
|
Soni D, Anjum Z, Raza K, Verma S. A Review on Picrosides Targeting NFκB and its Proteins for Treatment of Breast Cancer. Cell Biochem Biophys 2024; 82:575-591. [PMID: 38724755 DOI: 10.1007/s12013-024-01281-1] [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] [Accepted: 04/17/2024] [Indexed: 08/25/2024]
Abstract
Breast cancer is the most frequently diagnosed disease causing most deaths in women worldwide. Chemotherapy and neo-adjuvant therapy are the standard method of treatment in early stages of breast cancer. However drug resistance in breast cancer limit the use of these methods for treatment. Research focus is now shifted towards identifying natural phytochemicals with lower toxicity. This review illustrates the NF κB interaction with different signaling pathways in normal condition, breast cancer and other cancer and thus represent a potential target for treatment. No reports are available on the action of picrosides on NFκB and its associated proteins for anticancer activity. In the present review, potential interaction of picrosides with NF-κB and its associated proteins is reviewed for anticancer action. Further, an important facet of this review entails the ADMET analysis of Picroside, elucidating key ADMET properties which serves to underscore the crucial characteristics of Picroside as a potential drug for treating breast cancer. Furthermore, in silico analysis of Picrosides was executed in order to get potential binding modes between ligand (Picrosides II) and NFκB.
Collapse
Affiliation(s)
- Deepika Soni
- National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Zubina Anjum
- Department of Bioinformatics, Central University of South Bihar, Gaya, Bihar, India
| | - Khalid Raza
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Saurabh Verma
- Indian Council of Medical Research, HRD Division, V. Ramalingaswami Bhawan, Ansari Nagar, New Delhi, India.
| |
Collapse
|
2
|
Naldi L, Fibbi B, Anceschi C, Nardini P, Guasti D, Peri A, Marroncini G. Effects of Reduced Extracellular Sodium Concentrations on Cisplatin Treatment in Human Tumor Cells: The Role of Autophagy. Int J Mol Sci 2024; 25:4377. [PMID: 38673964 PMCID: PMC11050238 DOI: 10.3390/ijms25084377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Hyponatremia is the prevalent electrolyte imbalance in cancer patients, and it is associated with a worse outcome. Notably, emerging clinical evidence suggests that hyponatremia adversely influences the response to anticancer treatments. Therefore, this study aims to investigate how reduced extracellular [Na+] affects the responsiveness of different cancer cell lines (from human colon adenocarcinoma, neuroblastoma, and small cell lung cancer) to cisplatin and the underlying potential mechanisms. Cisplatin dose-response curves revealed higher IC50 in low [Na+] than normal [Na+]. Accordingly, cisplatin treatment was less effective in counteracting the proliferation and migration of tumor cells when cultured in low [Na+], as demonstrated by colony formation and invasion assays. In addition, the expression analysis of proteins involved in autophagosome-lysosome formation and the visualization of lysosomal areas by electron microscopy revealed that one of the main mechanisms involved in chemoresistance to cisplatin is the promotion of autophagy. In conclusion, our data first demonstrate that the antitumoral effect of cisplatin is markedly reduced in low [Na+] and that autophagy is an important mechanism of drug escape. This study indicates the role of hyponatremia in cisplatin chemoresistance and reinforces the recommendation to correct this electrolyte alteration in cancer patients.
Collapse
Affiliation(s)
- Laura Naldi
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Careggi University Hospital, University of Florence, 50139 Florence, Italy; (L.N.); (C.A.); (A.P.); (G.M.)
| | - Benedetta Fibbi
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Careggi University Hospital, University of Florence, 50139 Florence, Italy; (L.N.); (C.A.); (A.P.); (G.M.)
- Pituitary Diseases and Sodium Alterations Unit, Careggi University Hospital, 50139 Florence, Italy
| | - Cecilia Anceschi
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Careggi University Hospital, University of Florence, 50139 Florence, Italy; (L.N.); (C.A.); (A.P.); (G.M.)
| | - Patrizia Nardini
- Research Unit of Histology & Embryology, Department of Experimental & Clinical Medicine, University of Florence, 50134 Florence, Italy; (P.N.); (D.G.)
| | - Daniele Guasti
- Research Unit of Histology & Embryology, Department of Experimental & Clinical Medicine, University of Florence, 50134 Florence, Italy; (P.N.); (D.G.)
| | - Alessandro Peri
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Careggi University Hospital, University of Florence, 50139 Florence, Italy; (L.N.); (C.A.); (A.P.); (G.M.)
