1
|
Trejo-Solís C, Escamilla-Ramírez Á, Gómez-Manzo S, Castillo-Rodriguez RA, Palomares-Alonso F, Castillo-Pérez C, Jiménez-Farfán D, Sánchez-García A, Gallardo-Pérez JC. The pentose phosphate pathway (PPP) in the glioma metabolism: A potent enhancer of malignancy. Biochimie 2025; 232:117-126. [PMID: 39894336 DOI: 10.1016/j.biochi.2025.01.013] [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: 06/20/2024] [Revised: 01/23/2025] [Accepted: 01/31/2025] [Indexed: 02/04/2025]
Abstract
The glioma hallmark includes reprogramming metabolism to support biosynthetic and bioenergetic demands, as well as to maintain their redox equilibrium. It has been suggested that the pentose phosphate pathway (PPP) and glycolysis are directly involved in the dynamics and regulation of glioma cell proliferation and migration. The PPP is implicated in cellular redox homeostasis and the modulation of signaling pathways, which play a fundamental role in the progression of tumors to malignant grades, metastasis, and drug resistance. Several studies have shown that in glioblastoma cells, the activity, expression, and metabolic flux of some PPP enzymes increase, leading to heightened activity of the pathway. This generates higher levels of DNA, lipids, cholesterol, and amino acids, favoring rapid cell proliferation. Due to the crucial role played by the PPP in the development of glioma cells, enzymes from this pathway have been proposed as potential therapeutic targets. This review summarizes and highlights the role that the PPP plays in glioma cells and focuses on the key functions of the enzymes and metabolites generated by this pathway, as well as the regulation of the PPP. The studies described in this article enrich the understanding of the PPP as a therapeutic tool in the search for pharmacological targets for the development of a new generation of drugs to treat glioma.
Collapse
Affiliation(s)
- Cristina Trejo-Solís
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Unidad Periférica para el Estudio de la Neuroinflamación, Laboratorio de Neuropatologia Experimental, Instituto Nacional de Neurología y Neurocirugía, CDMX, 14269, Mexico.
| | | | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, CDMX, 04530, Mexico.
| | - Rosa Angélica Castillo-Rodriguez
- CICATA Unidad Morelos, Instituto Politécnico Nacional, Boulevard de la Tecnología, 1036 Z-1, P 2/2, Atlacholoaya, 62790, Xochitepec, Mexico.
| | - Francisca Palomares-Alonso
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Unidad Periférica para el Estudio de la Neuroinflamación, Laboratorio de Neuropatologia Experimental, Instituto Nacional de Neurología y Neurocirugía, CDMX, 14269, Mexico
| | - Carlos Castillo-Pérez
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Unidad Periférica para el Estudio de la Neuroinflamación, Laboratorio de Neuropatologia Experimental, Instituto Nacional de Neurología y Neurocirugía, CDMX, 14269, Mexico.
| | - Dolores Jiménez-Farfán
- Laboratorio de Inmunología, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, 04510, Ciudad de Mexico, Mexico.
| | - Aurora Sánchez-García
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Unidad Periférica para el Estudio de la Neuroinflamación, Laboratorio de Neuropatologia Experimental, Instituto Nacional de Neurología y Neurocirugía, CDMX, 14269, Mexico
| | - Juan Carlos Gallardo-Pérez
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología, 14080, Ciudad de Mexico, Mexico; Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Ciudad de México, Mexico.
| |
Collapse
|
2
|
Ul Hassan MH, Shahbaz M, Imran M, Momal U, Naeem H, Mujtaba A, Hussain M, Anwar MJ, Alsagaby SA, Al Abdulmonem W, Yehuala TF, Abdelgawad MA, El‐Ghorab AH, Selim S, Mostafa EM. Isoflavones: Promising Natural Agent for Cancer Prevention and Treatment. Food Sci Nutr 2025; 13:e70091. [PMID: 40078339 PMCID: PMC11896816 DOI: 10.1002/fsn3.70091] [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: 10/08/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Isoflavones are currently being investigated by researchers in order to demonstrate their ability to prevent the proliferation of cancer cells. The current review aimed to demonstrate the potential of isoflavones to eliminate cancerous cells in the stomach, liver, lung, breast, and prostate, as their anticancer properties are due to the ability to block the signaling pathways of the extracellular signal-controlled kinase (MAPK/ERK) and proteasome (PI3K/AKT/mTOR). Isoflavones can inhibit the cell division of various cancer cells. Isoflavones can block the androgen receptor (AR), a protein that is required for the growth and dissemination of prostate cancer. It initiates the caspase cascade and obstructs the production of new proteins to eliminate lung cancer cells. These inhibit colon cancer cells by entering their G2/M cell cycle phase and inducing apoptosis. These are also known to inhibit the production of cyclin-dependent kinase 2 and cyclin B1, two proteins that are related to an enhanced risk of colon cancer. These suppress the breakdown of cyclin B1 and CDK2 to stop the development of cancer. Preclinical evidence consistently supports the efficacy of isoflavones in suppressing tumor growth; however, human clinical trials show variability due to differences in bioavailability, metabolism, and dosage. Despite their promise as alternative or adjunctive cancer therapies, limitations such as low solubility, interindividual metabolic variations, and inconsistent clinical outcomes necessitate further large-scale, controlled trials. Future research should focus on improving bioavailability and exploring synergistic effects with conventional therapies.
Collapse
Affiliation(s)
- Muhammad Hammad Ul Hassan
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of AgricultureMultanPakistan
| | - Muhammad Shahbaz
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of AgricultureMultanPakistan
| | - Muhammad Imran
- Department of Food Science and TechnologyUniversity of NarowalNarowalPakistan
| | - Ushna Momal
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of AgricultureMultanPakistan
| | - Hammad Naeem
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of AgricultureMultanPakistan
- Post Harvest Research CentreAyub Agricultural Research InstituteFaisalabadPakistan
| | - Ahmed Mujtaba
- Department of Food Science and Technology, Faculty of Engineering Sciences and TechnologyHamdard University Islamabad CampusIslamabadPakistan
| | - Muzzamal Hussain
- Department of Food ScienceGovernment College University FaisalabadFaisalabadPakistan
| | - Muhammad Junaid Anwar
- Department of Food Science and Technology, Faculty of Food Science and NutritionBahauddin Zakariya UniversityMultanPakistan
| | - Suliman A. Alsagaby
- Department of Medical Laboratory Sciences, College of Applied Medical SciencesMajmaah UniversityAl‐MajmaahSaudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of MedicineQassim UniversityBuraidahSaudi Arabia
| | - Tadesse Fenta Yehuala
- Faculty of Chemical and Food Engineering, Bahir Dar Institute of TechnologyBahir Dar UniversityBahir Dar CityEthiopia
| | - Mohamed A. Abdelgawad
- Department of Pharmaceutical Chemistry, College of PharmacyJouf UniversitySakakaAljoufSaudi Arabia
| | - Ahmed H. El‐Ghorab
- Department of Chemistry, College of ScienceJouf UniversitySakakaSaudi Arabia
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesJouf UniversitySakakaSaudi Arabia
| | - Ehab M. Mostafa
- Department of Pharmacognosy, College of PharmacyJouf UniversitySakakaSaudi Arabia
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys)Al‐Azhar UniversityCairoEgypt
| |
Collapse
|
3
|
Pennisi G, Bruzzaniti P, Burattini B, Piaser Guerrato G, Della Pepa GM, Sturiale CL, Lapolla P, Familiari P, La Pira B, D’Andrea G, Olivi A, D’Alessandris QG, Montano N. Advancements in Telomerase-Targeted Therapies for Glioblastoma: A Systematic Review. Int J Mol Sci 2024; 25:8700. [PMID: 39201386 PMCID: PMC11354571 DOI: 10.3390/ijms25168700] [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: 06/25/2024] [Revised: 07/25/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Glioblastoma (GBM) is a primary CNS tumor that is highly lethal in adults and has limited treatment options. Despite advancements in understanding the GBM biology, the standard treatment for GBM has remained unchanged for more than a decade. Only 6.8% of patients survive beyond five years. Telomerase, particularly the hTERT promoter mutations present in up to 80% of GBM cases, represents a promising therapeutic target due to its role in sustaining telomere length and cancer cell proliferation. This review examines the biology of telomerase in GBM and explores potential telomerase-targeted therapies. We conducted a systematic review following the PRISMA-P guidelines in the MEDLINE/PubMed and Scopus databases, from January 1995 to April 2024. We searched for suitable articles by utilizing the terms "GBM", "high-grade gliomas", "hTERT" and "telomerase". We incorporated studies addressing telomerase-targeted therapies into GBM studies, excluding non-English articles, reviews, and meta-analyses. We evaluated a total of 777 records and 46 full texts, including 36 studies in the final review. Several compounds aimed at inhibiting hTERT transcription demonstrated promising preclinical outcomes; however, they were unsuccessful in clinical trials owing to intricate regulatory pathways and inadequate pharmacokinetics. Direct hTERT inhibitors encountered numerous obstacles, including a prolonged latency for telomere shortening and the activation of the alternative lengthening of telomeres (ALT). The G-quadruplex DNA stabilizers appeared to be potential indirect inhibitors, but further clinical studies are required. Imetelstat, the only telomerase inhibitor that has undergone clinical trials, has demonstrated efficacy in various cancers, but its efficacy in GBM has been limited. Telomerase-targeted therapies in GBM is challenging due to complex hTERT regulation and inadequate inhibitor pharmacokinetics. Our study demonstrates that, despite promising preclinical results, no Telomerase inhibitors have been approved for GBM, and clinical trials have been largely unsuccessful. Future strategies may include Telomerase-based vaccines and multi-target inhibitors, which may provide more effective treatments when combined with a better understanding of telomere dynamics and tumor biology. These treatments have the potential to be integrated with existing ones and to improve the outcomes for patients with GBM.
Collapse
Affiliation(s)
- Giovanni Pennisi
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
- Department of Neurosurgery, F. Spaziani Hospital, 03100 Frosinone, Italy; (B.L.P.); (G.D.)
| | - Placido Bruzzaniti
- Department of Neurosurgery, F. Spaziani Hospital, 03100 Frosinone, Italy; (B.L.P.); (G.D.)
