101
|
Yalçın E, Uzun A, Çavuşoğlu K. In vivo epiclorohidrine toxicity: cytogenetic, biochemical, physiological, and anatomical evidences. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:22400-22406. [PMID: 31154645 DOI: 10.1007/s11356-019-05518-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/17/2019] [Indexed: 06/09/2023]
Abstract
In this study, the toxic effects of epiclorohidrine (ECh) were investigated in vivo by Allium test. The toxic effects have been investigated in terms of physiological, cytogenetic, anatomical, and biochemical aspects. The changes in germination percentage, weight gain, and root length were investigated as physiological parameter; micronucleus (MN), mitotic index (MI), and chromosomal abnormality (CA) frequencies were as cytogenetic parameter. Oxidative stress indicators such as superoxide dismutase (SOD), catalase (CAT), and malondialdehyde (MDA) were analyzed for biochemical changes and also damages in root tip cells were evaluated as anatomical parameter. It was determined that germination percentage, weight gain, root length, and MI decreased; MN and CA frequencies were increased with the increase of ECh treatment dose. ECh treatment caused significant increase in SOD and CAT enzyme activities and MDA levels and these results indicated a stress formation. A variety of anatomical changes and damages were observed in the root tip cells induced by ECh. In conclusion, the toxic effects of ECh on A. cepa which is a model of eucaryotic cell were investigated in a multi-directional way and serious toxic effects of ECh treatment were determined.
Collapse
Affiliation(s)
- Emine Yalçın
- Science and Art Faculty, Departmant of Biology, Giresun University, Giresun, Turkey.
| | - Aytül Uzun
- Science and Technology Application and Research Center, Yozgat Bozok University, Yozgat, Turkey
| | - Kültiğin Çavuşoğlu
- Science and Art Faculty, Departmant of Biology, Giresun University, Giresun, Turkey
| |
Collapse
|
102
|
Hahn RZ, Antunes MV, Verza SG, Perassolo MS, Suyenaga ES, Schwartsmann G, Linden R. Pharmacokinetic and Pharmacogenetic Markers of Irinotecan Toxicity. Curr Med Chem 2019; 26:2085-2107. [PMID: 29932028 DOI: 10.2174/0929867325666180622141101] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 06/04/2018] [Accepted: 06/06/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Irinotecan (IRI) is a widely used chemotherapeutic drug, mostly used for first-line treatment of colorectal and pancreatic cancer. IRI doses are usually established based on patient's body surface area, an approach associated with large inter-individual variability in drug exposure and high incidence of severe toxicity. Toxic and therapeutic effects of IRI are also due to its active metabolite SN-38, reported to be up to 100 times more cytotoxic than IRI. SN-38 is detoxified by the formation of SN-38 glucuronide, through UGT1A1. Genetic polymorphisms in the UGT1A1 gene are associated to higher exposures to SN-38 and severe toxicity. Pharmacokinetic models to describe IRI and SN-38 kinetic profiles are available, with few studies exploring pharmacokinetic and pharmacogenetic-based dose individualization. The aim of this manuscript is to review the available evidence supporting pharmacogenetic and pharmacokinetic dose individualization of IRI in order to reduce the occurrence of severe toxicity during cancer treatment. METHODS The PubMed database was searched, considering papers published in the period from 1995-2017, using the keywords irinotecan, pharmacogenetics, metabolic genotyping, dose individualization, therapeutic drug monitoring, pharmacokinetics and pharmacodynamics, either alone or in combination, with original papers being selected based on the presence of relevant data. CONCLUSION The findings of this review confirm the importance of considering individual patient characteristics to select IRI doses. Currently, the most straightforward approach for IRI dose individualization is UGT1A1 genotyping. However, this strategy is sub-optimal due to several other genetic and environmental contributions to the variable pharmacokinetics of IRI and its active metabolite. The use of dried blood spot sampling could allow the clinical application of limited sampling and population pharmacokinetic models for IRI doses individualization.
Collapse
Affiliation(s)
- Roberta Zilles Hahn
- Laboratory of Analytical Toxicology, Institute of Health Sciences, Universidade Feevale, Novo Hamburgo- RS, Brazil.,Graduate Program on Toxicology and Analytical Toxicology, Universidade Feevale, Novo Hamburgo- RS, Brazil
| | - Marina Venzon Antunes
- Laboratory of Analytical Toxicology, Institute of Health Sciences, Universidade Feevale, Novo Hamburgo- RS, Brazil.,Graduate Program on Toxicology and Analytical Toxicology, Universidade Feevale, Novo Hamburgo- RS, Brazil
| | - Simone Gasparin Verza
- Graduate Program on Toxicology and Analytical Toxicology, Universidade Feevale, Novo Hamburgo- RS, Brazil
| | - Magda Susana Perassolo
- Graduate Program on Toxicology and Analytical Toxicology, Universidade Feevale, Novo Hamburgo- RS, Brazil
| | - Edna Sayuri Suyenaga
- Graduate Program on Toxicology and Analytical Toxicology, Universidade Feevale, Novo Hamburgo- RS, Brazil
| | | | - Rafael Linden
- Laboratory of Analytical Toxicology, Institute of Health Sciences, Universidade Feevale, Novo Hamburgo- RS, Brazil.,Graduate Program on Toxicology and Analytical Toxicology, Universidade Feevale, Novo Hamburgo- RS, Brazil
| |
Collapse
|
103
|
Dhas N, Parekh K, Pandey A, Kudarha R, Mutalik S, Mehta T. Two dimensional carbon based nanocomposites as multimodal therapeutic and diagnostic platform: A biomedical and toxicological perspective. J Control Release 2019; 308:130-161. [PMID: 31310783 DOI: 10.1016/j.jconrel.2019.07.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/10/2019] [Accepted: 07/12/2019] [Indexed: 12/12/2022]
Abstract
Graphene based nanocomposites have revolutionized cancer treatment, diagnosis and imaging owing to its good compatibility, elegant flexibility, high surface area, low mass density along with excellent combined additive effect of graphene with other nanomaterials. This review inculcates the type of graphene based nanocomposites and their fabrication techniques to improve its properties as photothermal and theranostic platform. With decades' efforts, many significant breakthroughs in the method of synthesis and characterization in addition to various functionalization options of graphene based nanocomposite have paved a solid foundation for their potential applications in the cancer therapy. This work intends to provide a thorough, up-to-date holistic discussion on correlation of breakthroughs with their biomedical applications and illustrate how to utilize these breakthroughs to address long-standing challenges in the clinical translation of nanomedicines. This review also emphasizes on graphene based nanocomposites based toxicity concerns pertaining to delivery platforms.
Collapse
Affiliation(s)
- Namdev Dhas
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Khushali Parekh
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Abhijeet Pandey
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Ritu Kudarha
- The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390002, India
| | - Srinivas Mutalik
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Tejal Mehta
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India.
| |
Collapse
|
104
|
Ren X, Boriero D, Chaiswing L, Bondada S, St Clair DK, Butterfield DA. Plausible biochemical mechanisms of chemotherapy-induced cognitive impairment ("chemobrain"), a condition that significantly impairs the quality of life of many cancer survivors. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1088-1097. [PMID: 30759363 PMCID: PMC6502692 DOI: 10.1016/j.bbadis.2019.02.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/05/2019] [Accepted: 02/07/2019] [Indexed: 12/13/2022]
Abstract
Increasing numbers of cancer patients survive and live longer than five years after therapy, but very often side effects of cancer treatment arise at same time. One of the side effects, chemotherapy-induced cognitive impairment (CICI), also called "chemobrain" or "chemofog" by patients, brings enormous challenges to cancer survivors following successful chemotherapeutic treatment. Decreased abilities of learning, memory, attention, executive function and processing speed in cancer survivors with CICI, are some of the challenges that greatly impair survivors' quality of life. The molecular mechanisms of CICI involve very complicated processes, which have been the subject of investigation over the past decades. Many mechanistic candidates have been studied including disruption of the blood-brain barrier (BBB), DNA damage, telomere shortening, oxidative stress and associated inflammatory response, gene polymorphism of neural repair, altered neurotransmission, and hormone changes. Oxidative stress is considered as a vital mechanism, since over 50% of FDA-approved anti-cancer drugs can generate reactive oxygen species (ROS) or reactive nitrogen species (RNS), which lead to neuronal death. In this review paper, we discuss these important candidate mechanisms, in particular oxidative stress and the cytokine, TNF-alpha and their potential roles in CICI.
Collapse
Affiliation(s)
- Xiaojia Ren
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Diana Boriero
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA; Department of Neurosciences, Biomedicine, and Movement Disorders, Section on Biological Chemistry, University of Verona, 37134 Verona, Italy
| | - Luksana Chaiswing
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - Subbarao Bondada
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Daret K St Clair
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - D Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
105
|
de Groot S, Pijl H, van der Hoeven JJM, Kroep JR. Effects of short-term fasting on cancer treatment. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:209. [PMID: 31113478 PMCID: PMC6530042 DOI: 10.1186/s13046-019-1189-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/22/2019] [Indexed: 12/14/2022]
Abstract
Growing preclinical evidence shows that short-term fasting (STF) protects from toxicity while enhancing the efficacy of a variety of chemotherapeutic agents in the treatment of various tumour types. STF reinforces stress resistance of healthy cells, while tumor cells become even more sensitive to toxins, perhaps through shortage of nutrients to satisfy their needs in the context of high proliferation rates and/or loss of flexibility to respond to extreme circumstances. In humans, STF may be a feasible approach to enhance the efficacy and tolerability of chemotherapy. Clinical research evaluating the potential of STF is in its infancy. This review focuses on the molecular background, current knowledge and clinical trials evaluating the effects of STF in cancer treatment. Preliminary data show that STF is safe, but challenging in cancer patients receiving chemotherapy. Ongoing clinical trials need to unravel if STF can also diminish toxicity and increase efficacy of chemotherapeutic regimes in daily practice.
Collapse
Affiliation(s)
- Stefanie de Groot
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, P.O. Box 9600, 2300RC, Leiden, The Netherlands
| | - Hanno Pijl
- Department of Endocrinology, Leiden University Medical Center, P.O. Box 9600, 2300RC, Leiden, The Netherlands
| | - Jacobus J M van der Hoeven
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, P.O. Box 9600, 2300RC, Leiden, The Netherlands
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, P.O. Box 9600, 2300RC, Leiden, The Netherlands.
| |
Collapse
|
106
|
Oyeleye SI, Adefegha SA, Dada FA, Okeke BM, Oboh G. Effect of
p
‐coumaric acid on the erectogenic enzyme activities and non‐protein thiol level in the penile tissue of normal and doxorubicin‐induced oxidative stress male rat. Andrologia 2019; 51:e13281. [DOI: 10.1111/and.13281] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 02/05/2019] [Accepted: 02/20/2019] [Indexed: 01/18/2023] Open
Affiliation(s)
- Sunday Idowu Oyeleye
- Functional Foods, Nutraceuticals and Phytomedicine Laboratory, Department of Biochemistry Federal University of Technology Akure Nigeria
- Department of Biomedical Technology Federal University of Technology Akure Nigeria
| | - Stephen Adeniyi Adefegha
- Functional Foods, Nutraceuticals and Phytomedicine Laboratory, Department of Biochemistry Federal University of Technology Akure Nigeria
| | - Felix Abayomi Dada
- Science Laboratory Technology Department (Biochemistry Unit) Federal Polytechnic Ede Ede Nigeria
| | - Bathlomew Maduka Okeke
- Functional Foods, Nutraceuticals and Phytomedicine Laboratory, Department of Biochemistry Federal University of Technology Akure Nigeria
| | - Ganiyu Oboh
- Functional Foods, Nutraceuticals and Phytomedicine Laboratory, Department of Biochemistry Federal University of Technology Akure Nigeria
| |
Collapse
|
107
|
Ren X, Keeney JTR, Miriyala S, Noel T, Powell DK, Chaiswing L, Bondada S, St Clair DK, Butterfield DA. The triangle of death of neurons: Oxidative damage, mitochondrial dysfunction, and loss of choline-containing biomolecules in brains of mice treated with doxorubicin. Advanced insights into mechanisms of chemotherapy induced cognitive impairment ("chemobrain") involving TNF-α. Free Radic Biol Med 2019; 134:1-8. [PMID: 30593843 PMCID: PMC6588453 DOI: 10.1016/j.freeradbiomed.2018.12.029] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 12/23/2018] [Indexed: 10/27/2022]
Abstract
Cancer treatments are developing fast and the number of cancer survivors could arise to 20 million in United State by 2025. However, a large fraction of cancer survivors demonstrate cognitive dysfunction and associated decreased quality of life both shortly, and often long-term, after chemotherapy treatment. The etiologies of chemotherapy induced cognitive impairment (CICI) are complicated, made more so by the fact that many anti-cancer drugs cannot cross the blood-brain barrier (BBB). Multiple related factors and confounders lead to difficulties in determining the underlying mechanisms. Chemotherapy induced, oxidative stress-mediated tumor necrosis factor-alpha (TNF-α) elevation was considered as one of the main candidate mechanisms underlying CICI. Doxorubicin (Dox) is a prototypical reactive oxygen species (ROS)-generating chemotherapeutic agent used to treat solid tumors and lymphomas as part of multi-drug chemotherapeutic regimens. We previously reported that peripheral Dox-administration leads to plasma protein damage and elevation of TNF-α in plasma and brain of mice. In the present study, we used TNF-α null (TNFKO) mice to investigate the role of TNF-α in Dox-induced, oxidative stress-mediated alterations in brain. We report that Dox-induced oxidative stress in brain is ameliorated and brain mitochondrial function assessed by the Seahorse-determined oxygen consumption rate (OCR) is preserved in brains of TNFKO mice. Further, we show that Dox-decreased the level of hippocampal choline-containing compounds and brain phospholipases activity are partially protected in TNFKO group in MRS study. Our results provide strong evidence that Dox-targeted mitochondrial damage and levels of brain choline-containing metabolites, as well as phospholipases changes decreased in the CNS are associated with oxidative stress mediated by TNF-α. These results are consistent with the notion that oxidative stress and elevated TNF-α in brain underlie the damage to mitochondria and other pathological changes that lead to CICI. The results are discussed with reference to our identifying a potential therapeutic target to protect against cognitive problems after chemotherapy.