- Pituitary Diseases and Sodium Alterations Unit, Careggi University Hospital, 50139 Florence, Italy
| | - Giada Marroncini
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Careggi University Hospital, University of Florence, 50139 Florence, Italy; (L.N.); (C.A.); (A.P.); (G.M.)
| |
Collapse
|
3
|
Roda E, De Luca F, Priori EC, Ratto D, Pinelli S, Corradini E, Mozzoni P, Poli D, Mazzini G, Bottone MG, Gatti AM, Marti M, Locatelli CA, Rossi P, Bottai D. The Designer Drug αPHP Affected Cell Proliferation and Triggered Deathly Mechanisms in Murine Neural Stem/Progenitor Cells. BIOLOGY 2023; 12:1225. [PMID: 37759624 PMCID: PMC10525791 DOI: 10.3390/biology12091225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/08/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
Increasing reports of neurological and psychiatric outcomes due to psychostimulant synthetic cathinones (SCs) have recently raised public concern. However, the understanding of neurotoxic mechanisms is still lacking, particularly for the under-investigated αPHP, one of the major MDPV derivatives. In particular, its effects on neural stem/progenitor cell cultures (NSPCs) are still unexplored. Therefore, in the current in vitro study, the effects of increasing αPHP concentrations (25-2000 μM), on cell viability/proliferation, morphology/ultrastructure, genotoxicity and cell death pathways, have been evaluated after exposure in murine NSPCs, using a battery of complementary techniques, i.e., MTT and clonogenic assay, flow cytometry, immunocytochemistry, TEM, and patch clamp. We revealed that αPHP was able to induce a dose-dependent significant decrease of the viability, proliferation and clonal capability of the NSPCs, paralleled by the resting membrane potential depolarization and apoptotic/autophagic/necroptotic pathway activation. Moreover, ultrastructural alterations were clearly observed. Overall, our current findings demonstrate that αPHP, damaging NSPCs and the morpho-functional fundamental units of adult neurogenic niches may affect neurogenesis, possibly triggering long-lasting, irreversible CNS damage. The present investigation could pave the way for a broadened understanding of SCs toxicology, needed to establish an appropriate treatment for NPS and the potential consequences for public health.
Collapse
Affiliation(s)
- Elisa Roda
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy (C.A.L.)
| | - Fabrizio De Luca
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Erica Cecilia Priori
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Daniela Ratto
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Silvana Pinelli
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Emilia Corradini
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Paola Mozzoni
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Diana Poli
- INAIL Research, Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Via Fontana Candida, 1, 00078 Monte Porzio Catone, Italy
| | - Giuliano Mazzini
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
- Institute of Molecular Genetics—CNR (National Research Council), 27100 Pavia, Italy
| | - Maria Grazia Bottone
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Anna Maria Gatti
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy (C.A.L.)
| | - Matteo Marti
- Department of Translational Medicine, Section of Legal Medicine, LTTA Center and University Center of Gender Medicine, University of Ferrara, 44121 Ferrara, Italy;
- Collaborative Centre for the Italian National Early Warning System, Department of Anti-Drug Policies, Presidency of the Council of Ministers, 44121 Ferrara, Italy
| | - Carlo Alessandro Locatelli
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy (C.A.L.)
| | - Paola Rossi
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (F.D.L.); (P.R.)
| | - Daniele Bottai
- Department of Pharmaceutical Sciences, Section of Pharmacology and Biosciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy;
| |
Collapse
|
4
|
Gaiaschi L, Favaron C, Casali C, Gola F, De Luca F, Ravera M, Roda E, Rossi P, Bottone MG. Study on the activation of cell death mechanisms: in search of new therapeutic targets in glioblastoma multiforme. Apoptosis 2023:10.1007/s10495-023-01857-x. [PMID: 37244884 DOI: 10.1007/s10495-023-01857-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2023] [Indexed: 05/29/2023]
Abstract
Malignant primary brain tumors remain among the most difficult cancers to treat, in particular, Glioblastoma Multiforme (GBM) is the deadliest brain tumor. The standard therapies currently used are not efficient enough in improving patients' survival and quality of life. Cisplatin (CDDP), a platinum-based drug, has shown efficacy against different solid neoplasms, but it is also associated to different forms of off-target toxicity. To overcome the limitation in the use of CDDP in the treatment of GBM patients, fourth generation platinum compounds are been synthesized, one of them is the Pt(IV)Ac-POA, a prodrug with a medium-chain fatty acid as axial ligand, which acts as a histone 3 deacetylase inhibitor. Moreover, recently, the antioxidant effects of medicinal mushrooms have been shown to induce a lowering of the toxicity of chemotherapy drugs, inducing greater therapeutic efficiency, thus the combined therapy of chemotherapy and micotherapy could be helpful in the treatment of GBM reducing the adverse effects of the former thanks to phytotherapy's antioxidant, anti-inflammatory, immunomodulatory and antitumoral activities. Here, through immunoblotting, ultrastructural and immunofluorescence analysis, we evaluated the contribution in the activation of different cell death pathway of Micotherapy U-Care, a medicinal blend supplement, used together with platinum-based compounds on human glioblastoma U251 cells.