- Department of Human Neurosciences, Division of Neurosurgery, Policlinico Umberto I University Hospital, Sapienza, University of Rome, 00157 Rome, Italy;
| | - Benedetta Burattini
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
| | - Giacomo Piaser Guerrato
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
| | - Giuseppe Maria Della Pepa
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
| | - Carmelo Lucio Sturiale
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
| | | | - Pietro Familiari
- Department of Human Neurosciences, Division of Neurosurgery, Policlinico Umberto I University Hospital, Sapienza, University of Rome, 00157 Rome, Italy;
| | - Biagia La Pira
- Department of Neurosurgery, F. Spaziani Hospital, 03100 Frosinone, Italy; (B.L.P.); (G.D.)
| | - Giancarlo D’Andrea
- Department of Neurosurgery, F. Spaziani Hospital, 03100 Frosinone, Italy; (B.L.P.); (G.D.)
| | - Alessandro Olivi
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
| | - Quintino Giorgio D’Alessandris
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
| | - Nicola Montano
- Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (G.P.); (B.B.); (G.P.G.); (G.M.D.P.); (C.L.S.); (A.O.); (Q.G.D.); (N.M.)
| |
Collapse
|
4
|
Ng GYQ, Hande MP. Use of peptide nucleic acid probe to determine telomere dynamics in improving chromosome analysis in genetic toxicology studies. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2024; 897:503773. [PMID: 39054004 DOI: 10.1016/j.mrgentox.2024.503773] [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: 02/10/2024] [Revised: 05/01/2024] [Accepted: 05/22/2024] [Indexed: 07/27/2024]
Abstract
Genetic toxicology, strategically located at the intersection of genetics and toxicology, aims to demystify the complex interplay between exogenous agents and our genetic blueprint. Telomeres, the protective termini of chromosomes, play instrumental roles in cellular longevity and genetic stability. Traditionally karyotyping and fluorescence in situ hybridisation (FISH), have been indispensable tools for chromosomal analysis following exposure to genotoxic agents. However, their scope in discerning nuanced molecular dynamics is limited. Peptide Nucleic Acids (PNAs) are synthetic entities that embody characteristics of both proteins and nucleic acids and have emerged as potential game-changers. This perspective report comprehensively examines the vast potential of PNAs in genetic toxicology, with a specific emphasis on telomere research. PNAs' superior resolution and precision make them a favourable choice for genetic toxicological assessments. The integration of PNAs in contemporary analytical workflows heralds a promising evolution in genetic toxicology, potentially revolutionizing diagnostics, prognostics, and therapeutic avenues. In this timely review, we attempted to assess the limitations of current PNA-FISH methodology and recommend refinements.
Collapse
Affiliation(s)
- Gavin Yong Quan Ng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Manoor Prakash Hande
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
5
|
Choudhury SD, Kumar P, Choudhury D. Bioactive nutraceuticals as G4 stabilizers: potential cancer prevention and therapy-a critical review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3585-3616. [PMID: 38019298 DOI: 10.1007/s00210-023-02857-z] [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: 08/14/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023]
Abstract
G-quadruplexes (G4) are non-canonical, four-stranded, nucleic acid secondary structures formed in the guanine-rich sequences, where guanine nucleotides associate with each other via Hoogsteen hydrogen bonding. These structures are widely found near the functional regions of the mammalian genome, such as telomeres, oncogenic promoters, and replication origins, and play crucial regulatory roles in replication and transcription. Destabilization of G4 by various carcinogenic agents allows oncogene overexpression and extension of telomeric ends resulting in dysregulation of cellular growth-promoting oncogenesis. Therefore, targeting and stabilizing these G4 structures with potential ligands could aid cancer prevention and therapy. The field of G-quadruplex targeting is relatively nascent, although many articles have demonstrated the effect of G4 stabilization on oncogenic expressions; however, no previous study has provided a comprehensive analysis about the potency of a wide variety of nutraceuticals and some of their derivatives in targeting G4 and the lattice of oncogenic cell signaling cascade affected by them. In this review, we have discussed bioactive G4-stabilizing nutraceuticals, their sources, mode of action, and their influence on cellular signaling, and we believe our insight would bring new light to the current status of the field and motivate researchers to explore this relatively poorly studied arena.
Collapse
Affiliation(s)
- Satabdi Datta Choudhury
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Prateek Kumar
- School of Basic Sciences, Indian Institute of Technology (IIT), Mandi, Himachal Pradesh, 175005, India
| | - Diptiman Choudhury
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
- Centre for Excellence in Emerging Materials, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
| |
Collapse
|
6
|
Verma P, Joshi H, Singh T, Sharma B, Sharma U, Ramniwas S, Rana R, Gupta M, Kaur G, Tuli HS. Temozolomide and flavonoids against glioma: from absorption and metabolism to exosomal delivery. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:41-57. [PMID: 37566307 DOI: 10.1007/s00210-023-02660-w] [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: 02/12/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023]
Abstract
Patients with glioblastoma multiforme and anaplastic astrocytoma are treated with temozolomide. Although it has been demonstrated that temozolomide increases GBM patient survival, it has also been connected to negative immune-related adverse effects. Numerous research investigations have shown that flavonoids have strong antioxidant and chemo-preventive effects. Consequently, it might lessen chemotherapeutic medicines' side effects while also increasing therapeutic effectiveness. The need for creating innovative, secure, and efficient drug carriers for cancer therapy has increased over time. Recent research indicates that exosomes have enormous potential to serve as carriers and cutting-edge drug delivery systems to the target cell. In recent years, researchers have been paying considerable attention to exosomes because of their favorable biodistribution, biocompatibility, and low immunogenicity. In the present review, the mechanistic information of the anti-glioblastoma effects of temozolomide and flavonoids coupled with their exosomal delivery to the targeted cell has been discussed. In addition, we discuss the safety aspects of temozolomide and flavonoids against glioma. The in-depth information of temozolomide and flavonoids action via exosomal delivery can unravel novel strategies to target Glioma.
Collapse
Affiliation(s)
- Priyanka Verma
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, India
| | - Hemant Joshi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Tejveer Singh
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
| | - Bunty Sharma
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, India
| | - Ujjawal Sharma
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bhatinda, 151001, India
| | - Seema Ramniwas
- University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, Mohali, 140413, India
| | - Rashmi Rana
- Department of Research, Sir Ganga Ram Hospital, New Delhi, 122016, India.
| | - Madhu Gupta
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Ginpreet Kaur
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle-West, Mumbai, 400056, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, India.
| |
Collapse
|
7
|
Anticancer Mechanism of Flavonoids on High-Grade Adult-Type Diffuse Gliomas. Nutrients 2023; 15:nu15040797. [PMID: 36839156 PMCID: PMC9964830 DOI: 10.3390/nu15040797] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/23/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
High-grade adult-type diffuse gliomas are the most common and deadliest malignant adult tumors of the central nervous system. Despite the advancements in the multimodality treatment of high-grade adult-type diffuse gliomas, the five-year survival rates still remain poor. The biggest challenge in treating high-grade adult-type diffuse gliomas is the intra-tumor heterogeneity feature of the glioma tumors. Introducing dietary flavonoids to the current high-grade adult-type diffuse glioma treatment strategies is crucial to overcome this challenge, as flavonoids can target several molecular targets. This review discusses the anticancer mechanism of flavonoids (quercetin, rutin, chrysin, apigenin, naringenin, silibinin, EGCG, genistein, biochanin A and C3G) through targeting molecules associated with high-grade adult-type diffuse glioma cell proliferation, apoptosis, oxidative stress, cell cycle arrest, migration, invasion, autophagy and DNA repair. In addition, the common molecules targeted by the flavonoids such as Bax, Bcl-2, MMP-2, MMP-9, caspase-8, caspase-3, p53, p38, Erk, JNK, p38, beclin-1 and LC3B were also discussed. Moreover, the clinical relevance of flavonoid molecular targets in high-grade adult-type diffuse gliomas is discussed with comparison to small molecules inhibitors: ralimetinib, AMG232, marimastat, hydroxychloroquine and chloroquine. Despite the positive pre-clinical results, further investigations in clinical studies are warranted to substantiate the efficacy and safety of the use of flavonoids on high-grade adult-type diffuse glioma patients.
Collapse
|
8
|
Crosstalk between xanthine oxidase (XO) inhibiting and cancer chemotherapeutic properties of comestible flavonoids- a comprehensive update. J Nutr Biochem 2022; 110:109147. [PMID: 36049673 DOI: 10.1016/j.jnutbio.2022.109147] [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: 06/03/2021] [Revised: 12/17/2021] [Accepted: 08/10/2022] [Indexed: 01/13/2023]
Abstract
Gout is an inflammatory disease caused by metabolic disorder or genetic inheritance. People throughout the world are strongly dependent on ethnomedicine for the treatment of gout and some receive satisfactory curative treatment. The natural remedies as well as established drugs derived from natural sources or synthetically made exert their action by mechanisms that are closely associated with anticancer treatment mechanisms regarding inhibition of xanthine oxidase, feedback inhibition of de novo purine synthesis, depolymerization and disappearance of microtubule, inhibition of NF-ĸB activation, induction of TRAIL, promotion of apoptosis, and caspase activation and proteasome inhibition. Some anti-gout and anticancer novel compounds interact with same receptors for their action, e.g., colchicine and colchicine analogues. Dietary flavonoids, i.e., chrysin, kaempferol, quercetin, fisetin, pelargonidin, apigenin, luteolin, myricetin, isorhamnetin, phloretinetc etc. have comparable IC50 values with established anti-gout drug and effective against both cancer and gout. Moreover, a noticeable number of newer anticancer compounds have already been isolated from plants that have been using by local traditional healers and herbal practitioners to treat gout. Therefore, the anti-gout plants might have greater potentiality to become selective candidates for screening of newer anticancer leads.
Collapse
|
9
|
Qi X, Jha SK, Jha NK, Dewanjee S, Dey A, Deka R, Pritam P, Ramgopal K, Liu W, Hou K. Antioxidants in brain tumors: current therapeutic significance and future prospects. Mol Cancer 2022; 21:204. [PMID: 36307808 PMCID: PMC9615186 DOI: 10.1186/s12943-022-01668-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/27/2022] [Indexed: 11/23/2022] Open
Abstract
Brain cancer is regarded among the deadliest forms of cancer worldwide. The distinct tumor microenvironment and inherent characteristics of brain tumor cells virtually render them resistant to the majority of conventional and advanced therapies. Oxidative stress (OS) is a key disruptor of normal brain homeostasis and is involved in carcinogenesis of different forms of brain cancers. Thus, antioxidants may inhibit tumorigenesis by preventing OS induced by various oncogenic factors. Antioxidants are hypothesized to inhibit cancer initiation by endorsing DNA repair and suppressing cancer progression by creating an energy crisis for preneoplastic cells, resulting in antiproliferative effects. These effects are referred to as chemopreventive effects mediated by an antioxidant mechanism. In addition, antioxidants minimize chemotherapy-induced nonspecific organ toxicity and prolong survival. Antioxidants also support the prooxidant chemistry that demonstrate chemotherapeutic potential, particularly at high or pharmacological doses and trigger OS by promoting free radical production, which is essential for activating cell death pathways. A growing body of evidence also revealed the roles of exogenous antioxidants as adjuvants and their ability to reverse chemoresistance. In this review, we explain the influences of different exogenous and endogenous antioxidants on brain cancers with reference to their chemopreventive and chemotherapeutic roles. The role of antioxidants on metabolic reprogramming and their influence on downstream signaling events induced by tumor suppressor gene mutations are critically discussed. Finally, the review hypothesized that both pro- and antioxidant roles are involved in the anticancer mechanisms of the antioxidant molecules by killing neoplastic cells and inhibiting tumor recurrence followed by conventional cancer treatments. The requirements of pro- and antioxidant effects of exogenous antioxidants in brain tumor treatment under different conditions are critically discussed along with the reasons behind the conflicting outcomes in different reports. Finally, we also mention the influencing factors that regulate the pharmacology of the exogenous antioxidants in brain cancer treatment. In conclusion, to achieve consistent clinical outcomes with antioxidant treatments in brain cancers, rigorous mechanistic studies are required with respect to the types, forms, and stages of brain tumors. The concomitant treatment regimens also need adequate consideration.
Collapse
Affiliation(s)
- Xuchen Qi
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing, 312000, Zhejiang, China.,Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310020, Zhejiang, China
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India. .,Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, 140413, India. .,Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, 248007, India.