Collapse
Affiliation(s)
- Xiaojia Ren
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Jeriel T R Keeney
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Sumitra Miriyala
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Teresa Noel
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - David K Powell
- Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky Medical Center, Lexington, KY 40536, USA
| | - Luksana Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Subbarao Bondada
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536 USA, USA; Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Daret K St Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky 40536, USA; Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536 USA, USA
| | - D Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536 USA, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
108
|
Frisch J, Angenendt A, Hoth M, Prates Roma L, Lis A. STIM-Orai Channels and Reactive Oxygen Species in the Tumor Microenvironment. Cancers (Basel) 2019; 11:E457. [PMID: 30935064 PMCID: PMC6520831 DOI: 10.3390/cancers11040457] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/22/2019] [Accepted: 03/27/2019] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment (TME) is shaped by cancer and noncancerous cells, the extracellular matrix, soluble factors, and blood vessels. Interactions between the cells, matrix, soluble factors, and blood vessels generate this complex heterogeneous microenvironment. The TME may be metabolically beneficial or unbeneficial for tumor growth, it may favor or not favor a productive immune response against tumor cells, or it may even favor conditions suited to hijacking the immune system for benefitting tumor growth. Soluble factors relevant for TME include oxygen, reactive oxygen species (ROS), ATP, Ca2+, H⁺, growth factors, or cytokines. Ca2+ plays a prominent role in the TME because its concentration is directly linked to cancer cell proliferation, apoptosis, or migration but also to immune cell function. Stromal-interaction molecules (STIM)-activated Orai channels are major Ca2+ entry channels in cancer cells and immune cells, they are upregulated in many tumors, and they are strongly regulated by ROS. Thus, STIM and Orai are interesting candidates to regulate cancer cell fate in the TME. In this review, we summarize the current knowledge about the function of ROS and STIM/Orai in cancer cells; discuss their interdependencies; and propose new hypotheses how TME, ROS, and Orai channels influence each other.
Collapse
Affiliation(s)
- Janina Frisch
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, Medical Faculty, Saarland University, 66421 Homburg, Germany.
- Center for Human and Molecular Biology, Saarland University, 66421 Homburg, Germany.
| | - Adrian Angenendt
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, Medical Faculty, Saarland University, 66421 Homburg, Germany.
| | - Markus Hoth
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, Medical Faculty, Saarland University, 66421 Homburg, Germany.
| | - Leticia Prates Roma
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, Medical Faculty, Saarland University, 66421 Homburg, Germany.
- Center for Human and Molecular Biology, Saarland University, 66421 Homburg, Germany.
| | - Annette Lis
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, Medical Faculty, Saarland University, 66421 Homburg, Germany.
| |
Collapse
|
109
|
The Effect of Taxifolin on Cisplatin-Induced Pulmonary Damage in Rats: A Biochemical and Histopathological Evaluation. Mediators Inflamm 2019; 2019:3740867. [PMID: 30992689 PMCID: PMC6434269 DOI: 10.1155/2019/3740867] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 12/21/2018] [Accepted: 01/01/2019] [Indexed: 12/27/2022] Open
Abstract
The effect of taxifolin on cisplatin-induced oxidative pulmonary damage was investigated biochemically and histopathologically in male albino Wistar rats. There were four groups, with six animals in each group: 50 mg/kg of taxifolin plus 2.5 mg/kg of cisplatin (TC) group, 2.5 mg/kg of cisplatin only (CIS) group, 50 mg/kg of taxifolin only (TG) group, and a healthy control group (HG). In terms of the experimental procedure, the animals in the TC and TG groups were first treated via oral gavage. The CIS and HG groups received distilled water as solvent, respectively. One hour later, the TC and CIS groups received cisplatin at a dose of 2.5 mg/kg (injected intraperitoneally). Taxifolin, cisplatin, and the distilled water were administered at the indicated dose and volume, using the same method daily for 14 d. At the end of this period, the animals were killed with a high dosage of thiopental anaesthesia (50 mg/kg). Blood and lung tissue samples were taken for biochemical (malondialdehyde (MDA), myeloperoxidase (MPO), total glutathione (tGSH), and 8-hydroxy-2 deoxyguanosine (8-OHdG)) analyses and histopathological examinations. The biochemical and histopathological results in the TC and HG groups were then compared with those in the CIS group. Cisplatin increased the levels of MDA, myeloperoxidase, and 8-OHdG, a marker of oxidative DNA damage, and reduced the amount of tGSH in the lung tissue. Moreover, severe alveolar damage, including oedema and extensive alveolar septal fibrosis, in addition to infiltration of polymorphic nuclear leucocytes and haemorrhagic foci, was observed in the CIS group. These histopathological findings demonstrate that taxifolin provides protection against pulmonary oxidative stress by preventing increases in oxidant parameters and decreases in antioxidants.
Collapse
|
110
|
Ornell KJ, Lozada D, Phan NV, Coburn JM. Controlling methacryloyl substitution of chondroitin sulfate: injectable hydrogels with tunable long-term drug release profiles. J Mater Chem B 2019; 7:2151-2161. [PMID: 32073574 DOI: 10.1039/c8tb03020k] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Drug delivery systems capable of local sustained release of small molecule therapeutics remain a critical need in many fields, including oncology. Here, a system to create tunable hydrogels capable of modulating the loading and release of cationic small molecule therapeutics was developed. Chondroitin sulfate (CS) is a sulfated glycosaminoglycan that has many promising properties, including biocompatibility, biodegradation and chemically modifiable groups for both covalent and non-covalent bonding. CS was covalently modified with photocrosslinkable methacryloyl groups (CSMA) to develop an injectable hydrogel fabrication. Utilizing anionic groups, cationic drugs can be adsorbed and released from the hydrogels. This study demonstrates the synthesis of CSMA with a varying degree of substitution (DS) to generate hydrogels with varying swelling properties, maximum injection force, and drug release kinetics. The DS of the synthesized CSMA ranged from 0.05 ± 0.02 (2 h reaction) to 0.28 ± 0.02 (24 h reaction) with a DS of 1 representing 100% modification. The altered DS resulted in changes in hydrogel properties with the swelling of 20% CSMA hydrogels ranging from 42 (2 h reaction) to 13 (24 h reaction) and injection forces ranging from 18 N (2 h reaction) to 94 N (24 h reaction). The release of sunitinib, an oncology therapeutic that inhibits intracellular signaling by targeting multiple receptor tyrosine kinases, ranged from 18 μg per day (2 h reaction) to 9 μg per day (24 h reaction). While decreasing the DS increased the hydrogel swelling and rate of therapeutic release, it also limited the hydrogel fabrication range to only those containing 10% or higher CSMA. Blended polymer systems with poly(vinyl alcohol)-methacrylate (PVAMA) were fabricated to stabilize the resulting hydrogels via attenuating the swelling properties. Release profiles previously unattainable with the pure CSMA hydrogels were achieved with the blended hydrogel formulations. Overall, these studies identify a method to formulate tunable CSMA and blended CSMA/PVAMA hydrogels capable of sustained release of cationic therapeutics over six weeks with applications in oncology therapeutics.
Collapse
Affiliation(s)
- Kimberly J Ornell
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 60 Prescott Street, Worcester, MA 01605, USA.
| | | | | | | |
Collapse
|
111
|
Grushevskaya HV, Krylova NG. Carbon Nanotubes as A High-Performance Platform for Target Delivery of Anticancer Quinones. Curr Pharm Des 2019; 24:5207-5218. [PMID: 30652640 DOI: 10.2174/1381612825666190117095132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 01/11/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND In spite of considerable efforts of researchers the cancer deseases remain to be incurable and a percentage of cancer deseases in the structure of mortality increases every year. At that, high systemic toxicity of antitumor drugs hampers their effective use. Because of this fact, the development of nanosystems for targeted delivery of antitumor drugs is one of the leading problem in nanomedicine and nanopharmacy. OBJECTIVE To critically examine the modern strategies for carbon nanotubes (CNTs)-based delivery of anticancer quinones and to summarize the mechanisms which can provide high effectiveness and multifunctionality of the CNT-based quinone delivery platform. RESULTS Quinones, including anthracycline antibiotics - doxorubicin and daunorubicin, are among the most prospective group of natural and syntetic compounds which exhibit high antitumor activity against different type of tumors. In this review, we focus on the possibilities of using CNTs for targeted delivery of antitumor compounds with quinoid moiety which is ordinarily characterized by high specific interaction with DNA molecules. Quinones can be non-covalently adsorbed on CNT surface due to their aromatic structure and π-conjugated system of double bonds. The characteristic features of doxorubicine-CNT complex are high loading efficiency, pH-dependent release in acidic tumor microenviroment, enough stability in biological fluid. Different types of CNT functionalization, targeting strategies and designs for multifunctional CNT-based doxorubicine delivery platform are disscussed. CONCLUSION Nanosystems based on functionalized CNTs are very promising platform for quinone delivery resulting in significant enhancement of cancer treatment efficiency. Functionalization of CNTs with the polymeric shell, especially DNA-based shells, can provide the greatest affinity and mimicry with biological structures.
Collapse
Affiliation(s)
- H V Grushevskaya
- Physics Department, Belarusian State University, 4 Nezavisimosti Ave., Minsk 220030, Belarus
| | - N G Krylova
- Physics Department, Belarusian State University, 4 Nezavisimosti Ave., Minsk 220030, Belarus
| |
Collapse
|
112
|
Hearn JM, Hughes GM, Romero-Canelón I, Munro AF, Rubio-Ruiz B, Liu Z, Carragher NO, Sadler PJ. Pharmaco-genomic investigations of organo-iridium anticancer complexes reveal novel mechanism of action. Metallomics 2019; 10:93-107. [PMID: 29131211 DOI: 10.1039/c7mt00242d] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Resistance to platinum drugs (used in >50% of cancer chemotherapies) is a clinical problem. Other precious metal complexes with distinct mechanisms of action might overcome this. Half-sandwich organometallic complexes containing arene or cyclopentadienyl (Cp) ligands show promise. We screened two iridium(iii) complexes [Ir(CpXbiph)(ppy)Cl] (ZL49, 1, ppy = phenylpyridine) and [Ir(CpXph)(azpyNMe2)Cl]PF6 (ZL109, 2, azpyNMe2 = N,N-dimethylphenylazopyridine) in 916 cancer cell lines from 28 tissue types. On average, complex 2 was 78× more potent than 1, 36× more active than cisplatin (CDDP), and strongly active (nanomolar) in patient-derived ovarian cancer cell lines. RNA sequencing of A2780 ovarian cells revealed upregulation of antioxidant responses (NRF2, AP-1) consistent with observed induction of reactive oxygen species (ROS). Protein microarrays, high content imaging and cell cycle analysis showed S/G2 arrest, and late-stage DNA damage response without p53 requirement. The triple-negative breast cancer cell line OCUB-M was highly sensitive to 2 as were cell lines with KIT mutations. Complex 2 exhibits a markedly different pattern of antiproliferative activity compared to the 253 drugs in the Sanger Cancer Genome database, but is most similar to osmium(ii) arene complexes which share the same azopyridine ligand. Redox modulation and DNA damage can provide a multi-targeting strategy, allowing compounds such as 2 to overcome cellular resistance to platinum anticancer drugs.
Collapse
|
113
|
Chen J, Ning C, Zhou Z, Yu P, Zhu Y, Tan G, Mao C. Nanomaterials as photothermal therapeutic agents. PROGRESS IN MATERIALS SCIENCE 2019; 99:1-26. [PMID: 30568319 PMCID: PMC6295417 DOI: 10.1016/j.pmatsci.2018.07.005] [Citation(s) in RCA: 416] [Impact Index Per Article: 69.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Curing cancer has been one of the greatest conundrums in the modern medical field. To reduce side-effects associated with the traditional cancer therapy such as radiotherapy and chemotherapy, photothermal therapy (PTT) has been recognized as one of the most promising treatments for cancer over recent years. PTT relies on ablation agents such as nanomaterials with a photothermal effect, for converting light into heat. In this way, elevated temperature could kill cancer cells while avoiding significant side effects on normal cells. This theory works because normal cells have a higher heat tolerance than cancer cells. Thus, nanomaterials with photothermal effects have attracted enormous attention due to their selectivity and non-invasive attributes. This review article summarizes the current status of employing nanomaterials with photothermal effects for anti-cancer treatment. Mechanisms of the photothermal effect and various factors affecting photothermal performance will be discussed. Efficient and selective PTT is believed to play an increasingly prominent role in cancer treatment. Moreover, merging PTT with other methods of cancer therapies is also discussed as a future trend.
