1
|
Lu J, He R, Liu Y, Zhang J, Xu H, Zhang T, Chen L, Yang G, Zhang J, Liu J, Chi H. Exploiting cell death and tumor immunity in cancer therapy: challenges and future directions. Front Cell Dev Biol 2024; 12:1416115. [PMID: 38887519 PMCID: PMC11180757 DOI: 10.3389/fcell.2024.1416115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Cancer remains a significant global challenge, with escalating incidence rates and a substantial burden on healthcare systems worldwide. Herein, we present an in-depth exploration of the intricate interplay between cancer cell death pathways and tumor immunity within the tumor microenvironment (TME). We begin by elucidating the epidemiological landscape of cancer, highlighting its pervasive impact on premature mortality and the pronounced burden in regions such as Asia and Africa. Our analysis centers on the pivotal concept of immunogenic cell death (ICD), whereby cancer cells succumbing to specific stimuli undergo a transformation that elicits robust anti-tumor immune responses. We scrutinize the mechanisms underpinning ICD induction, emphasizing the release of damage-associated molecular patterns (DAMPs) and tumor-associated antigens (TAAs) as key triggers for dendritic cell (DC) activation and subsequent T cell priming. Moreover, we explore the contributions of non-apoptotic RCD pathways, including necroptosis, ferroptosis, and pyroptosis, to tumor immunity within the TME. Emerging evidence suggests that these alternative cell death modalities possess immunogenic properties and can synergize with conventional treatments to bolster anti-tumor immune responses. Furthermore, we discuss the therapeutic implications of targeting the TME for cancer treatment, highlighting strategies to harness immunogenic cell death and manipulate non-apoptotic cell death pathways for therapeutic benefit. By elucidating the intricate crosstalk between cancer cell death and immune modulation within the TME, this review aims to pave the way for the development of novel cancer therapies that exploit the interplay between cell death mechanisms and tumor immunity and overcome Challenges in the Development and implementation of Novel Therapies.
Collapse
Affiliation(s)
- Jiaan Lu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Ru He
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Yang Liu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jinghan Zhang
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Heng Xu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Tianchi Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Li Chen
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| | - Jun Zhang
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| |
Collapse
|
2
|
Allegra A, Mirabile G, Caserta S, Stagno F, Russo S, Pioggia G, Gangemi S. Oxidative Stress and Chronic Myeloid Leukemia: A Balance between ROS-Mediated Pro- and Anti-Apoptotic Effects of Tyrosine Kinase Inhibitors. Antioxidants (Basel) 2024; 13:461. [PMID: 38671909 PMCID: PMC11047441 DOI: 10.3390/antiox13040461] [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: 02/20/2024] [Revised: 03/31/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The balanced reciprocal translocation t (9; 22) (q34; q11) and the BCR-ABL fusion gene, which produce p210 bcr-abl protein production with high tyrosine kinase activity, are characteristics of chronic myeloid leukemia, a myeloproliferative neoplasm. This aberrant protein affects several signaling pathways connected to both apoptosis and cell proliferation. It has been demonstrated that tyrosine kinase inhibitor treatment in chronic myeloid leukemia acts by inducing oxidative stress and, depending on its level, can activate signaling pathways responsible for either apoptosis or survival in leukemic cells. Additionally, oxidative stress and reactive oxygen species generation also mediate apoptosis through genomic activation. Furthermore, it was shown that oxidative stress has a role in both BCR-ABL-independent and BCR-ABL-dependent resistance pathways to tyrosine kinases, while patients with chronic myeloid leukemia were found to have a significantly reduced antioxidant level. The ideal environment for tyrosine kinase inhibitor therapy is produced by a favorable oxidative status. We discuss the latest studies that aim to manipulate the redox system to alter the apoptosis of cancerous cells.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood ‘Gaetano Barresi’, University of Messina, 98125 Messina, Italy; (G.M.); (S.C.); (F.S.); (S.R.)
| | - Giuseppe Mirabile
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood ‘Gaetano Barresi’, University of Messina, 98125 Messina, Italy; (G.M.); (S.C.); (F.S.); (S.R.)
| | - Santino Caserta
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood ‘Gaetano Barresi’, University of Messina, 98125 Messina, Italy; (G.M.); (S.C.); (F.S.); (S.R.)
| | - Fabio Stagno
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood ‘Gaetano Barresi’, University of Messina, 98125 Messina, Italy; (G.M.); (S.C.); (F.S.); (S.R.)
| | - Sabina Russo
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood ‘Gaetano Barresi’, University of Messina, 98125 Messina, Italy; (G.M.); (S.C.); (F.S.); (S.R.)
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy;
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy;
| |
Collapse
|
3
|
Solaimani F, Habibi E, Ghasemi M, Mahboubi S, Zamani E, Shaki F. The Protective Effects of Trametes Versicolor on Arsenic-Induced Male Reproductive Toxicity through Regulation of Oxidative Stress: A Biochemical and Histopathological Survey. Andrologia 2023; 2023:1-13. [DOI: 10.1155/2023/7579366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
Abstract
Exposure to arsenic is linked to a wide range of diseases, in particular male reproductive toxicities. Trametes versicolor is a traditional medicinal fungus with a remarkable potential for antioxidant activity. The purpose of this study was to examine the ameliorating effects of water and methanol extracts of T. versicolor on arsenic-induced male reproductive toxicities via the abrogation of oxidative stress. The mice were divided as follows: control: normal saline, As: arsenic (15 mg/kg), WE: water extract (400 mg/kg), ME: methanol extract (400 mg/kg), As + WE: arsenic (15 mg/kg) + water extract (100, 200, 400 mg/kg), As + ME: arsenic (15 mg/kg) + methanol extract (100, 200, 400 mg/kg), and positive control: arsenic (15 mg/kg) + vitamin C (500 mg/kg). Animals were treated via the intraperitoneal route. About 24 hr later, the mice were euthanized, and oxidative stress parameters (reactive oxygen species [ROS], lipid peroxidation, glutathione concentration, protein carbonylation, glutathione peroxidase, and superoxide dismutase activity), histopathological changes and sperm parameters (count, motility, and morphology) were examined in the testicular tissue. Arsenic caused significant pathological changes in the testicular tissue and sperm morphology and significantly reduced sperm count and motility. Moreover, arsenic mediated oxidative stress via significant increases in ROS generation, lipid peroxidation, and protein carbonyl content, as well as significant depletion in glutathione concentration and superoxide dismutase and glutathione peroxidase activities. Although, coadministration of water and methanol extracts of T. versicolor at 200 and 400 mg/kg counteracted arsenic-induced oxidative and histopathological damages and improved sperm parameters. Our study indicated that T. versicolor ameliorated arsenic-induced testis toxicity and sperm dysfunction via attenuation of oxidative damage.
Collapse
Affiliation(s)
- Fatemeh Solaimani
- Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
| | - Emran Habibi
- Medicinal Plants Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Pharmacognosy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Maryam Ghasemi
- Department of Pathology, Immunogenetics Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saba Mahboubi
- Department of Pharmacology and Toxicology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Ehsan Zamani
- Department of Pharmacology and Toxicology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Shaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
4
|
García-Flores N, Jiménez-Suárez J, Garnés-García C, Fernández-Aroca DM, Sabater S, Andrés I, Fernández-Aramburo A, Ruiz-Hidalgo MJ, Belandia B, Sanchez-Prieto R, Cimas FJ. P38 MAPK and Radiotherapy: Foes or Friends? Cancers (Basel) 2023; 15:861. [PMID: 36765819 PMCID: PMC9913882 DOI: 10.3390/cancers15030861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/16/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Over the last 30 years, the study of the cellular response to ionizing radiation (IR) has increased exponentially. Among the various signaling pathways affected by IR, p38 MAPK has been shown to be activated both in vitro and in vivo, with involvement in key processes triggered by IR-mediated genotoxic insult, such as the cell cycle, apoptosis or senescence. However, we do not yet have a definitive clue about the role of p38 MAPK in terms of radioresistance/sensitivity and its potential use to improve current radiotherapy. In this review, we summarize the current knowledge on this family of MAPKs in response to IR as well as in different aspects related to radiotherapy, such as their role in the control of REDOX, fibrosis, and in the radiosensitizing effect of several compounds.