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, West Bengal, 700032, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, West Bengal, 700032, India
| | - Rahul Deka
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Pingal Pritam
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Kritika Ramgopal
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Weiting Liu
- School of Nursing, Anhui University of Chinese Medicine, Hefei, 230001, Anhui, China.
| | - Kaijian Hou
- School of Nursing, Anhui University of Chinese Medicine, Hefei, 230001, Anhui, China. .,School of Public Health, Shantou University, Shantou, 515000, Guangdong, China.
| |
Collapse
|
10
|
Rasheed S, Rehman K, Shahid M, Suhail S, Akash MSH. Therapeutic potentials of genistein: New insights and perspectives. J Food Biochem 2022; 46:e14228. [PMID: 35579327 DOI: 10.1111/jfbc.14228] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 12/11/2022]
Abstract
Genistein, a polyphenolic isoflavone compound found abundantly in soy or soy-based products, is widely consumed in the Asian population. Genistein has poor bioavailability, to overcome this problem many advanced nano-drug delivery carrier systems are designed to enhance its water solubility and stability. However, further research is required to develop more efficient bioavailability improvement strategies. Genistein is a phytoestrogen which has been associated with reducing the risk of cancer, cardiovascular disorders, and diabetes mellitus. This plant-based bioactive compound possesses numerous biological activities such as anti-oxidant, anti-inflammatory, anti-obesity, anti-cancer, cardioprotective, and anti-diabetic activities to treat various disease states. Genistein has been used as an active therapeutic agent in many medications. Moreover, several clinical trials are in the ongoing stage to develop more efficient treatment therapies, especially for cancer treatment. This article highlights the protective and therapeutic benefits of genistein in the treatment of different ailments, and more specifically elaborates on the anti-cancer potential of genistein regarding various types of cancers. PRACTICAL APPLICATIONS: Genistein possesses versatile biological activities, including anti-diabetic, anti-inflammatory, anti-oxidant, anti-obesity, and anti-angiogenic. The most studied activity is anti-cancer. Currently, a number of pre-clinical and clinical trials are being carried out on anti-neoplastic and cytotoxic activities of genistein to develop novel therapeutic agents with excellent anti-cancer potential for the treatment of various kinds of cancer. Moreover, many bioavailability enhancement strategies have been developed to improve the bioavailability of genistein. Genistein shows significant hypoglycemic effects alone or in combination with other anti-diabetic agents. Genistein in combination with other chemotherapeutic agents is used for the treatment of prostate, bone, colorectal, glioma, breast, and bladder cancer.
Collapse
Affiliation(s)
- Sumbal Rasheed
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | - Kanwal Rehman
- Department of Pharmacy, The Women University, Multan, Pakistan
| | - Momina Shahid
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | - Shaleem Suhail
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | | |
Collapse
|
11
|
Mas-Bargues C, Borrás C, Viña J. The multimodal action of genistein in Alzheimer's and other age-related diseases. Free Radic Biol Med 2022; 183:127-137. [PMID: 35346775 DOI: 10.1016/j.freeradbiomed.2022.03.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/07/2023]
Abstract
Genistein is a phytoestrogen that, due to its structural similarity with estrogen, can both mimic and antagonize estrogen effects. Early analysis proved that at high concentrations, genistein inhibits breast cancer cell proliferation, thereby suggesting an anticancer activity. Since then, many discoveries have identified the genistein mechanism of action, including cell cycle arrest, apoptosis induction, as well as angiogenesis, and metastasis inhibition. In this review, we aim to discuss the multimodal action of genistein as an antioxidant, anti-inflammatory, anti-amyloid β, and autophagy promoter, which could be responsible for the genistein beneficial effect on Alzheimer's. Furthermore, we pinpoint the main signal transduction pathways that are known to be modulated by genistein. Genistein has thus several beneficial effects in several diseases, many of them associated with age, such as the above mentioned Alzheimer disease. Indeed, the beneficial effects of genistein for health promotion depend on each multimodality. In the context of geroscience, genistein has promising beneficial effects due to its multimodal action to treat age associated-diseases.
Collapse
Affiliation(s)
- Cristina Mas-Bargues
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, Valencia, 46010, Spain.
| | - Consuelo Borrás
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, Valencia, 46010, Spain.
| | - José Viña
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, Valencia, 46010, Spain
| |
Collapse
|
12
|
Harnessing oxidative stress for anti-glioma therapy. Neurochem Int 2022; 154:105281. [PMID: 35038460 DOI: 10.1016/j.neuint.2022.105281] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 12/22/2021] [Accepted: 01/10/2022] [Indexed: 02/06/2023]
Abstract
Glioma cells use intermediate levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) for growth and invasion, and suppressing these reactive molecules thus may compromise processes that are vital for glioma survival. Increased oxidative stress has been identified in glioma cells, in particular in glioma stem-like cells. Studies have shown that these cells harbor potent antioxidant defenses, although endogenous protection against nitrosative stress remains understudied. The enhancement of oxidative or nitrosative stress offers a potential target for triggering glioma cell death, but whether oxidative and nitrosative stresses can be combined for therapeutic effects requires further research. The optimal approach of harnessing oxidative stress for anti-glioma therapy should include the induction of free radical-induced oxidative damage and the suppression of antioxidant defense mechanisms selectively in glioma cells. However, selective induction of oxidative/nitrosative stress in glioma cells remains a therapeutic challenge, and research into selective drug delivery systems is ongoing. Because of multifactorial mechanisms of glioma growth, progression, and invasion, prospective oncological therapies may include not only therapeutic oxidative/nitrosative stress but also inhibition of oncogenic kinases, antioxidant molecules, and programmed cell death mediators.
Collapse
|
13
|
Abstract
The use of natural products has been increasing at a rapid pace, worldwide, with the aim to maintain a healthy lifestyle and to modify one's dietary habits. Ayurveda is a domain that has numerous wealth of information concerning medicinal plants and its part in controlling numerous ailments, such as neoplastic, cardiovascular, neurological plus immunological ailments. The use of such medicinal plants is important for preventing such diseases, especially "cancer" which is the succeeding foremost cause of mortality collectively. Even though abundant developments have been made in the management and control of cancer progression, substantial deficits and scope for advancement still continue to be unchanged. Several lethal adjacent consequences occur throughout the course of chemotherapy. Natural treatments, such as the use of plant-derived products in the treatment of cancer, might reduce the hostile side effects. Presently, a few plant-based products and its phytoconstituents are being utilized for the management of cancer. Here we have focused on numerous plant-derived phytochemicals and promising compounds from these plants to act as anticancer agents, along with their mechanisms of action.
Collapse
Affiliation(s)
- Nikhil Pandey
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Y B Tripathi
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
| |
Collapse
|
14
|
Genistein and Temozolomide-Loaded Polymeric Nanoparticles: A Synergistic Approach For Improved Anti-Tumor Efficacy Against Glioblastoma. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.07.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
15
|
Medeiros M, Candido MF, Valera ET, Brassesco MS. The multifaceted NF-kB: are there still prospects of its inhibition for clinical intervention in pediatric central nervous system tumors? Cell Mol Life Sci 2021; 78:6161-6200. [PMID: 34333711 PMCID: PMC11072991 DOI: 10.1007/s00018-021-03906-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/16/2022]
Abstract
Despite advances in the understanding of the molecular mechanisms underlying the basic biology and pathogenesis of pediatric central nervous system (CNS) malignancies, patients still have an extremely unfavorable prognosis. Over the years, a plethora of natural and synthetic compounds has emerged for the pharmacologic intervention of the NF-kB pathway, one of the most frequently dysregulated signaling cascades in human cancer with key roles in cell growth, survival, and therapy resistance. Here, we provide a review about the state-of-the-art concerning the dysregulation of this hub transcription factor in the most prevalent pediatric CNS tumors: glioma, medulloblastoma, and ependymoma. Moreover, we compile the available literature on the anti-proliferative effects of varied NF-kB inhibitors acting alone or in combination with other therapies in vitro, in vivo, and clinical trials. As the wealth of basic research data continues to accumulate, recognizing NF-kB as a therapeutic target may provide important insights to treat these diseases, hopefully contributing to increase cure rates and lower side effects related to therapy.
Collapse
Affiliation(s)
- Mariana Medeiros
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marina Ferreira Candido
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Elvis Terci Valera
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - María Sol Brassesco
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, FFCLRP-USP, University of São Paulo, Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirão Preto, São Paulo, CEP 14040-901, Brazil.
| |
Collapse
|
16
|
Sharifi-Rad J, Quispe C, Imran M, Rauf A, Nadeem M, Gondal TA, Ahmad B, Atif M, Mubarak MS, Sytar O, Zhilina OM, Garsiya ER, Smeriglio A, Trombetta D, Pons DG, Martorell M, Cardoso SM, Razis AFA, Sunusi U, Kamal RM, Rotariu LS, Butnariu M, Docea AO, Calina D. Genistein: An Integrative Overview of Its Mode of Action, Pharmacological Properties, and Health Benefits. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3268136. [PMID: 34336089 PMCID: PMC8315847 DOI: 10.1155/2021/3268136] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/11/2021] [Accepted: 06/28/2021] [Indexed: 12/15/2022]
Abstract
Genistein is an isoflavone first isolated from the brooming plant Dyer's Genista tinctoria L. and is widely distributed in the Fabaceae family. As an isoflavone, mammalian genistein exerts estrogen-like functions. Several biological effects of genistein have been reported in preclinical studies, such as the antioxidant, anti-inflammatory, antibacterial, and antiviral activities, the effects of angiogenesis and estrogen, and the pharmacological activities on diabetes and lipid metabolism. The purpose of this review is to provide up-to-date evidence of preclinical pharmacological activities with mechanisms of action, bioavailability, and clinical evidence of genistein. The literature was researched using the most important keyword "genistein" from the PubMed, Science, and Google Scholar databases, and the taxonomy was validated using The Plant List. Data were also collected from specialized books and other online resources. The main positive effects of genistein refer to the protection against cardiovascular diseases and to the decrease of the incidence of some types of cancer, especially breast cancer. Although the mechanism of protection against cancer involves several aspects of genistein metabolism, the researchers attribute this effect to the similarity between the structure of soy genistein and that of estrogen. This structural similarity allows genistein to displace estrogen from cellular receptors, thus blocking their hormonal activity. The pharmacological activities resulting from the experimental studies of this review support the traditional uses of genistein, but in the future, further investigations are needed on the efficacy, safety, and use of nanotechnologies to increase bioavailability and therapeutic efficacy.
