1
|
Sacli-Bilmez B, Bas A, Erşen Danyeli A, Yakicier MC, Pamir MN, Özduman K, Dinçer A, Ozturk-Isik E. Detecting IDH and TERTp mutations in diffuse gliomas using 1H-MRS with attention deep-shallow networks. Comput Biol Med 2025; 186:109736. [PMID: 39874812 DOI: 10.1016/j.compbiomed.2025.109736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Preoperative and noninvasive detection of isocitrate dehydrogenase (IDH) and telomerase reverse transcriptase gene promoter (TERTp) mutations in glioma is critical for prognosis and treatment planning. This study aims to develop deep learning classifiers to identify IDH and TERTp mutations using proton magnetic resonance spectroscopy (1H-MRS) and a one-dimensional convolutional neural network (1D-CNN) architecture. METHODS This study included 1H-MRS data from 225 adult patients with hemispheric diffuse glioma (117 IDH mutants and 108 IDH wild-type; 99 TERTp mutants and 100 TERTp wild-type). The spectra were processed using the LCModel, and multiple deep learning models, including a baseline, a deep-shallow network, and an attention deep-shallow network (ADSN), were trained to classify mutational subgroups of gliomas. The Gradient-weighted Class Activation Mapping (Grad-CAM) technique was used to interpret the models' decision-making process. RESULTS The ADSN model was the most effective for IDH mutation detection, achieving F1-scores of 93 % on the validation set and 88 % on the test set. For TERTp mutation detection, the ADSN model achieved F1-scores of 80 % in the validation set and 81 % in the test set, whereas TERTp-only gliomas were detected with F1-scores of 88 % in the validation set and 86 % in the test set using the same architecture. CONCLUSION Deep learning models accurately predicted the IDH and TERTp mutational subgroups of hemispheric diffuse gliomas by extracting relevant information from 1H-MRS spectra without the need for manual feature extraction.
Collapse
Affiliation(s)
- Banu Sacli-Bilmez
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey.
| | - Abdullah Bas
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey
| | - Ayça Erşen Danyeli
- Department of Pathology, Acibadem University, School of Medicine, Istanbul, Turkey; Center for Neuroradiological Applications and Research, Acibadem University, Istanbul, Turkey
| | | | - M Necmettin Pamir
- Center for Neuroradiological Applications and Research, Acibadem University, Istanbul, Turkey; Department of Neurosurgery, Acibadem University, School of Medicine, Istanbul, Turkey
| | - Koray Özduman
- Center for Neuroradiological Applications and Research, Acibadem University, Istanbul, Turkey; Department of Neurosurgery, Acibadem University, School of Medicine, Istanbul, Turkey
| | - Alp Dinçer
- Center for Neuroradiological Applications and Research, Acibadem University, Istanbul, Turkey; Department of Radiology, Acibadem University, School of Medicine, Istanbul, Turkey
| | - Esin Ozturk-Isik
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey; Center for Neuroradiological Applications and Research, Acibadem University, Istanbul, Turkey
| |
Collapse
|
2
|
Jiang R, Li L, Li M. Biomimetic Construction of Degradable DNAzyme-Loaded Nanocapsules for Self-Sufficient Gene Therapy of Pulmonary Metastatic Breast Cancer. ACS NANO 2023; 17:22129-22144. [PMID: 37925681 DOI: 10.1021/acsnano.3c09581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Pulmonary metastasis of breast cancer is the major cause of deaths of breast cancer patients, but the effective treatment of pulmonary metastases is still lacking at present. Herein, a degradable biomimetic DNAzyme biocapsule is developed with the poly(ethylenimine) (PEI)-DNAzyme complex encapsulated in a Mn2+/Zn2+-coordinated inositol hexaphosphate (IP6) capsule modified with the cRGD targeting peptide for high-efficiency gene therapy of both primary and pulmonary metastatic breast tumors. This DNAzyme biocapsule is degradable inside acidic lysosomes, leading to the release of DNAzyme and abundant Mn2+/Zn2+ for catalytic cleavage of EGR-1 mRNA. We find that PEI promotes the lysosomal escape of the released DNAzyme. Both in vitro and in vivo experiments demonstrate the apparent downregulation of EGR-1 and Bcl-2 protein expression after treatment with the DNAzyme biocapsule, thereby inducing apoptotic death of tumor cells. We further verify that the DNAzyme biocapsule exhibits potent therapeutic efficacy against both primary and pulmonary metastatic breast tumors with significant inhibition of peri-pulmonary metastasis. This study provides a promising effective strategy for constructing degradable DNAzyme-based platforms with self-supply of abundant metal ion cofactors for high-efficiency gene therapy of metastatic breast cancer.
