351
|
Schink M, Leipold E, Schirmeyer J, Schönherr R, Hoshi T, Heinemann SH. Reactive species modify NaV1.8 channels and affect action potentials in murine dorsal root ganglion neurons. Pflugers Arch 2016; 468:99-110. [PMID: 26383867 PMCID: PMC5165275 DOI: 10.1007/s00424-015-1735-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/29/2015] [Accepted: 09/07/2015] [Indexed: 01/10/2023]
Abstract
Dorsal root ganglion (DRG) neurons are important relay stations between the periphery and the central nervous system and are essential for somatosensory signaling. Reactive species are produced in a variety of physiological and pathophysiological conditions and are known to alter electric signaling. Here we studied the influence of reactive species on the electrical properties of DRG neurons from mice with the whole-cell patch-clamp method. Even mild stress induced by either low concentrations of chloramine-T (10 μM) or low-intensity blue light irradiation profoundly diminished action potential frequency but prolonged single action potentials in wild-type neurons. The impact on evoked action potentials was much smaller in neurons deficient of the tetrodotoxin (TTX)-resistant voltage-gated sodium channel NaV1.8 (NaV1.8(-/-)), the channel most important for the action potential upstroke in DRG neurons. Low concentrations of chloramine-T caused a significant reduction of NaV1.8 peak current and, at higher concentrations, progressively slowed down inactivation. Blue light had a smaller effect on amplitude but slowed down NaV1.8 channel inactivation. The observed effects were less apparent for TTX-sensitive NaV channels. NaV1.8 is an important reactive-species-sensitive component in the electrical signaling of DRG neurons, potentially giving rise to loss-of-function and gain-of-function phenomena depending on the type of reactive species and their effective concentration and time of exposure.
Collapse
Affiliation(s)
- Martin Schink
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena & Jena University Hospital, Hans-Knöll-Str. 2, 07745, Jena, Germany
| | - Enrico Leipold
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena & Jena University Hospital, Hans-Knöll-Str. 2, 07745, Jena, Germany
| | - Jana Schirmeyer
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena & Jena University Hospital, Hans-Knöll-Str. 2, 07745, Jena, Germany
| | - Roland Schönherr
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena & Jena University Hospital, Hans-Knöll-Str. 2, 07745, Jena, Germany
| | - Toshinori Hoshi
- Department of Physiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Stefan H Heinemann
- Center for Molecular Biomedicine, Department of Biophysics, Friedrich Schiller University Jena & Jena University Hospital, Hans-Knöll-Str. 2, 07745, Jena, Germany.
| |
Collapse
|
352
|
Inhibition of ATP synthesis by fenbufen and its conjugated metabolites in rat liver mitochondria. Toxicol In Vitro 2015; 31:23-9. [PMID: 26612354 DOI: 10.1016/j.tiv.2015.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/11/2015] [Accepted: 11/19/2015] [Indexed: 11/21/2022]
Abstract
Fenbufen is an arylpropionic acid derivative belonging to the group of non-steroidal anti-inflammatory drugs (NSAIDs). Even though fenbufen is considered a safe drug, some adverse reactions including hepatic events have been reported. To investigate whether mitochondrial damage could be involved in the drug induced liver injury (DILI) by fenbufen, the inhibitory effect of fenbufen and its conjugated metabolites on oxidative phosphorylation (ATP synthesis) in rat liver mitochondria was investigated. Fenbufen glucuronide (F-GlcA), fenbufen-N-acetyl cysteine-thioester (F-NAC) and fenbufen-S-glutathione thioester (F-SG) were found to be more potent inhibitors compared to parent fenbufen (F), whereas fenbufen-O-carnitine (F-carn), fenbufen-glycine (F-gly) and fenbufen-N-acetyl lysine amide (F-NAL) were less potent compared to fenbufen. Fenbufen-CoA thioester (F-CoA) was equally potent as fenbufen in inhibiting ATP synthesis. Fenbufen showed time and concentration dependent inhibition of ATP synthesis with Kinact of 4.4 min(-1) and KI of 0.88 μM and Kinact/KI ratio of 5.01 min(-1) μM(-1). Data show that fenbufen did not act through opening MPT pore, nor did incubation of mitochondria with reduced GSH and fenbufen show any protective effect on fenbufen mediated inhibition of oxidative phosphorylation. Inclusion of NADPH in mitochondrial preparations with fenbufen did not modulate the inhibitory effects, suggesting no role of CYP mediated oxidative metabolites on the ATP synthesis in isolated mitochondria. The results from the present experiments provide evidence that fenbufen and its metabolites could be involved in mitochondrial toxicity through inhibition of ATP synthesis.
Collapse
|
353
|
Farias CF, Massaoka MH, Girola N, Azevedo RA, Ferreira AK, Jorge SD, Tavares LC, Figueiredo CR, Travassos LR. Benzofuroxan derivatives N-Br and N-I induce intrinsic apoptosis in melanoma cells by regulating AKT/BIM signaling and display anti metastatic activity in vivo. BMC Cancer 2015; 15:807. [PMID: 26503030 PMCID: PMC4621849 DOI: 10.1186/s12885-015-1835-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 10/19/2015] [Indexed: 12/14/2022] Open
Abstract
Background Malignant melanoma is an aggressive type of skin cancer, and despite recent advances in treatment, the survival rate of the metastatic form remains low. Nifuroxazide analogues are drugs based on the substitution of the nitrofuran group by benzofuroxan, in view of the pharmacophore similarity of the nitro group, improving bioavailability, with higher intrinsic activity and less toxicity. Benzofuroxan activity involves the intracellular production of free-radical species. In the present work, we evaluated the antitumor effects of different benzofuroxan derivatives in a murine melanoma model. Methods B16F10-Nex2 melanoma cells were used to investigate the antitumor effects of Benzofuroxan derivatives in vitro and in a syngeneic melanoma model in C57Bl/6 mice. Cytotoxicity, morphological changes and reactive oxygen species (ROS) were assessed by a diphenyltetrasolium reagent, optical and fluorescence microscopy, respectively. Annexin-V binding and mitochondrial integrity were analyzed by flow cytometry. Western blotting and colorimetry identified cell signaling proteins. Results Benzofuroxan N-Br and N-I derivatives were active against murine and human tumor cell lines, exerting significant protection against metastatic melanoma in a syngeneic model. N-Br and N-I induce apoptosis in melanoma cells, evidenced by specific morphological changes, DNA condensation and degradation, and phosphatidylserine translocation in the plasma membrane. The intrinsic mitochondrial pathway in B16F10-Nex2 cells is suggested owing to reduced outer membrane potential in mitochondria, followed by caspase −9, −3 activation and cleavage of PARP. The cytotoxicity of N-Br and N-I in B16F10-Nex2 cells is mediated by the generation of ROS, inhibited by pre-incubation of the cells with N-acetylcysteine (NAC). The induction of ROS by N-Br and N-I resulted in the inhibition of AKT activation, an important molecule related to tumor cell survival, followed by upregulation of BIM. Conclusion We conclude that N-Br and N-I are promising agents aiming at cancer treatment. They may be useful in melanoma therapy as inducers of intrinsic apoptosis and by exerting significant antitumor activity against metastatic melanoma, as presently shown in syngeneic mice.
Collapse
Affiliation(s)
- C F Farias
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, Rua Botucatu 862, 8 andar, São Paulo, SP, 04023-062, Brazil.
| | - M H Massaoka
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, Rua Botucatu 862, 8 andar, São Paulo, SP, 04023-062, Brazil.
| | - N Girola
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, Rua Botucatu 862, 8 andar, São Paulo, SP, 04023-062, Brazil.
| | - R A Azevedo
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| | - A K Ferreira
- Experimental Physiopathology, Faculty of Medicine, University of São Paulo, São Paulo, SP, Brazil.
| | - S D Jorge
- Laboratory of Tumor Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| | - L C Tavares
- Department of Biochemical and Pharmaceutical Technology, University of São Paulo, São Paulo, SP, Brazil.
| | - C R Figueiredo
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, Rua Botucatu 862, 8 andar, São Paulo, SP, 04023-062, Brazil.
| | - L R Travassos
- Experimental Oncology Unit (UNONEX), Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, Rua Botucatu 862, 8 andar, São Paulo, SP, 04023-062, Brazil.
| |
Collapse
|
354
|
Angsutararux P, Luanpitpong S, Issaragrisil S. Chemotherapy-Induced Cardiotoxicity: Overview of the Roles of Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:795602. [PMID: 26491536 PMCID: PMC4602327 DOI: 10.1155/2015/795602] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 05/17/2015] [Indexed: 02/02/2023]
Abstract
Chemotherapy-induced cardiotoxicity is a serious complication that poses a serious threat to life and limits the clinical use of various chemotherapeutic agents, particularly the anthracyclines. Understanding molecular mechanisms of chemotherapy-induced cardiotoxicity is a key to effective preventive strategies and improved chemotherapy regimen. Although no reliable and effective preventive treatment has become available, numerous evidence demonstrates that chemotherapy-induced cardiotoxicity involves the generation of reactive oxygen species (ROS). This review provides an overview of the roles of oxidative stress in chemotherapy-induced cardiotoxicity using doxorubicin, which is one of the most effective chemotherapeutic agents against a wide range of cancers, as an example. Current understanding in the molecular mechanisms of ROS-mediated cardiotoxicity will be explored and discussed, with emphasis on cardiomyocyte apoptosis leading to cardiomyopathy. The review will conclude with perspectives on model development needed to facilitate further progress and understanding on chemotherapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Paweorn Angsutararux
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
355
|
Ghosh R, Alajbegovic A, Gomes AV. NSAIDs and Cardiovascular Diseases: Role of Reactive Oxygen Species. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:536962. [PMID: 26457127 PMCID: PMC4592725 DOI: 10.1155/2015/536962] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/03/2015] [Accepted: 03/03/2015] [Indexed: 12/24/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are the most commonly used drugs worldwide. NSAIDs are used for a variety of conditions including pain, rheumatoid arthritis, and musculoskeletal disorders. The beneficial effects of NSAIDs in reducing or relieving pain are well established, and other benefits such as reducing inflammation and anticancer effects are also documented. The undesirable side effects of NSAIDs include ulcers, internal bleeding, kidney failure, and increased risk of heart attack and stroke. Some of these side effects may be due to the oxidative stress induced by NSAIDs in different tissues. NSAIDs have been shown to induce reactive oxygen species (ROS) in different cell types including cardiac and cardiovascular related cells. Increases in ROS result in increased levels of oxidized proteins which alters key intracellular signaling pathways. One of these key pathways is apoptosis which causes cell death when significantly activated. This review discusses the relationship between NSAIDs and cardiovascular diseases (CVD) and the role of NSAID-induced ROS in CVD.
Collapse
Affiliation(s)
- Rajeshwary Ghosh
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
| | - Azra Alajbegovic
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
| | - Aldrin V. Gomes
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
- Department of Physiology and Membrane Biology, University of California, Davis, CA 95616, USA
| |
Collapse
|
356
|
Granados ML, Hudson LG, Samudio-Ruiz SL. Contributions of the Epidermal Growth Factor Receptor to Acquisition of Platinum Resistance in Ovarian Cancer Cells. PLoS One 2015; 10:e0136893. [PMID: 26351843 PMCID: PMC4564275 DOI: 10.1371/journal.pone.0136893] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 08/09/2015] [Indexed: 01/12/2023] Open
Abstract
Acquisition of platinum resistance following first line platinum/taxane therapy is commonly observed in ovarian cancer patients and prevents clinical effectiveness. There are few options to prevent platinum resistance; however, demethylating agents have been shown to resensitize patients to platinum therapy thereby demonstrating that DNA methylation is a critical contributor to the development of platinum resistance. We previously reported the Epidermal Growth Factor Receptor (EGFR) is a novel regulator of DNA methyltransferase (DNMT) activity and DNA methylation. Others have shown that EGFR activation is linked to cisplatin treatment and platinum resistance. We hypothesized that cisplatin induced activation of the EGFR mediates changes in DNA methylation associated with the development of platinum resistance. To investigate this, we evaluated EGFR signaling and DNMT activity after acute cisplatin exposure. We also developed an in vitro model of platinum resistance to examine the effects of EGFR inhibition on acquisition of cisplatin resistance. Acute cisplatin treatment activates the EGFR and downstream signaling pathways, and induces an EGFR mediated increase in DNMT activity. Cisplatin resistant cells also showed increased DNMT activity and global methylation. EGFR inhibition during repeated cisplatin treatments generated cells that were more sensitive to cisplatin and did not develop increases in DNA methylation or DNMT activity compared to controls. These findings suggest that activation of EGFR during platinum treatment contributes to the development of platinum resistance. Furthermore, EGFR inhibition may be an effective strategy at attenuating the development of platinum resistance thereby enhancing the effectiveness of chemotherapeutic treatment in ovarian cancer.