Collapse
Affiliation(s)
- Junqi Chen
- College of Material Science and Engineering, South China University of Technology, Guangzhou 510641, China
- Guangdong Key Laboratory for Biomedical Engineering, Guangzhou 510641, China
| | - Chengyun Ning
- College of Material Science and Engineering, South China University of Technology, Guangzhou 510641, China
- Guangdong Key Laboratory for Biomedical Engineering, Guangzhou 510641, China
| | - Zhengnan Zhou
- College of Material Science and Engineering, South China University of Technology, Guangzhou 510641, China
- Guangdong Key Laboratory for Biomedical Engineering, Guangzhou 510641, China
| | - Peng Yu
- College of Material Science and Engineering, South China University of Technology, Guangzhou 510641, China
- Guangdong Key Laboratory for Biomedical Engineering, Guangzhou 510641, China
| | - Ye Zhu
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, Oklahoma, United States
| | - Guoxin Tan
- Institute of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, Oklahoma, United States
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| |
Collapse
|
114
|
Li Y, Yang D, Wang Y, Li Z, Zhu C. Co-delivery doxorubicin and silybin for anti-hepatoma via enhanced oral hepatic-targeted efficiency. Int J Nanomedicine 2018; 14:301-315. [PMID: 30643408 PMCID: PMC6314320 DOI: 10.2147/ijn.s187888] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background To establish the combination of doxorubicin (DOX) and silybin (SLB) in oral hepatic-targeting liposomes with the goal of reducing cardiotoxic side effects and improve oral hepatoma treatment. Methods Distearoylphosphatidylethanolamine–polyethylene glycol–cholic acid-modified liposomes (CA-LP) were used to encapsulate DOX and SLB (CA-LP–DOX/SLB), and the hepatic targeting, efficacy against hepatoma and cardioprotective effects were evaluated by cell toxicity, scratch and apoptosis in vitro studies, and pharmacokinetics and pharmacodynamics in vivo studies. Results In vitro cell studies showed that CA-LP–DOX/SLB inhibited HepG2 cell proliferation and HCC97H cell migration, and protected H9c2 cells. In vivo pharmacokinetics demonstrated that the CA-LP–DOX/SLB-treated group showed higher liver accumulation and lower heart accumulation of DOX relative to those in the CA-LP–DOX and LP–DOX-treated groups. In vivo pharmacodynamic studies showed that the CA-LP–DOX/SLB-treated group not only efficiently inhibited growth but also induced significantly less tissue damage than that observed in the CA-LP–DOX-treated group. Conclusion Concurrent administration of DOX and SLB via CA-LP provided a viable strategy to mitigate acute DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Ying Li
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, People's Republic of China,
| | - Dandan Yang
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, People's Republic of China,
| | - Yian Wang
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, People's Republic of China,
| | - Zhan Li
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, People's Republic of China,
| | - Chunyan Zhu
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, People's Republic of China,
| |
Collapse
|
115
|
Rasool M, Malik A, Abdul Basit Ashraf M, Arooj M, Kiran A, Waquar S, Ayyaz U, Zahid A, Zaheer A, Jabbar A, Zain M, Raza A, Mehmood A, Batool Qaisrani T, Mirza Z, Hussein Al-Qahtani M, Karim S, Haque A. Role of diagnostic factors associated with antioxidative status and expression of matrix metalloproteinases (MMPs) in patients with cancer therapy induced ocular disorders. Saudi J Biol Sci 2018; 25:1724-1728. [PMID: 30591791 PMCID: PMC6303172 DOI: 10.1016/j.sjbs.2018.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/11/2018] [Accepted: 08/12/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Cancer patients when treated with different chemotherapeutic drugs often develop mild to severe sight threatening diseases during or after chemotherapy. The mechanism involved in the pathogenesis of ocular toxicities is poorly understood. Oxidative stress, inflammation and MMPs (angiogenic factor) are involved in the progression of chemotherapy related ocular disorders. MATERIALS AND METHODS The concentration of oxidative stress markers such as MDA, NO and levels of different antioxidant molecules such as SOD, CAT, GSH, GPx, GPr, VIT A, VIT E and VIT C present in the serum of chemotherapy treated patients (n = 50) and in normal persons (n = 20) were estimated by the direct spectrophotometric method while the concentration of TNF-α and MMP-9 activity were determined using human TNF-α and MMP-9 ELISA kits. RESULTS The concentration of SOD and CAT (0.356 ± 0.05 μg/dl and 1.26 ± 0.01 μmol/mol of protein) was significantly lower as compared to that (1.09 ± 0.03 μg/dl and 3.99 ± 0.04 μmol/mol of protein) in controls. The levels of GPx (0.06 ± 0.01 mmol/dl) in the cancer patients were much lower than those in the controls (0.78 ± 0.06 mmol/dl). Lower level of GSH (0.96 ± 0.003 μg/dl) in serum of the diseased group was observed as compared to healthy group (7.26 ± 1.40 μg/dl). The level of Vit A, Vit C and Vit E was lower in systemic circulation of cancer patients (109.99 ± 6.35 μg/ml, 1.26 ± 0.36 μg/ml and 1.29 ± 0.191 μg/ml) as compared to control subjects (166.35 ± 14.26 μg/ml, 3.25 ± 0.099 μg/ml and 6.354 ± 2.26 μg/ml) respectively. The concentration of nitric oxide was significantly higher in the cancer patients (45.26 ± 6.35 ng/ml) than that in the normal subjects (16.35 ± 3.26 ng/ml). The higher concentration of MDA (8.65 ± 3.26 nmol/ml) was observed in the patients than normal ones (1.254 ± 0.065 nmol/ml). The quantity of TNF-α was significantly higher in chemotherapy treated patients (32.68 ± 4.33 pg/ml) as compared to the control group (20.979 ± 1.98 pg/ml). Significantly higher concentration of MMP-9 (40.26 ± 3.26 ng/ml) was observed in the cancer patients than the controls (7.256 ± 1.95 ng/ml). CONCLUSION Lower levels of antioxidant enzymes and non-enzymatic small molecules and higher levels of oxidative stress and inflammatory clinical parameters such as NO, MDA, TNF-α and MMP-9 may be involved in the pathogenesis of systemic chemotherapy related ocular complications such as cataract, glaucoma, blepharitis, retinitis pigmentosa, macular degeneration, pterygium and retinal degeneration.
Collapse
Affiliation(s)
- Mahmood Rasool
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Arif Malik
- Institute of Molecular Biology and Biotechnology, the University of Lahore, Lahore, Pakistan
| | | | - Mahwish Arooj
- University College of Medicine and Dentistry, the University of Lahore, Lahore, Pakistan
| | - Asia Kiran
- Institute of Molecular Biology and Biotechnology, the University of Lahore, Lahore, Pakistan
| | - Sulayman Waquar
- Institute of Molecular Biology and Biotechnology, the University of Lahore, Lahore, Pakistan
| | - Ujala Ayyaz
- Institute of Molecular Biology and Biotechnology, the University of Lahore, Lahore, Pakistan
| | - Ayesha Zahid
- Institute of Molecular Biology and Biotechnology, the University of Lahore, Lahore, Pakistan
| | - Ahmad Zaheer
- National Institute for Biotechnology and Genetic Engineering, Faisalabad, Pakistan
| | - Abdul Jabbar
- Department of Biotechnology, Mirpur University of Science and Technology (MUST), Mirpur (AJK), Pakistan
| | - Maryam Zain
- Microbiology and Molecular Genetics Department, The Women University, Multan, Pakistan
| | - Amir Raza
- National Institute for Biotechnology and Genetic Engineering, Faisalabad, Pakistan
| | - Asim Mehmood
- Department of Biosciences, COMSATS University, Sahiwal Campus, Sahiwal, Pakistan
| | | | - Zeenat Mirza
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Sajjad Karim
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Absarul Haque
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
116
|
Park HS, Kim CJ, Kwak HB, No MH, Heo JW, Kim TW. Physical exercise prevents cognitive impairment by enhancing hippocampal neuroplasticity and mitochondrial function in doxorubicin-induced chemobrain. Neuropharmacology 2018; 133:451-461. [PMID: 29477301 DOI: 10.1016/j.neuropharm.2018.02.013] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 11/26/2022]
Abstract
Although chemotherapy increases the survival rate of patients with various cancers, such treatment can induce acute or long-term cognitive dysfunction a phenomenon known as post-chemotherapy cognitive impairment (PCCI) or "chemobrain." Exercise is known to positively affect brain function. Thus, the present study aimed to determine whether symptoms of chemobrain and disruptions in the neuroplasticity and functioning of hippocampal mitochondria can be prevented or relieved by exercise. Wistar rats were separated into the following groups: control, control plus exercise, chemobrain, and chemobrain plus exercise. For chemobrain induction, 2 mg/kg of doxorubicin (DOX) a widely utilized chemotherapeutic agent among patients with breast cancer was dissolved in saline and directly injected to the abdomen once every 4 weeks. The exercise groups were subjected to low-intensity treadmill, 6 days per week for 4 weeks. The Morris water maze and step-down avoidance tests were conducted to evaluate cognitive function, while neuroplasticity and mitochondrial function were assessed in the hippocampus and dentate gyrus. Decreased cognitive function were observed in the chemobrain group, along with decreases in levels of neurogenesis, brain derived neurotrophic factor (BDNF), tropomyosin-related kinase B (TrkB), Ca2+ retention in hippocampus. Rats of the chemobrain group also exhibited an increase in apoptosis, H2O2 emission and permeability transition pore by hippocampal mitochondria. However, exercise attenuated impairments in cognitive function, neuroplasticity, and mitochondrial function induced by DOX treatment. Therefore, the findings of the present study indicate that low-intensity exercise may assist in preventing cognitive dysfunction during or after chemotherapy in patients with various cancers, including breast cancer.
Collapse
Affiliation(s)
- Hye-Sang Park
- Department of Physiology, College of Medicine, KyungHee University, Seoul, Republic of Korea
| | - Chang-Ju Kim
- Department of Physiology, College of Medicine, KyungHee University, Seoul, Republic of Korea
| | - Hyo-Bum Kwak
- Department of Kinesiology, Art & Sports, InHa University, Incheon, Republic of Korea
| | - Mi-Hyun No
- Department of Kinesiology, Art & Sports, InHa University, Incheon, Republic of Korea
| | - Jun-Won Heo
- Department of Kinesiology, Art & Sports, InHa University, Incheon, Republic of Korea
| | - Tae-Woon Kim
- Department of Physiology, College of Medicine, KyungHee University, Seoul, Republic of Korea; Exercise Rehabilitation Research Institute, Department of Exercise & Health Science, SangMyung University, Seoul, Republic of Korea.
| |
Collapse
|
117
|
Farhood B, Mortezaee K, Goradel NH, Khanlarkhani N, Salehi E, Nashtaei MS, Najafi M, Sahebkar A. Curcumin as an anti-inflammatory agent: Implications to radiotherapy and chemotherapy. J Cell Physiol 2018; 234:5728-5740. [PMID: 30317564 DOI: 10.1002/jcp.27442] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 08/27/2018] [Indexed: 12/21/2022]
Abstract
Cancer is the second cause of death worldwide. Chemotherapy and radiotherapy are the most common modalities for the treatment of cancer. Experimental studies have shown that inflammation plays a central role in tumor resistance and the incidence of several side effects following both chemotherapy and radiotherapy. Inflammation resulting from radiotherapy and chemotherapy is responsible for adverse events such as dermatitis, mucositis, pneumonitis, fibrosis, and bone marrow toxicity. Chronic inflammation may also lead to the development of second cancer during years after treatment. A number of anti-inflammatory drugs such as nonsteroidal anti-inflammatory agents have been proposed to alleviate chronic inflammatory reactions after radiotherapy or chemotherapy. Curcumin is a well-documented herbal anti-inflammatory agents. Studies have proposed that curcumin can help management of inflammation during and after radiotherapy and chemotherapy. Curcumin targets various inflammatory mediators such as cyclooxygenase-2, inducible nitric oxide synthase, and nuclear factor κB (NF-κB), thereby attenuating the release of proinflammatory and profibrotic cytokines, and suppressing chronic production of free radicals, which culminates in the amelioration of tissue toxicity. Through modulation of NF-κB and its downstream signaling cascade, curcumin can also reduce angiogenesis, tumor growth, and metastasis. Low toxicity of curcumin is linked to its cytoprotective effects in normal tissues. This protective action along with the capacity of this phytochemical to sensitize tumor cells to radiotherapy and chemotherapy makes it a potential candidate for use as an adjuvant in cancer therapy. There is also evidence from clinical trials suggesting the potential utility of curcumin for acute inflammatory reactions during radiotherapy such as dermatitis and mucositis.