Collapse
Affiliation(s)
- Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Cristina Quispe
- Facultad de Ciencias de la Salud, Universidad Arturo Prat, Avda. Arturo Prat 2120, Iquique 1110939, Chile
| | - Muhammad Imran
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, Lahore, Pakistan
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar-, 23561 Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Nadeem
- Department of Environmental Sciences, COMSATS Institute of Information Technology, Vehari-, Pakistan
| | | | - Bashir Ahmad
- Center of Biotechnology and Microbiology, University of Peshawar, Peshawar-, 25120 KPK, Pakistan
| | - Muhammad Atif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72341, Saudi Arabia
| | | | - Oksana Sytar
- Department of Plant Biology Department, Institute of Biology, Taras Shevchenko National University of Kyiv, Volodymyrska Str., 64, Kyiv 01033, Ukraine
- Department of Plant Physiology, Slovak University of Agriculture, A. Hlinku 2, 94976 Nitra, Slovakia
| | - Oxana Mihailovna Zhilina
- Department of Organic Chemistry, Pyatigorsk Medical-Pharmaceutical Institute (PMPI), Branch of Volgograd State Medical University, Ministry of Health of Russia, Pyatigorsk 357532, Russia
| | - Ekaterina Robertovna Garsiya
- Department of Pharmacognosy, Botany and Technology of Phytopreparations, Pyatigorsk Medical-Pharmaceutical Institute (PMPI), Branch of Volgograd State Medical University, Ministry of Health of Russia, Pyatigorsk 357532, Russia
| | - Antonella Smeriglio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Domenico Trombetta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Daniel Gabriel Pons
- Grupo Multidisciplinar de Oncología Traslacional (GMOT), Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears (UIB), Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma 07122, Spain
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, University of Concepción, Concepción 4070386, Chile
- Unidad de Desarrollo Tecnológico, Universidad de Concepción UDT, Concepción 4070386, Chile
| | - Susana M. Cardoso
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ahmad Faizal Abdull Razis
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Usman Sunusi
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Biochemistry, Bayero University Kano, PMB 3011 Kano, Nigeria
| | - Ramla Muhammad Kamal
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Pharmacology, Federal University Dutse, PMB 7156 Dutse Jigawa State, Nigeria
| | - Lia Sanda Rotariu
- Banat's University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timisoara, Romania
| | - Monica Butnariu
- Banat's University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timisoara, Romania
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
17
|
Current Perspective on the Natural Compounds and Drug Delivery Techniques in Glioblastoma Multiforme. Cancers (Basel) 2021; 13:cancers13112765. [PMID: 34199460 PMCID: PMC8199612 DOI: 10.3390/cancers13112765] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Glioblastoma multiforme (GBM) is one of the belligerent neoplasia that metastasize to other brain regions and invade nearby healthy tissues. However, the treatments available are associated with some limitations, such as high variations in solid tumors and deregulation of multiple cellular pathways. The heterogeneity of the GBM tumor and its aggressive infiltration into the nearby tissues makes it difficult to treat. Hence, the development of multimodality therapy that can be more effective, novel, with fewer side effects, improving the prognosis for GBM is highly desired. This review evaluated the use of natural phytoconstituents as an alternative for the development of a new therapeutic strategy. The key aspects of GBM and the potential of drug delivery techniques were also assessed, for tumor site delivery with limited side-effects. These efforts will help to provide better therapeutic options to combat GBM in future. Abstract Glioblastoma multiforme (GBM) is one of the debilitating brain tumors, being associated with extremely poor prognosis and short median patient survival. GBM is associated with complex pathogenesis with alterations in various cellular signaling events, that participate in cell proliferation and survival. The impairment in cellular redox pathways leads to tumorigenesis. The current standard pharmacological regimen available for glioblastomas, such as radiotherapy and surgical resection following treatment with chemotherapeutic drug temozolomide, remains fatal, due to drug resistance, metastasis and tumor recurrence. Thus, the demand for an effective therapeutic strategy for GBM remains elusive. Hopefully, novel products from natural compounds are suggested as possible solutions. They protect glial cells by reducing oxidative stress and neuroinflammation, inhibiting proliferation, inducing apoptosis, inhibiting pro-oncogene events and intensifying the potent anti-tumor therapies. Targeting aberrant cellular pathways in the amelioration of GBM could promote the development of new therapeutic options that improve patient quality of life and extend survival. Consequently, our review emphasizes several natural compounds in GBM treatment. We also assessed the potential of drug delivery techniques such as nanoparticles, Gliadel wafers and drug delivery using cellular carriers which could lead to a novel path for the obliteration of GBM.
Collapse
|
18
|
Atiq A, Parhar I. Anti-neoplastic Potential of Flavonoids and Polysaccharide Phytochemicals in Glioblastoma. Molecules 2020; 25:E4895. [PMID: 33113890 PMCID: PMC7660188 DOI: 10.3390/molecules25214895] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023] Open
Abstract
Clinically, gliomas are classified into four grades, with grade IV glioblastoma multiforme being the most malignant and deadly, which accounts for 50% of all gliomas. Characteristically, glioblastoma involves the aggressive proliferation of cells and invasion of normal brain tissue, outcomes as poor patient prognosis. With the current standard therapy of glioblastoma; surgical resection and radiotherapy followed by adjuvant chemotherapy with temozolomide, it remains fatal, because of the development of drug resistance, tumor recurrence, and metastasis. Therefore, the need for the effective therapeutic option for glioblastoma remains elusive. Previous studies have demonstrated the chemopreventive role of naturally occurring pharmacological agents through preventing or reversing the initiation phase of carcinogenesis or arresting the cancer progression phase. In this review, we discuss the role of natural phytochemicals in the amelioration of glioblastoma, with the aim to improve therapeutic outcomes, and minimize the adverse side effects to improve patient's prognosis and enhancing their quality of life.
Collapse
Affiliation(s)
- Ayesha Atiq
- Brain Research Institute Monash Sunway (BRIMS), Jeffery Cheah School of Medicine, Monash University Malaysia, 47500 Bandar Sunway, Selangor, Malaysia;
| | - Ishwar Parhar
- Brain Research Institute Monash Sunway (BRIMS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
19
|
Zhang Z, Zhou L, Xie N, Nice EC, Zhang T, Cui Y, Huang C. Overcoming cancer therapeutic bottleneck by drug repurposing. Signal Transduct Target Ther 2020; 5:113. [PMID: 32616710 PMCID: PMC7331117 DOI: 10.1038/s41392-020-00213-8] [Citation(s) in RCA: 309] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
Ever present hurdles for the discovery of new drugs for cancer therapy have necessitated the development of the alternative strategy of drug repurposing, the development of old drugs for new therapeutic purposes. This strategy with a cost-effective way offers a rare opportunity for the treatment of human neoplastic disease, facilitating rapid clinical translation. With an increased understanding of the hallmarks of cancer and the development of various data-driven approaches, drug repurposing further promotes the holistic productivity of drug discovery and reasonably focuses on target-defined antineoplastic compounds. The "treasure trove" of non-oncology drugs should not be ignored since they could target not only known but also hitherto unknown vulnerabilities of cancer. Indeed, different from targeted drugs, these old generic drugs, usually used in a multi-target strategy may bring benefit to patients. In this review, aiming to demonstrate the full potential of drug repurposing, we present various promising repurposed non-oncology drugs for clinical cancer management and classify these candidates into their proposed administration for either mono- or drug combination therapy. We also summarize approaches used for drug repurposing and discuss the main barriers to its uptake.
Collapse
Affiliation(s)
- Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Na Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Tao Zhang
- The School of Biological Science and Technology, Chengdu Medical College, 610083, Chengdu, China.
- Department of Oncology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, Sichuan, China.
| | - Yongping Cui
- Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, and Cancer Institute, Shenzhen Bay Laboratory Shenzhen, 518035, Shenzhen, China.
- Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China.
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
| |
Collapse
|
20
|
Tuli HS, Tuorkey MJ, Thakral F, Sak K, Kumar M, Sharma AK, Sharma U, Jain A, Aggarwal V, Bishayee A. Molecular Mechanisms of Action of Genistein in Cancer: Recent Advances. Front Pharmacol 2019; 10:1336. [PMID: 31866857 PMCID: PMC6910185 DOI: 10.3389/fphar.2019.01336] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/18/2019] [Indexed: 01/13/2023] Open
Abstract
Background: Genistein is one among the several other known isoflavones that is found in different soybeans and soy products. The chemical name of genistein is 4',5,7-trihydroxyisoflavone. Genistein has drawn attention of scientific community because of its potential beneficial effects on human grave diseases, such as cancer. Mechanistic insight of genistein reveals its potential for apoptotic induction, cell cycle arrest, as well as antiangiogenic, antimetastatic, and anti-inflammatory effects. Objective: The purpose of this review is to unravel and analyze various molecular mechanisms of genistein in diverse cancer models. Data sources: English language literature was searched using various databases, such as PubMed, ScienceDirect, EBOSCOhost, Scopus, Web of Science, and Cochrane Library. Key words used in various combinations included genistein, cancer, anticancer, molecular mechanisms prevention, treatment, in vivo, in vitro, and clinical studies. Study selection: Study selection was carried out strictly in accordance with the statement of Preferred Reporting Items for Systematic Reviews and Meta-analyses. Data extraction: Four authors independently carried out the extraction of articles. Data synthesis: One hundred one papers were found suitable for use in this review. Conclusion: This review covers various molecular interactions of genistein with various cellular targets in cancer models. It will help the scientific community understand genistein and cancer biology and will provoke them to design novel therapeutic strategies.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, India
| | - Muobarak Jaber Tuorkey
- Division of Physiology, Zoology Department, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Falak Thakral
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, India
| | | | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University, Sadopur, India
| | - Anil Kumar Sharma
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, India
| | - Uttam Sharma
- Department of Animal Sciences, Central University of Punjab, Bathinda, India
| | - Aklank Jain
- Department of Animal Sciences, Central University of Punjab, Bathinda, India
| | - Vaishali Aggarwal
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| |
Collapse
|
21
|
Lin M, Han P, Li Y, Wang W, Lai D, Zhou L. Quinoa Secondary Metabolites and Their Biological Activities or Functions. Molecules 2019; 24:E2512. [PMID: 31324047 PMCID: PMC6651730 DOI: 10.3390/molecules24132512] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/03/2019] [Accepted: 07/03/2019] [Indexed: 02/07/2023] Open
Abstract
Quinoa (Chenopodium quinoa Willd.) was known as the "golden grain" by the native Andean people in South America, and has been a source of valuable food over thousands of years. It can produce a variety of secondary metabolites with broad spectra of bioactivities. At least 193 secondary metabolites from quinoa have been identified in the past 40 years. They mainly include phenolic acids, flavonoids, terpenoids, steroids, and nitrogen-containing compounds. These metabolites exhibit many physiological functions, such as insecticidal, molluscicidal and antimicrobial activities, as well as various kinds of biological activities such as antioxidant, cytotoxic, anti-diabetic and anti-inflammatory properties. This review focuses on our knowledge of the structures, biological activities and functions of quinoa secondary metabolites. Biosynthesis, development and utilization of the secondary metabolites especially from quinoa bran were prospected.
Collapse
Affiliation(s)
- Minyi Lin
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China
| | - Peipei Han
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China
| | - Yuying Li
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China
| | - Weixuan Wang
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China
| | - Daowan Lai
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China
| | - Ligang Zhou
- Department of Plant Pathology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
22
|
Ferrari SM, Antonelli A, Guidi P, Bernardeschi M, Scarcelli V, Fallahi P, Frenzilli G. Genotoxicity Evaluation of the Soybean Isoflavone Genistein in Human Papillary Thyroid Cancer Cells. Study of Its Potential Use in Thyroid Cancer Therapy. Nutr Cancer 2019; 71:1335-1344. [PMID: 31017483 DOI: 10.1080/01635581.2019.1604004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 03/29/2019] [Accepted: 03/29/2019] [Indexed: 12/19/2022]
Abstract
Genistein is one of the several known isoflavonic phytoestrogens found in a number of plants, with soybeans and soy products being the primary food source. The aim of the study is to evaluate if genistein is able to exert antineoplastic action in primary human papillary thyroid cancer (PTC) cells. Thyroid tissues were treated with genistein (1-10-50-100 µM). Cell viability, proliferation, DNA primary damage and chromosomal damage were evaluated. An antiproliferative effect was induced by the highest doses of genistein, and such an effect was synergistically enhanced by the cotreatment with the antineoplastic drug sorafenib. Comet assay did not show any genotoxic effect in terms of primary DNA damage at all the times (4 and 24 h) and tested doses. A reduction of hydrogen peroxide-induced DNA primary damage in primary thyrocytes from PTC cells pretreated with genistein was observed. Data suggest that genistein exerts antineoplastic action, does not induce genotoxic effects while reduces oxidative-induced DNA damage in primary thyrocytes from PTC cells, supporting its possible use in therapeutic intervention.