Collapse
Affiliation(s)
- Michaela L. Granados
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, United States of America
| | - Laurie G. Hudson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, United States of America
| | - Sabrina L. Samudio-Ruiz
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, United States of America
| |
Collapse
|
357
|
Singh P, Sharma R, McElhanon K, Allen CD, Megyesi JK, Beneš H, Singh SP. Sulforaphane protects the heart from doxorubicin-induced toxicity. Free Radic Biol Med 2015; 86:90-101. [PMID: 26025579 PMCID: PMC4554811 DOI: 10.1016/j.freeradbiomed.2015.05.028] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 05/04/2015] [Accepted: 05/19/2015] [Indexed: 01/08/2023]
Abstract
Cardiotoxicity is one of the major side effects encountered during cancer chemotherapy with doxorubicin (DOX) and other anthracyclines. Previous studies have shown that oxidative stress caused by DOX is one of the primary mechanisms for its toxic effects on the heart. Since the redox-sensitive transcription factor, Nrf2, plays a major role in protecting cells from the toxic metabolites generated during oxidative stress, we examined the effects of the phytochemical sulforaphane (SFN), a potent Nrf2-activating agent, on DOX-induced cardiotoxicity. These studies were carried out both in vitro and in vivo using rat H9c2 cardiomyoblast cells and wild type 129/sv mice, and involved SFN pretreatment followed by SFN administration during DOX exposure. SFN treatment protected H9c2 cells from DOX cytotoxicity and also resulted in restored cardiac function and a significant reduction in DOX-induced cardiomyopathy and mortality in mice. Specificity of SFN induction of Nrf2 and protection of H9c2 cells was demonstrated in Nrf2 knockdown experiments. Cardiac accumulation of 4-hydroxynonenal (4-HNE) protein adducts, due to lipid peroxidation following DOX-induced oxidative stress, was significantly attenuated by SFN treatment. The respiratory function of cardiac mitochondria isolated from mice exposed to DOX alone was repressed, while SFN treatment with DOX significantly elevated mitochondrial respiratory complex activities. Co-administration of SFN reversed the DOX-associated reduction in nuclear Nrf2 binding activity and restored cardiac expression of Nrf2-regulated genes at both the RNA and protein levels. Together, our results demonstrate for the first time that the Nrf2 inducer, SFN, has the potential to provide protection against DOX-mediated cardiotoxicity.
Collapse
Affiliation(s)
- Preeti Singh
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Rajendra Sharma
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kevin McElhanon
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Charles D Allen
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Judit K Megyesi
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA; Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Helen Beneš
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sharda P Singh
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA.
| |
Collapse
|
358
|
Barciszewska AM, Gurda D, Głodowicz P, Nowak S, Naskręt-Barciszewska MZ. A New Epigenetic Mechanism of Temozolomide Action in Glioma Cells. PLoS One 2015; 10:e0136669. [PMID: 26309255 PMCID: PMC4550362 DOI: 10.1371/journal.pone.0136669] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/05/2015] [Indexed: 01/15/2023] Open
Abstract
Temozolomide (TMZ) is an oral alkylating chemotherapeutic agent that prolongs the survival of patients with glioblastoma (GBM). Despite that high TMZ potential, progression of disease and recurrence are still observed. Therefore a better understanding of the mechanism of action of this drug is necessary and may allow more durable benefit from its anti-glioma properties. Using nucleotide post-labelling method and separation on thin-layer chromatography we measured of global changes of 5-methylcytosine (m5C) in DNA of glioma cells treated with TMZ. Although m5C is not a product of TMZ methylation reaction of DNA, we analysed the effects of the drug action on different glioma cell lines through global changes at the level of the DNA main epigenetic mark. The first effect of TMZ action we observed is DNA hypermethylation followed by global demethylation. Therefore an increase of DNA methylation and down regulation of some genes expression can be ascribed to activation of DNA methyltransferases (DNMTs). On the other hand hypomethylation is induced by oxidative stress and causes uncontrolled expression of pathologic protein genes. The results of brain tumours treatment with TMZ suggest the new mechanism of modulation epigenetic marker in cancer cells. A high TMZ concentration induced a significant increase of m5C content in DNA in the short time, but a low TMZ concentration at longer time hypomethylation is observed for whole range of TMZ concentrations. Therefore TMZ administration with low doses of the drug and short time should be considered as optimal therapy.
Collapse
Affiliation(s)
- Anna-Maria Barciszewska
- Department of Neurosurgery and Neurotraumatology, Karol Marcinkowski University of Medical Sciences, Przybyszewskiego 49, 60–355, Poznan, Poland
| | - Dorota Gurda
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61–704, Poznan, Poland
| | - Paweł Głodowicz
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61–704, Poznan, Poland
| | - Stanisław Nowak
- Department of Neurosurgery and Neurotraumatology, Karol Marcinkowski University of Medical Sciences, Przybyszewskiego 49, 60–355, Poznan, Poland
| | | |
Collapse
|
359
|
Dai F, Liu GY, Li Y, Yan WJ, Wang Q, Yang J, Lu DL, Ding DJ, Lin D, Zhou B. Insights into the importance for designing curcumin-inspired anticancer agents by a prooxidant strategy: The case of diarylpentanoids. Free Radic Biol Med 2015; 85:127-37. [PMID: 25912482 DOI: 10.1016/j.freeradbiomed.2015.04.017] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 04/09/2015] [Accepted: 04/13/2015] [Indexed: 12/20/2022]
Abstract
Developing anticancer agents by a prooxidant strategy has attracted increasing attention in recent years, although it is not conventional in medicinal chemistry and is completely opposite to antioxidant therapy. In this work, a panel of diarylpentanoids as the curcumin mono-carbonyl analogs were designed and synthesized, and their cytotoxic and proapoptotic mechanisms against human lung cancer A549 cells were investigated at the frontiers of chemistry and biology. It was found that compared with curcumin, the compounds (A1, B1, and C1) bearing two ortho substituents on the aromatic rings, especially A1, exhibit significantly increased cytotoxic and proapoptotic activities through a Michael acceptor unit-dependent prooxidant-mediated mechanism. The prooxidative ability is governed not only by their electrophilicity but also by their geometry, cellular uptake and metabolic stability, and TrxR-inhibitory activity. Mechanistic investigation reveals that the compound A1 could effectively and irreversibly modify the TrxR by virtue of the above optimal biochemical parameters, and convert this antioxidant enzyme into a reactive oxygen species (ROS) promoter, resulting in a burst of the intracellular ROS including H2O2 and O2(-)•. The ROS generation is associated with falling apart in the redox buffering system, and subsequently induces increases in Ca(2+) influx and oxidative stress, collapse of mitochondrial membrane potential, and activation of caspase-9 and caspase-3, ultimately leading to cell apoptosis. This work highlights the feasibility in designing curcumin-inspired anticancer agents by a prooxidant strategy, and gives us useful information on how to design them.
Collapse
Affiliation(s)
- Fang Dai
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Guo-Yun Liu
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Yan Li
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Wen-Jing Yan
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Qi Wang
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Jie Yang
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Dong-Liang Lu
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - De-Jun Ding
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Dong Lin
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China
| | - Bo Zhou
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, 222 Tianshui Street S., Lanzhou, Gansu 730000, China.
| |
Collapse
|
360
|
Luoma AM, Kuo F, Cakici O, Crowther MN, Denninger AR, Avila RL, Brites P, Kirschner DA. Plasmalogen phospholipids protect internodal myelin from oxidative damage. Free Radic Biol Med 2015; 84:296-310. [PMID: 25801291 DOI: 10.1016/j.freeradbiomed.2015.03.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 02/25/2015] [Accepted: 03/12/2015] [Indexed: 12/16/2022]
Abstract
Reactive oxygen species (ROS) are implicated in a range of degenerative conditions, including aging, neurodegenerative diseases, and neurological disorders. Myelin is a lipid-rich multilamellar sheath that facilitates rapid nerve conduction in vertebrates. Given the high energetic demands and low antioxidant capacity of the cells that elaborate the sheaths, myelin is considered intrinsically vulnerable to oxidative damage, raising the question whether additional mechanisms prevent structural damage. We characterized the structural and biochemical basis of ROS-mediated myelin damage in murine tissues from both central nervous system (CNS) and peripheral nervous system (PNS). To determine whether ROS can cause structural damage to the internodal myelin, whole sciatic and optic nerves were incubated ex vivo with a hydroxyl radical-generating system consisting of copper (Cu), hydrogen peroxide (HP), and ortho-phenanthroline (OP). Quantitative assessment of unfixed tissue by X-ray diffraction revealed irreversible compaction of myelin membrane stacking in both sciatic and optic nerves. Incubation in the presence of the hydroxyl radical scavenger sodium formate prevented this damage, implicating hydroxyl radical species. Myelin membranes are particularly enriched in plasmalogens, a class of ether-linked phospholipids proposed to have antioxidant properties. Myelin in sciatic nerve from plasmalogen-deficient (Pex7 knockout) mice was significantly more vulnerable to Cu/OP/HP-mediated ROS-induced compaction than myelin from WT mice. Our results directly support the role of plasmalogens as endogenous antioxidants providing a defense that protects ROS-vulnerable myelin.
Collapse
Affiliation(s)
- Adrienne M Luoma
- Biology Department, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467-3811, USA
| | - Fonghsu Kuo
- Biology Department, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467-3811, USA
| | - Ozgur Cakici
- Biology Department, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467-3811, USA
| | - Michelle N Crowther
- Biology Department, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467-3811, USA
| | - Andrew R Denninger
- Biology Department, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467-3811, USA
| | - Robin L Avila
- Biology Department, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467-3811, USA
| | - Pedro Brites
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Daniel A Kirschner
- Biology Department, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467-3811, USA.
| |
Collapse
|
361
|
Effect of hesperidin on mice bearing Ehrlich solid carcinoma maintained on doxorubicin. Tumour Biol 2015; 36:9267-75. [DOI: 10.1007/s13277-015-3655-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 06/09/2015] [Indexed: 12/26/2022] Open
|
362
|
Gayathri L, Dhivya R, Dhanasekaran D, Periasamy VS, Alshatwi AA, Akbarsha MA. Hepatotoxic effect of ochratoxin A and citrinin, alone and in combination, and protective effect of vitamin E: In vitro study in HepG2 cell. Food Chem Toxicol 2015; 83:151-63. [PMID: 26111808 DOI: 10.1016/j.fct.2015.06.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 06/02/2015] [Accepted: 06/08/2015] [Indexed: 10/23/2022]
Abstract
Ochratoxin A (OTA) and citrinin (CTN) are the most commonly co-occurring mycotoxins in a wide variety of food and feed commodities. The major target organ of these toxins is kidney but liver could also be a target organ. The combined toxicity of these two toxins in kidney cells has been studied but not in liver cell. In this study HepG2 cells were exposed to OTA and CTN, alone and in combination, with a view to compare the molecular and cellular mechanisms underlying OTA, CTN and OTA + CTN hepatotoxicity. OTA and CTN alone as well as in combination affected the viability of HepG2 cells in a dose-dependent manner. OTA + CTN, at a dose of 20% of IC50 of each, produced effect almost similar to that produced by either of the toxins at its IC50 concentration, indicating that the two toxins in combination act synergistically. The cytotoxicity of OTA + CTN on hepatocytes is mediated by increased level of intracellular ROS followed/accompanied by DNA strand breaks and mitochondria-mediated intrinsic apoptosis. Co-treatment of vitamin E (Vit E) with OTA, CTN and OTA + CTN reduced the levels of ROS and the cytotoxicity. But the genotoxic effect of OTA and OTA + CTN was not completely alleviated by Vit E treatment whereas the DNA damage as caused by CTN when treated alone was obviated, indicating that OTA induces DNA damage directly whereas CTN induces ROS-mediated DNA damage and OTA + CTN combination induces DNA damage not exclusively relying on but influenced by ROS generation. Taken together, these findings indicate that OTA and CTN in combination affect hepatocytes at very low concentrations and, thereby, pose a potential threat to public and animal health.