Collapse
Affiliation(s)
- Ludovica Gaiaschi
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy.
| | - Cristina Favaron
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Claudio Casali
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Federica Gola
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Fabrizio De Luca
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Mauro Ravera
- Department of Sciences and Technological Innovation (DiSIT), University of Piemonte Orientale "A. Avogadro", Viale Teresa Michel 11, 15121, Alessandria, Italy
| | - Elisa Roda
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
- Laboratory of Clinical and Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, ICS Maugeri Spa, IRCCS Pavia, Via Maugeri 10, Pavia, Italy
| | - Paola Rossi
- Laboratory of Neurophysiology and Integrated Physiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| | - Maria Grazia Bottone
- Laboratory of Cell Biology and Neurobiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Via Ferrata 9, 27100, Pavia, Italy
| |
Collapse
|
5
|
The power of a novel combined anticancer therapy: challenge and opportunity of micotherapy in the treatment of Glioblastoma Multiforme. Biomed Pharmacother 2022; 155:113729. [PMID: 36166961 DOI: 10.1016/j.biopha.2022.113729] [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: 07/20/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022] Open
Abstract
Glioblastoma (GBM) is the most common and mortal primary brain tumor in human. After standard therapies, that include surgical resection followed by radiotherapy and chemotherapy, it is difficult to completely remove the tumor and the development of relapses and resistance is almost inevitable. The chemotherapy now available also show important side effects, to overcame those limitation, new platinum-based drugs are being synthetized, Pt(IV)Ac-POA, (OC-6-44)-acetate-diamine-chloride(2-(2-propynyl)octanoato)platinum(IV), a prodrug having an Histone-3-DeAcetylase-Inhibitor as axial ligands, is one of them. Moreover, new compounds of plant origin are increasingly seen as potential sources of benefits in oncological treatments. The aim of the study is to investigate the possible contribution of micotherapy in the fight against GBM, its role in the metabolism of reactive oxygen species (ROS) and its synergic effect with a new platinum-based compound, Pt(IV)Ac-POA, on human glioblastoma U251 cells. Through cytofluorimetric and immunofluorescence analysis, the ability of the micotherapy in study to regulate the cell cycle was assessed, and its importance in controlling the cellular redox state was also revealed, opening to the possibility of a new therapy in which micotherapy can support the activity of new chemotherapy while reducing its side effects controlling inflammatory conditions in the microenvironment. Additionally, the combined therapy appeared able to induce regulated form of necrosis, such as ferroptosis, and to hinder the establishment of resistance mechanisms.
Collapse
|
6
|
Holsæter AM, Wizgird K, Karlsen I, Hemmingsen JF, Brandl M, Škalko-Basnet N. How docetaxel entrapment, vesicle size, zeta potential and stability change with liposome composition-A formulation screening study. Eur J Pharm Sci 2022; 177:106267. [PMID: 35872073 DOI: 10.1016/j.ejps.2022.106267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 12/01/2022]
Abstract
Limitations of the anticancer drug product Taxotere® have encouraged researchers to entrap the active ingredient docetaxel (DTX) into nanocarriers such as liposomes. However, until now no DTX-liposome formulation has reached the clinic. Hence, in the present study, different Soy-PC based DTX-liposome formulations were screened in an attempt to identify lipid-compositions with promising DTX-entrapment (DTX-EE). Various other quality attributes, such as vesicle size and morphology, poly dispersity index (PDI), zeta potential (ZP), stability and in vitro drug release were also investigated. In an initial study, the inclusion of charged lipids within the liposome bilayer was observed to have a positive effect on DTX-EE. Thus, cationic DOTAP (1,2-Dioleoyl-3-trimethylammonium-propane) and anionic DMPG (1,2-Dimyristoyl-sn-glycero-3-phospho-(1'-rac-glycerol) lipids were selected for further investigations. With anionic DMPG, only a temporary rise in EE was gained with ≥ 20% (w/w) DMPG in Soy-PC lipid-based liposomes, whereas a concentration-dependent increase in EE was observed with cationic DOTAP. A DTX-EE > 95% was obtained with only 5% (w/w) DOTAP in Soy-PC, while neutral liposomes formed from Soy-PC alone, gave 41.5% DTX-EE. In the stability study, a DOTAP concentration > 10% (w/w) in Soy-PC was found to facilitate a stable DTX-EE > 90% after 12 weeks storage. The positive effect of cationic lipids on the EE was confirmed when replacing cholesterol (CHOL), initially shown to suppress DTX-entrapment, with cationic 3ß-[N-(N',N'-dimethylaminoethane)-carbamoyl]Cholesterol (DC-CHOL). Here, DTX-EE was improved from 29.8% to 92.0% (w/w) with 10% (w/w) CHOL and DC-CHOL in Soy-PC, respectively. Finally, PEGylation of DOTAP-liposomes with DSPE-PEG2000 and DSPE-PEG750 reduced the DTX-EE relative to DOTAP-liposome with no PEGylation. As with the DMPG-liposomes, a temporarily raised affinity between DTX and liposomes was obtained with anionic DSPE-PEGylation of Soy-PC liposomes, however, this effect was not maintained after 4 weeks storage. However, in a dialysis set-up, cationic DOTAP-liposomes released DTX to a higher extent than PEGylated liposomes. Thus, the optimal formulation with regard to storage stability and in vivo performance need to be investigated further, applying conditions that are closer to mimic the in vivo-situation. Applying the Dual Asymmetric Centrifugation (DAC) method in liposome production appears favourable due to its good reproducibility. The observed increase in DTX entrapment with cationic lipids or PEGylation appears scalable into pilot manufacturing scale.