Collapse
Affiliation(s)
- Natalia García-Flores
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Jaime Jiménez-Suárez
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Cristina Garnés-García
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Diego M. Fernández-Aroca
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Sebastia Sabater
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Servicio de Oncología Radioterápica, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain
| | - Ignacio Andrés
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Servicio de Oncología Radioterápica, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain
| | - Antonio Fernández-Aramburo
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Servicio de Oncología Médica, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain
| | - María José Ruiz-Hidalgo
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Área de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Borja Belandia
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, 28029 Madrid, Spain
| | - Ricardo Sanchez-Prieto
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, 28029 Madrid, Spain
- Departamento de Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Francisco J. Cimas
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Área de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| |
Collapse
|
5
|
Sadeghinezhad S, Khodamoradi E, Diojan L, Taeb S, Najafi M. Radioprotective Mechanisms of Arbutin: A Systematic Review. Curr Drug Res Rev 2022; 14:132-138. [PMID: 35319405 DOI: 10.2174/2589977514666220321114415] [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: 09/21/2021] [Revised: 12/15/2021] [Accepted: 01/06/2022] [Indexed: 06/14/2023]
Abstract
PURPOSE Efforts to produce radioprotective agents of high potential are appropriate strategies for overcoming possible IR toxicity in organisms. The present research aims to evaluate the signaling pathways and mechanisms through which arbutin exerts radioprotective effects on organisms. METHODS The databases of PubMed, Web of Sciences, Google Scholar, and Scopus were searched to find studies that reported radioprotective effects for arbutin. Besides, the data were searched within the time period from 2010 to 2020. RESULTS Five research articles met our criteria, which were included in the analysis based on their relevance to the topic. The present systematic review provides conclusions about various mechanisms and pathways through which arbutin induces radioprotection. CONCLUSIONS Based on the relevant studies, various mechanisms can be proposed for inducing radioprotective effects by arbutin, including inhibition of oxidative stress, apoptosis, and inflammation.
Collapse
Affiliation(s)
- Shima Sadeghinezhad
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ehsan Khodamoradi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Loghman Diojan
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shahram Taeb
- Department of Radiology, School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
6
|
Apoptotic mechanisms of myricitrin isolated from Madhuca longifolia leaves in HL-60 leukemia cells. Mol Biol Rep 2021; 48:5327-5334. [PMID: 34156605 DOI: 10.1007/s11033-021-06500-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/15/2021] [Indexed: 10/21/2022]
Abstract
Myricitrin, a naturally occurring flavonoid in Madhuca longifolia, possesses several medicinal properties. Even though our earlier work revealed its role against the proliferation of acute myelogenous leukemia cells (HL-60), its molecular mechanisms have not yet been revealed. The current study aims to explore the molecular mechanisms of myricitrin (isolated from an ethnomedicinal drug Madhuca longifolia) to induce apoptosis in HL-60 cells. Treatment with IC-50 dose of myricitrin (353 µM) caused cellular shrinkage and cell wall damage in HL-60 cells compared to untreated control cells. Myricitrin treatment reduced the mitochondrial membrane potential (22.95%), increased DNA fragmentation (90.4%), inhibited the cell survival proteins (RAS, B-RAF, & BCL-2) and also induced pro-apoptotic proteins (p38, pro-caspase-3, pro-caspase-9 and caspase-3) in the HL-60 cells. The present study provides scientific evidence for the apoptosis caused by myricitrin in HL-60 leukemia cells. Hence, the phytochemical myricitrin could be considered as a potential candidate to develop an anticancer drug after checking its efficacy through suitable pre-clinical and clinical studies.
Collapse
|
7
|
Sarkar MK, Kar A, Jayaraman A, Kar Mahapatra S, Vadivel V. Vitexin isolated from Prosopis cineraria leaves induce apoptosis in K-562 leukemia cells via inhibition of the BCR-ABL-Ras-Raf pathway. J Pharm Pharmacol 2021; 74:103-111. [PMID: 34109977 DOI: 10.1093/jpp/rgab085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 05/14/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Leukemia is one of the severe cancer types all around the globe. Even though some chemotherapeutic drugs are available for treating leukemia, they have various side effects. As an alternative approach, herbal drugs are focused on current research to overcome leukemia. The present work was conducted to investigate the antileukemic mechanism of active phytochemical vitexin, which was isolated from ethno-medicine (Prosopis cineraria leaf) used by traditional healers of West Bengal, India. METHODS Antiproliferative mechanisms of selected phyto-compound against K-562 cells were evaluated using cellular uptake, morphological changes, DNA fragmentation, mitochondrial membrane potential and signaling pathways analysis. KEY FINDINGS Vitexin exhibited cytotoxicity by reducing mitochondrial membrane potential (32.40%) and causing DNA fragmentation (84.15%). The western blotting study indicated inhibition of cell survival proteins (BCR, ABL, H-RAS, N-RAS, K-RAS and RAF) and expression of apoptotic proteins (p38, BAX and caspase-9) in leukemia cells upon treatment with vitexin. CONCLUSIONS Based on the results, presently investigated phyto-compound vitexin could be considered for developing safe and natural drugs to treat leukemia after conducting suitable preclinical and clinical trials.
Collapse
Affiliation(s)
- Monaj Kumar Sarkar
- Chemical Biology Lab (ASK-II-409), School of Chemical and Biotechnology (SCBT), SASTRA Deemed University, Thanjavur, India
| | - Amrita Kar
- Medicinal Chemistry and Immunology Lab (ASK-II-406), School of Chemical and Biotechnology (SCBT), SASTRA Deemed University, Thanjavur, India
| | - Adithyan Jayaraman
- Medicinal Chemistry and Immunology Lab (ASK-II-406), School of Chemical and Biotechnology (SCBT), SASTRA Deemed University, Thanjavur, India
| | - Santanu Kar Mahapatra
- Medicinal Chemistry and Immunology Lab (ASK-II-406), School of Chemical and Biotechnology (SCBT), SASTRA Deemed University, Thanjavur, India
| | - Vellingiri Vadivel
- Chemical Biology Lab (ASK-II-409), School of Chemical and Biotechnology (SCBT), SASTRA Deemed University, Thanjavur, India
| |
Collapse
|
8
|
Vineetha RC, Hariharan S, Jaleel A, Chandran M, Nair RH. L-Ascorbic Acid and α-Tocopherol Synergistically Triggers Apoptosis Inducing Antileukemic Effects of Arsenic Trioxide via Oxidative Stress in Human Acute Promyelocytic Leukemia Cells. Front Oncol 2020; 10:65. [PMID: 32154163 PMCID: PMC7047343 DOI: 10.3389/fonc.2020.00065] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/14/2020] [Indexed: 01/14/2023] Open
Abstract
Chemosensitization is an effective strategy to overcome the drawbacks of arsenic trioxide (As2O3) treatment, which may be possible through the use of dietary supplements in combination. The present investigation evaluates the synergistic mechanism of action of vitamins, such as L-ascorbic acid (L-AA) and α-tocopherol (α-TOC) in As2O3 chemotherapy using human leukemia (HL-60) cells. In vitro assays on the cytotoxicity of As2O3 and vitamins and cellular apoptotic evidences were done; a proteomic investigation with mass spectrometry was also performed. The combination of L-AA and α-TOC potentiates As2O3 cytotoxicity in HL-60 cells, substantiated by depletion in antioxidant status, mitochondrial transmembrane potential, and inhibition of nuclear factor erythroid 2-related factor 2 and B-cell lymphoma 2 transcription factors. Mass spectrometry results showed decreased expression of proteins regulating cell cycle and translation in cells treated with As2O3, L-AA, and α-TOC when compared with As2O3-treated sample. In addition, this combination treatment identified numerous proteins associated with apoptosis and cell stress. HL-60 cells became more prone to As2O3 on exposure to L-AA and α-TOC, indicating that this combination may be a promising approach to increase the outcome of As2O3 chemotherapy.