Collapse
Affiliation(s)
- S M Ferrari
- Department of Clinical and Experimental Medicine - Section of Internal Medicine, University of Pisa , Pisa , Italy
| | - A Antonelli
- Department of Clinical and Experimental Medicine - Section of Internal Medicine, University of Pisa , Pisa , Italy
| | - P Guidi
- Department of Clinical and Experimental Medicine - Section of Applied Biology and Genetics, University of Pisa , Pisa , Italy
| | - M Bernardeschi
- Department of Clinical and Experimental Medicine - Section of Applied Biology and Genetics, University of Pisa , Pisa , Italy
| | - V Scarcelli
- Department of Clinical and Experimental Medicine - Section of Applied Biology and Genetics, University of Pisa , Pisa , Italy
| | - P Fallahi
- Department of Clinical and Experimental Medicine - Section of Internal Medicine, University of Pisa , Pisa , Italy
| | - G Frenzilli
- Department of Clinical and Experimental Medicine - Section of Applied Biology and Genetics, University of Pisa , Pisa , Italy
| |
Collapse
|
23
|
Inducers of Senescence, Toxic Compounds, and Senolytics: The Multiple Faces of Nrf2-Activating Phytochemicals in Cancer Adjuvant Therapy. Mediators Inflamm 2018; 2018:4159013. [PMID: 29618945 PMCID: PMC5829354 DOI: 10.1155/2018/4159013] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 12/19/2017] [Indexed: 12/18/2022] Open
Abstract
The reactivation of senescence in cancer and the subsequent clearance of senescent cells are suggested as therapeutic intervention in the eradication of cancer. Several natural compounds that activate Nrf2 (nuclear factor erythroid-derived 2-related factor 2) pathway, which is involved in complex cytoprotective responses, have been paradoxically shown to induce cell death or senescence in cancer. Promoting the cytoprotective Nrf2 pathway may be desirable for chemoprevention, but it might be detrimental in later stages and advanced cancers. However, senolytic activity shown by some Nrf2-activating compounds could be used to target senescent cancer cells (particularly in aged immune-depressed organisms) that escape immunosurveillance. We herein describe in vitro and in vivo effects of fifteen Nrf2-interacting natural compounds (tocotrienols, curcumin, epigallocatechin gallate, quercetin, genistein, resveratrol, silybin, phenethyl isothiocyanate, sulforaphane, triptolide, allicin, berberine, piperlongumine, fisetin, and phloretin) on cellular senescence and discuss their use in adjuvant cancer therapy. In light of available literature, it can be concluded that the meaning and the potential of adjuvant therapy with natural compounds in humans remain unclear, also taking into account the existence of few clinical trials mostly characterized by uncertain results. Further studies are needed to investigate the therapeutic potential of those compounds that display senolytic activity.
Collapse
|
24
|
Cytotoxic effect of Rosa canina extract on human colon cancer cells through repression of telomerase expression. J Pharm Anal 2017; 8:394-399. [PMID: 30595946 PMCID: PMC6308030 DOI: 10.1016/j.jpha.2017.12.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/14/2017] [Accepted: 12/12/2017] [Indexed: 11/29/2022] Open
Abstract
Rosa canina is a member of the genus Rosa that has long been used for medical objectives. Several studies have reported cytotoxic effects of different Rosa species, but there has been only limited investigation of the cytotoxic effect of R. canina. The purpose of the current study was to examine the potential effect of R. canina extract on cell viability, the cell cycle, apoptosis, and the expression of telomerase in human colon cancer (WiDr) cells. The cytotoxic effect of the extract was determined using MTT assay. The mechanism involved in the cytotoxic effect of the extract was then evaluated in terms of apoptosis and the cell cycle using flow cytometry. Mitochondrial membrane potential (MMP) was investigated using the fluorometric method, and expression levels of telomerase were studied using RT-PCR. R. canina extract exhibited a selective cytotoxic effect on WiDr cells compared with normal colon cells. The extract induced cell cycle arrest at the S phase and apoptosis via reduced MMP in WiDr cells. R. canina extract significantly repressed telomerase expressions at treatment times of 48 and 72 h in WiDr cells. Our results suggest that R. canina may have considerable potential for development as a novel natural product-based anticancer agent.
Collapse
|
25
|
Ardito F, Pellegrino MR, Perrone D, Troiano G, Cocco A, Lo Muzio L. In vitro study on anti-cancer properties of genistein in tongue cancer. Onco Targets Ther 2017; 10:5405-5415. [PMID: 29180873 PMCID: PMC5692202 DOI: 10.2147/ott.s133632] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose Tongue cancer is an extremely aggressive disease and is characterized by a poor prognosis. It is a complex disease to treat and current therapies have produced mediocre results with many side effects. Some facts suggest that natural essences can support traditional cancer therapy by carrying out a synergistic function with chemotherapy. Therefore, we evaluated the antitumor effects of genistein on tongue carcinoma cells. Methods Genistein 20, 50 and 100 µM were used for 24, 48 and 72 hours on 3 tongue carcinoma cell lines. xCELLigence system was used to evaluate the effects on cell adhesion, proliferation and to calculate IC50 values. Both MTT assay and Trypan blue assay were used to evaluate alterations in cell viability, scratch assay for cell migration and Western blot analysis for expression of some proteins. Results Cell adhesion was inhibited especially between 20 and 50 µM of genistein treatment. Proliferation was reduced by 50% for treatments with 20 µM at 24 hours, with 20 or 50 µM at 48 and 50 µM at 72 hours (P<0.0001). Viability tests confirmed a proportional reduction in concentration of genistein and duration of treatments. Even cell migration was reduced significantly (P<0.001). Genistein down-regulates vitronectin, OCT4 and survivin. Conclusion This in vitro study clarifies the anti-tumor effect of genistein on tongue carcinoma. In vivo studies are needed to confirm these data and develop a suitable delivery system that is capable of acting directly on tumor.
Collapse
Affiliation(s)
- Fatima Ardito
- Department of Clinical and Experimental Medicine, Foggia University, Foggia, Italy
| | - Mario R Pellegrino
- Department of Clinical and Experimental Medicine, Foggia University, Foggia, Italy
| | - Donatella Perrone
- Department of Clinical and Experimental Medicine, Foggia University, Foggia, Italy
| | - Giuseppe Troiano
- Department of Clinical and Experimental Medicine, Foggia University, Foggia, Italy
| | - Armando Cocco
- Department of Clinical and Experimental Medicine, Foggia University, Foggia, Italy
| | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, Foggia University, Foggia, Italy
| |
Collapse
|
26
|
Abstract
Bacteria and viruses possess circular DNA, whereas eukaryotes with typically very large DNA molecules have had to evolve into linear chromosomes to circumvent the problem of supercoiling circular DNA of that size. Consequently, such organisms possess telomeres to cap chromosome ends. Telomeres are essentially tandem repeats of any DNA sequence that are present at the ends of chromosomes. Their biology has been an enigmatic one, involving various molecules interacting dynamically in an evolutionarily well-trimmed fashion. Telomeres range from canonical hexameric repeats in most eukaryotes to unimaginably random retrotransposons, which attach to chromosome ends and reverse-transcribe to DNA in some plants and insects. Telomeres invariably associate with specialised protein complexes that envelop it, also regulating access of the ends to legitimate enzymes involved in telomere metabolism. They also transcribe into repetitive RNA which also seems to be playing significant roles in telomere maintenance. Telomeres thus form the intersection of DNA, protein, and RNA molecules acting in concert to maintain chromosome integrity. Telomere biology is emerging to appear ever more complex than previously envisaged, with the continual discovery of more molecules and interplays at the telomeres. This review also includes a section dedicated to the history of telomere biology, and intends to target the scientific audience new to the field by rendering an understanding of the phenomenon of chromosome end protection at large, with more emphasis on the biology of human telomeres. The review provides an update on the field and mentions the questions that need to be addressed.
Collapse
Affiliation(s)
- Shriram Venkatesan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore, Singapore.
| | - Aik Kia Khaw
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore, Singapore.
- Clinical Research Unit, Khoo Teck Puat Hospital, 768828 Singapore, Singapore.
| | - Manoor Prakash Hande
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore, Singapore.
- Tembusu College, National University of Singapore, 138598 Singapore, Singapore.
- VIT University, Vellore 632014, India.
- Mangalore University, Mangalore 574199, India.
| |
Collapse
|
27
|
Ilbeigi D, Nourbakhsh M, Khaghani S, Einollahi N, Kheiripour N, Gholinejad Z, Alaee M, Saberian M. Enterolactone Reduces Telomerase Activity and The Level of Its Catalytic Subunit in Breast Cancer Cells. CELL JOURNAL 2017; 19:37-43. [PMID: 28580306 PMCID: PMC5448327 DOI: 10.22074/cellj.2017.4705] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/22/2016] [Indexed: 11/04/2022]
Abstract
OBJECTIVE There is a positive correlation between higher serum phytoestrogen concentrations and lower risk of breast cancer. The activation of telomerase is crucial for the growth of cancer cells; therefore, the aim of this study was to examine the effects of enterolactone (ENL) and enterodiol (END) on this enzyme. MATERIALS AND METHODS In this experimental study, we performed the viability assay to determine the effects of different concentrations of ENL and END on cell viability, and the effective concentrations of these two compounds on cell growth. We used western blot analysis to evaluate human telomerase reverse transcriptase catalytic subunit (hTERT) expression and polymerase chain reaction (PCR)-ELISA based on the telomeric repeat amplification protocol (TRAP) assay for telomerase activity. RESULTS Both ENL and END, at 100 μM concentrations, significantly (P<0.05) reduced cell viability. However, only the 100 μM concentration of ENL significantly (P<0.05) decreased hTERT protein levels and telomerase activity. Lower concentrations of ENL did not have any significant effects on telomerase activity and hTERT protein levels. CONCLUSION High concentration of ENL decreased the viability of MCF-7 breast cancer cells and inhibited the expression and activity of telomerase in these cells. Although END could reduce breast cancer cell viability, it did not have any effect on telomerase expression and activity.