Collapse
Affiliation(s)
- Loganathan Gayathri
- Department of Microbiology, Bharathidasan University, Tiruchirappalli 620024, India; Mahatma Gandhi-Doerenkamp Center, Bharathidasan University, Tiruchirappalli 620024, India
| | - Rajakumar Dhivya
- Mahatma Gandhi-Doerenkamp Center, Bharathidasan University, Tiruchirappalli 620024, India
| | | | - Vaiyapuri S Periasamy
- Nanobiotechnology and Molecular Biology Research Laboratory, Department of Food Science and Nutrition, College of Food Science and Agriculture, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Ali A Alshatwi
- Nanobiotechnology and Molecular Biology Research Laboratory, Department of Food Science and Nutrition, College of Food Science and Agriculture, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Mohammad A Akbarsha
- Mahatma Gandhi-Doerenkamp Center, Bharathidasan University, Tiruchirappalli 620024, India; Nanobiotechnology and Molecular Biology Research Laboratory, Department of Food Science and Nutrition, College of Food Science and Agriculture, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia.
| |
Collapse
|
363
|
Nerimetla R, Krishnan S. Electrocatalysis by subcellular liver fractions bound to carbon nanostructures for stereoselective green drug metabolite synthesis. Chem Commun (Camb) 2015; 51:11681-4. [PMID: 26103056 DOI: 10.1039/c5cc03364k] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A novel, reusable, cofactor-free, and mediator-free human liver microsomal bioreactor constructed on carbon nanostructure electrodes for stereoselective green syntheses of drug metabolites and specialty chemicals is reported here for the first time. Drug metabolites are useful for examining pharmaceutical and pharmacological properties of new drugs under development.
Collapse
|
364
|
Onodera Y, Teramura T, Takehara T, Shigi K, Fukuda K. Reactive oxygen species induce Cox-2 expression via TAK1 activation in synovial fibroblast cells. FEBS Open Bio 2015; 5:492-501. [PMID: 26110105 PMCID: PMC4476901 DOI: 10.1016/j.fob.2015.06.001] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 05/15/2015] [Accepted: 06/02/2015] [Indexed: 11/05/2022] Open
Abstract
Oxidative stress in the arthritis joint is involved in generating mediators for inflammation. Oxidative stress-induced expression of Cox-2 was mediated by MAPKs and NF-κB. ROS-induced MAPKs and NF-κB were attenuated by inhibition of MAPKKK TAK1. Inhibition of TAK1 activity resulted in reduced expression of Cox-2 and PGE2. ROS-induced TAK1 activation and Cox-2 expression was inhibited by antioxidants N-acetyl cysteamine and hyaluronic acid.
Oxidative stress within the arthritis joint has been indicated to be involved in generating mediators for tissue degeneration and inflammation. COX-2 is a mediator in inflammatory action, pain and some catabolic reactions in inflamed tissues. Here, we demonstrated a direct relationship between oxidative stress and Cox-2 expression in the bovine synovial fibroblasts. Furthermore, we elucidated a novel mechanism, in which oxidative stress induced phosphorylation of MAPKs and NF-κB through TAK1 activation and resulted in increased Cox-2 and prostaglandin E2 expression. Finally, we demonstrated that ROS-induced Cox-2 expression was inhibited by supplementation of an antioxidant such as N-acetyl cysteamine and hyaluronic acid in vitro and in vivo. From these results, we conclude that oxidative stress is an important factor for generation of Cox-2 in synovial fibroblasts and thus its neutralization may be an effective strategy in palliative therapy for chronic joint diseases.
Collapse
Affiliation(s)
- Yuta Onodera
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Takeshi Teramura
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Toshiyuki Takehara
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kanae Shigi
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kanji Fukuda
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| |
Collapse
|
365
|
Wu L, Chen X, Huang L, Tian J, Ke F, Xu J, Chen Y, Zheng M. A Novobiocin Derivative, XN4, Inhibits the Proliferation of Chronic Myeloid Leukemia Cells by Inducing Oxidative DNA Damage. PLoS One 2015; 10:e0123314. [PMID: 25928540 PMCID: PMC4415758 DOI: 10.1371/journal.pone.0123314] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 03/02/2015] [Indexed: 12/22/2022] Open
Abstract
XN4 might induce DNA damage and apoptotic cell death through reactive oxygen species (ROS). The inhibition of proliferation of K562 and K562/G01 cells was measured by MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide). The mRNA levels of NADPH oxidase 1-5 (Nox1-5) genes were evaluated by qRT-PCR. The levels of extracellular reactive oxygen species (ROS), DNA damage, apoptosis, and cell cycle progression were examined by flow cytometry (FCM). Protein levels were analyzed by immunoblotting. XN4 significantly inhibited the proliferation of K562 and K562/G01 cells, with IC50 values of 3.75±0.07 µM and 2.63±0.43 µM, respectively. XN4 significantly increased the levels of Nox4 and Nox5 mRNA, stimulating the generation of intracellular ROS, inducing DNA damage and activating ATM-γ-H2AX signaling, which increased the number of cells in the S and G2/M phase of the cell cycle. Subsequently, XN4 induced apoptotic cell death by activating caspase-3 and PARP. Moreover, the above effects were all reversed by the ROS scavenger N-acetylcysteine (NAC). Additionally, XN4 can induce apoptosis in progenitor/stem cells isolated from CML patients’ bone marrow. In conclusion, XN4-induced DNA damage and cell apoptosis in CML cells is mediated by the generation of ROS.
Collapse
Affiliation(s)
- Lixian Wu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, P. R.China
- Institute of Materia Medica, FMU, Fuzhou, P. R.China
- Fuijan Key Laboratory of Natural Medicine pharmacology, FMU, Fuzhou, P. R.China
- * E-mail: (LW); (MZ)
| | - Xianling Chen
- Fujian Institute of Hematology, Union Hospital, FMU, Fuzhou, P. R.China
| | - Lisen Huang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, P. R.China
- Institute of Materia Medica, FMU, Fuzhou, P. R.China
- Fuijan Key Laboratory of Natural Medicine pharmacology, FMU, Fuzhou, P. R.China
| | - Jue Tian
- Department of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, P. R.China
- Institute of Materia Medica, FMU, Fuzhou, P. R.China
- Fuijan Key Laboratory of Natural Medicine pharmacology, FMU, Fuzhou, P. R.China
| | - Fang Ke
- Department of Pharmacochemistry, School of Pharmacy, FMU, Fuzhou, P. R.China
| | - Jianhua Xu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University (FMU), Fuzhou, P. R.China
- Institute of Materia Medica, FMU, Fuzhou, P. R.China
- Fuijan Key Laboratory of Natural Medicine pharmacology, FMU, Fuzhou, P. R.China
| | - Yuanzhong Chen
- Fujian Institute of Hematology, Union Hospital, FMU, Fuzhou, P. R.China
| | - Ming Zheng
- Department of Anatomy, School of Basic Medicine, FMU, Fuzhou, P. R.China
- * E-mail: (LW); (MZ)
| |
Collapse
|
366
|
Chen GG, Mao M, Qiu LZ, Liu QM. Gene transfection mediated by polyethyleneimine-polyethylene glycol nanocarrier prevents cisplatin-induced spiral ganglion cell damage. Neural Regen Res 2015; 10:425-31. [PMID: 25878591 PMCID: PMC4396105 DOI: 10.4103/1673-5374.153691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2014] [Indexed: 01/22/2023] Open
Abstract
Polyethyleneimine-polyethylene glycol (PEI-PEG), a novel nanocarrier, has been used for transfection and gene therapy in a variety of cells. In our previous study, we successfully carried out PEI-PEG-mediated gene transfer in spiral ganglion cells. It remains unclear whether PEI-PEG could be used for gene therapy with X-linked inhibitor of apoptosis protein (XIAP) in the inner ear. In the present study, we performed PEI-PEG-mediated XIAP gene transfection in the cochlea of Sprague-Dawley rats, via scala tympani fenestration, before daily cisplatin injections. Auditory brainstem reflex tests demonstrated the protective effects of XIAP gene therapy on auditory function. Immunohistochemical staining revealed XIAP protein expression in the cytoplasm of cells in the spiral ganglion, the organ of Corti and the stria vascularis. Reverse transcription-PCR detected high levels of XIAP mRNA expression in the cochlea. The present findings suggest that PEI-PEG nanocarrier-mediated XIAP gene transfection results in XIAP expression in the cochlea, prevents damage to cochlear spiral ganglion cells, and protects hearing.
Collapse
Affiliation(s)
- Guan-Gui Chen
- Department of Otorhinolaryngology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Min Mao
- Department of Otorhinolaryngology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Li-Zi Qiu
- Department of Otorhinolaryngology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Qi-Ming Liu
- Department of Otorhinolaryngology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
367
|
Hong Y, Sengupta S, Hur W, Sim T. Identification of Novel ROS Inducers: Quinone Derivatives Tethered to Long Hydrocarbon Chains. J Med Chem 2015; 58:3739-50. [DOI: 10.1021/jm501846y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Yeonsun Hong
- KU-KIST Graduate School of Converging Science and Technology, 145 Anam-ro, Seongbuk-gu, Seoul, 136-713, Republic of Korea
| | - Sandip Sengupta
- Chemical
Kinomics Research Center, Korea Institute of Science and Technology (KIST), 5 Hwarangro 14-gil, Seongbuk-gu, Seoul, 136-791, Republic of Korea
| | - Wooyoung Hur
- Chemical
Kinomics Research Center, Korea Institute of Science and Technology (KIST), 5 Hwarangro 14-gil, Seongbuk-gu, Seoul, 136-791, Republic of Korea
| | - Taebo Sim
- KU-KIST Graduate School of Converging Science and Technology, 145 Anam-ro, Seongbuk-gu, Seoul, 136-713, Republic of Korea
- Chemical
Kinomics Research Center, Korea Institute of Science and Technology (KIST), 5 Hwarangro 14-gil, Seongbuk-gu, Seoul, 136-791, Republic of Korea
| |
Collapse
|
368
|
Dahl JU, Gray MJ, Jakob U. Protein quality control under oxidative stress conditions. J Mol Biol 2015; 427:1549-63. [PMID: 25698115 PMCID: PMC4357566 DOI: 10.1016/j.jmb.2015.02.014] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 02/11/2015] [Accepted: 02/11/2015] [Indexed: 12/22/2022]
Abstract
Accumulation of reactive oxygen and chlorine species (RO/CS) is generally regarded to be a toxic and highly undesirable event, which serves as contributing factor in aging and many age-related diseases. However, it is also put to excellent use during host defense, when high levels of RO/CS are produced to kill invading microorganisms and regulate bacterial colonization. Biochemical and cell biological studies of how bacteria and other microorganisms deal with RO/CS have now provided important new insights into the physiological consequences of oxidative stress, the major targets that need protection, and the cellular strategies employed by organisms to mitigate the damage. This review examines the redox-regulated mechanisms by which cells maintain a functional proteome during oxidative stress. We will discuss the well-characterized redox-regulated chaperone Hsp33, and we will review recent discoveries demonstrating that oxidative stress-specific activation of chaperone function is a much more widespread phenomenon than previously anticipated. New members of this group include the cytosolic ATPase Get3 in yeast, the Escherichia coli protein RidA, and the mammalian protein α2-macroglobulin. We will conclude our review with recent evidence showing that inorganic polyphosphate (polyP), whose accumulation significantly increases bacterial oxidative stress resistance, works by a protein-like chaperone mechanism. Understanding the relationship between oxidative and proteotoxic stresses will improve our understanding of both host-microbe interactions and how mammalian cells combat the damaging side effects of uncontrolled RO/CS production, a hallmark of inflammation.
Collapse
Affiliation(s)
- Jan-Ulrik Dahl
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048, USA
| | - Michael J Gray
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048, USA
| | - Ursula Jakob
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048, USA.
| |
Collapse
|
369
|
Doherty KR, Talbert DR, Trusk PB, Moran DM, Shell SA, Bacus S. Structural and functional screening in human induced-pluripotent stem cell-derived cardiomyocytes accurately identifies cardiotoxicity of multiple drug types. Toxicol Appl Pharmacol 2015; 285:51-60. [PMID: 25841593 DOI: 10.1016/j.taap.2015.03.008] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 03/05/2015] [Accepted: 03/06/2015] [Indexed: 12/31/2022]
Abstract
Safety pharmacology studies that evaluate new drug entities for potential cardiac liability remain a critical component of drug development. Current studies have shown that in vitro tests utilizing human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CM) may be beneficial for preclinical risk evaluation. We recently demonstrated that an in vitro multi-parameter test panel assessing overall cardiac health and function could accurately reflect the associated clinical cardiotoxicity of 4 FDA-approved targeted oncology agents using hiPS-CM. The present studies expand upon this initial observation to assess whether this in vitro screen could detect cardiotoxicity across multiple drug classes with known clinical cardiac risks. Thus, 24 drugs were examined for their effect on both structural (viability, reactive oxygen species generation, lipid formation, troponin secretion) and functional (beating activity) endpoints in hiPS-CM. Using this screen, the cardiac-safe drugs showed no effects on any of the tests in our panel. However, 16 of 18 compounds with known clinical cardiac risk showed drug-induced changes in hiPS-CM by at least one method. Moreover, when taking into account the Cmax values, these 16 compounds could be further classified depending on whether the effects were structural, functional, or both. Overall, the most sensitive test assessed cardiac beating using the xCELLigence platform (88.9%) while the structural endpoints provided additional insight into the mechanism of cardiotoxicity for several drugs. These studies show that a multi-parameter approach examining both cardiac cell health and function in hiPS-CM provides a comprehensive and robust assessment that can aid in the determination of potential cardiac liability.