Collapse
Affiliation(s)
- Renting Jiang
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Linhu Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Ming Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan 410083, China
| |
Collapse
|
3
|
Dilworth L, Stennett D, Omoruyi F. Cellular and Molecular Activities of IP6 in Disease Prevention and Therapy. Biomolecules 2023; 13:972. [PMID: 37371552 DOI: 10.3390/biom13060972] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
IP6 (phytic acid) is a naturally occurring compound in plant seeds and grains. It is a poly-phosphorylated inositol derivative that has been shown to exhibit many biological activities that accrue benefits in health and diseases (cancer, diabetes, renal lithiasis, cardiovascular diseases, etc.). IP6 has been shown to have several cellular and molecular activities associated with its potential role in disease prevention. These activities include anti-oxidant properties, chelation of metal ions, inhibition of inflammation, modulation of cell signaling pathways, and modulation of the activities of enzymes and hormones that are involved in carbohydrate and lipid metabolism. Studies have shown that IP6 has anti-oxidant properties and can scavenge free radicals known to cause cellular damage and contribute to the development of chronic diseases such as cancers and cardiovascular diseases, as well as diabetes mellitus. It has also been shown to possess anti-inflammatory properties that may modulate immune responses geared towards the prevention of inflammatory conditions. Moreover, IP6 exhibits anti-cancer properties through the induction of cell cycle arrest, promoting apoptosis and inhibiting cancer cell growth. Additionally, it has been shown to have anti-mutagenic properties, which reduce the risk of malignancies by preventing DNA damage and mutations. IP6 has also been reported to have a potential role in bone health. It inhibits bone resorption and promotes bone formation, which may help in the prevention of bone diseases such as osteoporosis. Overall, IP6's cellular and molecular activities make it a promising candidate for disease prevention. As reported in many studies, its anti-inflammatory, anti-oxidant, and anti-cancer properties support its inclusion as a dietary supplement that may protect against the development of chronic diseases. However, further studies are needed to understand the mechanisms of action of this dynamic molecule and its derivatives and determine the optimal doses and appropriate delivery methods for effective therapeutic use.
Collapse
Affiliation(s)
- Lowell Dilworth
- Department of Pathology, The University of the West Indies, Mona Campus, Kingston 7, Jamaica
| | - Dewayne Stennett
- The Transitional Year Programme, University of Toronto, Toronto, ON M5S 2E8, Canada
| | - Felix Omoruyi
- Department of Life Sciences, Texas A&M University, Corpus Christi, TX 78412, USA
- Department of Health Sciences, Texas A&M University, Corpus Christi, TX 78412, USA
| |
Collapse
|
4
|
Badodi S, Pomella N, Zhang X, Rosser G, Whittingham J, Niklison-Chirou MV, Lim YM, Brandner S, Morrison G, Pollard SM, Bennett CD, Clifford SC, Peet A, Basson MA, Marino S. Inositol treatment inhibits medulloblastoma through suppression of epigenetic-driven metabolic adaptation. Nat Commun 2021; 12:2148. [PMID: 33846320 PMCID: PMC8042111 DOI: 10.1038/s41467-021-22379-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 03/12/2021] [Indexed: 12/11/2022] Open
Abstract
Deregulation of chromatin modifiers plays an essential role in the pathogenesis of medulloblastoma, the most common paediatric malignant brain tumour. Here, we identify a BMI1-dependent sensitivity to deregulation of inositol metabolism in a proportion of medulloblastoma. We demonstrate mTOR pathway activation and metabolic adaptation specifically in medulloblastoma of the molecular subgroup G4 characterised by a BMI1High;CHD7Low signature and show this can be counteracted by IP6 treatment. Finally, we demonstrate that IP6 synergises with cisplatin to enhance its cytotoxicity in vitro and extends survival in a pre-clinical BMI1High;CHD7Low xenograft model.