Collapse
Affiliation(s)
- Ann Mari Holsæter
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø 9037, Norway.
| | - Kristina Wizgird
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø 9037, Norway; Institute of Pharmaceutical Sciences, Department of Pharmaceutical Technology and Biopharmacy, Albert Ludwig University Freiburg, Freiburg 79085, Germany
| | - Iselin Karlsen
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø 9037, Norway
| | - Jeanette Frimand Hemmingsen
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø 9037, Norway; Drug Transport and Delivery, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense DK-5230, Denmark
| | - Martin Brandl
- Drug Transport and Delivery, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense DK-5230, Denmark
| | - Nataša Škalko-Basnet
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø 9037, Norway
| |
Collapse
|
7
|
Shakibaie M, Torabi-Shamsabad R, Forootanfar H, Amiri-Moghadam P, Amirheidari B, Adeli-Sardou M, Ameri A. Rapid microwave-assisted biosynthesis of platinum nanoparticles and evaluation of their antioxidant properties and cytotoxic effects against MCF-7 and A549 cell lines. 3 Biotech 2021; 11:511. [PMID: 34926109 DOI: 10.1007/s13205-021-03007-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
In this study, platinum nanoparticles (Pt NPs) were synthesized by a green method using an aqueous extract of Eucalyptus camaldulensis with assistance of microwave irradiation (850 W) and their physicochemical characteristics were studied by UV-visible spectroscopy, scanning electron microscopy (SEM), energy-dispersive X-ray spectroscopy (EDS), X-ray diffraction (XRD), and Fourier transform infrared spectroscopy (FTIR) analyses. Antioxidant activities, hemocompatibility, and cytotoxic effects of the prepared Pt NPs were then evaluated. The attained results showed that the newly formed Pt NPs possess a size range between 7.4 and 11.2 nm. These spherical-shaped NPs were slightly aggregated and held various functional groups on their surface. The antioxidant activity of Pt nanostructures was comparable to that of butylated hydroxyl anisole at concentrations higher than 320 µg/mL. At the same concentration of 640 μg/mL, the scavenging activities were 3.36 ± 0.9% (hexachloroplatinic acid) and 52.13 ± 0.43% (Pt NPs). The results of hemolytic assay revealed satisfactory hemocompatibility of the Pt NPs even at the concentration as high as 4 mg/mL (hemolysis percent equal to 3.5 ± 1.3%). The cytotoxicity studies revealed that MCF-7, A549, and 3T3 cell lines treated with hexachloroplatinic acid and cisplatin for 24 h and 48 h showed a higher percentage of cell death compared with the Pt NPs. After 24 h, for A549, 3T3, and MCF-7 cells exposed to Pt NPs, the cell viability was measured to be 80 ± 3.2%, 96 ± 1%, and 89 ± 2.6%, respectively, at concentration of 640 µg/mL. Further investigations are required to elucidate the mechanisms behind the biological activities of as-synthesized Pt NPs.
Collapse
Affiliation(s)
- Mojtaba Shakibaie
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Hamid Forootanfar
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Parinaz Amiri-Moghadam
- The Student Research Committee, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Bagher Amirheidari
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahboubeh Adeli-Sardou
- Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Atefeh Ameri
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
8
|
Maiuolo J, Gliozzi M, Carresi C, Musolino V, Oppedisano F, Scarano F, Nucera S, Scicchitano M, Bosco F, Macri R, Ruga S, Cardamone A, Coppoletta A, Mollace A, Cognetti F, Mollace V. Nutraceuticals and Cancer: Potential for Natural Polyphenols. Nutrients 2021; 13:nu13113834. [PMID: 34836091 PMCID: PMC8619660 DOI: 10.3390/nu13113834] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/20/2021] [Accepted: 10/25/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer is one of the leading causes of death globally, associated with multifactorial pathophysiological components. In particular, genetic mutations, infection or inflammation, unhealthy eating habits, exposition to radiation, work stress, and/or intake of toxins have been found to contribute to the development and progression of cancer disease states. Early detection of cancer and proper treatment have been found to enhance the chances of survival and healing, but the side effects of anticancer drugs still produce detrimental responses that counteract the benefits of treatment in terms of hospitalization and survival. Recently, several natural bioactive compounds were found to possess anticancer properties, capable of killing transformed or cancerous cells without being toxic to their normal counterparts. This effect occurs when natural products are associated with conventional treatments, thereby suggesting that nutraceutical supplementation may contribute to successful anticancer therapy. This review aims to discuss the current literature on four natural bioactive extracts mostly characterized by a specific polyphenolic profile. In particular, several activities have been reported to contribute to nutraceutical support in anticancer treatment: (1) inhibition of cell proliferation, (2) antioxidant activity, and (3) anti-inflammatory activity. On the other hand, owing to their attenuation of the toxic effect of current anticancer therapies, natural antioxidants may contribute to improving the compliance of patients undergoing anticancer treatment. Thus, nutraceutical supplementation, along with current anticancer drug treatment, may be considered for better responses and compliance in patients with cancer. It should be noted, however, that when data from studies with bioactive plant preparations are discussed, it is appropriate to ensure that experiments have been conducted in accordance with accepted pharmacological research practices so as not to disclose information that is only partially correct.