Collapse
Affiliation(s)
- Davod Ilbeigi
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences,
Tehran, Iran
| | - Mitra Nourbakhsh
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences,
Tehran, Iran
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular, Cellular Sciences Institute,
Tehran University of Medical Sciences, Tehran, Iran
| | - Shahnaz Khaghani
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences,
Tehran, Iran
| | - Nahid Einollahi
- Department of Clinical Laboratory Sciences, Faculty of Allied Medical Sciences, Tehran University of
Medical Sciences, Tehran, Iran
| | - Nejat Kheiripour
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences,
Tehran, Iran
| | - Zafar Gholinejad
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences,
Tehran, Iran
| | - Mohammad Alaee
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences,
Tehran, Iran
| | - Mostafa Saberian
- Department of Clinical Laboratory Sciences, Faculty of Allied Medical Sciences, Tehran University of
Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Yazdani Y, Sharifi Rad MR, Taghipour M, Chenari N, Ghaderi A, Razmkhah M. Genistein Suppression of Matrix Metalloproteinase 2 (MMP-2) and Vascular Endothelial Growth Factor (VEGF) Expression in Mesenchymal Stem Cell Like Cells Isolated from High and Low Grade Gliomas. Asian Pac J Cancer Prev 2016; 17:5303-5307. [PMID: 28125877 PMCID: PMC5454674 DOI: 10.22034/apjcp.2016.17.12.5303] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Objective: Brain tumors cause great mortality and morbidity worldwide, and success rates with surgical treatment remain very low. Several recent studies have focused on introduction of novel effective medical therapeutic approaches. Genistein is a member of the isoflavonoid family which has proved to exert anticancer effects. Here we assessed the effects of genistein on the expression of MMP-2 and VEGF in low and high grade gliomas in vitro. Materials and Methods: High and low grade glioma tumor tissue samples were obtained from a total of 16 patients, washed with PBS, cut into small pieces, digested with collagenase type I and cultured in DMEM containing 10% FBS. When cells reached passage 3, they were exposed to genistein and MMP-2 and VEGF gene transcripts were determined by quantitative real time PCR (qRT-PCR). Results: Expression of MMP-2 demonstrated 580-fold reduction in expression in low grade glioma cells post treatment with genistein compared to untreated cells (P value= 0.05). In cells derived from high grade lesions, expression of MMP-2 was 2-fold lower than in controls (P value> 0.05). Genistein caused a 4.7-fold reduction in VEGF transcript in high grade glioma cells (P value> 0.05) but no effects were evident in low grade glioma cells. Conclusion: Based on the data of the present study, low grade glioma cells appear much more sensitive to genistein and this isoflavone might offer an appropriate therapeutic intervention in these patients. Further investigation of this possibility is clearly warranted.
Collapse
Affiliation(s)
- Yasaman Yazdani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | | | | | | | | | | |
Collapse
|
29
|
Demir S, Turan I, Aliyazicioglu Y, Kilinc K, Yaman SO, Ayazoglu Demir E, Arslan A, Mentese A, Deger O. Morus RubraExtract Induces Cell Cycle Arrest and Apoptosis in Human Colon Cancer Cells Through Endoplasmic Reticulum Stress and Telomerase. Nutr Cancer 2016; 69:74-83. [DOI: 10.1080/01635581.2017.1247887] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
30
|
de Oliveira MR. Evidence for genistein as a mitochondriotropic molecule. Mitochondrion 2016; 29:35-44. [PMID: 27223841 DOI: 10.1016/j.mito.2016.05.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/13/2016] [Accepted: 05/16/2016] [Indexed: 12/19/2022]
Abstract
Genistein (4',5,7-trihydroxyisoflavone; C15H10O5), an isoflavone, has been investigated as an anti-cancer agent due to its ability to trigger cell death (both intrinsic and extrinsic apoptotic pathways) in different cancer cells in vitro and in vivo. Furthermore, genistein has been viewed as a mitochondriotropic molecule due to the direct effects this isoflavone induces in mitochondria, such as modulation of enzymatic activity of components of the oxidative phosphorylation system. Apoptosis triggering may also be mediated by genistein through activation of the mitochondria-dependent pathway by a mechanism associated with mitochondrial dysfunction (i.e., disruption of the mitochondrial membrane potential - MMP, release of cytochrome c, activation of the apoptosome, among others). Efforts have been made in order to elucidate how genistein coordinate these biochemical phenomena. Nonetheless, some areas of the mitochondria-associated research (mitochondrial biogenesis, redox biology of mitochondria, and mitochondria-associated bioenergetic parameters) need to be explored regarding the role of genistein as a mitochondria-targeted agent. This is a pharmacologically relevant issue due to the possibility of using genistein as a mitochondria-targeted drug in cases of cancer, neurodegeneration, cardiovascular, and endocrine disease, for example. The present review aims to describe, compare, and discuss relevant data about the effects of genistein upon mitochondria.
Collapse
Affiliation(s)
- Marcos Roberto de Oliveira
- Programa de Pós-Graduação em Química (PPGQ), Departamento de Química (DQ), Instituto de Ciências Exatas e da Terra (ICET), Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, CEP 78060-900 Cuiabá, MT, Brasil.
| |
Collapse
|
31
|
Russo M, Russo GL, Daglia M, Kasi PD, Ravi S, Nabavi SF, Nabavi SM. Understanding genistein in cancer: The "good" and the "bad" effects: A review. Food Chem 2016; 196:589-600. [PMID: 26593532 DOI: 10.1016/j.foodchem.2015.09.085] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 08/29/2015] [Accepted: 09/23/2015] [Indexed: 02/07/2023]
Abstract
Nowadays, diet and specific dietary supplements are seen as potential adjuvants to prevent different chronic diseases, including cancer, or to ameliorate pharmacological therapies. Soybean is one of the most important food components in Asian diet. A plethora of evidence supports the in vitro and in vivo anticancer effects of genistein, a soybean isoflavone. Major tumors affected by genistein here reviewed are breast, prostate, colon, liver, ovarian, bladder, gastric, brain cancers, neuroblastoma and chronic lymphocytic leukemia. However, it is not always clear if and when genistein is beneficial against tumors (the "good" effects), or the opposite, when the same molecule exerts adverse effects (the "bad" effects), favouring cancer cell proliferation. This review will critically evaluate this concept in the light of the different molecular mechanisms of genistein which occur when the molecule is administered at low doses (chemopreventive effects), or at high doses (pharmacological effects).
Collapse
Affiliation(s)
- Maria Russo
- Institute of Food Sciences, National Research Council, 83100 Avellino, Italy.
| | - Gian Luigi Russo
- Institute of Food Sciences, National Research Council, 83100 Avellino, Italy
| | - Maria Daglia
- Department of Drug Sciences, Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia, 27100 Pavia, Italy
| | - Pandima Devi Kasi
- Department of Biotechnology, Alagappa University, Karaikudi 630 004, Tamil Nadu, India.
| | - Sakthivel Ravi
- Department of Biotechnology, Alagappa University, Karaikudi 630 004, Tamil Nadu, India
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Ou Y, Xia R, Kong F, Zhang X, Yu S, Jiang L, Zheng L, Lin L. Overexpression of DEK is an indicator of poor prognosis in patients with gastric adenocarcinoma. Oncol Lett 2016; 11:1823-1828. [PMID: 26998084 DOI: 10.3892/ol.2016.4147] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 12/16/2015] [Indexed: 12/23/2022] Open
Abstract
Increased expression of the human DEK proto-oncogene (DEK) gene has been associated with numerous human malignancies. The DEK protein is associated with chromatin reconstruction and gene transcription, and is important in cell apoptosis. The present study aimed to elucidate the role of DEK with regard to gastric adenocarcinoma tumor progression and patient prognosis. DEK protein expression was analyzed using immunohistochemistry in 192 tumors paired with adjacent non-cancerous gastric mucosa that had been surgically resected from patients with primary gastric adenocarcinoma. The association between DEK expression and the clinicopathological characteristics of the patients was evaluated using the χ2 test and Fisher's exact test. The survival rates of the patients were calculated using the Kaplan-Meier method. Cox analysis evaluated the association between the expression of DEK and the survival rate of the patients. The DEK protein was expressed in 84 patients with gastric adenocarcinoma (43.8%) and in 20 of the paired normal gastric mucosa tissues (11.5%). The DEK expression rate was found to be associated with tumor size (P=0.006), tumor grade (P=0.023), lymph node metastasis (P=0.018), serous invasion (P=0.026), tumor stage (P=0.001) and Ki-67 expression (P=0.003). Furthermore, patients with gastric adenocarcinoma that expressed DEK had decreased disease-free (log-rank, 16.785; P<0.0001) and overall (log-rank, 15.759; P<0.0001) survival rates compared with patients without DEK expression. Patients with late-stage gastric adenocarcinoma that expressed DEK exhibited a lower overall survival rate compared with patients without DEK expression (P=0.002). Additional analysis revealed that DEK expression was an independent prognostic factor for the prognosis of gastric adenocarcinoma (hazard ratio, 0.556; 95% confidence interval, 0.337-0.918; P=0.022). From the results of the present study, it can be concluded that the detection of DEK protein expression in gastric adenocarcinoma tissues may be important for the diagnosis and prognosis of patients, and may be a targeted therapy for the treatment of gastric adenocarcinoma.
Collapse
Affiliation(s)
- Yingfu Ou
- Institute of Molecular Medicine, Medical College, Eastern Liaoning University, Dandong, Liaoning 118000, P.R. China
| | - Rongjun Xia
- Institute of Molecular Medicine, Medical College, Eastern Liaoning University, Dandong, Liaoning 118000, P.R. China
| | - Fanyong Kong
- Institute of Molecular Medicine, Medical College, Eastern Liaoning University, Dandong, Liaoning 118000, P.R. China
| | - Xiaokang Zhang
- Institute of Molecular Medicine, Medical College, Eastern Liaoning University, Dandong, Liaoning 118000, P.R. China; Department of Pathology, Dandong Central Hospital, Dandong, Liaoning 118000, P.R. China
| | - Shengjin Yu
- Institute of Molecular Medicine, Medical College, Eastern Liaoning University, Dandong, Liaoning 118000, P.R. China
| | - Lili Jiang
- Institute of Molecular Medicine, Medical College, Eastern Liaoning University, Dandong, Liaoning 118000, P.R. China
| | - Linlin Zheng
- Institute of Molecular Medicine, Medical College, Eastern Liaoning University, Dandong, Liaoning 118000, P.R. China
| | - Lijuan Lin
- Institute of Molecular Medicine, Medical College, Eastern Liaoning University, Dandong, Liaoning 118000, P.R. China
| |
Collapse
|
33
|
Amin ARMR, Karpowicz PA, Carey TE, Arbiser J, Nahta R, Chen ZG, Dong JT, Kucuk O, Khan GN, Huang GS, Mi S, Lee HY, Reichrath J, Honoki K, Georgakilas AG, Amedei A, Amin A, Helferich B, Boosani CS, Ciriolo MR, Chen S, Mohammed SI, Azmi AS, Keith WN, Bhakta D, Halicka D, Niccolai E, Fujii H, Aquilano K, Ashraf SS, Nowsheen S, Yang X, Bilsland A, Shin DM. Evasion of anti-growth signaling: A key step in tumorigenesis and potential target for treatment and prophylaxis by natural compounds. Semin Cancer Biol 2015; 35 Suppl:S55-S77. [PMID: 25749195 PMCID: PMC4561219 DOI: 10.1016/j.semcancer.2015.02.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 02/11/2015] [Accepted: 02/13/2015] [Indexed: 12/14/2022]
Abstract
The evasion of anti-growth signaling is an important characteristic of cancer cells. In order to continue to proliferate, cancer cells must somehow uncouple themselves from the many signals that exist to slow down cell growth. Here, we define the anti-growth signaling process, and review several important pathways involved in growth signaling: p53, phosphatase and tensin homolog (PTEN), retinoblastoma protein (Rb), Hippo, growth differentiation factor 15 (GDF15), AT-rich interactive domain 1A (ARID1A), Notch, insulin-like growth factor (IGF), and Krüppel-like factor 5 (KLF5) pathways. Aberrations in these processes in cancer cells involve mutations and thus the suppression of genes that prevent growth, as well as mutation and activation of genes involved in driving cell growth. Using these pathways as examples, we prioritize molecular targets that might be leveraged to promote anti-growth signaling in cancer cells. Interestingly, naturally occurring phytochemicals found in human diets (either singly or as mixtures) may promote anti-growth signaling, and do so without the potentially adverse effects associated with synthetic chemicals. We review examples of naturally occurring phytochemicals that may be applied to prevent cancer by antagonizing growth signaling, and propose one phytochemical for each pathway. These are: epigallocatechin-3-gallate (EGCG) for the Rb pathway, luteolin for p53, curcumin for PTEN, porphyrins for Hippo, genistein for GDF15, resveratrol for ARID1A, withaferin A for Notch and diguelin for the IGF1-receptor pathway. The coordination of anti-growth signaling and natural compound studies will provide insight into the future application of these compounds in the clinical setting.