Collapse
Affiliation(s)
- Bagher Farhood
- Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Nasser Hashemi Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Khanlarkhani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ensieh Salehi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Shabani Nashtaei
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Department of Radiology and Nuclear Medicine, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
118
|
Imam F, Al-Harbi NO, Al-Harbi MM, Ansari MA, Al-Asmari AF, Ansari MN, Al-Anazi WA, Bahashwan S, Almutairi MM, Alshammari M, Khan MR, Alsaad AM, Alotaibi MR. Apremilast prevent doxorubicin-induced apoptosis and inflammation in heart through inhibition of oxidative stress mediated activation of NF-κB signaling pathways. Pharmacol Rep 2018; 70:993-1000. [DOI: 10.1016/j.pharep.2018.03.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/01/2018] [Accepted: 03/27/2018] [Indexed: 12/25/2022]
|
119
|
Ingato D, Edson JA, Zakharian M, Kwon YJ. Cancer Cell-Derived, Drug-Loaded Nanovesicles Induced by Sulfhydryl-Blocking for Effective and Safe Cancer Therapy. ACS NANO 2018; 12:9568-9577. [PMID: 30130093 DOI: 10.1021/acsnano.8b05377] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Extracellular vesicles (EVs) pose great promise as therapeutic carriers due to their ideal size range and intrinsic biocompatibility. Limited scalability, poor quality control during production, and cumbersome isolation and purification processes have caused major setbacks in the progression of EV therapeutics to the clinic. Here, we overcome these setbacks by preparing cell-derived nanovesicles induced by sulfhydryl-blocking (NIbS), in the desirable size range for therapeutic delivery, that can be further loaded with the chemotherapeutic drug, doxorubicin (DOX), resulting in NIbS/DOX. Applicable to most cell types, this chemical blebbing approach enables efficient, quick, and simple harvest and purification as well as easily scalable production. Cellular uptake and intracellular release of DOX was improved using NIbS/DOX compared to a liposomal formulation. We also confirmed that in tumor-challenged C57BL/6 mice NIbS/DOX significantly slowed tumor growth and led to improved survival compared to treatment with free drug or liposomal drug. NIbS are a promising therapeutic carrier for improving cancer treatment outcomes since they are easy to prepare at a large scale, good candidates for drug loading, and capable of efficient administration of therapeutic agents with avoided nonspecific major distribution in vital organs. In addition, the utility of NIbS can be easily expanded to immunotherapy, gene therapy, and cell therapy when they are derived from applicable cell types.
Collapse
Affiliation(s)
- Dominique Ingato
- Department of Chemical Engineering and Materials Science , University of California , Irvine , California 92697 , United States
| | - Julius A Edson
- Department of Chemical Engineering and Materials Science , University of California , Irvine , California 92697 , United States
| | - Michael Zakharian
- Department of Molecular Biology and Biochemistry , University of California , Irvine , California 92697 , United States
| | - Young Jik Kwon
- Department of Chemical Engineering and Materials Science , University of California , Irvine , California 92697 , United States
- Department of Molecular Biology and Biochemistry , University of California , Irvine , California 92697 , United States
- Department of Pharmaceutical Sciences , University of California , Irvine , California 92697 , United States
- Department of Biomedical Engineering , University of California , Irvine , California 92697 , United States
| |
Collapse
|
120
|
Zuniga KE, Moran NE. Low Serum Carotenoids Are Associated with Self-Reported Cognitive Dysfunction and Inflammatory Markers in Breast Cancer Survivors. Nutrients 2018; 10:E1111. [PMID: 30126098 PMCID: PMC6116006 DOI: 10.3390/nu10081111] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/02/2018] [Accepted: 08/08/2018] [Indexed: 01/06/2023] Open
Abstract
Background: Dietary carotenoids may exert anti-inflammatory activities to reduce inflammation-driven cognitive impairments during cancer and cancer treatment. Our objective was to explore if cognitive function in breast cancer survivors (BCS) differs by serum carotenoid concentrations, and if blood carotenoids concentrations are associated with reduced systemic inflammation. Methods: Objective cognitive function and perceived cognitive impairment of 29 BCS and 38 controls were assessed cross-sectionally with the National Institutes of Health Toolbox Cognition Battery and The Functional Assessment of Cancer Therapy-Cognitive Function Questionnaire, respectively. Serum carotenoid and inflammatory marker (sTNF-RII, IL-6, IL-1ra, CRP) concentrations were measured. Results: Low-carotenoid BCS had more cognitive complaints compared to the low-carotenoid controls (Mdiff = -43.0, p < 0.001) and high-carotenoid controls (Mdiff = -44.5, p < 0.001). However, the cognitive complaints of high-carotenoid BCS were intermediate to and not different than the low-carotenoid BCS, or low- or high-carotenoid controls. BCS performed similarly to controls on all objective cognitive measures. Multiple linear regression, controlling for age and body mass index (BMI), demonstrated an inverse association between serum carotenoid concentrations and pro-inflammatory sTNFR-II (β = 0.404, p = 0.005) and IL-6 concentrations (β = -0.35, p = 0.001), but not IL-1ra or CRP. Conclusions: Higher serum carotenoid concentrations may convey cognitive and anti-inflammatory benefits in BCS. Future research should identify dietary components and patterns that support cognitive health in cancer survivors.
Collapse
Affiliation(s)
- Krystle E Zuniga
- Nutrition and Foods, Family and Consumer Sciences, Texas State University, 601 University Drive, San Marcos, TX 78666, USA.
| | - Nancy E Moran
- United States Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
121
|
Keeney JTR, Ren X, Warrier G, Noel T, Powell DK, Brelsfoard JM, Sultana R, Saatman KE, Clair DKS, Butterfield DA. Doxorubicin-induced elevated oxidative stress and neurochemical alterations in brain and cognitive decline: protection by MESNA and insights into mechanisms of chemotherapy-induced cognitive impairment ("chemobrain"). Oncotarget 2018; 9:30324-30339. [PMID: 30100992 PMCID: PMC6084398 DOI: 10.18632/oncotarget.25718] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 06/13/2018] [Indexed: 12/21/2022] Open
Abstract
Chemotherapy-induced cognitive impairment (CICI) is now widely recognized as a real and too common complication of cancer chemotherapy experienced by an ever-growing number of cancer survivors. Previously, we reported that doxorubicin (Dox), a prototypical reactive oxygen species (ROS)-producing anti-cancer drug, results in oxidation of plasma proteins, including apolipoprotein A-I (ApoA-I) leading to tumor necrosis factor-alpha (TNF-α)-mediated oxidative stress in plasma and brain. We also reported that co-administration of the antioxidant drug, 2-mercaptoethane sulfonate sodium (MESNA), prevents Dox-induced protein oxidation and subsequent TNF-α elevation in plasma. In this study, we measured oxidative stress in both brain and plasma of Dox-treated mice both with and without MESNA. MESNA ameliorated Dox-induced oxidative protein damage in plasma, confirming our prior studies, and in a new finding led to decreased oxidative stress in brain. This study also provides further functional and biochemical evidence of the mechanisms of CICI. Using novel object recognition (NOR), we demonstrated the Dox administration resulted in memory deficits, an effect that was rescued by MESNA. Using hydrogen magnetic resonance imaging spectroscopy (H1-MRS) techniques, we demonstrated that Dox administration led to a dramatic decrease in choline-containing compounds assessed by (Cho)/creatine ratios in the hippocampus in mice. To better elucidate a potential mechanism for this MRS observation, we tested the activities of the phospholipase enzymes known to act on phosphatidylcholine (PtdCho), a key component of phospholipid membranes and a source of choline for the neurotransmitter, acetylcholine (ACh). The activities of both phosphatidylcholine-specific phospholipase C (PC-PLC) and phospholipase D were severely diminished following Dox administration. The activity of PC-PLC was preserved when MESNA was co-administered with Dox; however, PLD activity was not protected. This study is the first to demonstrate the protective effects of MESNA on Dox-related protein oxidation, cognitive decline, phosphocholine (PCho) levels, and PC-PLC activity in brain and suggests novel potential therapeutic targets and strategies to mitigate CICI.
Collapse
Affiliation(s)
| | - Xiaojia Ren
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Govind Warrier
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Teresa Noel
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
| | - David K. Powell
- Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky Medical Center, Lexington, KY 40536, USA
| | - Jennifer M. Brelsfoard
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Rukhsana Sultana
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Kathryn E. Saatman
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Daret K. St. Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA
- Department of Radiation Medicine, University of Kentucky, Lexington, KY 40502, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40502, USA
| | - D. Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40502, USA
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
122
|
Bajic JE, Johnston IN, Howarth GS, Hutchinson MR. From the Bottom-Up: Chemotherapy and Gut-Brain Axis Dysregulation. Front Behav Neurosci 2018; 12:104. [PMID: 29872383 PMCID: PMC5972222 DOI: 10.3389/fnbeh.2018.00104] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 04/30/2018] [Indexed: 12/12/2022] Open
Abstract
The central nervous system and gastrointestinal tract form the primary targets of chemotherapy-induced toxicities. Symptoms associated with damage to these regions have been clinically termed chemotherapy-induced cognitive impairment and mucositis. Whilst extensive literature outlines the complex etiology of each pathology, to date neither chemotherapy-induced side-effect has considered the potential impact of one on the pathogenesis of the other disorder. This is surprising considering the close bidirectional relationship shared between each organ; the gut-brain axis. There are complex multiple pathways linking the gut to the brain and vice versa in both normal physiological function and disease. For instance, psychological and social factors influence motility and digestive function, symptom perception, and behaviors associated with illness and pathological outcomes. On the other hand, visceral pain affects central nociception pathways, mood and behavior. Recent interest highlights the influence of functional gut disorders, such as inflammatory bowel diseases and irritable bowel syndrome in the development of central comorbidities. Gut-brain axis dysfunction and microbiota dysbiosis have served as key portals in understanding the potential mechanisms associated with these functional gut disorders and their effects on cognition. In this review we will present the role gut-brain axis dysregulation plays in the chemotherapy setting, highlighting peripheral-to-central immune signaling mechanisms and their contribution to neuroimmunological changes associated with chemotherapy exposure. Here, we hypothesize that dysregulation of the gut-brain axis plays a major role in the intestinal, psychological and neurological complications following chemotherapy. We pay particular attention to evidence surrounding microbiota dysbiosis, the role of intestinal permeability, damage to nerves of the enteric and peripheral nervous systems and vagal and humoral mediated changes.
Collapse
Affiliation(s)
- Juliana E. Bajic
- Discipline of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Ian N. Johnston
- School of Psychology, The University of Sydney, Sydney, NSW, Australia
| | - Gordon S. Howarth
- School of Animal and Veterinary Sciences, University of Adelaide, Adelaide, SA, Australia
- Department of Gastroenterology, Women's and Children's Hospital, North Adelaide, SA, Australia
| | - Mark R. Hutchinson
- Discipline of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia
- Centre of Excellence for Nanoscale Biophotonics, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
123
|
Almeida D, Pinho R, Correia V, Soares J, Bastos MDL, Carvalho F, Capela JP, Costa VM. Mitoxantrone is More Toxic than Doxorubicin in SH-SY5Y Human Cells: A 'Chemobrain' In Vitro Study. Pharmaceuticals (Basel) 2018; 11:41. [PMID: 29734752 PMCID: PMC6027466 DOI: 10.3390/ph11020041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/28/2018] [Accepted: 04/29/2018] [Indexed: 12/20/2022] Open
Abstract
The potential neurotoxic effects of anticancer drugs, like doxorubicin (DOX) and mitoxantrone (MTX; also used in multiple sclerosis), are presently important reasons for concern, following epidemiological data indicating that cancer survivors submitted to chemotherapy may suffer cognitive deficits. We evaluated the in vitro neurotoxicity of two commonly used chemotherapeutic drugs, DOX and MTX, and study their underlying mechanisms in the SH-SY5Y human neuronal cell model. Undifferentiated human SH-SY5Y cells were exposed to DOX or MTX (0.13, 0.2 and 0.5 μM) for 48 h and two cytotoxicity assays were performed, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) reduction and the neutral red (NR) incorporation assays. Phase contrast microphotographs, Hoechst, and acridine orange/ethidium bromide stains were performed. Mitochondrial membrane potential was also assessed. Moreover, putative protective drugs, namely the antioxidants N-acetyl-l-cysteine (NAC; 1 mM) and 100 μM tiron, the inhibitor of caspase-3/7, Ac-DEVD-CHO (100 μM), and a protein synthesis inhibitor, cycloheximide (CHX; 10 nM), were tested to prevent DOX- or MTX-induced toxicity. The MTT reduction assay was also done in differentiated SH-SY5Y cells following exposure to 0.2 μM DOX or MTX. MTX was more toxic than DOX in both cytotoxicity assays and according to the morphological analyses. MTX also evoked a higher number of apoptotic nuclei than DOX. Both drugs, at the 0.13 μM concentration, caused mitochondrial membrane potential depolarization after a 48-h exposure. Regarding the putative neuroprotectors, 1 mM NAC was not able to prevent the cytotoxicity caused by either drug. Notwithstanding, 100 μM tiron was capable of partially reverting MTX-induced cytotoxicity in the NR uptake assay. One hundred μM Ac-DEVD-CHO and 10 nM cycloheximide (CHX) also partially prevented the toxicity induced by DOX in the NR uptake assay. MTX was more toxic than DOX in differentiated SH-SY5Y cells, while MTX had similar toxicity in differentiated and undifferentiated SH-SY5Y cells. In fact, MTX was the most neurotoxic drug tested and the mechanisms involved seem dissimilar among drugs. Thus, its toxicity mechanisms need to be further investigated as to determine the putative neurotoxicity for multiple sclerosis and cancer patients.