Collapse
Affiliation(s)
| | | | | | | | - Scott A Shell
- Quintiles, 777 Oakmont Lane Suite 100, Westmont, IL 60559,USA
| | - Sarah Bacus
- Quintiles, 777 Oakmont Lane Suite 100, Westmont, IL 60559,USA
| |
Collapse
|
370
|
Atli O, Ilgin S, Altuntas H, Burukoglu D. Evaluation of azithromycin induced cardiotoxicity in rats. Int J Clin Exp Med 2015; 8:3681-90. [PMID: 26064263 PMCID: PMC4443097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 02/28/2015] [Indexed: 06/04/2023]
Abstract
Although there are possible cardiovascular adverse effects associated with the azithromycin treatment according to some case reports and cohort studies, there is no experimental study evaluating cardiotoxicity in repeated pharmacological doses of this drug. In our study, 15 mg/kg and 30 mg/kg azithromycin were orally administered to rats for 14 days to evaluate the cardiotoxicity of this drug. ECGs of the azithromycin-treated and control animals were recorded. Blood samples were assayed to determine LDH and CK-MB levels. Additionally, CAT, SOD, GSH and MDA levels of heart tissues were measured. According to our ECG recordings, decreased heart rate, prolonged PR and QT intervals, QRS complex and T wave abnormalities were observed in 30 mg/kg azithromycin-administered group significantly when compared with control group. Plasma CK-MB and LDH levels were increased in 30 mg/kg azithromycin-administered group significantly when compared to the control group. In heart tissues, CAT, SOD and GSH levels were decreased while MDA levels were increased in both azithromycin-administered groups significantly when compared with the control group. In conclusion, our findings supported the possible cardiotoxicity risk with azithromycin treatment and also, oxidative stress, which was induced by azithromycin in our study, was thought to be occurred secondary to cardiac toxicity of the drug.
Collapse
Affiliation(s)
- Ozlem Atli
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Anadolu UniversityEskisehir, Turkey
| | - Sinem Ilgin
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Anadolu UniversityEskisehir, Turkey
| | - Hakan Altuntas
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Anadolu UniversityEskisehir, Turkey
| | - Dilek Burukoglu
- Department of Histology, Faculty of Medicine, Osmangazi UniversityEskisehir, Turkey
| |
Collapse
|
371
|
Hall SR, Blundon HL, Ladda MA, Robertson AW, Martinez-Farina CF, Jakeman DL, Goralski KB. Jadomycin breast cancer cytotoxicity is mediated by a copper-dependent, reactive oxygen species-inducing mechanism. Pharmacol Res Perspect 2015; 3:e00110. [PMID: 25729577 PMCID: PMC4324684 DOI: 10.1002/prp2.110] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 10/10/2014] [Indexed: 12/29/2022] Open
Abstract
Jadomycins are natural products biosynthesized by the bacteria Streptomyces venezuelae which kill drug-sensitive and multidrug-resistant breast cancer cells in culture. Currently, the mechanisms of jadomycin cytotoxicity are poorly understood; however, reactive oxygen species (ROS)–induced DNA cleavage is suggested based on bacterial plasmid DNA cleavage studies. The objective of this study was to determine if and how ROS contribute to jadomycin cytotoxicity in drug-sensitive MCF7 (MCF7-CON) and taxol-resistant MCF7 (MCF7-TXL) breast cancer cells. As determined using an intracellular, fluorescent, ROS-detecting probe, jadomycins B, S, SPhG, and F dose dependently increased intracellular ROS activity 2.5- to 5.9-fold. Cotreatment with the antioxidant N-acetyl cysteine lowered ROS concentrations to below baseline levels and decreased the corresponding cytotoxic potency of the four jadomycins 1.9- to 3.3-fold, confirming a ROS-mediated mechanism. Addition of CuSO4 enhanced, whereas addition of the Cu(II)-chelator d-penicillamine reduced, the ROS generation and cytotoxicity of each jadomycin. Specific inhibitors of the antioxidant enzymes, superoxide dismutase 1, glutathione S-transferase, and thioredoxin reductase, but not catalase, enhanced jadomycin-mediated ROS generation and anticancer activity. In conclusion, the results indicate that jadomycin cytotoxicity involves the generation of cytosolic superoxide via a Cu(II)-jadomycin reaction, a mechanism common to all jadomycins tested and observed in MCF7-CON and drug-resistant MCF7-TXL cells. The superoxide dismutase 1, glutathione, and peroxiredoxin/thioredoxin cellular antioxidant enzyme pathways scavenged intracellular ROS generated by jadomycin treatment. Blocking these antioxidant pathways could serve as a strategy to enhance jadomycin cytotoxic potency in drug-sensitive and multidrug-resistant breast cancers.
Collapse
Affiliation(s)
- Steven R Hall
- Department of Pharmacology, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada, B3H 4R2
| | - Heather L Blundon
- College of Pharmacy, Faculty of Health Professions, Dalhousie University Halifax, Nova Scotia, Canada, B3H 4R2
| | - Matthew A Ladda
- College of Pharmacy, Faculty of Health Professions, Dalhousie University Halifax, Nova Scotia, Canada, B3H 4R2
| | - Andrew W Robertson
- Department of Chemistry, Faculty of Sciences, Dalhousie University Halifax, Nova Scotia, Canada, B3H 4R2
| | - Camilo F Martinez-Farina
- Department of Chemistry, Faculty of Sciences, Dalhousie University Halifax, Nova Scotia, Canada, B3H 4R2
| | - David L Jakeman
- College of Pharmacy, Faculty of Health Professions, Dalhousie University Halifax, Nova Scotia, Canada, B3H 4R2 ; Department of Chemistry, Faculty of Sciences, Dalhousie University Halifax, Nova Scotia, Canada, B3H 4R2
| | - Kerry B Goralski
- Department of Pharmacology, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada, B3H 4R2 ; College of Pharmacy, Faculty of Health Professions, Dalhousie University Halifax, Nova Scotia, Canada, B3H 4R2
| |
Collapse
|
372
|
Hielscher A, Gerecht S. Hypoxia and free radicals: role in tumor progression and the use of engineering-based platforms to address these relationships. Free Radic Biol Med 2015; 79:281-91. [PMID: 25257256 PMCID: PMC4339408 DOI: 10.1016/j.freeradbiomed.2014.09.015] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 08/11/2014] [Accepted: 09/15/2014] [Indexed: 12/23/2022]
Abstract
Hypoxia is a feature of all solid tumors, contributing to tumor progression and therapy resistance. Through stabilization of the hypoxia-inducible factor 1 alpha (HIF-1α), hypoxia activates the transcription of a number of genes that sustain tumor progression. Since the seminal discovery of HIF-1α as a hypoxia-responsive master regulator of numerous genes and transcription factors, several groups have reported a novel mechanism whereby hypoxia mediates stabilization of HIF-1α. This process occurs as a result of hypoxia-generated reactive oxygen species (ROS), which, in turn, stabilize the expression of HIF-1α. As a result, a number of genes regulating tumor growth are expressed, fueling ongoing tumor progression. In this review, we outline a role for hypoxia in generating ROS and additionally define the mechanisms contributing to ROS-induced stabilization of HIF-1α.We further explore how ROS-induced HIF-1α stabilization contributes to tumor growth, angiogenesis, metastasis, and therapy response. We discuss a future outlook, describing novel therapeutic approaches for attenuating ROS production while considering how these strategies should be carefully selected when combining with chemotherapeutic agents. As engineering-based approaches have been more frequently utilized to address biological questions, we discuss opportunities whereby engineering techniques may be employed to better understand the physical and biochemical factors controlling ROS expression. It is anticipated that an improved understanding of the mechanisms responsible for the hypoxia/ROS/HIF-1α axis in tumor progression will yield the development of better targeted therapies.
Collapse
Affiliation(s)
- Abigail Hielscher
- Department of Biomedical Sciences, Georgia Philadelphia College of Osteopathic Medicine, Suwanee, GA 30024, USA; Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA.
| | - Sharon Gerecht
- Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
373
|
Fetoni AR, Eramo SLM, Paciello F, Rolesi R, Podda MV, Troiani D, Paludetti G. Curcuma longa (curcumin) decreases in vivo cisplatin-induced ototoxicity through heme oxygenase-1 induction. Otol Neurotol 2015; 35:e169-77. [PMID: 24608370 DOI: 10.1097/mao.0000000000000302] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
HYPOTHESIS To investigate whether curcumin may have in vivo protective effects against cisplatin ototoxicity by its direct scavenger activity and/or by curcumin-mediated upregulation of HO-1. BACKGROUND Cisplatin-induced ototoxicity is a major dose-limiting side effect in anticancer chemotherapy. A protective approach to decrease cisplatin ototoxicity without compromising its therapeutic efficacy remains a critical goal for anticancer therapy. Recent evidences indicate that curcumin exhibits antioxidant, anti-inflammatory, and chemosensitizer activities. METHODS In male adult Wistar rats, a curcumin dose of 200 mg/kg, selected from a dose-response curve, was injected 1 hour before cisplatin administration and once daily for the following 3 days. A single dose of cisplatin (16 mg/kg) was administered intraperitoneally. Rats were divided as follows: 1) control, 2) curcumin control, 3) vehicle control, 4) cisplatin, 5) cisplatin+ vehicle, and 6) curcumin+cisplatin. ABRs were measured before and at Days 3 and 5 after cisplatin administration. Rhodamine-phalloidin staining, 4-hydroxy-2-nonenal and heme-oxigenase-1 immunostainings, and Western blot analyses were performed to assess and quantify OHC loss, lipid peroxidation, and the endogenous response to cisplatin-induced damage and to curcumin protection. RESULTS Curcumin treatment attenuated hearing loss induced by cisplatin, increased OHC survival, decreased 4-HNE expression, and increased HO-1 expression. CONCLUSION This preclinical study demonstrates that systemic curcumin attenuates ototoxicity and provides molecular evidence for a role of HO-1 as an additional mediator in attenuating cisplatin-induced damage.
Collapse
Affiliation(s)
- Anna R Fetoni
- Department of Otolaryngology, Head and Neck Surgery; Institute of Human Physiology, Catholic University of Rome, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
374
|
Trivedi MS, Deth R. Redox-based epigenetic status in drug addiction: a potential contributor to gene priming and a mechanistic rationale for metabolic intervention. Front Neurosci 2015; 8:444. [PMID: 25657617 PMCID: PMC4302946 DOI: 10.3389/fnins.2014.00444] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 12/16/2014] [Indexed: 12/26/2022] Open
Abstract
Alcohol and other drugs of abuse, including psychostimulants and opioids, can induce epigenetic changes: a contributing factor for drug addiction, tolerance, and associated withdrawal symptoms. DNA methylation is a major epigenetic mechanism and it is one of more than 200 methylation reactions supported by methyl donor S-adenosylmethionine (SAM). Levels of SAM are controlled by cellular redox status via the folate and vitamin B12-dependent enzyme methionine synthase (MS). For example, under oxidative conditions MS is inhibited, diverting its substrate homocysteine (HCY) to the trans sulfuration pathway. Alcohol, dopamine, and morphine, can alter intracellular levels of glutathione (GSH)-based cellular redox status, subsequently affecting SAM levels and DNA methylation status. Here, existing evidence is presented in a coherent manner to propose a novel hypothesis implicating the involvement of redox-based epigenetic changes in drug addiction. Further, we discuss how a “gene priming” phenomenon can contribute to the maintenance of redox and methylation status homeostasis under various stimuli including drugs of abuse. Additionally, a new mechanistic rationale for the use of metabolic interventions/redox-replenishers as symptomatic treatment of alcohol and other drug addiction and associated withdrawal symptoms is also provided. Hence, the current review article strengthens the hypothesis that neuronal metabolism has a critical bidirectional coupling with epigenetic changes in drug addiction exemplified by the link between redox-based metabolic changes and resultant epigenetic consequences under the effect of drugs of abuse.