Collapse
Affiliation(s)
- Jessica Maiuolo
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Micaela Gliozzi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Cristina Carresi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Vincenzo Musolino
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Francesca Oppedisano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Federica Scarano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Saverio Nucera
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Miriam Scicchitano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Francesca Bosco
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Roberta Macri
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Stefano Ruga
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
| | - Antonio Cardamone
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
| | - Annarita Coppoletta
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
| | - Annachiara Mollace
- Medical Oncology 1, Regina Elena National Cancer Institute, IRCCS, 00144 Rome, Italy; (A.M.); (F.C.)
| | - Francesco Cognetti
- Medical Oncology 1, Regina Elena National Cancer Institute, IRCCS, 00144 Rome, Italy; (A.M.); (F.C.)
| | - Vincenzo Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
- IRCCS San Raffaele, Via di Valcannuta 247, 00133 Rome, Italy
- Correspondence:
| |
Collapse
|
9
|
Ruthenium Complexes as Promising Candidates against Lung Cancer. Molecules 2021; 26:molecules26154389. [PMID: 34361543 PMCID: PMC8348655 DOI: 10.3390/molecules26154389] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 11/17/2022] Open
Abstract
Lung cancer is one of the most common malignancies with the highest mortality rate and the second-highest incidence rate after breast cancer, posing a serious threat to human health. The accidental discovery of the antitumor properties of cisplatin in the early 1960s aroused a growing interest in metal-based compounds for cancer treatment. However, the clinical application of cisplatin is limited by serious side effects and drug resistance. Therefore, other transition metal complexes have been developed for the treatment of different malignant cancers. Among them, Ru(II/III)-based complexes have emerged as promising anticancer drug candidates due to their potential anticancer properties and selective cytotoxic activity. In this review, we summarized the latest developments of Ru(II/III) complexes against lung cancer, focusing mainly on the mechanisms of their biological activities, including induction of apoptosis, necroptosis, autophagy, cell cycle arrest, inhibition of cell proliferation, and invasion and metastasis of lung cancer cells.
Collapse
|
10
|
Cordani M, Somoza Á, Tafani M, Dando I, Kumar S. Editorial: Novel Cancer Treatments Based on Autophagy Modulation. Front Pharmacol 2021; 12:650559. [PMID: 33935758 PMCID: PMC8085590 DOI: 10.3389/fphar.2021.650559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/01/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Marco Cordani
- IMDEA Nanociencia, Ciudad Universitaria de Cantoblanco, Madrid, Spain
| | - Álvaro Somoza
- IMDEA Nanociencia, Ciudad Universitaria de Cantoblanco, Madrid, Spain
| | - Marco Tafani
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Ilaria Dando
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Suresh Kumar
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
11
|
Ferrari B, Roda E, Priori EC, De Luca F, Facoetti A, Ravera M, Brandalise F, Locatelli CA, Rossi P, Bottone MG. A New Platinum-Based Prodrug Candidate for Chemotherapy and Its Synergistic Effect With Hadrontherapy: Novel Strategy to Treat Glioblastoma. Front Neurosci 2021; 15:589906. [PMID: 33828444 PMCID: PMC8019820 DOI: 10.3389/fnins.2021.589906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is the most common tumor of the central nervous system. Current therapies, often associated with severe side effects, are inefficacious to contrast the GBM relapsing forms. In trying to overcome these drawbacks, (OC-6-44)-acetatodiamminedichlorido(2-(2-propynyl)octanoato)platinum(IV), also called Pt(IV)Ac-POA, has been recently synthesized. This new prodrug bearing as axial ligand (2-propynyl)octanoic acid (POA), a histone deacetylase inhibitor, has a higher activity due to (i) its high cellular accumulation by virtue of its high lipophilicity and (ii) the inhibition of histone deacetylase, which leads to the increased exposure of nuclear DNA, permitting higher platination and promoting cancer cell death. In the present study, we investigated the effects induced by Pt(IV)Ac-POA and its potential antitumor activity in human U251 glioblastoma cell line using a battery of complementary techniques, i.e., flow cytometry, immunocytochemistry, TEM, and Western blotting analyses. In addition, the synergistic effect of Pt(IV)Ac-POA associated with the innovative oncological hadrontherapy with carbon ions was investigated, with the aim to identify the most efficient anticancer treatment combination. Our in vitro data demonstrated that Pt(IV)Ac-POA is able to induce cell death, through different pathways, at concentrations lower than those tested for other platinum analogs. In particular, an enduring Pt(IV)Ac-POA antitumor effect, persisting in long-term treatment, was demonstrated. Interestingly, this effect was further amplified by the combined exposure to carbon ion radiation. In conclusion, Pt(IV)Ac-POA represents a promising prodrug to be incorporated into the treatment regimen for GBM. Moreover, the synergistic efficacy of the combined protocol using chemotherapeutic Pt(IV)Ac-POA followed by carbon ion radiation may represent a promising approach, which may overcome some typical limitations of conventional therapeutic protocols for GBM treatment.