Collapse
Affiliation(s)
- Sara Badodi
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nicola Pomella
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Xinyu Zhang
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gabriel Rosser
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - John Whittingham
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Maria Victoria Niklison-Chirou
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Therapeutic Innovation (CTI-Bath), Department of Pharmacy & Pharmacology, University of Bath, Bath, UK
| | - Yau Mun Lim
- UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Sebastian Brandner
- UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Gillian Morrison
- Centre for Regenerative Medicine & Cancer Research UK Edinburgh Centre, The University of Edinburgh, Edinburgh, UK
| | - Steven M Pollard
- Centre for Regenerative Medicine & Cancer Research UK Edinburgh Centre, The University of Edinburgh, Edinburgh, UK
| | - Christopher D Bennett
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Birmingham Women and Children's Hospital, Birmingham, UK
| | - Steven C Clifford
- Newcastle University Centre for Cancer, Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Andrew Peet
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Birmingham Women and Children's Hospital, Birmingham, UK
| | - M Albert Basson
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Silvia Marino
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
5
|
Thakur S, Goswami K, Rao P, Kaushik S, Singh BP, Kain P, Asthana S, Bhattacharjee S, Guchhait P, Eswaran SV. Fluoresceinated Aminohexanol Tethered Inositol Hexakisphosphate: Studies on Arabidopsis thaliana and Drosophila melanogaster and Docking with 2P1M Receptor. ACS OMEGA 2020; 5:9585-9597. [PMID: 32363311 PMCID: PMC7191843 DOI: 10.1021/acsomega.0c00961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 04/01/2020] [Indexed: 05/17/2023]
Abstract
Inositol hexakisphosphate (InsP6; phytic acid) is considered as the second messenger and plays a very important role in plants, animals, and human beings. It is the principal storage form of phosphorus in many plant tissues, especially in dry fruits, bran, and seeds. The resulting anion is a colorless species that plays a critical role in nutrition and is believed to cure many diseases. A fluoresceinated aminohexanol tethered inositol hexakisphosphate (III) had been synthesized earlier involving many complicated steps. We describe here a simple two-step synthesis of (III) and its characterization using different techniques such as matrix-assisted laser desorption ionization mass spectrometry, tandem mass spectrometry, and Fourier transform infrared, ultraviolet-visible, ultraviolet-fluorescence, 1H nuclear magnetic resonance (NMR), and two-dimensional NMR spectroscopies. The effect of (III) has been investigated in the model systems, Arabidopsis thaliana and Drosophila melanogaster. Using Schrodinger software, computational studies on the binding of (III) with the protein 2P1M (Auxin-receptor TIR1-adaptor ASK1 complex) has revealed strong binding propensity with this compound. These studies on the fluoresceinated tethered phytic acid could have far reaching implications on its efficacy for human health and treatment of diseases (cancer/tumor and glioblastoma) and for understanding phosphorous recycling in the environment, especially for plant systems.
Collapse
Affiliation(s)
- Sujeet
Kumar Thakur
- TERI
School of Advanced Studies, Plot No. 10, Vasant Kunj Institutional Area, Vasant
Kunj, Institutional Area, New Delhi 110070, India
| | - Krishnendu Goswami
- Regional
Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad, 121001 Haryana, India
| | - Pallavi Rao
- Amity
University, Noida, 201313 Uttar Pradesh, India
| | - Shivam Kaushik
- Regional
Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad, 121001 Haryana, India
| | - Bhanu Pratap Singh
- Translational
Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad, 121001 Haryana, India
| | - Pinky Kain
- Regional
Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad, 121001 Haryana, India
| | - Shailendra Asthana
- Translational
Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad, 121001 Haryana, India
| | - Saikat Bhattacharjee
- Regional
Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad, 121001 Haryana, India
| | - Prasenjit Guchhait
- Regional
Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad, 121001 Haryana, India
| | - Sambasivan V. Eswaran
- Teri
Deakin Nano Biotechnology Centre (TDNBC), Teri Gram, Gwal Pahari, Gurgaon- Faridabad Expressway, Gurugram, 122002 Haryana, India
| |
Collapse
|
6
|
Brehm MA, Windhorst S. New options of cancer treatment employing InsP 6. Biochem Pharmacol 2019; 163:206-214. [PMID: 30797871 DOI: 10.1016/j.bcp.2019.02.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/19/2019] [Indexed: 12/21/2022]
Abstract
Many mechanistic studies have been performed to analyze the cellular functions of the highly phosphorylated molecule inositol hexakisphosphate (InsP6) in health and disease. While the physiological intracellular functions are well described, the mechanism of potential pharmacological effects on cancer cell proliferation is still controversial. There are numerous studies demonstrating that a high InsP6 concentration (≥75 µM) inhibits growth of cancer cells in vitro and in vivo. Thus, there is no doubt that InsP6 exhibits anticancer activity but the mechanism underlying the cellular effects of extracellular InsP6 on cancer cells is far from being understood. In addition, studies on the inhibitory effect of InsP6 on cancer progression in animal models ignore aspects of its bioavailability. Here, we review and critically discuss the uptake mechanism and the intracellular involvement in signaling pathways of InsP6 in cancer cells. We take into account the controversial findings on InsP6 plasma concentration, which is a critical aspect of pharmacological accessibility of InsP6 for cancer treatment. Further, we discuss novel findings with respect to the effect of InsP6 on normal and immune cells as well as on platelet aggregate size. Our goal is to stimulate further mechanistic studies into novel directions considering previously disregarded aspects of InsP6. Only when we fully understand the mechanism underlying the anticancer activity of InsP6 novel and more efficient treatment options can be developed.