Collapse
Affiliation(s)
- Malav S Trivedi
- Department of Pharmaceutical Sciences, Northeastern University Boston, MA, USA
| | - Richard Deth
- Department of Pharmaceutical Sciences, Northeastern University Boston, MA, USA
| |
Collapse
|
375
|
Wang K, Yan R, Cooper KF, Strich R. Cyclin C mediates stress-induced mitochondrial fission and apoptosis. Mol Biol Cell 2015; 26:1030-43. [PMID: 25609094 PMCID: PMC4357504 DOI: 10.1091/mbc.e14-08-1315] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mitochondria are dynamic organelles that undergo constant fission and fusion cycles. In response to cellular damage, this balance is shifted dramatically toward fission. Cyclin C-Cdk8 kinase regulates transcription of diverse gene sets. Using knockout mouse embryonic fibroblasts (MEFs), we demonstrate that cyclin C directs the extensive mitochondrial scission induced by the anticancer drug cisplatin or oxidative stress. This activity is independent of transcriptional regulation, as Cdk8 is not required for this activity. Furthermore, adding purified cyclin C to unstressed permeabilized MEF cultures induced complete mitochondrial fragmentation that was dependent on the fission factors Drp1 and Mff. To regulate fission, a portion of cyclin C translocates from the nucleus to the cytoplasm, where it associates with Drp1 and is required for its enhanced mitochondrial activity in oxidatively stressed cells. In addition, although HeLa cells regulate cyclin C in a manner similar to MEF cells, U2OS osteosarcoma cultures display constitutively cytoplasmic cyclin C and semifragmented mitochondria. Finally, cyclin C, but not Cdk8, is required for loss of mitochondrial outer membrane permeability and apoptosis in cells treated with cisplatin. In conclusion, this study suggests that cyclin C connects stress-induced mitochondrial hyperfission and programmed cell death in mammalian cells.
Collapse
Affiliation(s)
- Kun Wang
- Department of Molecular Biology, Rowan University School of Osteopathic Medicine, Stratford, NJ 08055
| | - Ruilan Yan
- Department of Molecular Biology, Rowan University School of Osteopathic Medicine, Stratford, NJ 08055
| | - Katrina F Cooper
- Department of Molecular Biology, Rowan University School of Osteopathic Medicine, Stratford, NJ 08055
| | - Randy Strich
- Department of Molecular Biology, Rowan University School of Osteopathic Medicine, Stratford, NJ 08055
| |
Collapse
|
376
|
Sabiu S, Sunmonu TO, Ajani EO, Ajiboye TO. Combined administration of silymarin and vitamin C stalls acetaminophen-mediated hepatic oxidative insults in Wistar rats. REVISTA BRASILEIRA DE FARMACOGNOSIA-BRAZILIAN JOURNAL OF PHARMACOGNOSY 2015. [DOI: 10.1016/j.bjp.2014.11.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
377
|
Wang GW, Lv C, Shi ZR, Zeng RT, Dong XY, Zhang WD, Liu RH, Shan L, Shen YH. Abieslactone induces cell cycle arrest and apoptosis in human hepatocellular carcinomas through the mitochondrial pathway and the generation of reactive oxygen species. PLoS One 2014; 9:e115151. [PMID: 25502685 PMCID: PMC4263740 DOI: 10.1371/journal.pone.0115151] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/19/2014] [Indexed: 11/23/2022] Open
Abstract
Abieslactone is a triterpenoid lactone isolated from Abies plants. Previous studies have demonstrated that its derivative abiesenonic acid methyl ester possesses anti-tumor-promoting activity in vitro and in vivo. In the present study, cell viability assay demonstrated that abieslactone had selective cytotoxicity against human hepatoma cell lines. Immunostaining experiments revealed that abieslactone induced HepG2 and SMMC7721 cell apoptosis. Flow cytometry and western blot analysis showed that the apoptosis was associated with cell cycle arrest during the G1 phase, up-regulation of p53 and p21, and down-regulation of CDK2 and cyclin D1. Furthermore, our results revealed that induction of apoptosis through a mitochondrial pathway led to upregulation of Bax, down-regulation of Bcl-2, mitochondrial release of cytochrome c, reduction of mitochondrial membrane potential (MMP), and activation of caspase cascades (Casp-9 and -3). Activation of caspase cascades also resulted in the cleavage of PARP fragment. Involvement of the caspase apoptosis pathway was confirmed using caspase inhibitor Z-VAD-FMK pretreatment. Recent studies have shown that ROS is upstream of Akt signal in mitochondria-mediated hepatoma cell apoptosis. Our results showed that the accumulation of ROS was detected in HepG2 cells when treated with abieslactone, and ROS scavenger partly blocked the effects of abieslactone-induced HepG2 cell death. In addition, inactivation of total and phosphorylated Akt activities was found to be involved in abieslactone-induced HepG2 cell apoptosis. Therefore, our findings suggested that abieslactone induced G1 cell cycle arrest and caspase-dependent apoptosis via the mitochondrial pathway and the ROS/Akt pathway in HepG2 cells.
Collapse
Affiliation(s)
- Guo-Wei Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Chao Lv
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350108, PR China
| | - Zhi-Ran Shi
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Ren-Tao Zeng
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Xue-Yun Dong
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian 350108, PR China
| | - Wei-Dong Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Run-Hui Liu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
| | - Lei Shan
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
- * E-mail: (YHS); (LS)
| | - Yun-Heng Shen
- School of Pharmacy, Second Military Medical University, Shanghai 200433, PR China
- * E-mail: (YHS); (LS)
| |
Collapse
|
378
|
Yen YH, Farooqi AA, Li KT, Butt G, Tang JY, Wu CY, Cheng YB, Hou MF, Chang HW. Methanolic extracts of Solieria robusta inhibits proliferation of oral cancer Ca9-22 cells via apoptosis and oxidative stress. Molecules 2014; 19:18721-32. [PMID: 25405289 PMCID: PMC6271418 DOI: 10.3390/molecules191118721] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 11/07/2014] [Accepted: 11/10/2014] [Indexed: 11/16/2022] Open
Abstract
Many red algae-derived natural products are known to have anticancer effects. The biological functions of the red alga Solieria robusta from the Karachi coast (Pakistan) remain unclear. Here, we prepared a methanolic extracts of S. robusta (MESR) to examine its possible anti-oral cancer effects and the corresponding mechanism of action. Cell viability of MESR-incubated oral cancer Ca9-22 cells was dose-responsively decreased (p<0.001). According to a propidium iodide (PI)-based assay the cell cycle distribution was dramatically changed, especially for subG1 accumulation. Annexin V/PI assay of apoptosis using flow cytometry also showed that MESR-incubated Ca9-22 cells were dose-responsively increased (p<0.001). For evaluation of oxidative stress in MESR-incubated Ca9-22 cells, we found that reactive oxygen species (ROS) were overexpressed dose- and time-responsively and mitochondrial depolarization was also increased (p<0.001). Taken together, MESR showed inhibitory effects on oral cancer proliferation coupled with apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Yii-Huei Yen
- Department of Dentistry, Ten Chan General Hospital, Chung-Li 32043, Taiwan.
| | - Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore 54000, Pakistan.
| | - Kun-Tzu Li
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Ghazala Butt
- Department of Botany, Government College University, Lahore, Katchery Road Lahore 54000, Pakistan.
| | - Jen-Yang Tang
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan.
| | - Yuan-Bin Cheng
- Department of Dentistry, Ten Chan General Hospital, Chung-Li 32043, Taiwan.
| | - Ming-Feng Hou
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
379
|
Wang LF, Lin YS, Huang NC, Yu CY, Tsai WL, Chen JJ, Kubota T, Matsuoka M, Chen SR, Yang CS, Lu RW, Lin YL, Chang TH. Hydroxychloroquine-inhibited dengue virus is associated with host defense machinery. J Interferon Cytokine Res 2014; 35:143-56. [PMID: 25321315 DOI: 10.1089/jir.2014.0038] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Hydroxychloroquine (HCQ) is an antimalarial drug also used in treating autoimmune diseases. Its antiviral activity was demonstrated in restricting HIV infection in vitro; however, the clinical implications remain controversial. Infection with dengue virus (DENV) is a global public health problem, and we lack an antiviral drug for DENV. Here, we evaluated the anti-DENV potential of treatment with HCQ. Immunofluorescence assays demonstrated that HCQ could inhibit DENV serotype 1-4 infection in vitro. RT-qPCR analysis of HCQ-treated cells showed induced expression of interferon (IFN)-related antiviral proteins and certain inflammatory cytokines. Mechanistic study suggested that HCQ activated the innate immune signaling pathways of IFN-β, AP-1, and NFκB. Knocking down mitochondrial antiviral signaling protein (MAVS), inhibiting TANK binding kinase 1 (TBK1)/inhibitor-κB kinase ɛ (IKKɛ), and blocking type I IFN receptor reduced the efficiency of HCQ against DENV-2 infection. Furthermore, HCQ significantly induced cellular production of reactive oxygen species (ROS), which was involved in the host defense system. Suppression of ROS production attenuated the innate immune activation and anti-DENV-2 effect of HCQ. In summary, HCQ triggers the host defense machinery by inducing ROS- and MAVS-mediated innate immune activation against DENV infection and may be a candidate drug for DENV infection.
Collapse
Affiliation(s)
- Li-Fong Wang
- 1 Division of Allergy, Immunology, and Rheumatology, Kaohsiung Veterans General Hospital , Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
380
|
Zhang X, Wang H, Ma Z, Wu B. Effects of pharmaceutical PEGylation on drug metabolism and its clinical concerns. Expert Opin Drug Metab Toxicol 2014; 10:1691-702. [DOI: 10.1517/17425255.2014.967679] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
381
|
The cytotoxicity of benzaldehyde nitrogen mustard-2-pyridine carboxylic acid hydrazone being involved in topoisomerase IIα inhibition. BIOMED RESEARCH INTERNATIONAL 2014; 2014:527042. [PMID: 24995306 PMCID: PMC4066686 DOI: 10.1155/2014/527042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/12/2014] [Indexed: 11/18/2022]
Abstract
The antitumor property of iron chelators and aromatic nitrogen mustard derivatives has been well documented. Combination of the two pharmacophores in one molecule in drug designation is worth to be explored. We reported previously the syntheses and preliminary cytotoxicity evaluation of benzaldehyde nitrogen mustard pyridine carboxyl acid hydrazones (BNMPH) as extended study, more tumor cell lines (IC50 for HepG2: 26.1 ± 3.5 μM , HCT-116: 57.5 ± 5.3 μM, K562: 48.2 ± 4.0 μM, and PC-12: 19.4 ± 2.2 μM) were used to investigate its cytotoxicity and potential mechanism. In vitro experimental data showed that the BNMPH chelating Fe2+ caused a large number of ROS formations which led to DNA cleavage, and this was further supported by comet assay, implying that ROS might be involved in the cytotoxicity of BNMPH. The ROS induced changes of apoptosis related genes, but the TFR1 and NDRG1 metastatic genes were not obviously regulated, prompting that BNMPH might not be able to deprive Fe2+ of ribonucleotide reductase. The BNMPH induced S phase arrest was different from that of iron chelators (G1) and alkylating agents (G2). BNMPH also exhibited its inhibition of human topoisomerase IIα. Those revealed that the cytotoxic mechanism of the BNMPH could stem from both the topoisomerase II inhibition, ROS generation and DNA alkylation.