Collapse
Affiliation(s)
- Daniela Almeida
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Rita Pinho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Verónica Correia
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Jorge Soares
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Maria de Lourdes Bastos
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - João Paulo Capela
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- FP-ENAS (Unidade de Investigação UFP em Energia, Ambiente e Saúde), CEBIMED (Centro de Estudos em Biomedicina), Faculdade de Ciências da Saúde, Universidade Fernando Pessoa, 4249-004 Porto, Portugal.
| | - Vera Marisa Costa
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
124
|
Qiu Y, Yu Q, Liu Y, Tang J, Wang X, Lu Z, Xu Z, He Q. Dual Receptor Targeting Cell Penetrating Peptide Modified Liposome for Glioma and Breast Cancer Postoperative Recurrence Therapy. Pharm Res 2018; 35:130. [PMID: 29700620 DOI: 10.1007/s11095-018-2399-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/31/2018] [Indexed: 02/05/2023]
Abstract
PURPOSE Cell penetrating peptides (CPPs) were widely used as motifs for drug delivery to tumor. In former study, an RGD reverse sequence dGR was used to develop active-targeting liposome R8dGR-Lip, which showed well penetrating ability and treatment efficiency on glioma model. However, recurrence after tumor resection caused by post-operative residual cancer cells was a huge obstacle in tumor treatment. In consideration of the effective anti-cancer effect of PTX-R8dGR-Lip when treating glioma in former study, we decide to evaluate its pharmacodynamics on tumor resection models, which were more invasive and resistant. METHOD In vitro, the effectiveness of PTX-R8dGR-Lip in reducing tumor initiating cell (TIC) was investigated using mammosphere formation. In vivo, the inhibition efficiency of PTX-R8dGR-Lip on C6 glioma recurrence and 4 T1 breast cancer recurrence model were evaluated, including tumor bioluminescence imaging, survival rate and immumohistochemical staining, etc.. RESULTS C6 mammosphere formation rate of PTX-R8dGR-Lip group was 48.06 ± 2.72%, and 4 T1 mammosphere formation rate of PTX-R8dGR-Lip group was 39.51 ± 4.02% when PBS group was set as 100%. C6 and 4 T1 bioluminescent tumor resected model were established, then effectiveness of different PTX-loaded preparations were evaluated on these two models. PTX-R8dGR-Lip could obviously inhibit tumor recurrence, prolong survival rate and reduce tumor tissue invasion. CONCLUSION PTX-R8dGR-Lip could reduce post-operative recurrence rate, prolong survival time, and decrease the proliferation of residual cancer cells through regulating the expression of recurrence-related cytokines.
Collapse
Affiliation(s)
- Yue Qiu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Qianwen Yu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yayuan Liu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jiajing Tang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Xuhui Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Zhengze Lu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Zhuping Xu
- Ophthalmology Department, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Qin He
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
125
|
Repka CP, Hayward R. Effects of an Exercise Intervention on Cancer-Related Fatigue and Its Relationship to Markers of Oxidative Stress. Integr Cancer Ther 2018; 17:503-510. [PMID: 29649913 PMCID: PMC6041925 DOI: 10.1177/1534735418766402] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background: Although the underlying mechanisms of cancer-related fatigue (CRF) are not fully characterized, treatment-associated oxidative stress may play a role. The purpose of this study was to determine the effect of an exercise intervention on the relationship between CRF and oxidative stress. Methods: Upon cessation of radiation or chemotherapy, 8 cancer patients participated in a 10-week exercise intervention (EX), while 7 continued standard care (CON). Blood draws and fatigue questionnaires were administered to cancer patients before and after the intervention as well as to 7 age-matched individuals with no cancer history. Changes in plasma 8-hydroxy-deoxyguanosine (8-OHdG), protein carbonyls, antioxidant capacity, and fatigue were compared between groups. Correlations between CRF and oxidative stress were evaluated. Results: Mean total fatigue scores decreased significantly (5.0 ± 2.2 to 2.6 ± 1.5, P < .05) in EX, but not in CON. Antioxidant capacity significantly increased (+41%; P < .05) and protein carbonyls significantly decreased (−36%; P < .05) in EX, but not in CON. Increases in antioxidant capacity were significantly correlated with reductions in affective (r = −.49), sensory (r = −.47), and cognitive fatigue (r = −.58). Changes in total (r = .46) and affective (r = .47) fatigue exhibited significant correlations with changes in 8-OHdG over time, while behavioral (r = .46) and sensory (r = .47) fatigue changes were significantly correlated with protein carbonyls. Conclusions: Oxidative stress may be implicated in CRF, while improved antioxidant capacity following an exercise intervention may play a role in mitigating CRF in cancer survivors.
Collapse
Affiliation(s)
- Chris P Repka
- 1 Department of Health Sciences, Northern Arizona University, Flagstaff, AZ, USA.,2 Cancer Rehabilitation Institute, University of Northern Colorado, Greeley, CO, USA
| | - Reid Hayward
- 2 Cancer Rehabilitation Institute, University of Northern Colorado, Greeley, CO, USA
| |
Collapse
|
126
|
Chemotherapeutic Drugs and Mitochondrial Dysfunction: Focus on Doxorubicin, Trastuzumab, and Sunitinib. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7582730. [PMID: 29743983 PMCID: PMC5878876 DOI: 10.1155/2018/7582730] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/23/2018] [Accepted: 02/06/2018] [Indexed: 02/07/2023]
Abstract
Many cancer therapies produce toxic side effects whose molecular mechanisms await full elucidation. The most feared and studied side effect of chemotherapeutic drugs is cardiotoxicity. Also, skeletal muscle physiology impairment has been recorded after many chemotherapeutical treatments. However, only doxorubicin has been extensively studied for its side effects on skeletal muscle. Chemotherapeutic-induced adverse side effects are, in many cases, mediated by mitochondrial damage. In particular, trastuzumab and sunitinib toxicity is mainly associated with mitochondria impairment and is mostly reversible. Vice versa, doxorubicin-induced toxicity not only includes mitochondria damage but can also lead to a more robust and extensive cell injury which is often irreversible and lethal. Drugs interfering with mitochondrial functionality determine the depletion of ATP reservoirs and lead to subsequent reversible contractile dysfunction. Mitochondrial damage includes the impairment of the respiratory chain and the loss of mitochondrial membrane potential with subsequent disruption of cellular energetic. In a context of increased stress, AMPK has a key role in maintaining energy homeostasis, and inhibition of the AMPK pathway is one of the proposed mechanisms possibly mediating mitochondrial toxicity due to chemotherapeutics. Therapies targeting and protecting cell metabolism and energy management might be useful tools in protecting muscular tissues against the toxicity induced by chemotherapeutic drugs.
Collapse
|
127
|
Redox control in cancer development and progression. Mol Aspects Med 2018; 63:88-98. [PMID: 29501614 DOI: 10.1016/j.mam.2018.02.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 02/26/2018] [Accepted: 02/26/2018] [Indexed: 02/07/2023]
Abstract
Cancer is the leading cause of death worldwide after cardiovascular diseases. This has been the case for the last few decades despite there being an increase in the number of cancer treatments. One reason for the apparent lack of drug effectiveness might be, at least in part, due to unspecificity for tumors; which often leads to substantial side effects. One way to improve the treatment of cancer is to increase the specificity of the treatment in accordance with the concept of individualized medicine. This will help to prevent further progression of an existing cancer or even to reduce the tumor burden. Alternatively it would be much more attractive and efficient to prevent the development of cancer in the first place. Therefore, it is important to understand the risk factors and the mechanisms of carcinogenesis in detail. One such risk factor, often associated with tumorigenesis and tumor progression, is an increased abundance of reactive oxygen species (ROS) arising from an imbalance of ROS-producing and -eliminating components. A surplus of ROS can induce oxidative damage of macromolecules including proteins, lipids and DNA. In contrast, ROS are essential for an adequate signal transduction and are known to regulate crucial cellular processes like cellular quiescence, differentiation and even apoptosis. Therefore, regulated ROS-formation at physiological levels can inhibit tumor formation and progression. With this review we provide an overview on the current knowledge of redox control in cancer development and progression.
Collapse
|
128
|
Afsar T, Razak S, Almajwal A, Khan MR. Acacia hydaspica R. Parker ameliorates cisplatin induced oxidative stress, DNA damage and morphological alterations in rat pulmonary tissue. Altern Ther Health Med 2018; 18:49. [PMID: 29394892 PMCID: PMC5797377 DOI: 10.1186/s12906-018-2113-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 01/25/2018] [Indexed: 12/20/2022]
Abstract
Background Cisplatin (CP) drug is platinum compounds used for the treatment of various human malignancies. However, adverse outcomes related to CP restrict its usage. Acacia hydaspica is a natural shrub with various pharmacological properties. The current investigation aimed to assess the protective potential of A. hydaspica polyphenol rich ethyl acetate extract (AHE) against cisplatin (CP) induced pulmonary toxicity. Methods Rats were divided into six groups. Group 1 served as control (saline); Group 2 (drug control) recieved single dose of CP (7.5 mg/kg i.p.) on 1st day; Group 3 (extract control) (400 mg/kg bw, p.o.) received AHE for one week; Group 4 (Post-treated) and Group 5 (pretreated) received AHE (400 mg/kg bw/day, p.o) for 7 days after and before CP (7.5 mg/kg b.w., i.p.) respectively; Group 6 (Standard control) received silymarin (100 mg/kg b.w/7 days) before CP. At the end of dosing rats were sacrificed and pulmonary tissue samples were processed for the evaluation of antioxidant enzymes, oxidative stress markers, genotoxicity and histopathological alterations. Results CP caused body weights loss and increase pulmonary tissue weight. The CP significantly increases oxidative stress markers and decreases tissue antioxidant enzyme levels. Furthermore, CP induced deleterious changes in the microanatomy of pulmonary tissue by rupturing the alveolar septa, thickening of alveolar walls, and injuring the cells with subsequent collapse of blood vessels. AHE pretreatment returned MDA, NO, H2O2 production and improved tissue antioxidant enzyme levels to near normalcy. The histological observations evidenced that AHE effectively rescues the lungs from CP-mediated oxidative damage. CP induction in rats also caused DNA fragmentation which was restored by AHE treatment. Our results suggest that pretreatment more significantly improve CP induced deleterious effects compared with post treatment indicating protective effect. Potency of AHE pretreatment is similar to silymarin. Conclusion These findings demonstrated that A. hydaspica AHE extract might serve as potential adjuvant that prevents CP persuaded pulmonary toxicity due to its intrinsic antioxidant potential and polyphenolic constituents.
Collapse
|
129
|
Epigenetic and microenvironmental alterations in bone marrow associated with ROS in experimental aplastic anemia. Eur J Cell Biol 2018; 97:32-43. [DOI: 10.1016/j.ejcb.2017.11.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/04/2017] [Accepted: 11/20/2017] [Indexed: 12/22/2022] Open
|
130
|
Rizk HA, Masoud MA, Maher OW. Prophylactic effects of ellagic acid and rosmarinic acid on doxorubicin-induced neurotoxicity in rats. J Biochem Mol Toxicol 2017; 31. [PMID: 28815802 DOI: 10.1002/jbt.21977] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 08/01/2017] [Indexed: 12/25/2022]
Abstract
Doxorubicin (DOX) is a chemotherapeutic agent widely used in human malignancies. Its long-term use cause neurobiological side effects. The aim of the present study was to investigate the prophylactic effect exerted by daily administration of ellagic acid (EA) and rosmarinic acid (RA) on DOX-induced neurotoxicity in rats. Our data showed that DOX-induced significant elevation of brain malondialdehyde, tumor necrosis factor-alpha (TNF-α), inducible nitric oxide synthase (iNOS), caspase-3, and cholinesterase associated with significant reduction in reduced glutathione, monoamines namely serotonin, dopamine, as well as norepinephrine. Concomitant administration of EA (10 mg/kg/day, p.o. for 14 days) and/or RA (75 mg/kg/day, p.o. for 14 days) with DOX significantly mitigated the neural changes induced by DOX. Meanwhile, treatment ameliorated pro-inflammatory cytokines as TNF-α, iNOS, and attenuated oxidative stress biomarkers as well as brain monoamines. In conclusion, EA and RA can effectively protect against DOX-induced neurotoxicity, and the mechanisms underlying the neuroprotective effect are potentially associated with its antioxidant, anti-inflammatory, and antiapoptotic properties.