Collapse
Affiliation(s)
- Radhakrishnan Chandraprabha Vineetha
- Physiology Research Laboratory, School of Biosciences, Mahatma Gandhi University, Kottayam, India.,Laboratory of Cytogenetics and Molecular Diagnostics, Division of Cancer Research, Regional Cancer Centre, Trivandrum, India
| | - Sreedharan Hariharan
- Laboratory of Cytogenetics and Molecular Diagnostics, Division of Cancer Research, Regional Cancer Centre, Trivandrum, India
| | - Abdul Jaleel
- Proteomics Facility, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | - Mahesh Chandran
- Proteomics Facility, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | | |
Collapse
|
9
|
da F Iwahara LK, de Oliveira MS, de Alencar MAV. Evaluation of the effect of three constituent metals of monazita on the radiosensibility of human osteoblasts. JOURNAL OF ENVIRONMENTAL RADIOACTIVITY 2019; 208-209:106011. [PMID: 31288125 DOI: 10.1016/j.jenvrad.2019.106011] [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: 08/09/2018] [Revised: 06/17/2019] [Accepted: 07/03/2019] [Indexed: 06/09/2023]
Abstract
Thorium has gained notoriety in recent years, as a potential source of nuclear energy, substituting uranium in power plants. Monazite is an important source of thorium, as well of uranium and rare earths elements. Workers involved in the extraction and manipulation of this mineral are occupationally exposed to a range of metal mixtures containing thorium and to ionizing radiation. As an osteotropic substance, thorium is mostly deposited in bone tissue and may interfere in cellular radiosensitivity. A human osteoblast cell line was used to evaluate the effects of thorium (Th), cerium (Ce) and lanthanum (La) on cell radiosensivity, using proliferation as indicator. Assays were performed using cell cultures exposed to metals alone and metals combined with ionizing radiation. No stimulus of proliferation was observed when samples were exposed to metals or radiation alone. On the other hand, the metals were able to influence cell radiosensivity, in a concentration-dependent manner when metals and radiation were applied simultaneously. Samples irradiated and exposed to metals combinations revealed an interaction between them in all the tested arrangements (Th-Ce, Th-La, Th-Ce-La). All interactions proved to be of the antagonist type relative to the proliferation indicator, with a higher degree seen for the Th-Ce association. Such results showed the possibility that metal mixtures together with radiation may produce combined effects on osteoblasts, through modifications on the degree of radiosensivity. The results indicate the possibility of an enhancement in occupational risk for workers that manipulate monazite byproducts. Thus, the development of risk assessment models that include the evaluation of mixtures and their cytotoxic and radiotoxic effects on tissues and organs must be highlighted.
Collapse
Affiliation(s)
- Lucas K da F Iwahara
- Dosimetry Division, Radiation Protection and Dosimetry Institute, Av. Salvador Allende, s/no. Barra da Tijuca, Rio de Janeiro, CEP 22783-127l, Brazil.
| | - Monica S de Oliveira
- Dosimetry Division, Radiation Protection and Dosimetry Institute, Av. Salvador Allende, s/no. Barra da Tijuca, Rio de Janeiro, CEP 22783-127l, Brazil
| | - Marcus A V de Alencar
- Dosimetry Division, Radiation Protection and Dosimetry Institute, Av. Salvador Allende, s/no. Barra da Tijuca, Rio de Janeiro, CEP 22783-127l, Brazil
| |
Collapse
|
10
|
Hu Z, Tie Y, Lv G, Zhu J, Fu H, Zheng X. Transcriptional activation of miR-320a by ATF2, ELK1 and YY1 induces cancer cell apoptosis under ionizing radiation conditions. Int J Oncol 2018; 53:1691-1702. [PMID: 30066913 DOI: 10.3892/ijo.2018.4497] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/06/2018] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs or miRs) play important roles in numerous cellular processes, including development, proliferation, tumorigenesis and apoptosis. It has been reported that miRNA expression is induced by ionizing radiation (IR) in cancer cells. However, the underlying molecular mechanisms are not yet fully understood. In this study, endogenous miR‑320a and its primary precursor (pri‑miR‑320a) were assayed by reverse transcription‑quantitative PCR (RT‑qPCR). Luciferase activities were measured using a dual‑luciferase reporter assay system. Western blot analysis was used to determine the protein expressions of upstream and downstream genes of miR‑320a. Cell apoptosis was evaluated by Annexin V apoptosis assay and cell proliferation was measured using the trypan blue exclusion method. The results revealed that miR‑320a expression increased linearly with the IR dose and treatment duration. Three transcription factors, activating transcription factor 2 (ATF2), ETS transcription factor (ELK1) and YY1 transcription factor (YY1), were activated by p38 mitogen‑activated protein kinase (MAPK) and mitogen‑activated protein kinase 8 (JNK) and by upregulated miR‑320a expression under IR conditions. In addition, it was identified that X‑linked inhibitor of apoptosis (XIAP) was an miR‑320a target gene during the IR response. By targeting XIAP, miR‑320a induced apoptosis and inhibited the proliferation of the cancer cells. On the whole, the results of this study demonstrated that miRNA‑320a, regulated by the p38 MAPK/JNK pathway, enhanced the radiosensitivity of cancer cells by inhibiting XIAP and this may thus prove to be a potential therapeutic approach with which to overcome radioresistance in cancer treatment.
Collapse
Affiliation(s)
- Zheng Hu
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Yi Tie
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Guixiang Lv
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Jie Zhu
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Hanjiang Fu
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Xiaofei Zheng
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| |
Collapse
|
11
|
Moloudi K, Neshasteriz A, Hosseini A, Eyvazzadeh N, Shomali M, Eynali S, Mirzaei E, Azarnezhad A. Synergistic Effects of Arsenic Trioxide and Radiation: Triggering the Intrinsic Pathway of Apoptosis. IRANIAN BIOMEDICAL JOURNAL 2017; 21:330-7. [PMID: 28459147 PMCID: PMC5548965 DOI: 10.18869/acadpub.ibj.21.5.330] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 10/18/2016] [Accepted: 01/01/2017] [Indexed: 12/26/2022]
Abstract
Background Arsenic trioxide (ATO) has been reported as an effective anti-cancer and a US Food and Drug Administration (FDA) approved drug for treatment of some cancers. The aim of this study was to determine the underlying apoptosis molecular and cellular mechanisms of ATO in the presence or absence of ionizing radiation (IR) in vitro in the glioblastoma multiforme (GBM) cell line, U87MG. Methods Cells were treated by different concentrations of ATO either in presence or absence of IR. Viability and apoptosis pathway of both treated and control groups were evaluated using MTT assay and the expression analysis of Bax, Bcl-2, and caspase-3 genes, respectively. All treatments were performed on 100-μm diameter spheroids. Results Results showed a significant reduction in the survival of the cells in all treated groups. As expected, cell survival was much less in combination treatment than treatment with only ATO. Moreover, combination therapy made Bax and caspase-3 up-regulated and Bcl-2 down-regulated. Conclusion ATO and radiation had a synergistic apoptotic effect on GBM cells by up-regulation of caspase-3 and alteration of the Bax-Bcl-2 balance; therefore, ATO may act as a potential anti-cancer agent against GBM cells through triggering the mitochondrial pathway of apoptosis.