Collapse
|
382
|
In vitro/in vivo screening of oxidative homeostasis and damage to DNA, protein, and lipids using UPLC/MS-MS. Anal Bioanal Chem 2014; 406:5465-76. [DOI: 10.1007/s00216-014-7983-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/12/2014] [Accepted: 06/16/2014] [Indexed: 12/23/2022]
|
383
|
Kim HJ, Oh GS, Shen A, Lee SB, Choe SK, Kwon KB, Lee S, Seo KS, Kwak TH, Park R, So HS. Augmentation of NAD(+) by NQO1 attenuates cisplatin-mediated hearing impairment. Cell Death Dis 2014; 5:e1292. [PMID: 24922076 PMCID: PMC4611728 DOI: 10.1038/cddis.2014.255] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 04/18/2014] [Accepted: 05/09/2014] [Indexed: 12/20/2022]
Abstract
Cisplatin (cis-diaminedichloroplatinum-II) is an extensively used chemotherapeutic agent, and one of its most adverse effects is ototoxicity. A number of studies have demonstrated that these effects are related to oxidative stress and DNA damage. However, the precise mechanism underlying cisplatin-associated ototoxicity is still unclear. The cofactor nicotinamide adenine dinucleotide (NAD(+)) has emerged as a key regulator of cellular energy metabolism and homeostasis. Here, we demonstrate for the first time that, in cisplatin-mediated ototoxicity, the levels and activities of SIRT1 are suppressed by the reduction of intracellular NAD(+) levels. We provide evidence that the decrease in SIRT1 activity and expression facilitated by increasing poly(ADP-ribose) transferase (PARP)-1 activation and microRNA-34a through p53 activation aggravates cisplatin-mediated ototoxicity. Moreover, we show that the induction of cellular NAD(+) levels using β-lapachone (β-Lap), whose intracellular target is NQO1, prevents the toxic effects of cisplatin through the regulation of PARP-1 and SIRT1 activity. These results suggest that direct modulation of cellular NAD(+) levels by pharmacological agents could be a promising therapeutic approach for protection from cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- H-J Kim
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea
| | - G-S Oh
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea
| | - A Shen
- 1] Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea [2] BK21plus Program and Department of Smart Life-Care Convergence, Wonkwang University Graduate School, Jeonbuk, Republic of Korea
| | - S-B Lee
- 1] Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea [2] BK21plus Program and Department of Smart Life-Care Convergence, Wonkwang University Graduate School, Jeonbuk, Republic of Korea
| | - S-K Choe
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea
| | - K-B Kwon
- 1] Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea [2] BK21plus Program and Department of Smart Life-Care Convergence, Wonkwang University Graduate School, Jeonbuk, Republic of Korea [3] Department of Oriental Medical Physiology, College of Korean Medicine, Wonkwang University, Jeonbuk, Republic of Korea
| | - S Lee
- Life Science Research Center, KT&G Life Sciences, Suwon, Republic of Korea
| | - K-S Seo
- Life Science Research Center, KT&G Life Sciences, Suwon, Republic of Korea
| | - T H Kwak
- Life Science Research Center, KT&G Life Sciences, Suwon, Republic of Korea
| | - R Park
- 1] Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea [2] BK21plus Program and Department of Smart Life-Care Convergence, Wonkwang University Graduate School, Jeonbuk, Republic of Korea
| | - H-S So
- 1] Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Republic of Korea [2] BK21plus Program and Department of Smart Life-Care Convergence, Wonkwang University Graduate School, Jeonbuk, Republic of Korea
| |
Collapse
|
384
|
Bhattacharyya A, Chattopadhyay R, Mitra S, Crowe SE. Oxidative stress: an essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol Rev 2014; 94:329-54. [PMID: 24692350 DOI: 10.1152/physrev.00040.2012] [Citation(s) in RCA: 1395] [Impact Index Per Article: 139.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Reactive oxygen species (ROS) are generated as by-products of normal cellular metabolic activities. Superoxide dismutase, glutathione peroxidase, and catalase are the enzymes involved in protecting cells from the damaging effects of ROS. ROS are produced in response to ultraviolet radiation, cigarette smoking, alcohol, nonsteroidal anti-inflammatory drugs, ischemia-reperfusion injury, chronic infections, and inflammatory disorders. Disruption of normal cellular homeostasis by redox signaling may result in cardiovascular, neurodegenerative diseases and cancer. ROS are produced within the gastrointestinal (GI) tract, but their roles in pathophysiology and disease pathogenesis have not been well studied. Despite the protective barrier provided by the mucosa, ingested materials and microbial pathogens can induce oxidative injury and GI inflammatory responses involving the epithelium and immune/inflammatory cells. The pathogenesis of various GI diseases including peptic ulcers, gastrointestinal cancers, and inflammatory bowel disease is in part due to oxidative stress. Unraveling the signaling events initiated at the cellular level by oxidative free radicals as well as the physiological responses to such stress is important to better understand disease pathogenesis and to develop new therapies to manage a variety of conditions for which current therapies are not always sufficient.
Collapse
|
385
|
Oxytocin improves follicular reserve in a cisplatin-induced gonadotoxicity model in rats. BIOMED RESEARCH INTERNATIONAL 2014; 2014:703691. [PMID: 24959584 PMCID: PMC4053297 DOI: 10.1155/2014/703691] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 04/11/2014] [Accepted: 04/23/2014] [Indexed: 11/30/2022]
Abstract
Cisplatin (CP), an antitumor agent, has been shown to cause ovarian injury and dysfunction in both animal and human studies. The present study was conducted to investigate the protective effect of oxytocin (OT) on CP-induced ovarian toxicity in rats. Twenty-one adult female rats were included in the study. Fourteen rats were administered intraperitoneally CP (2 mg/kg/day) twice a week for 5 weeks. Control group (n = 7) did not receive any treatment. Following treatment, CP-received rats were randomly divided into two groups and treated with either saline (1 mL/kg/day, n = 7) or OT (160 μg/kg/day, n = 7) for 5 weeks. Then, ovarian toxicity and effects of OT were evaluated by histomorphological and biochemical analysis. Our findings revealed a significant reduction in the number of follicles at each grade in saline-treated group. AMH level was significantly lower in saline group compared to control (P < 0.0005). OT treatment significantly attenuated CP toxicity in ovaries and increased AMH levels compared to saline group (P < 0.005). Also, administration of OT lessened lipid peroxidation and prevented glutathione depletion in CP-treated rats (P < 0.05). These results indicated that OT could lessen the CP-induced ovarian damage and improve follicular reserve by preventing oxidative damage.
Collapse
|
386
|
Prooxidant mechanisms in toxicology. BIOMED RESEARCH INTERNATIONAL 2014; 2014:308625. [PMID: 24772419 PMCID: PMC3977463 DOI: 10.1155/2014/308625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 01/20/2014] [Indexed: 01/03/2023]
|
387
|
Phase II enzyme induction by a carotenoid, lutein, in a PC12D neuronal cell line. Biochem Biophys Res Commun 2014; 446:535-40. [DOI: 10.1016/j.bbrc.2014.02.135] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 02/28/2014] [Indexed: 12/31/2022]
|
388
|
Tang Y, Chen R, Huang Y, Li G, Huang Y, Chen J, Duan L, Zhu BT, Thrasher JB, Zhang X, Li B. Natural compound Alternol induces oxidative stress-dependent apoptotic cell death preferentially in prostate cancer cells. Mol Cancer Ther 2014; 13:1526-36. [PMID: 24688053 DOI: 10.1158/1535-7163.mct-13-0981] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Prostate cancers at the late stage of castration resistance are not responding well to most of current therapies available in clinic, reflecting a desperate need of novel treatment for this life-threatening disease. In this study, we evaluated the anticancer effect of a recently isolated natural compound, Alternol, in multiple prostate cancer cell lines with the properties of advanced prostate cancers in comparison to prostate-derived nonmalignant cells. As assessed by trypan blue exclusion assay, significant cell death was observed in all prostate cancer cell lines except DU145 but not in nonmalignant (RWPE-1 and BPH1) cells. Further analyses revealed that Alternol-induced cell death was an apoptotic response in a dose- and time-dependent manner, as evidenced by the appearance of apoptosis hallmarks such as caspase-3 processing and PARP cleavage. Interestingly, Alternol-induced cell death was completely abolished by reactive oxygen species scavengers N-acetylcysteine and dihydrolipoic acid. We also demonstrated that the proapoptotic Bax protein was activated after Alternol treatment and was critical for Alternol-induced apoptosis. Animal xenograft experiments in nude mice showed that Alternol treatment largely suppressed tumor growth of PC-3 xenografts but not Bax-null DU-145 xenografts in vivo. These data suggest that Alternol might serve as a novel anticancer agent for patients with late-stage prostate cancer.
Collapse
Affiliation(s)
- Yuzhe Tang
- Authors' Affiliations: Department of Urology, Military Postgraduate Medical College, Chinese People's Liberation Army General Hospital, Beijing, China; Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas; Department of Pharmacology, Three Gorges University College of Medicine, Yichang, China; Strand Biotechnology Institute of Research, Shantou, China; Department of Pharmacology & Toxicology, The University of Kansas Medical Center, Kansas City, KansasAuthors' Affiliations: Department of Urology, Military Postgraduate Medical College, Chinese People's Liberation Army General Hospital, Beijing, China; Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas; Department of Pharmacology, Three Gorges University College of Medicine, Yichang, China; Strand Biotechnology Institute of Research, Shantou, China; Department of Pharmacology & Toxicology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Ruibao Chen
- Authors' Affiliations: Department of Urology, Military Postgraduate Medical College, Chinese People's Liberation Army General Hospital, Beijing, China; Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas; Department of Pharmacology, Three Gorges University College of Medicine, Yichang, China; Strand Biotechnology Institute of Research, Shantou, China; Department of Pharmacology & Toxicology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Yan Huang
- Authors' Affiliations: Department of Urology, Military Postgraduate Medical College, Chinese People's Liberation Army General Hospital, Beijing, China; Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas; Department of Pharmacology, Three Gorges University College of Medicine, Yichang, China; Strand Biotechnology Institute of Research, Shantou, China; Department of Pharmacology & Toxicology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Guodong Li
- Authors' Affiliations: Department of Urology, Military Postgraduate Medical College, Chinese People's Liberation Army General Hospital, Beijing, China; Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas; Department of Pharmacology, Three Gorges University College of Medicine, Yichang, China; Strand Biotechnology Institute of Research, Shantou, China; Department of Pharmacology & Toxicology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Yiling Huang
- Authors' Affiliations: Department of Urology, Military Postgraduate Medical College, Chinese People's Liberation Army General Hospital, Beijing, China; Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas; Department of Pharmacology, Three Gorges University College of Medicine, Yichang, China; Strand Biotechnology Institute of Research, Shantou, China; Department of Pharmacology & Toxicology, The University of Kansas Medical Center, Kansas City, KansasAuthors' Affiliations: Department of Urology, Military Postgraduate Medical College, Chinese People's Liberation Army General Hospital, Beijing, China; Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas; Department of Pharmacology, Three Gorges University College of Medicine, Yichang, China; Strand Biotechnology Institute of Research, Shantou, China; Department of Pharmacology & Toxicology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Jiepeng Chen
- Authors' Affiliations: Department of Urology, Military Postgraduate Medical College, Chinese People's Liberation Army General Hospital, Beijing, China; Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas; Department of Pharmacology, Three Gorges University College of Medicine, Yichang, China; Strand Biotechnology Institute of Research, Shantou, China; Department of Pharmacology & Toxicology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Lili Duan
- Authors' Affiliations: Department of Urology, Military Postgraduate Medical College, Chinese People's Liberation Army General Hospital, Beijing, China; Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas; Department of Pharmacology, Three Gorges University College of Medicine, Yichang, China; Strand Biotechnology Institute of Research, Shantou, China; Department of Pharmacology & Toxicology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Bao-Ting Zhu
- Authors' Affiliations: Department of Urology, Military Postgraduate Medical College, Chinese People's Liberation Army General Hospital, Beijing, China; Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas; Department of Pharmacology, Three Gorges University College of Medicine, Yichang, China; Strand Biotechnology Institute of Research, Shantou, China; Department of Pharmacology & Toxicology, The University of Kansas Medical Center, Kansas City, Kansas
| | - J Brantley Thrasher
- Authors' Affiliations: Department of Urology, Military Postgraduate Medical College, Chinese People's Liberation Army General Hospital, Beijing, China; Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas; Department of Pharmacology, Three Gorges University College of Medicine, Yichang, China; Strand Biotechnology Institute of Research, Shantou, China; Department of Pharmacology & Toxicology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Xu Zhang
- Authors' Affiliations: Department of Urology, Military Postgraduate Medical College, Chinese People's Liberation Army General Hospital, Beijing, China; Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas; Department of Pharmacology, Three Gorges University College of Medicine, Yichang, China; Strand Biotechnology Institute of Research, Shantou, China; Department of Pharmacology & Toxicology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Benyi Li
- Authors' Affiliations: Department of Urology, Military Postgraduate Medical College, Chinese People's Liberation Army General Hospital, Beijing, China; Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas; Department of Pharmacology, Three Gorges University College of Medicine, Yichang, China; Strand Biotechnology Institute of Research, Shantou, China; Department of Pharmacology & Toxicology, The University of Kansas Medical Center, Kansas City, KansasAuthors' Affiliations: Department of Urology, Military Postgraduate Medical College, Chinese People's Liberation Army General Hospital, Beijing, China; Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas; Department of Pharmacology, Three Gorges University College of Medicine, Yichang, China; Strand Biotechnology Institute of Research, Shantou, China; Department of Pharmacology & Toxicology, The University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
389
|
Environmental pollutants and lifestyle factors induce oxidative stress and poor prenatal development. Reprod Biomed Online 2014; 29:17-31. [PMID: 24813750 DOI: 10.1016/j.rbmo.2014.03.002] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/13/2013] [Accepted: 03/06/2014] [Indexed: 01/20/2023]
Abstract
Developmental toxicity caused by exposure to a mixture of environmental pollutants has become a major health concern. Human-made chemicals, including xenoestrogens, pesticides and heavy metals, as well as unhealthy lifestyle behaviours, mainly tobacco smoking, alcohol consumption and medical drug abuse, are major factors that adversely influence prenatal development and increase susceptibility of offspring to diseases. There is evidence to suggest that the developmental toxicological mechanisms of chemicals and lifestyle factors involve the generation of reactive oxygen species (ROS) and cellular oxidative damage. Overproduction of ROS induces oxidative stress, a state where increased ROS generation overwhelms antioxidant protection and subsequently leads to oxidative damage of cellular macromolecules. Data on the involvement of oxidative stress in the mechanism of developmental toxicity following exposure to environmental pollutants are reviewed in an attempt to provide an updated basis for future studies on the toxic effect of such pollutants, particularly the notion of increased risk for developmental toxicity due to combined and cumulative exposure to various environmental pollutants. The aims of such studies are to better understand the mechanisms by which environmental pollutants adversely affect conceptus development and to elucidate the impact of cumulative exposures to multiple pollutants on post-natal development and health outcomes. Developmental toxicity caused by exposure to mixture of environmental pollutants has become a major health concern. Human-made chemicals, including xenoestrogens, pesticides and heavy metals, as well as unhealthy lifestyle behaviors, mainly tobacco smoking, alcohol consumption and medical drug abuse, are major factors that adversely influence prenatal development and increase the susceptibility of offspring to development complications and diseases. There is evidence to suggest that the developmental toxicological mechanisms of human-made chemicals and unhealthy lifestyle factors involve the generation of reactive oxygen species (ROS) and cellular oxidative damage. Overproduction of ROS induces oxidative stress, a state where increased generation of ROS overwhelms antioxidant protection and subsequently leads to oxidative damage of cellular macromolecules. Exposure to various environmental pollutants induces synergic and cumulative dose-additive adverse effects on prenatal development, pregnancy outcomes and neonate health. Data from the literature on the involvement of oxidative stress in the mechanism of developmental toxicity following in vivo exposure to environmental pollutants will be reviewed in an attempt to provide an updated basis for future studies on the toxic effect of such pollutants, particularly the notion of increased risk for developmental toxicity due to combined and cumulative exposure to various environmental pollutants. The aims of such studies are to better understand the mechanisms by which environmental pollutants adversely affect conceptus development and to elucidate the impact of cumulative exposures to multiple pollutants on postnatal development and health outcomes.