Collapse
Affiliation(s)
- Maria A Brehm
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabine Windhorst
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany.
| |
Collapse
|
7
|
Review of Natural Product-Derived Compounds as Potent Antiglioblastoma Drugs. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8139848. [PMID: 29181405 PMCID: PMC5664208 DOI: 10.1155/2017/8139848] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/17/2017] [Accepted: 09/17/2017] [Indexed: 12/28/2022]
Abstract
Common care for glioblastoma multiforme (GBM) is a surgical resection followed by radiotherapy and temozolomide- (TMZ-) based chemotherapy. Unfortunately, these therapies remain inadequate involving severe mortality and recurrence. Recently, new approaches discovering combinations of multiple inhibitors have been proposed along with the identification of key driver mutations that are specific to each patient. To date, this approach is still limited by the lack of effective therapy. Hopefully, novel compounds derived from natural products are suggested as potential solutions. Inhibitory effects of natural products on angiogenesis and metastasis and cancer suppressive effect of altering miRNA expression are provident discoveries. Angelica sinensis accelerates apoptosis by their key substances influencing factors of apoptosis pathways. Brazilin displays antitumor features by making influence on reactive oxygen species (ROS) intensity. Sargassum serratifolium, flavonoids, and so on have antimetastasis effect. Ficus carica controls miRNA that inhibits translation of certain secretory pathway proteins during the UPR. Serratia marcescens and patupilone (EPO 906) are physically assessed materials through clinical trials related to GBM progression. Consequently, our review puts emphasis on the potential of natural products in GBM treatment by regulating multiple malignant cancer-related pathway solving pending problem such as reducing toxicity and side effect.
Collapse
|
8
|
Silva EO, Bracarense APFRL. Phytic Acid: From Antinutritional to Multiple Protection Factor of Organic Systems. J Food Sci 2016; 81:R1357-62. [PMID: 27272247 DOI: 10.1111/1750-3841.13320] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 03/04/2016] [Accepted: 03/22/2016] [Indexed: 02/07/2023]
Abstract
Several studies have shown the benefits of natural antioxidants on health and food preservation. Phytic acid (IP6) is a natural antioxidant that is found mainly in cereals and vegetables and, for a long period of time, was considered an antinutritional factor. However, in vitro and in vivo studies have demonstrated its beneficial effects in the prevention and treatment of several pathological conditions and cancer. Despite the numerous benefits of IP6, the signs and intracellular interactions mediated by this antioxidant remain poorly understood. This review describes the main chemical and biological aspects of IP6, as well as its actions in the prevention and treatment of various diseases.
Collapse
Affiliation(s)
- Elisângela O Silva
- Laboratory of Animal Pathology, Univ. Estadual de Londrina, Rodovia Celso Garcia Cid, km 380, 86057-990, Londrina, PR, Brazil
| | - Ana Paula F R L Bracarense
- Laboratory of Animal Pathology, Univ. Estadual de Londrina, Rodovia Celso Garcia Cid, km 380, 86057-990, Londrina, PR, Brazil
| |
Collapse
|
9
|
Mörén L, Wibom C, Bergström P, Johansson M, Antti H, Bergenheim AT. Characterization of the serum metabolome following radiation treatment in patients with high-grade gliomas. Radiat Oncol 2016; 11:51. [PMID: 27039175 PMCID: PMC4818859 DOI: 10.1186/s13014-016-0626-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 03/22/2016] [Indexed: 11/26/2022] Open
Abstract
Background Glioblastomas progress rapidly making response evaluation using MRI insufficient since treatment effects are not detectable until months after initiation of treatment. Thus, there is a strong need for supplementary biomarkers that could provide reliable and early assessment of treatment efficacy. Analysis of alterations in the metabolome may be a source for identification of new biomarker patterns harboring predictive information. Ideally, the biomarkers should be found within an easily accessible compartment such as the blood. Method Using gas-chromatographic- time-of-flight-mass spectroscopy we have analyzed serum samples from 11 patients with glioblastoma during the initial phase of radiotherapy. Fasting serum samples were collected at admittance, on the same day as, but before first treatment and in the morning after the second and fifth dose of radiation. The acquired data was analyzed and evaluated by chemometrics based bioinformatics methods. Our findings were compared and discussed in relation to previous data from microdialysis in tumor tissue, i.e. the extracellular compartment, from the same patients. Results We found a significant change in metabolite pattern in serum comparing samples taken before radiotherapy to samples taken during early radiotherapy. In all, 68 metabolites were lowered in concentration following treatment while 16 metabolites were elevated in concentration. All detected and identified amino acids and fatty acids together with myo-inositol, creatinine, and urea were among the metabolites that decreased in concentration during treatment, while citric acid was among the metabolites that increased in concentration. Furthermore, when comparing results from the serum analysis with findings in tumor extracellular fluid we found a common change in metabolite patterns in both compartments on an individual patient level. On an individual metabolite level similar changes in ornithine, tyrosine and urea were detected. However, in serum, glutamine and glutamate were lowered after treatment while being elevated in the tumor extracellular fluid. Conclusion Cross-validated multivariate statistical models verified that the serum metabolome was significantly changed in relation to radiation in a similar pattern to earlier findings in tumor tissue. However, all individual changes in tissue did not translate into changes in serum. Our study indicates that serum metabolomics could be of value to investigate as a potential marker for assessing early response to radiotherapy in malignant glioma. Electronic supplementary material The online version of this article (doi:10.1186/s13014-016-0626-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lina Mörén
- Department of Chemistry, Computational Life Science Cluster, Umeå University, SE 901 87, Umeå, Sweden. .,Department of Chemistry, Umeå University, SE 90187, Umeå, Sweden.