Collapse
Affiliation(s)
- Kave Moloudi
- Radiation Sciences Department, Faculty of allied Medicine school, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Neshasteriz
- Radiation Sciences Department, Faculty of allied Medicine school, Iran University of Medical Sciences, Tehran, Iran
| | - Arshad Hosseini
- Department of Medical Biotechnology, Faculty of allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nazila Eyvazzadeh
- Radiation Research Center, Faculty of Paramedicine, AJA University of Medical sciences, Tehran, Iran
| | - Mehdi Shomali
- Radiology Department, Faculty of allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Eynali
- Medical physics and Biomedical Engineering Department, school of Medicine, Tran University of Medical Sciences, Tehran, Iran
| | - Elahe Mirzaei
- Microbiology Department, Faculty of Science, Islamic Azad University, Tehran, Iran
| | - Asaad Azarnezhad
- Cellular & Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Ma S, Huang D, Zhai M, Yang L, Peng S, Chen C, Feng X, Weng Q, Zhang B, Xu M. Isolation of a novel bio-peptide from walnut residual protein inducing apoptosis and autophagy on cancer cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 15:413. [PMID: 26593407 PMCID: PMC4656182 DOI: 10.1186/s12906-015-0940-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 11/17/2015] [Indexed: 01/10/2023]
Abstract
Background Walnut is unique because they have a perfect balance of n-6 and n-3 polyunsaturated fatty acids. The increasing market demand of walnut lipids results in the large amount of the oil extraction residue. The walnut residue is rich in nutritional proteins, and the uneconomic use of the by-product discouraged the development of walnut industry. Anticancer peptides have recently received attention as alternative chemotherapeutic agents that overcome the limits of current drugs. The aim of this study was to investigate whether anticancer bioactive peptide is contained in walnut. Methods Walnut residual protein was hydrolyzed separately by five different proteases. The sequential purification of the hydrolysates was carried out by ultra-filtration, gel filtration chromatography and RP-HPLC to obtain a cancer cell growth inhibitory peptide. Cell cycle distribution, Annexin V-FITC/PI double staining, TUNEL assay, western blot and immunofluorescence for LC3-II assay were used to detect apoptosis and autophagy on cells. Cytokine production was measured by ELISA kits, macrophage phagocytosis was measured by neutral red uptake assay, nitric oxide production was measured by Griess reagent. Results The hydrolysates of walnut residual protein produced by papain under the optimal conditions (5 % substrate concentration and an enzyme-substrate ratio of 10 % at temperature 60 C for 3 h), showed significant growth inhibitory activity on MCF-7. The amino acid sequence of the purified peptide was identified as CTLEW with a molecular weight of 651.2795 Da. It is a novel bio-peptide with an amphiphilic structure. CTLEW induced both apoptosis and autophagy on MCF-7 cells, inhibited the cancer cells growth of Caco-2 and HeLa significantly, but did not show any cytotoxic activity against non-cancerous IEC-6 cells. Moreover, the bio-peptide enhanced proliferation and IL-2 secretion of spleen lymphocytes, promoted phagocytosis and NO production of macrophages. Conclusion These results suggested that a novel bio-peptide, CTLEW inducing apoptosis and autophagy on MCF-7 cells can be released from walnut residual protein through papain hydrolyzing under the certain condition. The bio-peptide shows selective inhibition towards cancer cells growth and immunomodulatory activity.
Collapse
|
13
|
Ciarcia R, Damiano S, Puzio MV, Montagnaro S, Pagnini F, Pacilio C, Caparrotti G, Bellan C, Garofano T, Polito MS, Giordano A, Florio S. Comparison of Dasatinib, Nilotinib, and Imatinib in the Treatment of Chronic Myeloid Leukemia. J Cell Physiol 2015; 231:680-7. [PMID: 26235483 DOI: 10.1002/jcp.25118] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 07/30/2015] [Indexed: 11/06/2022]
Abstract
To overcome the drug resistance phenomenon induced by Imatibib (IM), in clinical practice, are often used second generation of tyrosine kinase inhibitors as Nilotinib (NIL); a such potent inhibitor of the BCR/ABL kinase and Dasatinib (DAS), a inhibitor of BCR/ABL kinase, and inhibitor SrC family kinase. In this study we evaluated the in vivo effect of DAS, NIL, and IM on intracellular calcium concentration, oxidative stress, and apoptosis in peripheral blood leukocytes of 45 newly diagnosed patients with chronic myeloid leukaemia (CML-PBM). Our data demonstrated that treatment with DAS and NIL showed an higher modulating potential than IM on intracellular calcium concentration by inhibiting the thapsigargin, a sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) inhibitor, and Lithium (Li) an inositol 1,4,5-triphosphate (InsP3) receptor inhibitor activities. Moreover our data demonstrated that NIL and DAS have significantly increased apoptosis more than IM by involving both intracellular calcium signaling as well as oxidative stress. The acquisition of the oxidative stress and calcium channels receptors values data could help the hematologist to modulate and improve the treatment of chronic myeloid leukaemia (CML) pathology.
Collapse
Affiliation(s)
- Roberto Ciarcia
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Sara Damiano
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Maria Valeria Puzio
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Serena Montagnaro
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Francesco Pagnini
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| | - Carmen Pacilio
- Clinical Trials Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione G. Pascale-IRCCS, Naples, Italy
| | - Giuseppe Caparrotti
- Department of Haematology, ASL Caserta (CE), Hospital Moscati, Aversa (CE), Italy
| | - Cristiana Bellan
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | | | - Maria Sole Polito
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Antonio Giordano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy.,AORN Dei Colli Monaldi UOC, Oncology, Naples, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Salvatore Florio
- Department of Veterinary Medicine and Animal Productions, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
14
|
Meng G, Liu J, Lin S, Guo Z, Xu L. Microcystin-LR-caused ROS generation involved in p38 activation and tau hyperphosphorylation in neuroendocrine (PC12) cells. ENVIRONMENTAL TOXICOLOGY 2015; 30:366-374. [PMID: 24142891 DOI: 10.1002/tox.21914] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 09/23/2013] [Accepted: 10/02/2013] [Indexed: 06/02/2023]
Abstract
Microcystin-LR (MC-LR), a potent specific hepatotoxin produced by cyanobacteria, has recently been reported to show neurotoxicity. Our previous study demonstrated that MC-LR caused the reorganization of cytoskeleton architectures and hyperphosphorylation of the cytoskeletal-associated proteins tau and HSP27 in neuroendocrine PC12 cell line by direct PP2A inhibition and indirect p38 mitogen-activated protein kinase (MAPK) activation. It has been shown that oxidative stress is extensively associated with MC-LR toxicity, mainly resulting from an excessive production of reactive oxygen species (ROS). However, the mechanisms by which ROS mediates the cytotoxic action of MC-LR are unclear. In the present study, we investigated whether ROS might play a critical role in MC-LR-induced hyperphosphorylation of microtubule-associated protein tau and the activation of the MAPKs in PC12 cell line. The results showed that MC-LR had time- and concentration-dependent effects on ROS generation, p38-MAPK activation and tau phosphorylation. The time-course studies indicated similar biphasic changes in ROS generation and tau hyperphosphorylation, which started to increase within 1 h and reached the maximum level at 3 h followed by a decrease after prolonged treatment. Furthermore, pretreatment with the antioxidants, N-acetylcysteine and vitamin C, significantly decreased MC-LR-induced ROS generation and effectively attenuated p38-MAPK activation as well as tau hyperphosphorylation. Taken together, these findings suggest that ROS generation triggered by MC-LR is a key intracellular event that contributes to an induction of p38-MAPK activation and tau phosphorylation, and that blockade of this ROS-mediated redox-sensitive signal cascades may attenuate the toxic effects of MC-LR.