Collapse
|
390
|
Li Y, Dai F, Jin XL, Ma MM, Wang YH, Ren XR, Zhou B. An effective strategy to develop active cinnamic acid-directed antioxidants based on elongating the conjugated chains. Food Chem 2014; 158:41-7. [PMID: 24731312 DOI: 10.1016/j.foodchem.2014.02.092] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 02/07/2014] [Accepted: 02/18/2014] [Indexed: 12/15/2022]
Abstract
To optimize antioxidant activity and lipophilicity of cinnamic acid derivatives (CAs) including ferulic acid, sinapic acid, 3,4-dimethoxycinnamic acid, and p-hydroxycinnamic acid, four analogs bearing an additional double bond between their aromatic ring and propenoic acid moiety were designed and synthesized based on the conjugated chain elongation strategy. The antioxidant performance of the CAs were investigated by 2,2'-diphenyl-1-picrylhydrazyl (DPPH)-scavenging, ferric reducing/antioxidant power, cyclic voltammetry, DNA strand breakage-inhibiting and anti-haemolysis activity assays. It was found that CAs with elongation of conjugated chains display increased DPPH-scavenging, DNA strand breakage-inhibiting and anti-haemolysis activities as compared to their parent molecules, due to their improved hydrogen atom-donating ability and lipophilicity. Overall, this work highlights an effective strategy to develop potential CA-directed antioxidants by elongating their conjugated chain.
Collapse
Affiliation(s)
- Yan Li
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Fang Dai
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, Gansu 730000, China.
| | - Xiao-Ling Jin
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Meng-Meng Ma
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Yi-Hua Wang
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Xiao-Rong Ren
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Bo Zhou
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou, Gansu 730000, China.
| |
Collapse
|
391
|
Cesari IM, Carvalho E, Figueiredo Rodrigues M, Mendonça BDS, Amôedo ND, Rumjanek FD. Methyl jasmonate: putative mechanisms of action on cancer cells cycle, metabolism, and apoptosis. Int J Cell Biol 2014; 2014:572097. [PMID: 24648844 PMCID: PMC3933403 DOI: 10.1155/2014/572097] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Revised: 11/06/2013] [Accepted: 11/07/2013] [Indexed: 12/29/2022] Open
Abstract
Methyl jasmonate (MJ), an oxylipid that induces defense-related mechanisms in plants, has been shown to be active against cancer cells both in vitro and in vivo, without affecting normal cells. Here we review most of the described MJ activities in an attempt to get an integrated view and better understanding of its multifaceted modes of action. MJ (1) arrests cell cycle, inhibiting cell growth and proliferation, (2) causes cell death through the intrinsic/extrinsic proapoptotic, p53-independent apoptotic, and nonapoptotic (necrosis) pathways, (3) detaches hexokinase from the voltage-dependent anion channel, dissociating glycolytic and mitochondrial functions, decreasing the mitochondrial membrane potential, favoring cytochrome c release and ATP depletion, activating pro-apoptotic, and inactivating antiapoptotic proteins, (4) induces reactive oxygen species mediated responses, (5) stimulates MAPK-stress signaling and redifferentiation in leukemia cells, (6) inhibits overexpressed proinflammatory enzymes in cancer cells such as aldo-keto reductase 1 and 5-lipoxygenase, and (7) inhibits cell migration and shows antiangiogenic and antimetastatic activities. Finally, MJ may act as a chemosensitizer to some chemotherapics helping to overcome drug resistant. The complete lack of toxicity to normal cells and the rapidity by which MJ causes damage to cancer cells turn MJ into a promising anticancer agent that can be used alone or in combination with other agents.
Collapse
Affiliation(s)
- Italo Mario Cesari
- Laboratório de Bioquímica e Biologia Molecular do Câncer, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Prédio CCS, Bloco E, Sala 22, Ilha do Fundão, Cidade Universitária, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Erika Carvalho
- Laboratório de Bioquímica e Biologia Molecular do Câncer, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Prédio CCS, Bloco E, Sala 22, Ilha do Fundão, Cidade Universitária, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Mariana Figueiredo Rodrigues
- Laboratório de Bioquímica e Biologia Molecular do Câncer, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Prédio CCS, Bloco E, Sala 22, Ilha do Fundão, Cidade Universitária, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Bruna dos Santos Mendonça
- Laboratório de Bioquímica e Biologia Molecular do Câncer, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Prédio CCS, Bloco E, Sala 22, Ilha do Fundão, Cidade Universitária, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Nivea Dias Amôedo
- Laboratório de Bioquímica e Biologia Molecular do Câncer, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Prédio CCS, Bloco E, Sala 22, Ilha do Fundão, Cidade Universitária, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Franklin David Rumjanek
- Laboratório de Bioquímica e Biologia Molecular do Câncer, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, Prédio CCS, Bloco E, Sala 22, Ilha do Fundão, Cidade Universitária, 21941-902 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
392
|
Miura S, Shibazaki M, Kasai S, Yasuhira S, Watanabe A, Inoue T, Kageshita Y, Tsunoda K, Takahashi K, Akasaka T, Masuda T, Maesawa C. A Somatic Mutation of the KEAP1 Gene in Malignant Melanoma Is Involved in Aberrant NRF2 Activation and an Increase in Intrinsic Drug Resistance. J Invest Dermatol 2014; 134:553-556. [DOI: 10.1038/jid.2013.343] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
393
|
Azad G, Singh V, Mandal P, Singh P, Golla U, Baranwal S, Chauhan S, Tomar RS. Ebselen induces reactive oxygen species (ROS)-mediated cytotoxicity in Saccharomyces cerevisiae with inhibition of glutamate dehydrogenase being a target. FEBS Open Bio 2014; 4:77-89. [PMID: 24490132 PMCID: PMC3907691 DOI: 10.1016/j.fob.2014.01.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 01/03/2014] [Accepted: 01/03/2014] [Indexed: 12/12/2022] Open
Abstract
Ebselen is a synthetic, lipid-soluble seleno-organic compound. The high electrophilicity of ebselen enables it to react with multiple cysteine residues of various proteins. Despite extensive research on ebselen, its target molecules and mechanism of action remains less understood. We performed biochemical as well as in vivo experiments employing budding yeast as a model organism to understand the mode of action of ebselen. The growth curve analysis and FACS (florescence activated cell sorting) assays revealed that ebselen exerts growth inhibitory effects on yeast cells by causing a delay in cell cycle progression. We observed that ebselen exposure causes an increase in intracellular ROS levels and mitochondrial membrane potential, and that these effects were reversed by addition of antioxidants such as reduced glutathione (GSH) or N-acetyl-l-cysteine (NAC). Interestingly, a significant increase in ROS levels was noticed in gdh3-deleted cells compared to wild-type cells. Furthermore, we showed that ebselen inhibits GDH function by interacting with its cysteine residues, leading to the formation of inactive hexameric GDH. Two-dimensional gel electrophoresis revealed protein targets of ebselen including CPR1, the yeast homolog of Cyclophilin A. Additionally, ebselen treatment leads to the inhibition of yeast sporulation. These results indicate a novel direct connection between ebselen and redox homeostasis.
Collapse
Key Words
- CypA, Cyclophilin A
- DCFH-DA, 2,7-dichlorodihydrofluorescein diacetate
- Ebselen
- FACS, florescence activated cell sorting
- GDH, glutamate dehydrogenase
- GSH, glutathione
- Glutamate dehydrogenase
- Histone clipping
- Mitochondrial membrane potential
- NAC, N-acetyl-l-cysteine
- Ni-NTA, nickel-nitrilotriacetic acid
- ROS levels
- ROS, reactive oxygen species
- SOD, superoxide dismutase
- Yeast sporulation
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Raghuvir S. Tomar
- Laboratory of Chromatin Biology, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal 462023, India
| |
Collapse
|
394
|
Thushara RM, Hemshekhar M, Kemparaju K, Rangappa KS, Devaraja S, Girish KS. Therapeutic drug-induced platelet apoptosis: an overlooked issue in pharmacotoxicology. Arch Toxicol 2013; 88:185-98. [PMID: 24363025 DOI: 10.1007/s00204-013-1185-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 12/11/2013] [Indexed: 11/25/2022]
Abstract
The surfacing of the applied fields of biology such as, biotechnology, pharmacology and drug discovery was a boon to the modern man. However, it had its share of disadvantages too. The indiscriminate use of antibiotics and other biological drugs resulted in numerous adverse reactions including thrombocytopenia. One of the reasons for drug-induced thrombocytopenia could be attributed to an enhanced rate of platelet apoptosis, which is a less investigated aspect. The present essay sheds light on the adverse (pro-apoptotic) effects of some of the commonly used drugs and antibiotics on platelets viz. cisplatin, aspirin, vancomycin and balhimycin. Furthermore, the undesirable reactions resulting from chemotherapy could be attributed at least to some extent to the systemic stress induced by microparticles, which in turn are the byproducts of platelet apoptosis. Thereby, the essay aims to highlight the challenges in the emerging trend of cross-disciplinary implications, i.e., drug-induced platelet apoptosis, which is a nascent field. Thus, the different mechanisms through which drugs induce platelet apoptosis are discussed, which also opens up a new perspective through which the adverse effects of commonly used drugs could be dealt. The drug-associated platelet toxicity is of grave concern and demands immediate attention. Besides, it would also be appealing to examine the platelet pro-apoptotic effects of other commonly used therapeutic drugs.
Collapse
Affiliation(s)
- R M Thushara
- Department of Studies in Biochemistry, University of Mysore, Mysore, 570006, India
| | | | | | | | | | | |
Collapse
|
395
|
Meierjohann S. Oxidative stress in melanocyte senescence and melanoma transformation. Eur J Cell Biol 2013; 93:36-41. [PMID: 24342719 DOI: 10.1016/j.ejcb.2013.11.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 11/15/2013] [Accepted: 11/15/2013] [Indexed: 10/26/2022] Open
Abstract
Melanoma is a severe type of skin cancer with a high metastasis potential and poor survival rates once metastasized. The causes of melanoma formation are multifactorial and not fully understood. Several signaling cascades such as the RAS/RAF/ERK1/2 pathway, the PI3K/AKT pathway, RAC1 and NF-κB are involved in melanoma initiation and progression. Reactive oxygen species (ROS) are induced by these signal transduction cascades, and they play a fundamental role in melanomagenic processes. Cells derived from the melanocytic lineage are particularly sensitive to an increase in ROS, and thus, melanoma cells rely on efficient antioxidant measures. This review summarizes the causes and consequences of ROS generation in melanocytes and melanoma and discusses the potential of pro-oxidant therapy in melanoma treatment.