| | - Carl Wibom
- Department of Radiation Sciences, Oncology, Umeå University, SE 901 85, Umeå, Sweden
| | - Per Bergström
- Department of Radiation Sciences, Oncology, Umeå University, SE 901 85, Umeå, Sweden
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umeå University, SE 901 85, Umeå, Sweden
| | - Henrik Antti
- Department of Chemistry, Computational Life Science Cluster, Umeå University, SE 901 87, Umeå, Sweden
| | - A Tommy Bergenheim
- Department of Clinical Neuroscience, Neurosurgery, Umeå University, SE 901 85, Umeå, Sweden
| |
Collapse
|
10
|
Benisi-Kohansal S, Shayanfar M, Mohammad-Shirazi M, Tabibi H, Sharifi G, Saneei P, Esmaillzadeh A. Adherence to the Dietary Approaches to Stop Hypertension-style diet in relation to glioma: a case–control study. Br J Nutr 2016; 115:1108-1116. [DOI: 10.1017/s0007114515005504] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AbstractData on the association of adherence to the Dietary Approaches to Stop Hypertension (DASH)-style and glioma are scarce. We aimed to examine the association between adherence to the DASH-style diet and glioma in Iranian adults. In this study, 128 pathologically confirmed cases of glioma were recruited from hospitals and 256 age- and sex-matched controls were enrolled from other wards of the hospital. Dietary intakes were assessed using a 126-item validated FFQ. Adherence to the DASH-style diet was followed considering the healthy and non-healthy foods emphasised in the DASH dietary pattern. After controlling for potential confounders, individuals with the greatest adherence to the DASH diet were 72 % less likely to have glioma compared with those with the lowest adherence (OR 0·28; 95 % CI 0·13, 0·57). Individuals with the highest consumption of fruits had lower odds for having glioma compared with those with the lowest intake (OR 0·31; 95 % CI 0·14, 0·68). A protective association was also observed between consumption of legumes and nuts and risk of glioma (OR 0·23; 95 % CI 0·10, 0·53). We found a significant positive association between red and processed meat (OR 2·60; 95 % CI 1·16, 5·81) and salt intakes (OR 2·87; 95 % CI 1·30, 6·34) and risk of glioma, after taking all potential confounders into account. Adherence to the DASH-style dietary pattern was inversely associated with glioma. In addition, some components of the DASH diet, including red meats and salt intakes, were positively associated with glioma. Consumption of nuts and legumes as well as fruits was inversely associated with glioma. Prospective cohort studies are required to confirm our findings.