Collapse
Affiliation(s)
- Beatrice Ferrari
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Elisa Roda
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy.,Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Erica Cecilia Priori
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Fabrizio De Luca
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Angelica Facoetti
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy
| | - Mauro Ravera
- Department of Sciences and Technological Innovation (DiSIT), University of Piemonte Orientale "A. Avogadro", Alessandria, Italy
| | - Federico Brandalise
- Department of Fundamental Neurosciences (NEUFO), University of Geneva, Geneva, Switzerland
| | - Carlo Alessandro Locatelli
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Paola Rossi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Maria Grazia Bottone
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
12
|
Alvarez-Meythaler JG, Garcia-Mayea Y, Mir C, Kondoh H, LLeonart ME. Autophagy Takes Center Stage as a Possible Cancer Hallmark. Front Oncol 2020; 10:586069. [PMID: 33194736 PMCID: PMC7643020 DOI: 10.3389/fonc.2020.586069] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer remains one of the leading causes of death worldwide, despite significant advances in cancer research and improvements in anticancer therapies. One of the major obstacles to curing cancer is the difficulty of achieving the complete annihilation of resistant cancer cells. The resistance of cancer cells may not only be due to intrinsic factors or factors acquired during the evolution of the tumor but may also be caused by chemotherapeutic treatment failure. Conversely, autophagy is a conserved cellular process in which intracellular components, such as damaged organelles, aggregated or misfolded proteins and macromolecules, are degraded or recycled to maintain cellular homeostasis. Importantly, autophagy is an essential mechanism that plays a key role in tumor initiation and progression. Depending on the cellular context and microenvironmental conditions, autophagy acts as a double-edged sword, playing a role in inducing apoptosis or promoting cell survival. In this review, we propose several scenarios in which autophagy could contribute to cell survival or cell death. Moreover, a special focus on novel promising targets and therapeutic strategies based on autophagic resistant cells is presented.
Collapse
Affiliation(s)
- Jose G. Alvarez-Meythaler
- Biomedical Research in Cancer Stem Cells Laboratory, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Yoelsis Garcia-Mayea
- Biomedical Research in Cancer Stem Cells Laboratory, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Cristina Mir
- Biomedical Research in Cancer Stem Cells Laboratory, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Hiroshi Kondoh
- Geriatric Unit, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Matilde E. LLeonart
- Biomedical Research in Cancer Stem Cells Laboratory, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
- Spanish Biomedical Research Network Center in Oncology, CIBERONC, Barcelona, Spain
| |
Collapse
|
13
|
[Pt(O,O'-acac)(γ-acac)(DMS)]: Alternative Strategies to Overcome Cisplatin-Induced Side Effects and Resistance in T98G Glioma Cells. Cell Mol Neurobiol 2020; 41:563-587. [PMID: 32430779 DOI: 10.1007/s10571-020-00873-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Cisplatin (CDDP) is one of the most effective chemotherapeutic agents, used for the treatment of diverse tumors, including neuroblastoma and glioblastoma. CDDP induces cell death through different apoptotic pathways. Despite its clinical benefits, CDDP causes several side effects and drug resistance.[Pt(O,O'-acac)(γ-acac)(DMS)], namely PtAcacDMS, a new platinum(II) complex containing two acetylacetonate (acac) and a dimethylsulphide (DMS) in the coordination sphere of metal, has been recently synthesized and showed 100 times higher cytotoxicity than CDDP. Additionally, PtAcacDMS was associated to a decreased neurotoxicity in developing rat central nervous system, also displaying great antitumor and antiangiogenic activity both in vivo and in vitro. Thus, based on the knowledge that several chemotherapeutics induce cancer cell death through an aberrant increase in [Ca2+]i, in the present in vitro study we compared CDDP and PtAcacDMS effects on apoptosis and intracellular Ca2+ dynamics in human glioblastoma T98G cells, applying a battery of complementary techniques, i.e., flow cytometry, immunocytochemistry, electron microscopy, Western blotting, qRT-PCR, and epifluorescent Ca2+ imaging. The results confirmed that (i) platinum compounds may induce cell death through an aberrant increase in [Ca2+]i and (ii) PtAcacDMS exerted stronger cytotoxic effect than CDDP, associated to a larger increase in resting [Ca2+]i. These findings corroborate the use of PtAcacDMS as a promising approach to improve Pt-based chemotherapy against gliomas, either by inducing a chemosensitization or reducing chemoresistance in cell lineages resilient to CDDP treatment.