Collapse
Affiliation(s)
- Guanmin Meng
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, 234 Gucui Road, Hangzhou, 310012, China; Department of Biochemistry, School of Medicine, Zhejiang University, 866th Yu Hang Tang Road, Hangzhou, 310058, China
| | | | | | | | | |
Collapse
|
15
|
Luo DL, Yang D, Shu ML, Deng MM. Anticancer mechanisms of resveratrol in liver cancer. Shijie Huaren Xiaohua Zazhi 2014; 22:4769-4773. [DOI: 10.11569/wcjd.v22.i31.4769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Primary liver cancer is one of the most common malignant tumors. Because primary liver cancer has a high degree of malignancy and the early diagnosis is very difficult, it has a very poor prognosis and is associated with high mortality. Resveratrol is found in a variety of natural plants. Recent studies found that resveratrol has significant effects against liver cancer cells in vivo without obvious side effect, so it may become one of the most promising anticancer agents. This article will review the advances in understanding the anticancer mechanisms of resveratrol in liver cancer.
Collapse
|
16
|
Pyrroloquinoline quinone protects mouse brain endothelial cells from high glucose-induced damage in vitro. Acta Pharmacol Sin 2014; 35:1402-10. [PMID: 25283505 DOI: 10.1038/aps.2014.4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 01/14/2014] [Indexed: 12/31/2022]
Abstract
AIM To investigate the effects of pyrroloquinoline quinone (PQQ), an oxidoreductase cofactor, on high glucose-induced mouse endothelial cell damage in vitro. METHODS Mouse brain microvascular endothelial bEND.3 cells were exposed to different glucose concentrations (5.56, 25 and 40 mmol/L) for 24 or 48 h. The cell viability was examined using MTT assay. Flow cytometry was used to analyze the apoptosis and ROS levels in the cells. MitoTracker Green staining was used to examine the mitochondria numbers in the cells. Western blot analysis was used to analyze the expression of HIF-1α and the proteins in JNK pathway. RESULTS Treatment of bEND.3 cells with high glucose significantly decreased the cell viability, while addition of PQQ (1 and 10 μmol/L) reversed the high glucose-induced cell damage in a concentration-dependent manner. Furthermore, PQQ (100 μmol/L) significantly suppressed the high glucose-induced apoptosis and ROS production in the cells. PQQ significantly reversed the high glucose-induced reduction in both the mitochondrial membrane potential and mitochondria number in the cells. The high glucose treatment significantly increased the expression of HIF-1α and JNK phosphorylation in the cells, and addition of PQQ led to a further increase of HIF-1α level and a decrease of JNK phosphorylation. Addition of JNK inhibitor SP600125 (10 μmol/L) also significantly suppressed high glucose-induced apoptosis and JNK phosphorylation in bEND.3 cells. CONCLUSION PQQ protects mouse brain endothelial cells from high glucose damage in vitro by suppressing intracellular ROS and apoptosis via inhibiting JNK signaling pathway.
Collapse
|
17
|
Ghani S, Khan N, Koriyama C, Akiba S, Yamamoto M. N‑acetyl‑L‑cysteine reduces arsenite‑induced cytotoxicity through chelation in U937 monocytes and macrophages. Mol Med Rep 2014; 10:2961-6. [PMID: 25310083 DOI: 10.3892/mmr.2014.2612] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 05/14/2014] [Indexed: 11/06/2022] Open
Abstract
In the present study, in order to clarify the preventive mechanism of N‑acetyl‑L‑cysteine (NAC) on arsenite‑induced apoptosis in U937 cells, which lack functional p53, the cytotoxicity among U937 cells [monocytes and 12‑O‑tetradecanoylphorbol‑13‑acetate (TPA)‑treated macrophages] receiving NAC treatment at different times post arsenite treatment was examined. TPA‑treated macrophages were more resistant to arsenite‑induced apoptosis than monocytes, which may be associated with the induction of Bcl‑2 expression. Pretreatment with 20 mM NAC prior to arsenite exposure suppressed apoptosis up to 75% in the monocytes and 100% in the macrophages. However, 6‑h NAC pretreatment and subsequent washing out of NAC from the culture medium prior to arsenite treatment did not inhibit the arsenite‑induced apoptosis. Post‑treatment by NAC up to 1 h following arsenite exposure almost completely inhibited the cytotoxic effects of arsenite in U937 monocytes and macrophages. The results of the current study indicate that the preventive mechanism of NAC on arsenite‑induced apoptosis in U937 monocytes and macrophages mainly involves chelation of arsenite in culture medium.
Collapse
Affiliation(s)
- Sidra Ghani
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima 890‑8544, Japan
| | - Noureen Khan
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima 890‑8544, Japan
| | - Chihaya Koriyama
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima 890‑8544, Japan
| | - Suminori Akiba
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Kagoshima 890‑8544, Japan
| | - Megumi Yamamoto
- Integrated Physiology Section, Department of Basic Medical Science, National Institute for Minamata Disease, Minamata, Kumamoto 867‑0008, Japan
| |
Collapse
|
18
|
Yu R, Wang D, Ren X, Zeng L, Liu Y. The growth-inhibitory and apoptosis-inducing effect of deferoxamine combined with arsenic trioxide on HL-60 xenografts in nude mice. Leuk Res 2014; 38:1085-90. [PMID: 24908354 DOI: 10.1016/j.leukres.2014.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 04/27/2014] [Accepted: 05/03/2014] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The aim of this study is to investigate the growth-inhibitory and apoptosis-inducing effect of deferoxamine (DFO) combined with arsenic trioxide (ATO) on the human HL-60 xenografts in nude mice and its mechanism. METHOD The highly tumorigenic leukemia cell line HL-60 cells were inoculated subcutaneously into nude mice to establish a human leukemia xenograft model. The HL-60 xenograft nude mice models were randomly divided into four groups: control (Normal saline, NS), 50mg/kg DFO, 3mg/kg ATO, the combined treatment (50mg/kg DFO+1.5mg/kg ATO) once HL-60 cells were inoculated. Tumor sizes, growth curves, inhibitory rates, cell apoptosis, and the expression of apoptosis related markers were measured to evaluate the tumor growth. RESULTS Xenografted tumors were observed in all nude mice since the 5th day of inoculation. The inhibitory rates of tumor weight were 2.67%, 10.69%, and 25.57% in DFO, ATO and combination therapy groups, respectively. The combination of DFO with ATO induces significantly more tumor cell apoptosis than either agent alone (p<0.05). The expression of NF-κBp65 and survivin proteins decreased significantly while the expression of Caspase-3 and Bax increased in the combination therapy group (p<0.05). Double immunofluorescence for Caspase-3 and NFκBp65 demonstrated an inverse relationship between Caspase-3-positive areas and NFκBp65-positive areas, as well as the co-localization of Bax and survivin in xenografted tumor cells. CONCLUSIONS Combination of DFO and ATO has synergistic effects on tumor growth inhibition and apoptosis-inducing in vivo with no significant side effects. The DFO and ATO can up-regulate the expression of Caspase-3 and Bax, and down-regulate the expression of NF-κBp65 and survivin, especially for their combination.