Collapse
|
11
|
Bizzarri M, Dinicola S, Bevilacqua A, Cucina A. Broad Spectrum Anticancer Activity of Myo-Inositol and Inositol Hexakisphosphate. Int J Endocrinol 2016; 2016:5616807. [PMID: 27795708 PMCID: PMC5067332 DOI: 10.1155/2016/5616807] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/06/2016] [Indexed: 02/06/2023] Open
Abstract
Inositols (myo-inositol and inositol hexakisphosphate) exert a wide range of critical activities in both physiological and pathological settings. Deregulated inositol metabolism has been recorded in a number of diseases, including cancer, where inositol modulates different critical pathways. Inositols inhibit pRB phosphorylation, fostering the pRB/E2F complexes formation and blocking progression along the cell cycle. Inositols reduce PI3K levels, thus counteracting the activation of the PKC/RAS/ERK pathway downstream of PI3K activation. Upstream of that pathway, inositols disrupt the ligand interaction between FGF and its receptor as well as with the EGF-transduction processes involving IGF-II receptor and AP-1 complexes. Additionally, Akt activation is severely impaired upon inositol addition. Downregulation of both Akt and ERK leads consequently to NF-kB inhibition and reduced expression of inflammatory markers (COX-2 and PGE2). Remarkably, inositol-induced downregulation of presenilin-1 interferes with the epithelial-mesenchymal transition and reduces Wnt-activation, β-catenin translocation, Notch-1, N-cadherin, and SNAI1 release. Inositols interfere also with the cytoskeleton by upregulating Focal Adhesion Kinase and E-cadherin and decreasing Fascin and Cofilin, two main components of pseudopodia, leading hence to invasiveness impairment. This effect is reinforced by the inositol-induced inhibition on metalloproteinases and ROCK1/2 release. Overall, these effects enable inositols to remodel the cytoskeleton architecture.
Collapse
Affiliation(s)
- Mariano Bizzarri
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Systems Biology Group Lab, Sapienza University of Rome, Rome, Italy
- *Mariano Bizzarri:
| | - Simona Dinicola
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy
- Department of Surgery “Pietro Valdoni”, Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy
| | - Arturo Bevilacqua
- Department of Psychology, Section of Neuroscience, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy
| | - Alessandra Cucina
- Department of Surgery “Pietro Valdoni”, Sapienza University of Rome, Via A. Scarpa 14, 00161 Rome, Italy
- Azienda Policlinico Umberto I, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
12
|
de Lima EM, Kanunfre CC, de Andrade LF, Granato D, Rosso ND. Cytotoxic effect of inositol hexaphosphate and its Ni(II) complex on human acute leukemia Jurkat T cells. Toxicol In Vitro 2015; 29:2081-8. [PMID: 26335902 DOI: 10.1016/j.tiv.2015.08.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 08/26/2015] [Accepted: 08/28/2015] [Indexed: 01/22/2023]
Abstract
Inositol hexaphosphate (InsP6) is present in cereals, legumes, nuts and seed oils and is biologically active against some tumor and cancer cells. Herein, this study aimed at evaluating the cellular toxicity, antiproliferative activity and effects on cell cycle progression of free InsP6 and InsP6-Ni(II) of leukemic T (Jurkat) and normal human cells. Treatments with InsP6 at concentrations between 1.0 and 4.0mM significantly decreased the viability of Jurkat cells, but showed no cytotoxic effect on normal human lymphocytes. Treatment with InsP6-Ni(II) complex at concentrations between 0.05 and 0.30 mM showed an anti-proliferative dose and a time-dependent effect, with significantly reduced cell viability of Jurkat cells but showed no cytotoxic effect on normal human lymphocytes as compared to the control. Ni(II) free ion was toxic to normal cells while InsP6-Ni(II) had no cytotoxic effect. The InsP6-Ni(II) complex potentiated (up to 10×) the antiproliferative effect of free InsP6 on Jurkat cells. The cytometric flow assay showed that InsP6 led to an accumulation of cells in the G0/G1 phase of the cell cycle, accompanied by a decrease in the number of cells in S and G2/M phases, whereas InsP6-Ni(II) has led to an accumulation of cells in the S and G2/M phases. Our findings showed that InsP6-Ni(II) potentiates cytotoxic effects of InsP6 on Jurkat cells and may be a potential adjuvant in the treatment of cancer.
Collapse
Affiliation(s)
- Eliane May de Lima
- Departamento de Química, Universidade Estadual de Ponta Grossa, Av. Carlos Cavalcanti, 4748, 84030-900 Ponta Grossa, Brazil
| | - Carla Cristine Kanunfre
- Departamento de Biologia Celular, Universidade Estadual de Ponta Grossa, Av. Carlos Cavalcanti, 4748, 84030-900 Ponta Grossa, Brazil
| | - Lucas Ferrari de Andrade
- Departamento de Biologia Celular, Universidade Federal do Paraná, Rua Francisco H. dos Santos, s/n, 81531-990 Curitiba, Brazil
| | - Daniel Granato
- Departamento de Engenharia de Alimentos, Universidade Estadual de Ponta Grossa, Brazil. Av. Carlos Cavalcanti, 4748, 84030-900 Ponta Grossa, Brazil
| | - Neiva Deliberali Rosso
- Departamento de Química, Universidade Estadual de Ponta Grossa, Av. Carlos Cavalcanti, 4748, 84030-900 Ponta Grossa, Brazil.