Collapse
Affiliation(s)
- Svenja Meierjohann
- University of Wurzburg, Department of Physiological Chemistry I, Biocenter, Am Hubland, 97074, Wurzburg, Germany; Comprehensive Cancer Center Mainfranken, University Clinic Würzburg, 97078, Würzburg, Germany.
| |
Collapse
|
396
|
Zheng J, Piao MJ, Keum YS, Kim HS, Hyun JW. Fucoxanthin Protects Cultured Human Keratinocytes against Oxidative Stress by Blocking Free Radicals and Inhibiting Apoptosis. Biomol Ther (Seoul) 2013; 21:270-6. [PMID: 24244811 PMCID: PMC3819899 DOI: 10.4062/biomolther.2013.030] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 05/05/2013] [Accepted: 05/08/2013] [Indexed: 11/29/2022] Open
Abstract
Fucoxanthin is an important carotenoid derived from edible brown seaweeds and is used in indigenous herbal medicines. The aim of the present study was to examine the cytoprotective effects of fucoxanthin against hydrogen peroxide-induced cell damage. Fucoxanthin decreased the level of intracellular reactive oxygen species, as assessed by fluorescence spectrometry performed after staining cultured human HaCaT keratinocytes with 2',7'-dichlorodihydrofl uorescein diacetate. In addition, electron spin resonance spectrometry showed that fucoxanthin scavenged hydroxyl radical generated by the Fenton reaction in a cell-free system. Fucoxanthin also inhibited comet tail formation and phospho-histone H2A.X expression, suggesting that it prevents hydrogen peroxideinduced cellular DNA damage. Furthermore, the compound reduced the number of apoptotic bodies stained with Hoechst 33342, indicating that it protected keratinocytes against hydrogen peroxide-induced apoptotic cell death. Finally, fucoxanthin prevented the loss of mitochondrial membrane potential. These protective actions were accompanied by the down-regulation of apoptosispromoting mediators (i.e., B-cell lymphoma-2-associated x protein, caspase-9, and caspase-3) and the up-regulation of an apoptosis inhibitor (B-cell lymphoma-2). Taken together, the results of this study suggest that fucoxanthin defends keratinocytes against oxidative damage by scavenging ROS and inhibiting apoptosis.
Collapse
Affiliation(s)
- Jian Zheng
- School of Medicine and Institute for Nuclear Science and Technology, Jeju National University, Jeju 690-756
| | | | | | | | | |
Collapse
|
397
|
Nordgren KKS, Wallace KB. Keap1 redox-dependent regulation of doxorubicin-induced oxidative stress response in cardiac myoblasts. Toxicol Appl Pharmacol 2013; 274:107-16. [PMID: 24211725 DOI: 10.1016/j.taap.2013.10.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 10/22/2013] [Accepted: 10/24/2013] [Indexed: 11/18/2022]
Abstract
Doxorubicin (DOX) is a widely prescribed treatment for a broad scope of cancers, but clinical utility is limited by the cumulative, dose-dependent cardiomyopathy that occurs with repeated administration. DOX-induced cardiotoxicity is associated with the production of reactive oxygen species (ROS) and oxidation of lipids, DNA and proteins. A major cellular defense mechanism against such oxidative stress is activation of the Keap1/Nrf2-antioxidant response element (ARE) signaling pathway, which transcriptionally regulates expression of antioxidant genes such as Nqo1 and Gstp1. In the present study, we address the hypothesis that an initial event associated with DOX-induced oxidative stress is activation of the Keap1/Nrf2-dependent expression of antioxidant genes and that this is regulated through drug-induced changes in redox status of the Keap1 protein. Incubation of H9c2 rat cardiac myoblasts with DOX resulted in a time- and dose-dependent decrease in non-protein sulfhydryl groups. Associated with this was a near 2-fold increase in Nrf2 protein content and enhanced transcription of several of the Nrf2-regulated down-stream genes, including Gstp1, Ugt1a1, and Nqo1; the expression of Nfe2l2 (Nrf2) itself was unaltered. Furthermore, both the redox status and the total amount of Keap1 protein were significantly decreased by DOX, with the loss of Keap1 being due to both inhibited gene expression and increased autophagic, but not proteasomal, degradation. These findings identify the Keap1/Nrf2 pathway as a potentially important initial response to acute DOX-induced oxidative injury, with the primary regulatory events being the oxidation and autophagic degradation of the redox sensor Keap1 protein.
Collapse
Affiliation(s)
- Kendra K S Nordgren
- University of Minnesota Medical School Duluth, 1035 University Dr., 252 SMed, Duluth, MN 55812, USA.
| | - Kendall B Wallace
- University of Minnesota Medical School Duluth, 1035 University Dr., 252 SMed, Duluth, MN 55812, USA.
| |
Collapse
|
398
|
Chen YC, Huang WJ, Hsu JL, Yu CC, Wang WT, Guh JH. A novel hydroxysuberamide derivative potentiates MG132-mediated anticancer activity against human hormone refractory prostate cancers--the role of histone deacetylase and endoplasmic reticulum stress. Prostate 2013; 73:1270-80. [PMID: 23813634 DOI: 10.1002/pros.22641] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 12/18/2012] [Indexed: 01/05/2023]
Abstract
BACKGROUND Histone deacetylase (HDAC) inhibitors are successful for treatment of advanced cutaneous T-cell lymphoma but only show modest effect in solid tumors. Approaches for HDAC inhibitors to improve activity against solid tumors are necessary. METHODS Sulforhodamine B assay and flow cytometric analysis detected cell proliferation and cell-cycle progression, respectively. Protein expression was determined by Western blotting. Comet assay and DNA end-binding activity of Ku proteins detected DNA damage and DNA repair activity, respectively. siRNA technique was used for knockdown of specific cellular target. RESULTS WJ25591 displayed inhibitory activity against HDAC1 and cell proliferation in human hormone-refractory prostate cancers PC-3 and DU-145. WJ25591 caused an arrest of cell-cycle at both G1- and G2-phase and increased protein expressions of p21 and cyclin E, followed by cell apoptosis. WJ25591-induced Bcl-2 down-regulation and activation of caspase-9, -8, and -3, suggesting apoptotic execution through both intrinsic and extrinsic apoptotic pathways. WJ25591 also significantly inhibited DNA repair activity but not directly induced DNA damage. Moreover, the proteasome inhibitor MG-132 dramatically sensitized WJ25591-induced cell apoptosis. The siRNA technique demonstrated that endoplasmic reticulum (ER) stress, in particular CHOP/GADD153 up-regulation, contributed to the synergistic effect. CONCLUSIONS The data suggest that WJ25591 inhibited HDAC activity, leading to cell-cycle arrest and inhibition of DNA repair. Caspase cascades are subsequently triggered to execute cell apoptosis. MG-132 dramatically sensitizes WJ25591-mediated apoptosis, at least partly, through ER stress response. The data also reveal that combination of HDAC inhibitors and proteasome inhibitors may be a potential strategy against hormone-refractory prostate cancers.
Collapse
Affiliation(s)
- Yi-Cheng Chen
- College of Medicine, School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
399
|
Coşkun N, Hatipoğlu MT, Ozoğul C, Korkmaz C, Akyol SN, Mıcılı SC, Arık GS, Erdoğan D. The protective effects of acetyl L-carnitine on testis gonadotoxicity induced by Cisplatin in rats. Balkan Med J 2013; 30:235-41. [PMID: 25207106 DOI: 10.5152/balkanmedj.2013.7340] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 02/14/2013] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Cisplatin, an effective antineoplastic agent, damages normal cells in a manner related to chemotherapy. Acetyl L-carnitine protects cells against mitochondrial and nuclear damage induced by chemotherapy. AIMS Animal experiment. STUDY DESIGN The aim of this study was to examine the protective effects of acetyl L-carnitine on cisplatin-induced gonadotoxicity in testicular structures. Twenty-four male Wistar albino rats were divided into four Groups (n=6): Group 1 (control) was administered saline; Group 2 was administered acetyl L-carnitine; Group 3 was administered cisplatin; and Group 4 was pre-treated with acetyl L-carnitine before cisplatin administration. METHODS After 72hr of treatment with cisplatin, the rats were sacrificed, and the testicular tissues were removed. Morphometric, histomorphologic and immunohistochemical analyses were conducted. RESULTS At the end of the experiment, Group 3 was characterised by statistically significant weight loss, a degenerative appearance of the seminiferous tubules in the peripheral region, separation of spermatogenic cell series from the tubular wall, cellular debris in the lumen and central interstitial oedema. Sperm morphology appeared to be abnormal. Tubular diameter and wall thickness decreased, and the number of TUNEL- and active caspase-positive cells increased compared with the other Groups. The histological findings in Group 4 were better than those in Group 3. CONCLUSION It was concluded that the prophylactic use of acetyl L-carnitine protects against cisplatin-induced testicular tissue damage.
Collapse
Affiliation(s)
- Neslihan Coşkun
- Department of Histology and Embriyology, Gazi University School of Medicine, Ankara, Turkey
| | - M Tahir Hatipoğlu
- Department of Histology and Embriyology, Gazi University School of Medicine, Ankara, Turkey
| | - Candan Ozoğul
- Department of Histology and Embriyology, Gazi University School of Medicine, Ankara, Turkey
| | - Cem Korkmaz
- IVF Laboratories, Gülhane Military Medical Academy, Ankara, Turkey
| | - Seda Nur Akyol
- Department of Histology and Embriyology, Gazi University School of Medicine, Ankara, Turkey
| | - Serap Cilaker Mıcılı
- Department of Histology and Embriyology, Dokuz Eylul University School of Medicine, İzmir, Turkey
| | - Gülistan Sanem Arık
- Department of Histology and Embriyology, Gazi University School of Medicine, Ankara, Turkey
| | - Deniz Erdoğan
- Department of Histology and Embriyology, Gazi University School of Medicine, Ankara, Turkey
| |
Collapse
|
400
|
Koch M, Krieger ML, Stölting D, Brenner N, Beier M, Jaehde U, Wiese M, Royer HD, Bendas G. Overcoming chemotherapy resistance of ovarian cancer cells by liposomal cisplatin: molecular mechanisms unveiled by gene expression profiling. Biochem Pharmacol 2013; 85:1077-90. [PMID: 23396090 DOI: 10.1016/j.bcp.2013.01.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 01/29/2013] [Accepted: 01/30/2013] [Indexed: 12/12/2022]
Abstract
Previously we reported that liposomal cisplatin (CDDP) overcomes CDDP resistance of ovarian A2780cis cancer cells (Krieger et al., Int. J. Pharm. 389, 2010, 10-17). Here we find that the cytotoxic activity of liposomal CDDP is not associated with detectable DNA platination in resistant ovarian cancer cells. This suggests that the mode of action of liposomal CDDP is different from the free drug. To gain insight into mechanisms of liposomal CDDP activity, we performed a transcriptome analysis of untreated A2780cis cells, and A2780cis cells in response to exposure with IC50 values of free or liposomal CDDP. A process network analysis of upregulated genes showed that liposomal CDDP induced a highly different gene expression profile in comparison to the free drug. p53 was identified as a key player directing transcriptional responses to free or liposomal CDDP. The free drug induced expression of essential genes of the intrinsic (mitochondrial) apoptosis pathway (BAX, BID, CASP9) most likely through p38MAPK activation. In contrast, liposomal CDDP induced expression of genes from DNA damage pathways and several genes of the extrinsic pathway of apoptosis (TNFRSF10B-DR5, CD70-TNFSF7). It thus appears that liposomal CDDP overcomes CDDP resistance by inducing DNA damage and in consequence programmed cell death by the extrinsic pathway. Predictions from gene expression data with respect to apoptosis activation were confirmed at the protein level by an apoptosis antibody array. This sheds new light on liposomal drug carrier approaches in cancer and suggests liposomal CDDP as promising strategy for the treatment of CDDP resistant ovarian carcinomas.
Collapse
Affiliation(s)
- Martin Koch
- Pharmaceutical Institute, Rheinische Friedrich Wilhelms University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|