Collapse
Affiliation(s)
- Hanan A Rizk
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - Marwa A Masoud
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - Omar W Maher
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| |
Collapse
|
131
|
Geary DC. Evolution of Human Sex-Specific Cognitive Vulnerabilities. QUARTERLY REVIEW OF BIOLOGY 2017. [DOI: 10.1086/694934] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
132
|
Cho JH, Han JS. Phospholipase D and Its Essential Role in Cancer. Mol Cells 2017; 40:805-813. [PMID: 29145720 PMCID: PMC5712509 DOI: 10.14348/molcells.2017.0241] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/16/2017] [Accepted: 11/11/2017] [Indexed: 11/27/2022] Open
Abstract
The role of phospholipase D (PLD) in cancer development and management has been a major area of interest for researchers. The purpose of this mini-review is to explore PLD and its distinct role during chemotherapy including anti-apoptotic function. PLD is an enzyme that belongs to the phospholipase super family and is found in a broad range of organisms such as viruses, yeast, bacteria, animals, and plants. The function and activity of PLD are widely dependent on and regulated by neurotransmitters, hormones, small monomeric GTPases, and lipids. A growing body of research has shown that PLD activity is significantly increased in cancer tissues and cells, indicating that it plays a critical role in signal transduction, cell proliferation, and anti-apoptotic processes. In addition, recent studies show that PLD is a downstream transcriptional target of proteins that contribute to inflammation and carcinogenesis such as Sp1, NFκB, TCF4, ATF-2, NFATc2, and EWS-Fli. Thus, compounds that inhibit expression or activity of PLD in cells can be potentially useful in reducing inflammation and sensitizing resistant cancers during chemotherapy.
Collapse
Affiliation(s)
- Ju Hwan Cho
- Arthur G. James Cancer Hospital Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 4321,
USA
| | - Joong-Soo Han
- Biomedical Research Institute and Department of Biochemistry & Molecular Biology, College of Medicine, Hanyang University, Seoul 04763,
Korea
| |
Collapse
|
133
|
Silva FB, Romero WG, Carvalho ALRDA, Souza GAA, Claudio ERG, Abreu GR. Effects of treatment with chemotherapy and/or tamoxifen on the biomarkers of cardiac injury and oxidative stress in women with breast cancer. Medicine (Baltimore) 2017; 96:e8723. [PMID: 29381962 PMCID: PMC5708961 DOI: 10.1097/md.0000000000008723] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 10/18/2017] [Accepted: 10/21/2017] [Indexed: 12/03/2022] Open
Abstract
There has been an increase in deaths from cardiovascular diseases following breast cancer therapy. Evidence has shown that this outcome is, in part, associated with cardiotoxicity induced by the chemotherapeutic drugs and the increase in oxidative stress. The aim of this study was to evaluate the effects of chemotherapy and hormone therapy with tamoxifen on the biomarkers of cardiac injury and oxidative stress in women with breast cancer.Thirty women were followed-up for 1 year and were divided into 3 groups according to the treatment protocol: women treated only with tamoxifen and clinical follow up for 12 months (Tam, n = 10); women treated only with chemotherapy for 6 months with clinical follow up for an additional 6-month period (Chemo, n = 10); and women who received chemotherapy for 6 months followed by a 6-month period only with tamoxifen therapy and clinical follow up (Chemo + Tam, n = 10). Analysis of the blood levels of cardiac troponin I (cTnI), advanced oxidation protein products (AOPP) and the activity of the plasmatic isoform of the antioxidant enzyme glutathione peroxidase (GPx) was performed before treatment (T0) and at 6 (T6) and 12 (T12) months after treatment.The Chemo group showed higher levels of cTnI (0.065 ± 0.006 ng/mL, P < .05) and AOPP (4.99 ± 0.84 μmol/L, P < .05) and reduced GPx activity (24.4 ± 1.1 nM/min/mL, P < .05) at T12 than the Tam group (cTnI: 0.031 ± 0.001 ng/mL; AOPP: 1.40 ± 0.10 μmol/L; GPx: 28.0 ± 0.7 nM/min/mL) and Chemo + Tam group (cTnI: 0.037 ± 0.002 ng/mL; AOPP: 2.53 ± 0.30 μmol/L; GPx: 29.5 ± 1.0 nM/min/mL).These data support the hypothesis that long-term oxidative stress after chemotherapy may have an impact on cardiovascular diseases and that tamoxifen has cardioprotective effects.
Collapse
Affiliation(s)
| | - Walckiria Garcia Romero
- Department of Nursing, Health Sciences Center, Federal University of Espírito Santo, Vitória-ES, Brazil
| | | | | | | | | |
Collapse
|
134
|
Chemotherapeutic agents induce mitochondrial superoxide production and toxicity but do not alter respiration in skeletal muscle in vitro. Mitochondrion 2017; 42:33-49. [PMID: 29079447 DOI: 10.1016/j.mito.2017.10.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 09/21/2017] [Accepted: 10/23/2017] [Indexed: 12/31/2022]
Abstract
Chemotherapeutic agents (CAs) can independently promote skeletal muscle dysfunction, fatigue and wasting with mitochondrial toxicity implicated as a possible mechanism. Thus, we aimed to characterise the effects of various CAs on mitochondrial function, viability and oxidant production in C2C12 myoblasts and myotubes. All CAs significantly reduced the viable mitochondrial pool but did not affect mitochondrial functional parameters. Doxorubicin and oxaliplatin increased oxidant production in myotubes while all CAs, except for irinotecan, increased oxidant production in myoblasts and reduced myotube diameter. Our data demonstrate CAs mito-toxic effects, highlighting the potential for mitochondria-protective therapeutics to address chemotherapy-induced skeletal muscle damage.
Collapse
|
135
|
Chemotherapy-Induced Tissue Injury: An Insight into the Role of Extracellular Vesicles-Mediated Oxidative Stress Responses. Antioxidants (Basel) 2017; 6:antiox6040075. [PMID: 28956814 PMCID: PMC5745485 DOI: 10.3390/antiox6040075] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 09/21/2017] [Accepted: 09/22/2017] [Indexed: 12/16/2022] Open
Abstract
The short- and long-term side effects of chemotherapy limit the maximum therapeutic dose and impair quality of life of survivors. Injury to normal tissues, especially chemotherapy-induced cardiomyopathy, is an unintended outcome that presents devastating health impacts. Approximately half of the drugs approved by the Food and Drug Administration for cancer treatment are associated with the generation of reactive oxygen species, and Doxorubicin (Dox) is one of them. Dox undergoes redox cycling by involving its quinone structure in the production of superoxide free radicals, which are thought to be instrumental to the role it plays in cardiomyopathy. Dox-induced protein oxidation changes protein function, translocation, and aggregation that are toxic to cells. To maintain cellular homeostasis, oxidized proteins can be degraded intracellularly by ubiquitin-proteasome pathway or by autophagy, depending on the redox status of the cell. Alternatively, the cell can remove oxidized proteins by releasing extracellular vesicles (EVs), which can be transferred to neighboring or distant cells, thereby instigating an intercellular oxidative stress response. In this article, we discuss the role of EVs in oxidative stress response, the potential of EVs as sensitive biomarkers of oxidative stress, and the role of superoxide dismutase in attenuating EV-associated oxidative stress response resulting from chemotherapy.
Collapse
|
136
|
Jeelani R, Khan SN, Shaeib F, Kohan-Ghadr HR, Aldhaheri SR, Najafi T, Thakur M, Morris R, Abu-Soud HM. Cyclophosphamide and acrolein induced oxidative stress leading to deterioration of metaphase II mouse oocyte quality. Free Radic Biol Med 2017; 110:11-18. [PMID: 28499912 PMCID: PMC6854673 DOI: 10.1016/j.freeradbiomed.2017.05.006] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 05/03/2017] [Accepted: 05/07/2017] [Indexed: 12/30/2022]
Abstract
Cyclophosphamide (CTX) is a chemotherapeutic agent widely used to treat ovarian, breast, and hematological cancers as well as autoimmune disorders. Such chemotherapy is associated with reproductive failure and premature ovarian insufficiency. The mechanism by which CTX and/or its main metabolite, acrolein, affect female fertility remains unclear, but it is thought to be caused by an overproduction of reactive oxygen species (ROS). Here, we investigated the effect of CTX on metaphase II mouse oocytes obtained from treated animals (120mg/kg, 24h of single treatment), and oocytes directly exposed to increasing concentrations of CTX and acrolein (n=480; 0, 5, 10, 25, 50, and 100μM) with and without cumulus cells (CCs) for 45min which correlates to the time of maximum peak plasma concentrations after administration. Oocytes were fixed and subjected to indirect immunofluorescence and were scored based on microtubule spindle structure (MT) and chromosomal alignment (CH). Generation of ROS was evaluated using the Cellular Reactive Oxygen Species Detection Assay Kit. Deterioration of oocyte quality was noted when oocytes were obtained from CTX treated mice along with CTX and acrolein treated oocytes in a dose-dependent manner as shown by an increase in poor scores. Acrolein had an impact at a significantly lower level as compared to CTX, plateau at 10μM versus 50μM, respectively. These variation is are associated with the higher amount of ROS generated with acrolein exposure as compared to CTX (p<0.05). Utilization of antioxidant therapy and acrolein scavengers may mitigate the damaging effects of these compounds and help women undergoing such treatment.
Collapse
Affiliation(s)
- Roohi Jeelani
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Sana N Khan
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Faten Shaeib
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Hamid-Reza Kohan-Ghadr
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Sarah R Aldhaheri
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Tohid Najafi
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Mili Thakur
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA; Division of Genetic, Genomic and Metabolic Disorders, Department of Pediatrics and Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Robert Morris
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA; Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Husam M Abu-Soud
- Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
137
|
Farías JG, Molina VM, Carrasco RA, Zepeda AB, Figueroa E, Letelier P, Castillo RL. Antioxidant Therapeutic Strategies for Cardiovascular Conditions Associated with Oxidative Stress. Nutrients 2017; 9:nu9090966. [PMID: 28862654 PMCID: PMC5622726 DOI: 10.3390/nu9090966] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/17/2017] [Accepted: 08/18/2017] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress (OS) refers to the imbalance between the generation of reactive oxygen species (ROS) and the ability to scavenge these ROS by endogenous antioxidant systems, where ROS overwhelms the antioxidant capacity. Excessive presence of ROS results in irreversible damage to cell membranes, DNA, and other cellular structures by oxidizing lipids, proteins, and nucleic acids. Oxidative stress plays a crucial role in the pathogenesis of cardiovascular diseases related to hypoxia, cardiotoxicity and ischemia-reperfusion. Here, we describe the participation of OS in the pathophysiology of cardiovascular conditions such as myocardial infarction, anthracycline cardiotoxicity and congenital heart disease. This review focuses on the different clinical events where redox factors and OS are related to cardiovascular pathophysiology, giving to support for novel pharmacological therapies such as omega 3 fatty acids, non-selective betablockers and microRNAs.
Collapse
Affiliation(s)
- Jorge G Farías
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4780000, Chile.
| | - Víctor M Molina
- Unidad de Cuidados Intensivos, Hospital de Niños Roberto del Río, Santiago 7500922, Chile.
- Unidad de Cuidados Intensivos Pediátricos, Hospital Clínico Pontificia Universidad Católica de Chile, Santiago 7500922, Chile.
| | - Rodrigo A Carrasco
- Laboratorio de Investigación Biomédica, Departamento de Medicina Interna, Hospital del Salvador, Santiago 7500922, Chile.
- Departamento de Cardiología, Clínica Alemana, Santiago 7500922, Chile.
| | - Andrea B Zepeda
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4780000, Chile.
| | - Elías Figueroa
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4780000, Chile.
- Núcleo de Investigación en Producción Alimentaria, BIOACUI, Escuela de Acuicultura, Universidad Católica de Temuco, Temuco 4780000, Chile.
| | - Pablo Letelier
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4780000, Chile.
- School of Health Sciences, Universidad Católica de Temuco, Temuco 4780000, Chile.
| | - Rodrigo L Castillo
- Laboratorio de Investigación Biomédica, Departamento de Medicina Interna, Hospital del Salvador, Santiago 7500922, Chile.
- Programa de Fisiopatología Oriente, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile.
| |
Collapse
|
138
|
Dewanjee S, Joardar S, Bhattacharjee N, Dua TK, Das S, Kalita J, Manna P. Edible leaf extract of Ipomoea aquatica Forssk. (Convolvulaceae) attenuates doxorubicin-induced liver injury via inhibiting oxidative impairment, MAPK activation and intrinsic pathway of apoptosis. Food Chem Toxicol 2017; 105:322-336. [PMID: 28478100 DOI: 10.1016/j.fct.2017.05.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/22/2017] [Accepted: 05/02/2017] [Indexed: 12/17/2022]
Abstract
Ipomoea aquatica Forssk. (Convolvulaceae) is an aquatic vegetable traditionally employed against toxic effects of xenobiotics. The present study has been designed to investigate the molecular mechanism underlying the beneficial role of the edible (aqueous) leaf extract of I. aquatica (AEIA) against doxorubicin (Dox)-induced liver injury. AEIA exhibited a dose-dependent (∼400 μg/ml) increase in cell viability against Dox (1 μM) in isolated rodent hepatocytes. AEIA (400 μg/ml) prevented the Dox-induced increase in ROS, redox imbalance, and activation of mitogen activated protein kinases (MAPK) and intrinsic pathway of apoptosis in hepatocytes. In the in vivo assay, administration of AEIA (100 mg/kg, p.o.) against Dox (3 mg/kg, i.p.) also reduced the oxidative impairment, DNA fragmentation, ATP formation, and up-regulated the mitochondrial co-enzymes Qs in the liver tissues of Wistar rats. Histological assessments were in agreement with the biochemical findings. Substantial quantities of phyto-antioxidants in AEIA may mediate its beneficial function against Dox-induced liver injury.