| |
Collapse
|
13
|
Ahmadipour F, Noordin MI, Mohan S, Arya A, Paydar M, Looi CY, Keong YS, Siyamak EN, Fani S, Firoozi M, Yong CL, Sukari MA, Kamalidehghan B. Koenimbin, a natural dietary compound of Murraya koenigii (L) Spreng: inhibition of MCF7 breast cancer cells and targeting of derived MCF7 breast cancer stem cells (CD44(+)/CD24(-/low)): an in vitro study. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:1193-208. [PMID: 25759564 PMCID: PMC4346015 DOI: 10.2147/dddt.s72127] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Inhibition of breast cancer stem cells has been shown to be an effective therapeutic strategy for cancer prevention. The aims of this work were to evaluate the efficacy of koenimbin, isolated from Murraya koenigii (L) Spreng, in the inhibition of MCF7 breast cancer cells and to target MCF7 breast cancer stem cells through apoptosis in vitro. METHODS Koenimbin-induced cell viability was evaluated using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. Nuclear condensation, cell permeability, mitochondrial membrane potential, and cytochrome c release were observed using high-content screening. Cell cycle arrest was examined using flow cytometry, while human apoptosis proteome profiler assays were used to investigate the mechanism of apoptosis. Protein expression levels of Bax, Bcl2, and heat shock protein 70 were confirmed using Western blotting. Caspase-7, caspase-8, and caspase-9 levels were measured, and nuclear factor kappa B (NF-κB) activity was assessed using a high-content screening assay. Aldefluor™ and mammosphere formation assays were used to evaluate the effect of koenimbin on MCF7 breast cancer stem cells in vitro. The Wnt/β-catenin signaling pathway was investigated using Western blotting. RESULTS Koenimbin-induced apoptosis in MCF7 cells was mediated by cell death-transducing signals regulating the mitochondrial membrane potential by downregulating Bcl2 and upregulating Bax, due to cytochrome c release from the mitochondria to the cytosol. Koenimbin induced significant (P<0.05) sub-G0 phase arrest in breast cancer cells. Cytochrome c release triggered caspase-9 activation, which then activated caspase-7, leading to apoptotic changes. This form of apoptosis is closely associated with the intrinsic pathway and inhibition of NF-κB translocation from the cytoplasm to the nucleus. Koenimbin significantly (P<0.05) decreased the aldehyde dehydrogenase-positive cell population in MCF7 cancer stem cells and significantly (P<0.01) decreased the size and number of MCF7 cancer stem cells in primary, secondary, and tertiary mammospheres in vitro. Koenimbin also significantly (P<0.05) downregulated the Wnt/β-catenin self-renewal pathway. CONCLUSION Koenimbin has potential for future chemoprevention studies, and may lead to the discovery of further cancer management strategies by reducing cancer resistance and recurrence and improving patient survival.
Collapse
Affiliation(s)
- Fatemeh Ahmadipour
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Syam Mohan
- Medical Research Center, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Aditya Arya
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mohammadjavad Paydar
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Chung Yeng Looi
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yeap Swee Keong
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| | - Ebrahimi Nigjeh Siyamak
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| | - Somayeh Fani
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Maryam Firoozi
- Department of Medical Genetics, National Institute for Genetic Engineering and Biotechnology, Tehran, Iran
| | - Chung Lip Yong
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Behnam Kamalidehghan
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
14
|
Carminati PO, Donaires FS, Marques MM, Donadi EA, Passos GAS, Sakamoto-Hojo ET. Cisplatin associated with LY294002 increases cytotoxicity and induces changes in transcript profiles of glioblastoma cells. Mol Biol Rep 2013; 41:165-77. [DOI: 10.1007/s11033-013-2849-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Accepted: 10/29/2013] [Indexed: 02/03/2023]
|
15
|
Karmakar S, Choudhury SR, Banik NL, Ray SK. N-(4-Hydroxyphenyl) Retinamide Potentiated Anti-tumor Efficacy of Genistein in Human Ewing's Sarcoma Xenografts. World J Oncol 2011; 2:53-63. [PMID: 21822457 PMCID: PMC3151019 DOI: 10.4021/wjon301w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Ewing’s sarcoma is a pediatric tumor that mainly occurs in soft tissues and bones. New therapeutic strategies are urgently needed for treatment of Ewing’s sarcoma. We examined for the first time the efficacy of N-(4-hydroxyphenyl) retinamide (4-HPR) and genistein (GST) alone and also in combination for controlling growth of human Ewing’s sarcoma SK-N-MC and RD-ES xenografts. Methods Efficacy of combination therapy was evaluated using histopathological parameters. Molecular mechanisms of combination therapy were detected using Western blotting and immunofluorescence microscopy. Results Histopathological examination of tumor sections showed that control group maintained characteristic growth of tumors, 4-HPR alone inhibited proliferation of tumor cells, GST alone induced apoptosis to some extent, and combination of 4-HPR and GST significantly induced apoptosis in both Ewing’s sarcoma xenografts. Time-dependent reductions in body weight, tumor volume, and tumor weight were also found. Combination therapy increased Bax : Bcl-2 ratio to trigger mitochondrial release of Smac/Diablo into the cytosol to downregulate the baculovirus inhibitor-of-apoptosis repeat containing (BIRC) proteins such as BIRC-2 and BIRC-3 and thereby promote apoptosis. Activation of caspase-3 and mitochondrial release of apoptosis-inducing factor (AIF) occurred in course of apoptosis. Downregulation of the survival factor NF-κB and the angiogenic factors VEGF and FGF2 and increase in caspase-3 activity controlled tumor growth. In situ immunofluorescent labelings showed overexpression of calpain, caspase-12 and caspase-3, and AIF in xenografts, indicating induction of cysteine proteases and AIF for apoptosis. Conclusions Results revealed that combination of 4-HPR and GST could be highly effective treatment for inhibiting Ewing’s sarcomas in vivo.