Collapse
Affiliation(s)
| | - Phillip A Karpowicz
- Department of Biological Sciences, University of Windsor, 401 Sunset Ave., Room 327, Windsor, Ontario, N9B 3P4, Canada
| | | | - Jack Arbiser
- Winship Cancer Institute of Emory University, Atlanta, GA, USA; Atlanta Veterans Administration Health Center, Atlanta, GA, USA
| | - Rita Nahta
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Zhuo G Chen
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Jin-Tang Dong
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Omer Kucuk
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | | | | | - Shijun Mi
- Albert Einstein College of Medicine, New York, NY, USA
| | - Ho-Young Lee
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | | | | | | | | | - Amr Amin
- UAE University, Al Ain, United Arab Emirates; Faculty of Science, Cairo University, Cairo, Egypt
| | - Bill Helferich
- University of Illinois at Urbana Champaign, Urbana Champaign, IL, USA
| | | | | | - Sophie Chen
- Ovarian and Prostate Cancer Research Laboratory, Guildford, Surrey, United Kingdom
| | | | | | | | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | | | | | | | | | | | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Medical School, Mayo Graduate School, Mayo Clinic, Rochester, MN, USA
| | - Xujuan Yang
- University of Illinois at Urbana Champaign, Urbana Champaign, IL, USA
| | | | - Dong M Shin
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| |
Collapse
|
34
|
Casey SC, Amedei A, Aquilano K, Azmi AS, Benencia F, Bhakta D, Bilsland AE, Boosani CS, Chen S, Ciriolo MR, Crawford S, Fujii H, Georgakilas AG, Guha G, Halicka D, Helferich WG, Heneberg P, Honoki K, Keith WN, Kerkar SP, Mohammed SI, Niccolai E, Nowsheen S, Vasantha Rupasinghe HP, Samadi A, Singh N, Talib WH, Venkateswaran V, Whelan RL, Yang X, Felsher DW. Cancer prevention and therapy through the modulation of the tumor microenvironment. Semin Cancer Biol 2015; 35 Suppl:S199-S223. [PMID: 25865775 PMCID: PMC4930000 DOI: 10.1016/j.semcancer.2015.02.007] [Citation(s) in RCA: 265] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 02/06/2023]
Abstract
Cancer arises in the context of an in vivo tumor microenvironment. This microenvironment is both a cause and consequence of tumorigenesis. Tumor and host cells co-evolve dynamically through indirect and direct cellular interactions, eliciting multiscale effects on many biological programs, including cellular proliferation, growth, and metabolism, as well as angiogenesis and hypoxia and innate and adaptive immunity. Here we highlight specific biological processes that could be exploited as targets for the prevention and therapy of cancer. Specifically, we describe how inhibition of targets such as cholesterol synthesis and metabolites, reactive oxygen species and hypoxia, macrophage activation and conversion, indoleamine 2,3-dioxygenase regulation of dendritic cells, vascular endothelial growth factor regulation of angiogenesis, fibrosis inhibition, endoglin, and Janus kinase signaling emerge as examples of important potential nexuses in the regulation of tumorigenesis and the tumor microenvironment that can be targeted. We have also identified therapeutic agents as approaches, in particular natural products such as berberine, resveratrol, onionin A, epigallocatechin gallate, genistein, curcumin, naringenin, desoxyrhapontigenin, piperine, and zerumbone, that may warrant further investigation to target the tumor microenvironment for the treatment and/or prevention of cancer.
Collapse
Affiliation(s)
- Stephanie C Casey
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Asfar S Azmi
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Fabian Benencia
- Department of Biomedical Sciences, Ohio University, Athens, OH, United States
| | - Dipita Bhakta
- School of Chemical and Biotechnology, SASTRA University, Thanjavur 613401, Tamil Nadu, India
| | - Alan E Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Chandra S Boosani
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Laboratory, Guildford, Surrey, United Kingdom
| | | | - Sarah Crawford
- Department of Biology, Southern Connecticut State University, New Haven, CT, United States
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Gunjan Guha
- School of Chemical and Biotechnology, SASTRA University, Thanjavur 613401, Tamil Nadu, India
| | | | - William G Helferich
- University of Illinois at Urbana-Champaign, Champaign-Urbana, IL, United States
| | - Petr Heneberg
- Charles University in Prague, Third Faculty of Medicine, Prague, Czech Republic
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sid P Kerkar
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | | | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Graduate School, Mayo Medical School, Mayo Clinic, Rochester, MN, United States
| | - H P Vasantha Rupasinghe
- Department of Environmental Sciences, Faculty of Agriculture, Dalhousie University, Nova Scotia, Canada
| | | | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Wamidh H Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science University, Amman, Jordan
| | | | - Richard L Whelan
- Mount Sinai Roosevelt Hospital, Icahn Mount Sinai School of Medicine, New York City, NY, United States
| | - Xujuan Yang
- University of Illinois at Urbana-Champaign, Champaign-Urbana, IL, United States
| | - Dean W Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
35
|
Samadi AK, Bilsland A, Georgakilas AG, Amedei A, Amin A, Bishayee A, Azmi AS, Lokeshwar BL, Grue B, Panis C, Boosani CS, Poudyal D, Stafforini DM, Bhakta D, Niccolai E, Guha G, Vasantha Rupasinghe HP, Fujii H, Honoki K, Mehta K, Aquilano K, Lowe L, Hofseth LJ, Ricciardiello L, Ciriolo MR, Singh N, Whelan RL, Chaturvedi R, Ashraf SS, Shantha Kumara HMC, Nowsheen S, Mohammed SI, Keith WN, Helferich WG, Yang X. A multi-targeted approach to suppress tumor-promoting inflammation. Semin Cancer Biol 2015; 35 Suppl:S151-S184. [PMID: 25951989 PMCID: PMC4635070 DOI: 10.1016/j.semcancer.2015.03.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 03/13/2015] [Accepted: 03/16/2015] [Indexed: 12/15/2022]
Abstract
Cancers harbor significant genetic heterogeneity and patterns of relapse following many therapies are due to evolved resistance to treatment. While efforts have been made to combine targeted therapies, significant levels of toxicity have stymied efforts to effectively treat cancer with multi-drug combinations using currently approved therapeutics. We discuss the relationship between tumor-promoting inflammation and cancer as part of a larger effort to develop a broad-spectrum therapeutic approach aimed at a wide range of targets to address this heterogeneity. Specifically, macrophage migration inhibitory factor, cyclooxygenase-2, transcription factor nuclear factor-κB, tumor necrosis factor alpha, inducible nitric oxide synthase, protein kinase B, and CXC chemokines are reviewed as important antiinflammatory targets while curcumin, resveratrol, epigallocatechin gallate, genistein, lycopene, and anthocyanins are reviewed as low-cost, low toxicity means by which these targets might all be reached simultaneously. Future translational work will need to assess the resulting synergies of rationally designed antiinflammatory mixtures (employing low-toxicity constituents), and then combine this with similar approaches targeting the most important pathways across the range of cancer hallmark phenotypes.
Collapse
Affiliation(s)
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates; Faculty of Science, Cairo University, Cairo, Egypt
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, Miami, FL, United States
| | - Asfar S Azmi
- Department of Pathology, Wayne State Univeristy, Karmanos Cancer Center, Detroit, MI, USA
| | - Bal L Lokeshwar
- Department of Urology, University of Miami, Miller School of Medicine, Miami, FL, United States; Miami Veterans Administration Medical Center, Miami, FL, United States
| | - Brendan Grue
- Department of Environmental Science, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Carolina Panis
- Laboratory of Inflammatory Mediators, State University of West Paraná, UNIOESTE, Paraná, Brazil
| | - Chandra S Boosani
- Department of BioMedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Deepak Poudyal
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Diana M Stafforini
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Dipita Bhakta
- School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | | | - Gunjan Guha
- School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | - H P Vasantha Rupasinghe
- Department of Environmental Sciences, Faculty of Agriculture and Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Kapil Mehta
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada.
| | - Lorne J Hofseth
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Richard L Whelan
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - S Salman Ashraf
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - H M C Shantha Kumara
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Graduate School, Mayo Medical School, Mayo Clinic, Rochester, MN, United States
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | | | - Xujuan Yang
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| |
Collapse
|
36
|
Feitelson MA, Arzumanyan A, Kulathinal RJ, Blain SW, Holcombe RF, Mahajna J, Marino M, Martinez-Chantar ML, Nawroth R, Sanchez-Garcia I, Sharma D, Saxena NK, Singh N, Vlachostergios PJ, Guo S, Honoki K, Fujii H, Georgakilas AG, Bilsland A, Amedei A, Niccolai E, Amin A, Ashraf SS, Boosani CS, Guha G, Ciriolo MR, Aquilano K, Chen S, Mohammed SI, Azmi AS, Bhakta D, Halicka D, Keith WN, Nowsheen S. Sustained proliferation in cancer: Mechanisms and novel therapeutic targets. Semin Cancer Biol 2015; 35 Suppl:S25-S54. [PMID: 25892662 PMCID: PMC4898971 DOI: 10.1016/j.semcancer.2015.02.006] [Citation(s) in RCA: 480] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 02/20/2015] [Accepted: 02/23/2015] [Indexed: 02/08/2023]
Abstract
Proliferation is an important part of cancer development and progression. This is manifest by altered expression and/or activity of cell cycle related proteins. Constitutive activation of many signal transduction pathways also stimulates cell growth. Early steps in tumor development are associated with a fibrogenic response and the development of a hypoxic environment which favors the survival and proliferation of cancer stem cells. Part of the survival strategy of cancer stem cells may manifested by alterations in cell metabolism. Once tumors appear, growth and metastasis may be supported by overproduction of appropriate hormones (in hormonally dependent cancers), by promoting angiogenesis, by undergoing epithelial to mesenchymal transition, by triggering autophagy, and by taking cues from surrounding stromal cells. A number of natural compounds (e.g., curcumin, resveratrol, indole-3-carbinol, brassinin, sulforaphane, epigallocatechin-3-gallate, genistein, ellagitannins, lycopene and quercetin) have been found to inhibit one or more pathways that contribute to proliferation (e.g., hypoxia inducible factor 1, nuclear factor kappa B, phosphoinositide 3 kinase/Akt, insulin-like growth factor receptor 1, Wnt, cell cycle associated proteins, as well as androgen and estrogen receptor signaling). These data, in combination with bioinformatics analyses, will be very important for identifying signaling pathways and molecular targets that may provide early diagnostic markers and/or critical targets for the development of new drugs or drug combinations that block tumor formation and progression.