Collapse
|
14
|
Zhang Y, Zheng J, Jiang Y, Huang X, Fang L. Neglected, Drug-Induced Platinum Accumulation Causes Immune Toxicity. Front Pharmacol 2020; 11:1166. [PMID: 32903504 PMCID: PMC7438596 DOI: 10.3389/fphar.2020.01166] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/17/2020] [Indexed: 02/05/2023] Open
Abstract
Previous studies only focused on different adverse reactions caused by various platinum drugs, but not on common immunotoxicity caused by the accumulation of elemental platinum. Here, we determined the serum platinum concentrations of cancer patients after a metabolism period of platinum drug chemotherapy, in addition to hematological indices and subsequent immune-related adverse reactions, then analyzed the correlations between platinum accumulation, immune cell levels, and immune-toxicity. We chose the day before the next round of chemotherapy as the specified time point for blood sampling. Samples were collected at five time points, separately in oxaliplatin and cisplatin groups. The median serum platinum concentrations in all patients was 294.8 (205.6, 440.3) μg/L, and was approximately two-fold greater in the cisplatin group than in the oxaliplatin group (429.3 vs. 211.7 μg/L). The platinum level of both groups peaked at the third time point, with the average of females being higher than males (383.9 vs. 266.5 μg/L), and was positively correlated with leukocyte and platelet counts, but negatively correlated with erythrocyte counts and concentration of hemoglobin. The risks of anemia and adverse reactions were individually increased by 0.002- and 0.007-fold for every μg/L increase of platinum concentration. To our knowledge, this is the first study on the relationship between platinum accumulation, immune cell levels and toxicity, showing that drug-induced platinum accumulation may interfere with immune cells and thus increase the risk of toxicity.
Collapse
Affiliation(s)
- Yuling Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou, China
| | - Jieting Zheng
- Pharmacy Department, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yi Jiang
- Digestive Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Xuchun Huang
- Department of Clinical Laboratory Medicine, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Ling Fang
- Pharmacy Intravenous Admixture Service, Cancer Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Ling Fang,
| |
Collapse
|
15
|
Wu H, Chen L, Zhu F, Han X, Sun L, Chen K. The Cytotoxicity Effect of Resveratrol: Cell Cycle Arrest and Induced Apoptosis of Breast Cancer 4T1 Cells. Toxins (Basel) 2019; 11:toxins11120731. [PMID: 31847250 PMCID: PMC6950385 DOI: 10.3390/toxins11120731] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022] Open
Abstract
Resveratrol, a natural polyterpenoid, can scavenge reactive oxygen species in vivo to carry out the functions of antioxidation and antiaging. Resveratrol’s anti-cancer capability has attracted widespread attention, but its molecular mechanism has not been systematically explained. In this study, by comparing the activity of normal cell lines and cancer cell lines after treating with resveratrol, it was found that resveratrol has more significant cytotoxicity in cancer cell lines. Resveratrol could play a toxic role through inducing apoptosis of the cancer cell in a time- and concentration-dependent manner. A total of 330 significantly differential genes were identified through large-scale transcriptome sequencing, among which 103 genes were upregulated and 227 genes were downregulated. Transcriptome and qRT-PCR data proved that a large number of genes related to cell cycle were differentially expressed after the treatment of resveratrol. The changes of cell cycle phases at different time points after treating with resveratrol were further detected, and it was found that the cells were arrested in the S phase because of the percentage of cells in S phase increased and cells in G1/G0 phase decreased. In conclusion, resveratrol can inhibit the proliferation of 4T1 cancer cells by inhibiting cell cycle and inducing apoptosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Keping Chen
- Correspondence: ; Tel./Fax: +86-511-88791923
| |
Collapse
|
16
|
Molina V, Rodríguez-Vázquez L, Martí J. Patterns of Apoptosis and Autophagy Activation After Hydroxyurea Exposure in the Rat Cerebellar External Granular Layer: an Immunoperoxidase and Ultrastructural Analysis. Neurotox Res 2019; 37:93-99. [DOI: 10.1007/s12640-019-00094-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/24/2019] [Accepted: 07/30/2019] [Indexed: 12/14/2022]
|
17
|