Collapse
Affiliation(s)
- Surajit Karmakar
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | | | | | | |
Collapse
|
16
|
Wibom C, Surowiec I, Mörén L, Bergström P, Johansson M, Antti H, Bergenheim AT. Metabolomic patterns in glioblastoma and changes during radiotherapy: a clinical microdialysis study. J Proteome Res 2010; 9:2909-19. [PMID: 20302353 DOI: 10.1021/pr901088r] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We employed stereotactic microdialysis to sample extracellular fluid intracranially from glioblastoma patients, before and during the first five days of conventional radiotherapy treatment. Microdialysis catheters were implanted in the contrast enhancing tumor as well as in the brain adjacent to tumor (BAT). Reference samples were collected subcutaneously from the patients' abdomen. The samples were analyzed by gas chromatography-time-of-flight mass spectrometry (GC-TOF MS), and the acquired data was processed by hierarchical multivariate curve resolution (H-MCR) and analyzed with orthogonal partial least-squares (OPLS). To enable detection of treatment-induced alterations, the data was processed by individual treatment over time (ITOT) normalization. One-hundred fifty-one metabolites were reliably detected, of which 67 were identified. We found distinct metabolic differences between the intracranially collected samples from tumor and the BAT region. There was also a marked difference between the intracranially and the subcutaneously collected samples. Furthermore, we observed systematic metabolic changes induced by radiotherapy treatment among both tumor and BAT samples. The metabolite patterns affected by treatment were different between tumor and BAT, both containing highly discriminating information, ROC values of 0.896 and 0.821, respectively. Our findings contribute to increased molecular knowledge of basic glioblastoma pathophysiology and point to the possibility of detecting metabolic marker patterns associated to early treatment response.
Collapse
Affiliation(s)
- Carl Wibom
- Institution for Radiation Sciences, Department of Oncology, Umeå University Hospital, Umeå, Sweden
| | | | | | | | | | | | | |
Collapse
|
17
|
Eberhard M, Föller M, Lang F. Effect of phytic acid on suicidal erythrocyte death. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2010; 58:2028-2033. [PMID: 20058927 DOI: 10.1021/jf903666b] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Phytic acid, an anticarcinogenic food component, stimulates apoptosis of tumor cells. Similar to apoptosis, human erythrocytes may undergo suicidal death or eryptosis, characterized by cell membrane scrambling and cell shrinkage. Triggers of eryptosis include energy depletion. Phytate intake could cause anemia, an effect attributed to iron complexation. The present experiments explored whether phytic acid influences eryptosis. Supernatant hemoglobin concentration was determined to reveal hemolysis, annexin V-binding in FACS analysis was utilized to identify erythrocytes with scrambled cell membrane, forward scatter in FACS analysis was taken as a measure of cell volume, and a luciferin-luciferase assay was employed to determine erythrocyte ATP content. As a result, phytic acid (>or=1 mM) did not lead to significant hemolysis, but significantly increased the percentage of annexin V-binding erythrocytes, significantly decreased forward scatter, and significantly decreased cellular ATP content. In conclusion, phytic acid stimulates suicidal human erythrocyte death, an effect paralleling its proapoptotic effect on nucleated cells.
Collapse
Affiliation(s)
- Matthias Eberhard
- Department of Physiology, Eberhard-Karls-University of Tubingen, Gmelinstrasse 5, D-72076 Tuebingen, Germany
| | | | | |
Collapse
|