Collapse
Affiliation(s)
- Mark A Feitelson
- Department of Biology, Temple University, Philadelphia, PA, United States.
| | - Alla Arzumanyan
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Rob J Kulathinal
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Stacy W Blain
- Department of Pediatrics, State University of New York, Downstate Medical Center, Brooklyn, NY, United States
| | - Randall F Holcombe
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, United States
| | - Jamal Mahajna
- MIGAL-Galilee Technology Center, Cancer Drug Discovery Program, Kiryat Shmona, Israel
| | - Maria Marino
- Department of Science, University Roma Tre, V.le G. Marconi, 446, 00146 Rome, Italy
| | - Maria L Martinez-Chantar
- Metabolomic Unit, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Technology Park of Bizkaia, Bizkaia, Spain
| | - Roman Nawroth
- Department of Urology, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Isidro Sanchez-Garcia
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Salamanca, Spain
| | - Dipali Sharma
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Neeraj K Saxena
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Neetu Singh
- Tissue and Cell Culture Unit, CSIR-Central Drug Research Institute, Council of Scientific & Industrial Research, Lucknow, India
| | | | - Shanchun Guo
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara 634-8521, Japan
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara 634-8521, Japan
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Zografou 15780, Athens, Greece
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, UK
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, UAE University, Al-Ain, United Arab Emirates
| | - S Salman Ashraf
- Department of Chemistry, College of Science, UAE University, Al-Ain, United Arab Emirates
| | - Chandra S Boosani
- Department of BioMedical Sciences, Creighton University, Omaha, NE, United States
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Maria Rosa Ciriolo
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Sophie Chen
- Department of Research and Development, Ovarian and Prostate Cancer Research Trust Laboratory, Guildford, Surrey GU2 7YG, United Kingdom
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - Asfar S Azmi
- Department of Pathology, Karmonas Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Dorota Halicka
- Brander Cancer Research Institute, Department of Pathology, New York Medical College, Valhalla, NY, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, UK
| | - Somaira Nowsheen
- Mayo Graduate School, Mayo Medical School, Mayo Clinic Medical Scientist Training Program, Rochester, MN, United States
| |
Collapse
|
37
|
Abstract
Epigenetic mechanisms play a pivotal role in the expression of genes and can be influenced by both the quality and quantity of diet. Dietary compounds such as sulforaphane (SFN) found in cruciferous vegetables and epigallocatechin-3-gallate (EGCG) in green tea exhibit the ability to affect various epigenetic mechanisms such as DNA methyltransferase (DNMT) inhibition, histone modifications via histone deacetylase (HDAC), histone acetyltransferase (HAT) inhibition, or noncoding RNA expression. Regulation of these epigenetic mechanisms has been shown to have notable influences on the formation and progression of various neoplasms. We have shown that an epigenetic diet can influence both cellular longevity and carcinogenesis through the modulation of certain key genes that encode telomerase and p16. Caloric restriction (CR) can also play a crucial role in aging and cancer. Reductions in caloric intake have been shown to increase both the life- and health-span in a variety of animal models. Moreover, restriction of glucose has been demonstrated to decrease the incidence of age-related diseases such as cancer and diabetes. A diet rich in compounds such as genistein, SFN and EGCG can positively modulate the epigenome and lead to many health benefits. Also, reducing the quantity of calories and glucose in the diet can confer an increased health-span, including reduced cancer incidence.
Collapse
Affiliation(s)
- Michael Daniel
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA
| | - Trygve O Tollefsbol
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, 1530 3rd Avenue South, Birmingham, AL 35294, USA Comprehensive Cancer Center, University of Alabama at Birmingham, 1802 6th Avenue South, Birmingham, AL 35294, USA Nutrition Obesity Research Center, University of Alabama at Birmingham, 1675 University Boulevard, Birmingham, AL 35294, USA Comprehensive Diabetes Center, University of Alabama at Birmingham, 1825 University Boulevard, Birmingham, AL 35294, USA
| |
Collapse
|
38
|
Khaw AK, Sameni S, Venkatesan S, Kalthur G, Hande MP. Plumbagin alters telomere dynamics, induces DNA damage and cell death in human brain tumour cells. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2015; 793:86-95. [PMID: 26520377 DOI: 10.1016/j.mrgentox.2015.06.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 06/08/2015] [Indexed: 02/08/2023]
Abstract
Natural plant products may possess much potential in palliative therapy and supportive strategies of current cancer treatments with lesser cytotoxicity to normal cells compared to conventional chemotherapy. In the current study, anti-cancer properties of plumbagin, a plant-derived naphthoquinone, on brain cancer cells were determined. Plumbagin treatment resulted in the induction of DNA damage, cell cycle arrest and apoptosis, followed by suppression of the colony forming ability of the brain tumour cells. These effects were substantiated by upregulation of PTEN, TNFRSF1A and downregulation of E2F1 genes, along with a drop in MDM2, cyclin B1, survivin and BCL2 protein expression. Plumbagin induced elevated levels of caspase-3/7 activity as well. For the first time, we show here that plumbagin inhibits telomerase in brain tumour cells and results in telomere shortening following chronic long-term treatment. This observation implies considerable cytotoxicity of plumbagin towards cancer cells with higher telomerase activity. Collectively, our findings suggest plumbagin as a potential chemotherapeutic phytochemical in brain tumour treatment modalities.
Collapse
Affiliation(s)
- Aik Kia Khaw
- Genome Stability Laboratory, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Safoura Sameni
- Genome Stability Laboratory, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Shriram Venkatesan
- Genome Stability Laboratory, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Guruprasad Kalthur
- Genome Stability Laboratory, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Division of Clinical Embryology, Department of Obstetrics and Gynaecology, Kasturba Medical College, Manipal University, Manipal, 576104, India
| | - M Prakash Hande
- Genome Stability Laboratory, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Tembusu College, National University of Singapore, Singapore 138597, Singapore.
| |
Collapse
|
39
|
Wang Y, Sun C, Mao A, Zhang X, Zhou X, Wang Z, Zhang H. Radiosensitization to X-ray radiation by telomerase inhibitor MST-312 in human hepatoma HepG2 cells. Life Sci 2015; 123:43-50. [PMID: 25596016 DOI: 10.1016/j.lfs.2014.12.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 12/03/2014] [Accepted: 12/16/2014] [Indexed: 12/15/2022]
Abstract
AIMS Previous studies in malignant cells have shown that irradiation-induced upregulation of telomerase activity, not only protected damaged telomeres, but also contributed to DNA damage repair by chromosomal healing and increased resistance to irradiation. The purpose of the present study was to investigate the radiosensitizing effect of telomerase inhibitor MST-312 and the corresponding mechanism in the human hepatoma cell line HepG2. MAIN METHODS Cell proliferation, telomerase activity, cell cycle distribution, DNA damage and repair, expression of p53, mitochondrial membrane potential, and cell apoptosis were measured with the MTT assay, real-time fluorescent quantitative PCR, flow cytometry, immunofluorescence, western blots, JC-1 staining, and Hoechst 33258 staining, respectively. KEY FINDINGS MST-312 effectively inhibited telomerase activity and showed relative weak toxicity to HepG2 cells at 4 μM. Compared with irradiation alone, 4 μM MST-312 pretreatment, followed by X-ray treatment, significantly reduced clonogenic potential. Aggravated DNA damage and increased sub-G1 cell fractions were observed. Further investigation found that homologous recombination (HR) repair protein Rad51 foci nuclear formation was blocked, and expression of p53 was elevated. These led to the collapse of mitochondrial membrane potential, and enhanced the apoptotic rate. SIGNIFICANCE These data demonstrated that disturbances of telomerase function could enhance the radiosensitivity of HepG2 cells to X-ray irradiation by impairing HR repair processes. In addition, telomerase inhibitor MST-312 may be useful as an adjuvant treatment in combination with irradiation.
Collapse
Affiliation(s)
- Yali Wang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, People'sRepublic of China; Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences, Lanzhou 730000, People's Republic of China; Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, People's Republic of China; School of Pharmacy, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Chao Sun
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, People'sRepublic of China; Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences, Lanzhou 730000, People's Republic of China; Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, People's Republic of China
| | - Aihong Mao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, People'sRepublic of China; Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences, Lanzhou 730000, People's Republic of China; Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, People's Republic of China
| | - Xin Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, People'sRepublic of China; Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences, Lanzhou 730000, People's Republic of China; Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, People's Republic of China
| | - Xin Zhou
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, People'sRepublic of China; Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences, Lanzhou 730000, People's Republic of China; Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, People's Republic of China
| | - Zhenhua Wang
- College of Life Sciences, Yantai University, Yantai 264005, People's Republic of China
| | - Hong Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, People'sRepublic of China; Key Laboratory of Heavy Ion Radiation Medicine of Chinese Academy of Sciences, Lanzhou 730000, People's Republic of China; Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, People's Republic of China.
| |
Collapse
|
40
|
Gurung RL, Lim HK, Venkatesan S, Lee PSW, Hande MP. Targeting DNA-PKcs and telomerase in brain tumour cells. Mol Cancer 2014; 13:232. [PMID: 25307264 PMCID: PMC4213508 DOI: 10.1186/1476-4598-13-232] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 10/07/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Patients suffering from brain tumours such as glioblastoma and medulloblastoma have poor prognosis with a median survival of less than a year. Identifying alternative molecular targets would enable us to develop different therapeutic strategies for better management of these tumours. METHODS Glioblastoma (MO59K and KNS60) and medulloblastoma cells (ONS76) were used in this study. Telomerase inhibitory effects of MST-312, a chemically modified-derivative of epigallocatechin gallate, in the cells were assessed using telomere repeat amplification protocol. Gene expression analysis following MST-312 treatment was done by microarray. Telomere length was measured by telomere restriction fragments analysis. Effects of MST-312 on DNA integrity were evaluated by single cell gel electrophoresis, immunofluorescence assay and cytogenetic analysis. Phosphorylation status of DNA-PKcs was measured with immunoblotting and effects on cell proliferation were monitored with cell titre glow and trypan blue exclusion following dual inhibition. RESULTS MST-312 showed strong binding affinity to DNA and displayed reversible telomerase inhibitory effects in brain tumour cells. In addition to the disruption of telomere length maintenance, MST-312 treatment decreased brain tumour cell viability, induced cell cycle arrest and double strand breaks (DSBs). DNA-PKcs activation was observed in telomerase-inhibited cells presumably as a response to DNA damage. Impaired DNA-PKcs in MO59J cells or in MO59K cells treated with DNA-PKcs inhibitor, NU7026, caused a delay in the repair of DSBs. In contrast, MST-312 did not induce DSBs in telomerase negative osteosarcoma cells (U2OS). Combined inhibition of DNA-PKcs and telomerase resulted in an increase in telomere signal-free chromosomal ends in brain tumour cells as well. Interestingly, continual exposure of brain tumour cells to telomerase inhibitor led to population of cells, which displayed resistance to telomerase inhibition-mediated cell arrest. DNA-PKcs ablation in these cells, however, confers higher cell sensitivity to telomerase inhibition, inducing cell death. CONCLUSIONS Efficient telomerase inhibition was achieved with acute exposure to MST-312 and this resulted in subtle but significant increase in DSBs. Activation of DNA-PKcs might indicate the requirement of NHEJ pathway in the repair telomerase inhibitor induced DNA damage. Therefore, our results suggest a potential strategy in combating brain tumour cells with dual inhibition of telomerase and NHEJ pathway.
Collapse
Affiliation(s)
| | | | | | | | - M Prakash Hande
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, Singapore 117597, Singapore.
| |
Collapse
|