Collapse
Affiliation(s)
- Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| | - Swarnalata Joardar
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Niloy Bhattacharjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Tarun K Dua
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Subhadip Das
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Jatin Kalita
- Biological Science and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat, Assam 785006, India
| | - Prasenjit Manna
- Biological Science and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat, Assam 785006, India.
| |
Collapse
|
139
|
Chen T, Shen HM, Deng ZY, Yang ZZ, Zhao RL, Wang L, Feng ZP, Liu C, Li WH, Liu ZJ. A herbal formula, SYKT, reverses doxorubicin‑induced myelosuppression and cardiotoxicity by inhibiting ROS‑mediated apoptosis. Mol Med Rep 2017; 15:2057-2066. [PMID: 28260045 PMCID: PMC5364961 DOI: 10.3892/mmr.2017.6272] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 01/17/2017] [Indexed: 12/03/2022] Open
Abstract
Doxorubicin (DOX) is an antineoplastic drug widely used for the treatment of various types of cancer; however, it can induce severe side effects, such as myelosuppression and cardiotoxicity. Sanyang Xuedai (SYKT) is a natural medicine originating from an ancient prescription of the Dai nationality in Southwest China. With eight Chinese herbal medicines, including sanguis draconis, radix et rhizoma notoginseng, radix et rhizoma glycyrrhizae and radix angelicae sinensis as the primary ingredients, SYKT has been reported to possess numerous biological functions. The present study investigated whether SYKT can confer protection against DOX-induced myelosuppression and cardiotoxicity, and explored the potential mechanism involved. Mice were treated with DOX, SYKT or a combination of the two; hematopoietic functions were assessed by measuring the number of peripheral blood cells, cluster of differentiation CD34+/CD44+ bone marrow cells and apoptotic cells. Myocardial enzymes, including aspartate aminotransferase, lactate dehydrogenase, creatine kinase (CK) and its isoform CK-MB, were assessed using a biochemical analyzer. The apoptotic rate of cardiomyocytes was assessed using flow cytometry. Histopathological analysis was conducted using hematoxylin-eosin staining. Intracellular reactive oxygen species (ROS) production was evaluated using a dichlorofluorescein intensity assay. The mice treated with DOX exhibited a reduced survival rate, reduced peripheral blood and CD34+/CD44+ cell counts, elevated myocardial enzymes and apoptotic indices in bone marrow cells and cardiomyocytes, all of which were effectively prevented by SYKT co-administration. Furthermore, bone marrow cells and myocytes from mice treated with DOX demonstrated increased dichlorofluorescein intensity, which was attenuated by SYKT. Notably, SYKT did not interfere with the effects of DOX on tumor volume or the induction of tumor cell apoptosis in tumor-bearing mice. The present study indicated that SYKT may counteract DOX-induced myelosuppression and cardiotoxicity through inhibiting ROS-mediated apoptosis. These findings suggested that SYKT may have potential as a means to counteract the potentially fatal hematopoietic and cardiac complications associated with DOX treatment.
Collapse
Affiliation(s)
- Ting Chen
- Department of Nuclear Medicine, Tumor Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical College, Kunming, Yunnan 650118, P.R. China
| | - Hong-Mei Shen
- Department of Combination of Chinese Traditional and Western Medicine, Tumor Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical College, Kunming, Yunnan 650118, P.R. China
| | - Zhi-Yong Deng
- Department of Nuclear Medicine, Tumor Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical College, Kunming, Yunnan 650118, P.R. China
| | - Zuo-Zhang Yang
- Department of Orthopaedics, Tumor Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical College, Kunming, Yunnan 650118, P.R. China
| | - Rui-Lian Zhao
- Department of Combination of Chinese Traditional and Western Medicine, Tumor Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical College, Kunming, Yunnan 650118, P.R. China
| | - Li Wang
- The Institute of Molecular and Clinical Medicine of Kunming Medical College, Kunming, Yunnan 650500, P.R. China
| | - Zhi-Ping Feng
- Department of Nuclear Medicine, Tumor Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical College, Kunming, Yunnan 650118, P.R. China
| | - Chao Liu
- Department of Nuclear Medicine, Tumor Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical College, Kunming, Yunnan 650118, P.R. China
| | - Wen-Hui Li
- Department of Radiotherapy, Tumor Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical College, Kunming, Yunnan 650118, P.R. China
| | - Zhi-Jie Liu
- The Institute of Molecular and Clinical Medicine of Kunming Medical College, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
140
|
Dysregulation of cytokine mediated chemotherapy induced cognitive impairment. Pharmacol Res 2017; 117:267-273. [DOI: 10.1016/j.phrs.2017.01.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 01/03/2017] [Indexed: 11/19/2022]
|
141
|
Repka CP, Hayward R. Oxidative Stress and Fitness Changes in Cancer Patients after Exercise Training. Med Sci Sports Exerc 2016; 48:607-14. [PMID: 26587845 DOI: 10.1249/mss.0000000000000821] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
INTRODUCTION The purpose of this study was to determine the effect of an exercise intervention (EX) on muscular strength, cardiorespiratory fitness (CRF), and oxidative stress in cancer survivors compared with a nonexercising cancer control group (CON). METHODS Fifteen cancer patients and seven age-matched individuals with no history of cancer (NC) participated in this study. A blood draw and assessments of muscular strength and CRF were administered to cancer survivors within 6 wk of completing radiation or chemotherapy, and again 10 wk later. Eight cancer patients completed a 10-wk supervised exercise intervention, whereas seven continued standard care. Baseline oxidative stress was compared between cancer patients and the NC group. Changes in plasma protein carbonyls, 8-OHdG, and Trolox equivalent antioxidant capacity were compared between groups using repeated-measures ANOVA, and correlations between fitness and oxidative stress changes were evaluated. RESULTS Baseline antioxidant capacity was significantly lower, and plasma protein carbonyls were significantly higher in cancer patients compared with NC (P = 0.001). EX had a significant increase in antioxidant capacity (P < 0.001) and decrease in protein carbonyls (P = 0.023), whereas CON did not. Improvements in composite arm (41%, P = 0.002) and leg strength (34%, P = 0.008), isometric handgrip strength (11%, P = 0.015), and V˙O2peak (16%, P = 0.018) were significant in EX but not in CON. 8-OHdG changes were significantly correlated with changes in V˙O2peak (r = -0.89, P < 0.001), arm strength (r = -0.67, P = 0.004), and leg strength (r = -0.56, P = 0.019). CONCLUSION A whole-body exercise intervention for cancer survivors may be an effective method of concurrently increasing muscular strength, CRF, and antioxidant capacity while decreasing markers of oxidative stress.
Collapse
Affiliation(s)
- Chris P Repka
- 1Department of Health Sciences, Northern Arizona University, Flagstaff, AZ; 2School of Sport and Exercise Science and the Rocky Mountain Cancer Rehabilitation Institute, University of Northern Colorado, Greeley, CO
| | | |
Collapse
|
142
|
Yao YF, Liu X, Li WJ, Shi ZW, Yan YX, Wang LF, Chen M, Xie MY. (-)-Epigallocatechin-3-gallate alleviates doxorubicin-induced cardiotoxicity in sarcoma 180 tumor-bearing mice. Life Sci 2016; 180:151-159. [PMID: 27956351 DOI: 10.1016/j.lfs.2016.12.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 11/23/2016] [Accepted: 12/08/2016] [Indexed: 12/18/2022]
Abstract
AIMS (-)-Epigallocatechin-3-gallate (EGCG), a major green tea polyphenol compound, plays an important role in the prevention of cardiovascular disease and cancer. The present study aimed to investigate the effects of EGCG on doxorubicin (DOX)-induced cardiotoxicity in Sarcoma 180 (S180) tumor-bearing mice. MAIN METHODS S180 tumor-bearing mice were established by subcutaneous inoculation of S180 cells attached to the axillary region. The extent of myocardial injury was accessed by the amount of lactate dehydrogenase (LDH) released in serum. Heart tissue was morphologically studied with transmission electron microscopy. Apoptosis, reactive oxygen species (ROS) generation, mitochondrial membrane potential (ΔѰm) as well as calcium concentration were measured by flow cytometric analysis. Expression levels of manganese superoxide dismutase (MnSOD) were analyzed by Western blot. KEY FINDINGS Results showed that the combination with EGCG and DOX significantly inhibited tumor growth and enhanced induction of apoptosis compared with DOX alone. Moreover, administration of EGCG could suppress DOX-induced cardiotoxicity as evidenced by alleviating LDH release and apoptosis in cardiomyocyte. EGCG-evoked cardioprotection was in association with the increase of ΔѰm and MnSOD expression. EGCG was also found to attenuate ROS generation and myocardial calcium overload in Sarcoma 180 tumor-bearing mice subjected to DOX. SIGNIFICANCE EGCG alleviated DOX-induced cardiotoxicity possibly in part mediated by increasing of MnSOD and Ѱm, reducing myocardial calcium overload and subsequently attenuating the apoptosis and LDH release. Our findings suggest that co-administration of EGCG and DOX have potential as a feasible strategy to mitigate cardiotoxicity of DOX without compromising its chemotherapeutic value.
Collapse
Affiliation(s)
- Yu-Fei Yao
- Chinese Liberation Army No. 94 Hospital, No. 1028, Jinggangshan Avenue, Nanchang 330000, China; The Great Wall Affiliated Hospital, Nanchang University, No. 1028, Jinggangshan Avenue, Nanchang 330000, China
| | - Xiang Liu
- School of Basic Medical Sciences, Nanchang University, No. 999 Xuefu Road, Nanchang 330031, China
| | - Wen-Juan Li
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Zi-Wei Shi
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China
| | - Yu-Xin Yan
- School of Basic Medical Sciences, Nanchang University, No. 999 Xuefu Road, Nanchang 330031, China
| | - Le-Feng Wang
- School of Basic Medical Sciences, Nanchang University, No. 999 Xuefu Road, Nanchang 330031, China
| | - Ming Chen
- School of Basic Medical Sciences, Nanchang University, No. 999 Xuefu Road, Nanchang 330031, China
| | - Ming-Yong Xie
- State Key Laboratory of Food Science and Technology, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| |
Collapse
|
143
|
Identification of aberrant DNA methylation involved in chemoradiation-resistant HCT116 cells via methylation-specific microarray. Mol Cell Toxicol 2016. [DOI: 10.1007/s13273-016-0039-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
144
|
Guinan EM, Doyle SL, O’Neill L, Dunne MR, Foley EK, O’Sullivan J, Reynolds JV, Hussey J. Effects of a multimodal rehabilitation programme on inflammation and oxidative stress in oesophageal cancer survivors: the ReStOre feasibility study. Support Care Cancer 2016; 25:749-756. [DOI: 10.1007/s00520-016-3455-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 10/10/2016] [Indexed: 12/27/2022]
|
145
|
Román DA, Pizarro I, Rivera L, Ávila J, Cortés P. Magnesium, zinc, arsenic, selenium and platinum urinary excretion from cancer patients of Antofagasta region, Chile: multi-metal approach. JRSM Open 2016; 7:2054270416660932. [PMID: 27757244 PMCID: PMC5052785 DOI: 10.1177/2054270416660932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Objectives To evaluate the short-term 24 h urinary excretion of platinum, arsenic, selenium, magnesium and zinc in patients with lung cancer and with cancer other than lungs treated with cisplatin or/and carboplatin from Antofagasta, Chile. Design Urine measurements of Pt and Se were made by inductively coupled plasma optical emission spectrometry, As by hydride-generation atomic absorption spectrometry and Mg and Zn by means of flame furnace atomic absorption spectrometry. Setting All samples were provided by the Oncological Centre of Antofagasta Regional Hospital (Region of Antofagasta, Chile). Participants Ninety 24-h urine samples from cancer patients after the infusion of Pt-base drugs and 10 24-h urine samples from cancer patients not treated with metal-base drugs. Main outcome measures Concentrations of Pt, Se, As, Zn and Mg coming from 24-h urine samples. Results Pt excreted was not significantly different between patients with lung and other cancers treated with cisplatin. The excretion of Mg, Zn and Se was greater than As. Then, Pt favours the excretion of essential elements. For lung and other types of cancers treated with drugs without Pt, excretion of Mg, Zn and Se was also greater than that of As, suggesting antagonism Mg-Zn-Se–anti-cancer drug relationship. Conclusions The amounts of Mg, Zn and Se excreted were greater than for As either with or without Pt-containing drugs, suggesting antagonist Mg-Zn-Se–anti-cancer drug relationships. The excretion of As, Mg, Zn and Se is induced by Pt. Knowledge obtained can contribute to understanding the arsenic cancer mechanism and the As-Mg-Zn-Se-Pt inter-element association for lung cancer and other types of cancer.