Collapse
Affiliation(s)
- Runhong Yu
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Key Discipline Laboratory of Clinical Medicine, Zhengzhou 450052, China
| | - Dao Wang
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Key Discipline Laboratory of Clinical Medicine, Zhengzhou 450052, China
| | - Xiuhua Ren
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Li Zeng
- Department of Laboratory Medicine, Zhengzhou University, Zhengzhou 450052, China
| | - Yufeng Liu
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou University, Key Discipline Laboratory of Clinical Medicine, Zhengzhou 450052, China.
| |
Collapse
|
19
|
Nuta O, Moquet J, Bouffler S, Lloyd D, Sepai O, Rothkamm K. Impact of long-term exposure to sodium arsenite on cytogenetic radiation damage. Mutagenesis 2014; 29:123-9. [PMID: 24452505 DOI: 10.1093/mutage/get070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The aim of this work was to investigate the impact of long-term exposure to low concentrations of sodium arsenite on the cellular response to ionising radiation. Human lymphoblastoid GM1899a cells were cultured in the presence of sodium arsenite for up to six months. Following chemical exposure, acute challenge doses of X-rays were given and chromosome damage (dicentrics, acentric fragments, translocations, micronuclei) as well as cell growth and changes in cell cycle kinetics were determined. Initial short-term chemical exposures determined 8 ng/ml (60 nM) sodium arsenite as a suitable concentration for chronic exposures, which is below the current World Health Organization limit for arsenic in drinking water. At this concentration, cell growth was slightly, but consistently, slower than in untreated cultures throughout the six-month exposure period. Long-term exposure to the chemical induced no dicentrics and did not significantly alter the yield of dicentrics induced by 1 Gy acute X-irradiation. Similar results were obtained for chromosome translocations. In contrast, exposure to 8 ng/ml sodium arsenite induced significant levels of acentric fragments and micronuclei. Fragment/micronuclei data in combined treatment samples compared with single treatments were consistent with an additive effect of chemical and radiation exposure. As for X-rays, micronuclei induced by sodium arsenite tended to show no centromere in situ hybridisation signal, indicating that they represent structural aberrations rather than mis-segregated chromosomes. Similar results were obtained in human peripheral lymphocytes following short-term exposure to sodium arsenite or X-rays. Overall, an additive effect was observed for all combined exposures. Cellular radiation responses therefore seem to operate without any modulatory effects from chronic low level exposure to sodium arsenite in the systems analysed here.
Collapse
Affiliation(s)
- Otilia Nuta
- Public Health England, Centre for Radiation, Chemical and Environmental Hazards, Chilton, Didcot, Oxon, OX11 0RQ, UK
| | | | | | | | | | | |
Collapse
|
20
|
Nandakumar V, Vettriselvi V, Doble M. Toxicity of high glycolic poly(dl-lactic-co-glycolic acid) stabilized ruthenium nanoparticles against human promyelocytic leukemia cells. RSC Adv 2014. [DOI: 10.1039/c3ra47142j] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
21
|
Liu X, Gao Y, Yao H, Zhou L, Pei J, Sun L, Wang J, Sun D. p38 and Extracellular Signal-Regulated Kinases Activations have Opposite Effects on Primary-Cultured Rat Cerebellar Granule Neurons Exposed to Sodium Arsenite. J Biochem Mol Toxicol 2013; 28:143-8. [DOI: 10.1002/jbt.21546] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 11/06/2013] [Accepted: 11/24/2013] [Indexed: 12/20/2022]
Affiliation(s)
- Xiaona Liu
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| | - Yanhui Gao
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| | - Hongju Yao
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| | - Lingwang Zhou
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| | - Junrui Pei
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| | - Liyan Sun
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| | - Jing Wang
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| | - Dianjun Sun
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| |
Collapse
|
22
|
Liu X, Gao Y, Yao H, Zhou L, Sun D, Wang J. Neuroglobin involvement in the course of arsenic toxicity in rat cerebellar granule neurons. Biol Trace Elem Res 2013; 155:439-46. [PMID: 24057451 DOI: 10.1007/s12011-013-9810-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 08/29/2013] [Indexed: 12/16/2022]
Abstract
Exposure to arsenic in drinking water results in a widespread environmental problem in the world, and the brain is a major target. Neuroglobin is a vertebrate heme protein regarded as playing neuroprotective role in hypoxia or oxidative stress. In this study, we investigated the toxic effects of sodium arsenite (NaAsO2) on primary cultured rat cerebellar granule neurons (CGNs) and detected neuroglobin (Ngb) expression in rat CGNs exposed to NaAsO2. Our results show that apoptosis was obviously induced by NaAsO2 treatment in rat CGNs by annexin V-fluorescein isothiocyanate assay. Intracellular reactive oxygen species generation increased significantly in the cells exposed to NaAsO2, and the apoptotic effects could be partially reversed by antioxidant N-acetyl-L-cysteine. Ngb protein and mRNA expression were significantly downregulated in rat CGNs shortly after NaAsO2 exposure and then upregulated after a longer time of exposure. Furthermore, mRNA expression changed more than protein expression and the toxic effect of NaAsO2 on Ngb expression is dose dependent. Higher Ngb expression was also detected in rat cerebellum, but not in other parts (cerebrum, hippocampus, and midbrain) of the brain exposed to NaAsO2 for 16 weeks. Taken together, cytotoxic effects of NaAsO2 on rat CGNs is induced at least partly by oxidative stress and Ngb may influence the course of arsenic toxicity in rat CGNs and rat cerebellum.
Collapse
Affiliation(s)
- Xiaona Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618104), Harbin Medical University, 157# Baojian Road, Harbin, 150081, People's Republic of China
| | | | | | | | | | | |
Collapse
|
23
|
Wang L, Zhang XM, Li Z, Liu XJ, Chai J, Zhang GY, Cheng YF. Overexpression of nuclear β-catenin in rectal adenocarcinoma is associated with radioresistance. World J Gastroenterol 2013; 19:6876-6882. [PMID: 24187464 PMCID: PMC3812488 DOI: 10.3748/wjg.v19.i40.6876] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 07/29/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the association between nuclear β-catenin overexpression in rectal adenocarcinoma and radioresistance.
METHODS: A retrospective analysis was conducted. The analysis involved 136 patients with locally advanced rectal adenocarcinoma who underwent short-course preoperative radiotherapy and radical resection. The expression of β-catenin in both pretreatment biopsy specimens and resected primary tumor tissues was examined by immunohistochemistry. The correlation of β-catenin expression with radioresistance was evaluated using the tumor regression grading (TRG) system. The relationship between β-catenin expression and clinicopathological characteristics was also analyzed. Univariate and logistic multivariate regression analyses were adopted to determine the independent factors of radioresistance.
RESULTS: Nuclear β-catenin overexpression was more evident in radioresistant rectal adenocarcinoma than in radiosensitive rectal adenocarcinoma (57.6% vs 16.7%, P < 0.001). Nuclear β-catenin was overexpressed in favor of poor TRG (≤ 2), whereas membrane β-catenin was expressed in favor of good TRG (≥ 3). Nuclear β-catenin expression in tumor cell differentiation (P = 0.018), lymph node metastasis (P = 0.022), and TRG (P < 0.001) showed significant differences. Univariate analyses demonstrated that radioresistance is associated with nuclear β-catenin overexpression (P < 0.001). In addition, logistic multivariate regression analysis indicated that only three factors, namely, tumor size (P < 0.001), tumor cell differentiation (P < 0.001), and nuclear β-catenin overexpression (P < 0.001), are associated with radioresistance. By using radioresistance as a prediction target, nuclear β-catenin-based prediction alone achieved 83% accuracy, 65% sensitivity, and 88% specificity.
CONCLUSION: Nuclear β-catenin overexpression may be a valuable candidate to predict the response of rectal adenocarcinoma to preoperative radiotherapy.