New Platinum-Based Prodrug Pt(IV)Ac-POA: Antitumour Effects in Rat C6 Glioblastoma Cells. Neurotox Res 2019; 37:183-197. [PMID: 31240667 DOI: 10.1007/s12640-019-00076-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 03/30/2019] [Accepted: 06/13/2019] [Indexed: 02/07/2023]
Abstract
Gliomas are the most frequent primary tumours of the nervous system, characterised by high degree of malignancy, widespread invasion and high-rate proliferation. Cisplatin and analogue are currently employed in clinical trials as active chemotherapeutic agents for the systemic treatment of this type of malignancy. Despite therapy benefits, clinical use of these agents is hampered by severe side effects including neurotoxicity. Therefore, the aim of the present study was to analyse the effect of a new compound of platinum(IV) conjugate, named Pt(IV)Ac-POA, which can generate a synergistic antineoplastic action when released along with cisplatin, after a specific reduction reaction within tumour cells. To assess the effects of the novel compound on rat C6 glioma cells, cell cycle and cell death activation analyses were carried out using flow cytometry. Morphological changes and activation of different cell death pathways were evaluated by both transmission electron microscopy and immunofluorescence microscopy. Protein expression was investigated by western blotting analysis. The novel compound Pt(IV)Ac-POA, bearing as axial ligand (2-propynyl)octanoic acid (POA), which is a histone deacetylase inhibitor (HDACi), acts as a prodrug in tumour cells, inducing cell death through different pathways at a concentration lower than those tested for other platinum analogues. The current results showed that Pt(IV)Ac-POA could represent a promising improvement of Pt-based chemotherapy against gliomas, either inducing a chemosensitisation and reducing chemoresistance.
Collapse
|
18
|
Antonaci G, Cossa LG, Muscella A, Vetrugno C, De Pascali SA, Fanizzi FP, Marsigliante S. [Pt( O,O'-acac)(γ-acac)(DMS)] Induces Autophagy in Caki-1 Renal Cancer Cells. Biomolecules 2019; 9:biom9030092. [PMID: 30845773 PMCID: PMC6468382 DOI: 10.3390/biom9030092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/23/2019] [Accepted: 02/26/2019] [Indexed: 01/12/2023] Open
Abstract
We have demonstrated the cytotoxic effects of [Pt(O,O′-acac)(γ-acac)(dimethyl sulfide (DMS))] on various immortalized cell lines, in primary cultures, and in murine xenograft models in vivo. Recently, we also showed that [Pt(O,O′-acac)(γ-acac)(DMS)] is able to kill Caki-1 renal cells both in vivo and in vitro. In the present paper, apoptotic and autophagic effects of [Pt(O,O′-acac)(γ-acac)(DMS)] and cisplatin were studied and compared using Caki-1 cancerous renal cells. The effects of cisplatin include activation of caspases, proteolysis of enzyme poly ADP ribose polymerase (PARP), control of apoptosis modulators B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax), and BH3-interacting domain death agonist (Bid), and cell cycle arrest in G2/M phase. Conversely, [Pt(O,O′-acac)(γ-acac)(DMS)] did not induce caspase activation, nor chromatin condensation or DNA fragmentation. The effects of [Pt(O,O′-acac)(γ-acac)(DMS)] include microtubule-associated proteins 1A/1B light chain 3B (LC3)-I to LC3-II conversion, Beclin-1 and Atg-3, -4, and -5 increase, Bcl-2 decrease, and monodansylcadaverine accumulation in autophagic vacuoles. [Pt(O,O′-acac)(γ-acac)(DMS)] also modulated various kinases involved in intracellular transduction regulating cell fate. [Pt(O,O′-acac)(γ-acac)(DMS)] inhibited the phosphorylation of mammalian target of rapmycin (mTOR), p70S6K, and AKT, and increased the phosphorylation of c-Jun N-terminal kinase (JNK1/2), a kinase activity pattern consistent with autophagy induction. In conclusion, while in past reports the high cytotoxicity of [Pt(O,O′-acac)(γ-acac)(DMS)] was always attributed to its ability to trigger an apoptotic process, in this paper we show that Caki-1 cells die as a result of the induction of a strong autophagic process.
Collapse
Affiliation(s)
- Giovanna Antonaci
- Laboratory of Cell Physiology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Luca Giulio Cossa
- Laboratory of Cell Physiology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Antonella Muscella
- Laboratory of Cell Pathology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Carla Vetrugno
- Laboratory of Cell Pathology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Sandra Angelica De Pascali
- Laboratory of General Inorganic Chemistry, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Francesco Paolo Fanizzi
- Laboratory of General Inorganic Chemistry, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| | - Santo Marsigliante
- Laboratory of Cell Physiology, Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy.
| |
Collapse
|