Collapse
Affiliation(s)
- D A Román
- Department of Chemistry, Faculty of Basic Science, Bioinorganic and Environmental Analytical Chemistry Laboratory, University of Antofagasta, Antofagasta 124000, Chile
| | - I Pizarro
- Department of Chemistry, Faculty of Basic Science, Bioinorganic and Environmental Analytical Chemistry Laboratory, University of Antofagasta, Antofagasta 124000, Chile
| | - L Rivera
- Department of Chemistry, Faculty of Basic Science, Bioinorganic and Environmental Analytical Chemistry Laboratory, University of Antofagasta, Antofagasta 124000, Chile
| | - J Ávila
- Department of Chemistry, Faculty of Basic Science, Bioinorganic and Environmental Analytical Chemistry Laboratory, University of Antofagasta, Antofagasta 124000, Chile
| | - P Cortés
- Department of Chemistry, Faculty of Basic Science, Bioinorganic and Environmental Analytical Chemistry Laboratory, University of Antofagasta, Antofagasta 124000, Chile
| |
Collapse
|
146
|
Pimentel-Gutiérrez HJ, Bobadilla-Morales L, Barba-Barba CC, Ortega-De-La-Torre C, Sánchez-Zubieta FA, Corona-Rivera JR, González-Quezada BA, Armendáriz-Borunda JS, Silva-Cruz R, Corona-Rivera A. Curcumin potentiates the effect of chemotherapy against acute lymphoblastic leukemia cells via downregulation of NF-κB. Oncol Lett 2016; 12:4117-4124. [PMID: 27895780 DOI: 10.3892/ol.2016.5217] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 05/16/2016] [Indexed: 11/06/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) accounts for 30% of all pediatric cancers. Currently available treatments exhibit toxicity and certain patients may develop resistance. Thus, less toxic and chemoresistance-reversal agents are required. In the present study, the potential effect of curcumin, a component of Curcuma longa, as a pharmacological co-adjuvant of several chemotherapeutic agents against ALL, including prednisone, 6-mercaptopurine, dexamethasone, cyclophosphamide, l-asparaginase, vincristine, daunorubicin, doxorubicin, methotrexate and cytarabine, was investigated in the REH ALL cell line cultures treated in combination with chemotherapeutic agents and curcumin. The results of cell viability, gene expression and activation of NF-κB and caspase 3 indicated that curcumin potentiates the anticancer effects of the aforementioned chemotherapeutic agents in the REH ALL cell line. Following treatment with the above chemotherapeutic agents, curcumin enhanced caspase-3 activation and downregulated nuclear factor-kappa B (NF-κB) activation. Curcumin also downregulated the oxidative stress induced by certain chemotherapies. Notably, curcumin did not affect the gene expression of cell survival proteins such as B-cell lymphoma (Bcl)-2, Bcl-extra large, survivin, c-Myc and cyclin D1, which are regulated by the NF-κB transcription factor. In conclusion, curcumin has the potential to improve the effect of chemotherapeutic agents against ALL.
Collapse
Affiliation(s)
- Helia Judith Pimentel-Gutiérrez
- Cytogenetics, Genotoxicity and Biomonitoring Laboratory, Human Genetic Institute 'Dr. Enrique Corona Rivera', PhD Program in Molecular Biology, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco 44340, México; Cytogenetics Unit, Pediatric Hematology and Oncology Service, Pediatric Division, Civil Hospital of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Lucina Bobadilla-Morales
- Cytogenetics, Genotoxicity and Biomonitoring Laboratory, Human Genetic Institute 'Dr. Enrique Corona Rivera', PhD Program in Molecular Biology, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco 44340, México; Cytogenetics Unit, Pediatric Hematology and Oncology Service, Pediatric Division, Civil Hospital of Guadalajara, Guadalajara, Jalisco 44340, México
| | - César Cenobio Barba-Barba
- Cytogenetics Unit, Pediatric Hematology and Oncology Service, Pediatric Division, Civil Hospital of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Citlalli Ortega-De-La-Torre
- Cytogenetics Unit, Pediatric Hematology and Oncology Service, Pediatric Division, Civil Hospital of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Fernando Antonio Sánchez-Zubieta
- Cytogenetics Unit, Pediatric Hematology and Oncology Service, Pediatric Division, Civil Hospital of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Jorge Román Corona-Rivera
- Cytogenetics, Genotoxicity and Biomonitoring Laboratory, Human Genetic Institute 'Dr. Enrique Corona Rivera', PhD Program in Molecular Biology, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco 44340, México; Cytogenetics Unit, Pediatric Hematology and Oncology Service, Pediatric Division, Civil Hospital of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Betsy Annel González-Quezada
- Cytogenetics, Genotoxicity and Biomonitoring Laboratory, Human Genetic Institute 'Dr. Enrique Corona Rivera', PhD Program in Molecular Biology, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Juan S Armendáriz-Borunda
- Molecular Biology and Gene Therapy Institute, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Rocío Silva-Cruz
- Cytogenetics, Genotoxicity and Biomonitoring Laboratory, Human Genetic Institute 'Dr. Enrique Corona Rivera', PhD Program in Molecular Biology, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco 44340, México
| | - Alfredo Corona-Rivera
- Cytogenetics, Genotoxicity and Biomonitoring Laboratory, Human Genetic Institute 'Dr. Enrique Corona Rivera', PhD Program in Molecular Biology, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco 44340, México; Cytogenetics Unit, Pediatric Hematology and Oncology Service, Pediatric Division, Civil Hospital of Guadalajara, Guadalajara, Jalisco 44340, México
| |
Collapse
|
147
|
Pharmacokinetics and pharmacodynamics evaluation of a thermosensitive chitosan based hydrogel containing liposomal doxorubicin. Eur J Pharm Sci 2016; 92:137-45. [DOI: 10.1016/j.ejps.2016.07.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 06/22/2016] [Accepted: 07/02/2016] [Indexed: 11/23/2022]
|
148
|
Kwatra M, Jangra A, Mishra M, Sharma Y, Ahmed S, Ghosh P, Kumar V, Vohora D, Khanam R. Naringin and Sertraline Ameliorate Doxorubicin-Induced Behavioral Deficits Through Modulation of Serotonin Level and Mitochondrial Complexes Protection Pathway in Rat Hippocampus. Neurochem Res 2016; 41:2352-2366. [PMID: 27209303 DOI: 10.1007/s11064-016-1949-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 04/02/2016] [Accepted: 05/06/2016] [Indexed: 01/16/2023]
Abstract
The present study was designed to investigate the neuroprotective effect of naringin (NR) alone as well as its combination with sertraline (SRT) against doxorubicin (DOX)-induced neurobehavioral and neurochemical anomalies. DOX (15 mg/kg; i.p.) administration caused behavioral alterations, oxidative stress, neuroinflammation, mitochondrial dysfunction and monoamines alteration in male Wistar rats. NR (50 and 100 mg/kg; i.p.) and SRT (5 mg/kg; i.p.) treatment significantly attenuated DOX-induced anxiety and depressive-like behavior as evident from elevated plus maze (EPM) and modified forced swimming test (mFST), respectively. NR treatment significantly attenuated DOX-induced raised plasma corticosterone (CORT), tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β) levels in the hippocampus (HC). Furthermore, we found that combination of NR and SRT regimen ameliorated DOX-induced behavioral anomalies through modulation of the 5-HT level and mitochondrial complexes protection pathway along with alleviation of oxidative stress in the HC region. Therefore, NR treatment alone or in combination with SRT could be beneficial against DOX-induced neurotoxicity.
Collapse
Affiliation(s)
- Mohit Kwatra
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), New Delhi, 110062, India.
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Guwahati, Assam, 781032, India.
| | - Ashok Jangra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Guwahati, Assam, 781032, India
| | - Murli Mishra
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Yogita Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Guwahati, Assam, 781032, India
| | - Sahabuddin Ahmed
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Guwahati, Guwahati, Assam, 781032, India
| | - Pinaki Ghosh
- Department of Pharmacology, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Erandwane, Pune, Maharashtra, 411038, India
| | - Vikas Kumar
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
| | - Divya Vohora
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
| | - Razia Khanam
- Neurobehavioral Pharmacology Laboratory, Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
- Department of Pharmacology, Gulf Medical University, Ajman, United Arab Emirates
| |
Collapse
|
149
|
Involvement of p53 in insulin-like growth factor binding protein-3 regulation in the breast cancer cell response to DNA damage. Oncotarget 2016; 6:26583-98. [PMID: 26378048 PMCID: PMC4694938 DOI: 10.18632/oncotarget.5612] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 08/26/2015] [Indexed: 11/25/2022] Open
Abstract
Chemotherapy drugs that induce apoptosis by causing DNA double-strand breaks, upregulate the tumor suppressor p53. This study investigated the regulation of the growth-regulatory protein insulin-like growth factor binding protein-3 (IGFBP-3), a p53 target, by DNA-damaging agents in breast cancer cells. IGFBP-3 was upregulated 1.4- to 13-fold in response to doxorubicin and etoposide in MCF-10A, Hs578T, MCF-7 and T47D cells, which express low to moderate basal levels of IGFBP-3. In contrast, IGFBP-3 was strongly downregulated by these agents in cells with high basal levels of IGFBP-3 (MDA-MB-231, MDA-MB-436 and MDA-MB-468). In MDA-MB-468 cells containing the R273H p53 mutation, reported to display gain-of-function properties, chemotherapy-induced suppression of IGFBP-3 was not reversed by the p53 reactivating drug, PRIMA-1, or by p53 silencing, suggesting that the decrease in IGFBP-3 following DNA damage is not a mutant p53 gain-of-function response. SiRNA-mediated downregulation of endogenous IGFBP-3 modestly attenuated doxorubicin-induced apoptosis in MDA-MB-468 and Hs578T cells. IGFBP-3 downregulation in some breast cancer cell lines in response to DNA-damaging chemotherapy may have clinical implications because suppression of IGFBP-3 may modulate the apoptotic response. These observations provide further evidence that endogenous IGFBP-3 plays a role in breast cancer cell responsiveness to DNA damaging therapy.
Collapse
|
150
|
Supriya R, Tam BT, Pei XM, Lai CW, Chan LW, Yung BY, Siu PM. Doxorubicin Induces Inflammatory Modulation and Metabolic Dysregulation in Diabetic Skeletal Muscle. Front Physiol 2016; 7:323. [PMID: 27512375 PMCID: PMC4961708 DOI: 10.3389/fphys.2016.00323] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/14/2016] [Indexed: 11/13/2022] Open
Abstract
Anti-cancer agent doxorubicin (DOX) has been demonstrated to worsen insulin signaling, engender muscle atrophy, trigger pro-inflammation, and induce a shift to anaerobic glycolytic metabolism in skeletal muscle. The myotoxicity of DOX in diabetic skeletal muscle remains largely unclear. This study examined the effects of DOX on insulin signaling, muscle atrophy, pro-/anti-inflammatory microenvironment, and glycolysis metabolic regulation in skeletal muscle of db/db diabetic and db/+ non-diabetic mice. Non-diabetic db/+ mice and diabetic db/db mice were randomly assigned to the following groups: db/+CON, db/+DOX, db/dbCON, and db/dbDOX. Mice in db/+DOX and db/dbDOX groups were intraperitoneally injected with DOX at a dose of 15 mg per kg body weight whereas mice in db/+CON and db/dbCON groups were injected with the same volume of saline instead of DOX. Gastrocnemius was immediately harvested, weighed, washed with cold phosphate buffered saline, frozen in liquid nitrogen, and stored at -80°C for later analysis. The effects of DOX on diabetic muscle were neither seen in insulin signaling markers (Glut4, pIRS1Ser(636∕639), and pAktSer(473)) nor muscle atrophy markers (muscle mass, MuRF1 and MAFbx). However, DOX exposure resulted in enhancement of pro-inflammatory favoring microenvironment (as indicated by TNF-α, HIFα and pNFκBp65) accompanied by diminution of anti-inflammatory favoring microenvironment (as indicated by IL15, PGC1α and pAMPKβ1Ser108). Metabolism of diabetic muscle was shifted to anaerobic glycolysis after DOX exposure as demonstrated by our analyses of PDK4, LDH and pACCSer(79). Our results demonstrated that there might be a link between inflammatory modulation and the dysregulation of aerobic glycolytic metabolism in DOX-injured diabetic skeletal muscle. These findings help to understand the pathogenesis of DOX-induced myotoxicity in diabetic muscle.
Collapse
Affiliation(s)
- Rashmi Supriya
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University Hong Kong, China
| | - Bjorn T Tam
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University Hong Kong, China
| | - Xiao M Pei
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University Hong Kong, China
| | - Christopher W Lai
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University Hong Kong, China
| | - Lawrence W Chan
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University Hong Kong, China
| | - Benjamin Y Yung
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University Hong Kong, China
| | - Parco M Siu
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University Hong Kong, China
| |
Collapse
|