Collapse
|
24
|
Chowdhury AA, Chaudhuri J, Biswas N, Manna A, Chatterjee S, Mahato SK, Chaudhuri U, Jaisankar P, Bandyopadhyay S. Synergistic apoptosis of CML cells by buthionine sulfoximine and hydroxychavicol correlates with activation of AIF and GSH-ROS-JNK-ERK-iNOS pathway. PLoS One 2013; 8:e73672. [PMID: 24040019 PMCID: PMC3767640 DOI: 10.1371/journal.pone.0073672] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 07/22/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Hydroxychavicol (HCH), a constituent of Piper betle leaf has been reported to exert anti-leukemic activity through induction of reactive oxygen species (ROS). The aim of the study is to optimize the oxidative stress -induced chronic myeloid leukemic (CML) cell death by combining glutathione synthesis inhibitor, buthionine sulfoximine (BSO) with HCH and studying the underlying mechanism. MATERIALS AND METHODS Anti-proliferative activity of BSO and HCH alone or in combination against a number of leukemic (K562, KCL22, KU812, U937, Molt4), non-leukemic (A549, MIA-PaCa2, PC-3, HepG2) cancer cell lines and normal cell lines (NIH3T3, Vero) was measured by MTT assay. Apoptotic activity in CML cell line K562 was detected by flow cytometry (FCM) after staining with annexin V-FITC/propidium iodide (PI), detection of reduced mitochondrial membrane potential after staining with JC-1, cleavage of caspase- 3 and poly (ADP)-ribose polymerase proteins by western blot analysis and translocation of apoptosis inducing factor (AIF) by confocal microscopy. Intracellular reduced glutathione (GSH) was measured by colorimetric assay using GSH assay kit. 2',7'-dichlorodihydrofluorescein diacetate (DCF-DA) and 4-amino-5-methylamino-2',7'-difluorofluorescein (DAF-FM) were used as probes to measure intracellular increase in ROS and nitric oxide (NO) levels respectively. Multiple techniques like siRNA transfection and pharmacological inhibition were used to understand the mechanisms of action. RESULTS Non-apoptotic concentrations of BSO significantly potentiated HCH-induced apoptosis in K562 cells. BSO potentiated apoptosis-inducing activity of HCH in CML cells by caspase-dependent as well as caspase-independent but apoptosis inducing factor (AIF)-dependent manner. Enhanced depletion of intracellular GSH induced by combined treatment correlated with induction of ROS. Activation of ROS- dependent JNK played a crucial role in ERK1/2 activation which subsequently induced the expression of inducible nitric oxide synthase (iNOS). iNOS- mediated production of NO was identified as an effector molecule causing apoptosis of CML cells. CONCLUSION/SIGNIFICANCE BSO synergizes with HCH in inducing apoptosis of CML cells through the GSH-ROS-JNK-ERK-iNOS pathway.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis Inducing Factor/metabolism
- Blotting, Western
- Buthionine Sulfoximine/pharmacology
- Cell Line, Tumor
- Chlorocebus aethiops
- Drug Synergism
- Eugenol/analogs & derivatives
- Eugenol/pharmacology
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Glutathione/metabolism
- Hep G2 Cells
- Humans
- JNK Mitogen-Activated Protein Kinases/metabolism
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mice
- Microscopy, Confocal
- NIH 3T3 Cells
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type II/metabolism
- Reactive Oxygen Species/metabolism
- Signal Transduction/drug effects
- U937 Cells
- Vero Cells
Collapse
Affiliation(s)
- Avik Acharya Chowdhury
- Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB), Kolkata, India
| | - Jaydeep Chaudhuri
- Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB), Kolkata, India
| | - Nabendu Biswas
- Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB), Kolkata, India
| | - Anirban Manna
- Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB), Kolkata, India
| | | | | | - Utpal Chaudhuri
- The Institute of Hematology and Transfusion Medicine, Medical College, Kolkata, India
| | | | - Santu Bandyopadhyay
- Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB), Kolkata, India
| |
Collapse
|
25
|
Kim KA, Lee SA, Kim KH, Lee KS, Lee JC. Acteoside inhibits irradiation-mediated decreases in the viability and DNA synthesis of MC3T3-E1 cells. Food Sci Biotechnol 2013. [DOI: 10.1007/s10068-013-0154-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
26
|
Hu XM, Yuan B, Tanaka S, Zhou Q, Onda K, Toyoda H, Hirano T. Involvement of oxidative stress associated with glutathione depletion and p38 mitogen-activated protein kinase activation in arsenic disulfide-induced differentiation in HL-60 cells. Leuk Lymphoma 2013; 55:392-404. [DOI: 10.3109/10428194.2013.802779] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
27
|
The synthetic flavonoid WYC02-9 inhibits colorectal cancer cell growth through ROS-mediated activation of MAPK14 pathway. Life Sci 2013; 92:1081-92. [PMID: 23624232 DOI: 10.1016/j.lfs.2013.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Revised: 03/11/2013] [Accepted: 04/16/2013] [Indexed: 12/12/2022]
Abstract
AIM Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide. In this study, we explored the anti-cancer activity of WYC02-9, a synthetic protoapigenone, on human HCT116 CRC cells. MAIN METHODS The anti-cancer activity of WYC02-9 and its underlying mechanisms were analyzed using XTT cell proliferation assays, colony formation assays, FACS analysis, annexin V staining, immunoblotting analysis, reactive oxygen species (ROS) generation assays, soft agar assays, a nude mice xenograft study and immunohistochemistry assays. KEY FINDINGS Data showed that WYC02-9 suppressed CRC cell growth by arresting cells at G2/M and inducing cell death via apoptotic pathways. Further analysis demonstrated that WYC02-9-induced apoptosis was mediated by the activation of a ROS-mediated MAPK14 pathway. An in vivo xenograft study revealed that WYC02-9 enhanced MAP2K3/6 and MAPK14 phosphorylation, induced apoptosis, and suppressed CRC tumor growth. SIGNIFICANCE WYC02-9 exerts its anti-tumor effect via ROS/MAPK14-induced apoptosis and has the potential to be developed as a chemotherapeutic agent for CRC.
Collapse
|
28
|
Qiu J, Yang G, Shen Z, Xie Y, Wang L. hPEBP4 as a predictive marker for the pathological response of rectal cancer to preoperative radiotherapy. Int J Colorectal Dis 2013; 28:241-6. [PMID: 22801881 DOI: 10.1007/s00384-012-1534-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/01/2012] [Indexed: 02/04/2023]
Abstract
PURPOSE The ability to predict tumor sensitivity toward radiotherapy is important in the personalized application of preoperative radiotherapy for patients with rectal cancer. The aim of the present study was to test the human phosphatidylethanolamine-binding protein 4 (hPEBP4) as a predictive marker of tumor response to preoperative radiotherapy in patients with rectal cancer. METHOD A retrospective analysis was conducted, consisting of 86 patients with locally advanced rectal cancer who underwent short-course preoperative radiotherapy (20 Gy in five fractions for 1 week) followed by a radical resection. Both pretreatment biopsy specimens and resected primary tumor tissue were collected. Immunohistochemistry and tumor regression grading system were used to evaluate the expression of hPEBP4 in the pretreatment biopsy specimens and the response of rectal cancer to radiotherapy, respectively. Expression of hPEBP4 was correlated with tumor regression in the resected specimen and the clinical outcome of the patients as well. RESULTS We found that high expression of hPEBP4 was associated with radioresistance in both univariate and multivariate analyses, and patients with a high hPEBP4 expression had a poorer progression-free survival than those with low hPEBP4 expression. CONCLUSION Our study revealed the independent predictive values of hPEBP4 in response of rectal cancer to preoperative radiotherapy, which suggests that upregulating hPEBP4 might be a potential mechanism by which rectal cancer cells avoid the destructive effects of radiotherapy.
Collapse
Affiliation(s)
- Jianming Qiu
- Department of Colorectal Surgery, The Third People's Hospital of Hangzhou, Hangzhou, China.
| | | | | | | | | |
Collapse
|
29
|
Main-Group Medicinal Chemistry Including Li and Bi*. COMPREHENSIVE INORGANIC CHEMISTRY II 2013. [PMCID: PMC7152213 DOI: 10.1016/b978-0-08-097774-4.00338-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Main-group element compounds were among the first developed in the modern era as pharmaceutical preparations for the treatment of a wide variety of human ailments; it is now recognized that many of these elements exist in traditional medicine of many societies, for example, arsenic. The use of main-group element compounds in contemporary medicine continues for the treatment of, for example, depression (Li), stomach ulcers (Bi), cancer (As and Ga), and leishmaniasis (Sb). Not surprisingly, new compounds of these elements, and other main-group elements, continue to be investigated for their potential use in new therapies. In this chapter, the use of main-group elements as therapeutic agents is outlined and also, where understood, comments on biological targets and mechanisms of action. Further, key advances in new potential applications of main-group element compounds in medicine are evaluated.
Collapse
|
30
|
Galimberti S, Guerrini F, Salvi F, Petrini I, Gioia D, Messa E, Palumbo GA, Cilloni D, Petrini M, Levis A. Arsenic trioxide and ascorbic acid interfere with the BCL2 family genes in patients with myelodysplastic syndromes: an ex-vivo study. J Hematol Oncol 2012; 5:53. [PMID: 22964015 PMCID: PMC3465246 DOI: 10.1186/1756-8722-5-53] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 08/27/2012] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Arsenic Trioxide (ATO) is effective in about 20% of patients with myelodysplasia (MDS); its mechanisms of action have already been evaluated in vitro, but the in vivo activity is still not fully understood. Since ATO induces apoptosis in in vitro models, we compared the expression of 93 apoptotic genes in patients' bone marrow before and after ATO treatment. For this analysis, we selected 12 patients affected by MDS who received ATO in combination with Ascorbic Acid in the context of the Italian clinical trial NCT00803530, EudracT Number 2005-001321-28. METHODS Real-time PCR quantitative assays for genes involved in apoptosis were performed using TaqMan® Assays in 384-Well Microfluidic Cards "TaqMan® Human Apoptosis Array".Quantitative RT-PCR for expression of EVI1 and WT1 genes was also performed. Gene expression values (Ct) were normalized to the median expression of 3 housekeeping genes present in the card (18S, ACTB and GAPDH). RESULTS ATO treatment induced up-regulation of some pro-apoptotic genes, such as HRK, BAK1, CASPASE-5, BAD, TNFRSF1A, and BCL2L14 and down-regulation of ICEBERG. In the majority of cases with stable disease, apoptotic gene expression profile did not change, whereas in cases with advanced MDS more frequently pro-apoptotic genes were up-regulated. Two patients achieved a major response: in the patient with refractory anemia the treatment down-regulated 69% of the pro-apoptotic genes, whereas 91% of the pro-apoptotic genes were up-regulated in the patient affected by refractory anemia with excess of blasts-1. Responsive patients showed a higher induction of BAD than those with stable disease. Finally, WT1 gene expression was down-regulated by the treatment in responsive cases. CONCLUSIONS These results represent the basis for a possible association of ATO with other biological compounds able to modify the apoptotic pathways, such as inhibitors of the BCL2 family.
Collapse
Affiliation(s)
- Sara Galimberti
- Department of Oncology, Transplant, New Advances in Medicine, Section of Hematology, University of Pisa, Pisa, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Arsenic trioxide inhibits Ewing's sarcoma cell invasiveness by targeting p38(MAPK) and c-Jun N-terminal kinase. Anticancer Drugs 2012; 23:108-18. [PMID: 21946058 DOI: 10.1097/cad.0b013e32834bfd68] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Ewing's sarcoma is the second most frequent primary malignant bone tumor, mainly affecting children and young adults. The notorious metastatic capability of this tumor aggravates patient mortality and remains a problem to be overcome. We investigated the effect of arsenic trioxide (As₂O₃) on the metastasis capability of Ewing's sarcoma cells. We performed 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyl-2H-tetrazolium bromide assays to choose appropriate concentrations of As₂O₃ for the experiments. Migration, invasion, and adhesion assays were performed to assess the effect of As₂O₃ on the metastasis of Ewing's sarcoma. Immunofluorescent staining was used to observe cytoskeleton reorganization in Ewing's sarcoma cells treated with As₂O₃. Changes in matrix metalloproteinase-9 expression and the mitogen-activated protein kinase (MAPK) pathway were investigated using western blot. Inhibitors of p38(MAPK) (sb202190) and c-Jun NH₂-terminal kinase (JNK, sp600125) were used in invasion assays to determine the effect of p38(MAPK) and JNK. We found that As₂O₃ may markedly inhibit the migration and invasion capacity of Ewing's sarcoma cells with structural rearrangements of the actin cytoskeleton. The expressions of matrix metalloproteinase-9, phosphor-p38(MAPK), and phosphor-JNK were suppressed by As₂O₃ treatment in a dose-dependent manner. The inhibitors of p38(MAPK) (sb202190) and JNK (sp600125) enhanced the inhibition induced by As₂O₃, which was counteracted by anisomycin, an activating agent of p38(MAPK) and JNK. Taken together, our results demonstrate that As₂O₃ can inhibit the metastasis capability of RD-ES and A-673 cells and may have new therapeutic value for Ewing's sarcoma.
Collapse
|
32
|
Health and cellular impacts of air pollutants: from cytoprotection to cytotoxicity. Biochem Res Int 2012; 2012:493894. [PMID: 22550588 PMCID: PMC3328890 DOI: 10.1155/2012/493894] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 01/17/2012] [Accepted: 01/18/2012] [Indexed: 12/11/2022] Open
Abstract
Air pollution as one of the ravages of our modern societies is primarily linked to urban centers, industrial activities, or road traffic. These atmospheric pollutants have been incriminated in deleterious health effects by numerous epidemiological and in vitro studies. Environmental air pollutants are a heterogeneous mixture of particles suspended into a liquid and gaseous phase which trigger the disruption of redox homeostasis—known under the term of cellular oxidative stress—in relation with the establishment of inflammation and cell death via necrosis, apoptosis, or autophagy. Activation or repression of the apoptotic process as an adaptative response to xenobiotics might lead to either acute or chronic toxicity. The purpose of this paper is to highlight the central role of oxidative stress induced by air pollutants and to focus on the subsequent cellular impacts ranging from cytoprotection to cytotoxicity by decreasing or stimulating apoptosis, respectively.
Collapse
|
33
|
Qiu J, Zhu G, Chen X, Shao C, Gu S. Combined effects of γ-irradiation and cadmium exposures on osteoblasts in vitro. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2012; 33:149-157. [PMID: 22209727 DOI: 10.1016/j.etap.2011.12.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 11/09/2011] [Accepted: 12/05/2011] [Indexed: 05/31/2023]
Abstract
The combined effects of γ-irradiation and cadmium (Cd) exposures on osteoblasts were observed in the present study. Osteoblasts were exposed to γ-irradiation (0.5 Gy) and Cd (0-0.5 μmol/L). Cell viability, alkaline phosphatase (ALP) activity, mineralization ability, cell apoptosis and genes expression of ALP, osteocalcin (OC) and caspase 3 were observed. Low concentrations of Cd exposure had no obvious influence on cell viability, ALP activity and apoptosis. However, low levels of Cd exposure combined with γ-irradiation induced more toxic effects on osteoblasts than those treated with Cd or irradiation alone. High concentrations of Cd combined with irradiation exposure induced more significant inhibition in cell viability, ALP activity and mineralization ability than those exposed to Cd or irradiation alone. Meanwhile, OC and ALP mRNA expression of cells treated with Cd combined with irradiation were down-regulated more significantly than those treated with Cd or irradiation alone. Cd combined with γ-irradiation could obviously enhance osteoblast apoptosis and up-regulated caspase 3 mRNA expression compared with those treated with Cd or irradiation alone. This study indicated that ionizing irradiation can enhance Cd toxic effects on osteoblast viability and differentiation and apoptosis may play an important role in this progress.
Collapse
Affiliation(s)
- Jing Qiu
- Department of Environmental Epidemiology and Bone Toxicology, Institute of Radiation Medicine, Fudan University, China
| | | | | | | | | |
Collapse
|
34
|
Zhai JX, Zhang ZX, Feng YJ, Ding SS, Wang XH, Zou LW, Ye DQ. PDTC attenuate LPS-induced kidney injury in systemic lupus erythematosus-prone MRL/lpr Mice. Mol Biol Rep 2012; 39:6763-71. [DOI: 10.1007/s11033-012-1501-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 01/24/2012] [Indexed: 02/06/2023]
|