1
|
Mahadik SR, Reddy ART, Choudhary K, Nama L, Jamdade MS, Singh S, Murti K, Kumar N. Arsenic induced cardiotoxicity: An approach for molecular markers, epigenetic predictors and targets. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 111:104558. [PMID: 39245244 DOI: 10.1016/j.etap.2024.104558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/15/2024] [Accepted: 08/31/2024] [Indexed: 09/10/2024]
Abstract
Arsenic, a ubiquitous environmental toxicant, has been acknowledged as a significant issue for public health due to its widespread pollution of drinking water and food supplies. The present review aimed to study the toxicity associated with the cardiac system. Prolonged exposure to arsenic has been associated with several harmful health outcomes, especially cardiotoxicity. Arsenic-induced cardiotoxicity encompasses a range of cardiovascular abnormalities, including cardiac arrhythmias, ischemic heart disease, and cardiomyopathy. To tackle this toxicity, understanding the molecular markers, epigenetic predictors, and targets involved in arsenic-induced cardiotoxicity is essential for creating preventative and therapeutic approaches. For preventive measures against this heavy metal poisoning of groundwater, it is crucial to regularly monitor water quality, re-evaluate scientific findings, and educate the public about the possible risks. This review thoroughly summarised what is currently known in this field, highlighting the key molecular markers, epigenetic modifications, and potential therapeutic targets associated with arsenic-induced cardiotoxicity.
Collapse
Affiliation(s)
- Sakshi Ramesh Mahadik
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India
| | - Annem Ravi Teja Reddy
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India
| | - Khushboo Choudhary
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India
| | - Lokesh Nama
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India
| | - Mohini Santosh Jamdade
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India
| | - Krishna Murti
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali, Bihar 844102, India.
| |
Collapse
|
2
|
Hwang JH, An GJ, Kim CH, Chung HY, Lim KM. Trivalent arsenicals induce skin toxicity through thiol depletion. Toxicol Appl Pharmacol 2024; 492:117115. [PMID: 39357682 DOI: 10.1016/j.taap.2024.117115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/13/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Arsenic, a widespread environmental contaminant, is highly toxic to human health. Arsenic exposure is associated with the occurrence of skin lesions and diseases. This study investigated the dermal toxicity of trivalent arsenicals (AsIII and MMAIII) and its underlying mechanism using human keratinocyte cell line and ex vivo porcine skin. AsIII and MMAIII induced concentration-dependent cell apoptosis and necrosis in HaCaT cells, which was confirmed in ex vivo porcine skin. AsIII and MMAIII increased reactive oxygen species generation and GSH depletion. Interestingly, radical scavenger antioxidants such as Vitamin C failed to mitigate arsenic-induced cytotoxicity, while thiol-containing compounds effectively alleviated it, suggesting a key role of thiol depletion in the trivalent arsenical-induced dermal toxicity. DMSA showed the strongest protective effects against AsIII and MMAIII-induced cytotoxicity in HaCaT cells. Of note, DMSA restored arsenical-induced tissue damage, and reduced the apoptosis in ex vivo porcine skin, highlighting its potential use to alleviate arsenic-induced skin lesions and diseases.
Collapse
Affiliation(s)
- Jee-Hyun Hwang
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Gwang Jin An
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Chang-Hwan Kim
- Chem-Bio Technology Center, Agency for Defense Development, Republic of Korea
| | - Han Young Chung
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea.
| | - Kyung-Min Lim
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
3
|
Unsal V, Cicek M, Aktepe N, Oner E. Morin attenuates arsenic-induced toxicity in 3T3 embryonic fibroblast cells by suppressing oxidative stress, inflammation, and apoptosis: In vitro and silico evaluations. Toxicol Res (Camb) 2024; 13:tfae113. [PMID: 39036522 PMCID: PMC11260228 DOI: 10.1093/toxres/tfae113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/13/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
This study aims to investigate the curative effects of Morin, a flavonoid, against arsenic toxicity in 3T3 embryonic fibroblast cells and its effect on the molecular mechanisms of cells. The cytotoxicity and viability of the cells were measured by MTT and LDH tests. Arsenic (0.74 μM) was used to trigger toxicity and Morin (50 μM) was used for treatment. The levels of oxidative stress biomarkers and the activities of antioxidant enzymes were measured by spectrophotometric method, and inflammatory markers were measured by ELISA method. While mRNA expression levels of Bax, Bcl-2 levels, and Caspase-3 activity were measured by qRT-PCR technique, TUNEL staining was performed to detect DNA breaks and DAPI staining to visualize nuclear changes. Protein structures were retrieved from the protein data bank. OpenBabel and Autodock programs were used for the molecular docking study. Morin rescued the 3T3 embryonic fibroblast cells exposed to arsenic. However, Arsenic decreased the activities of antioxidant enzymes in cells and significantly increased oxidative stress, inflammation, and apoptosis. Morin treatment reduced oxidative damage and TNF-α and IL-1β levels. Arsenic-induced Caspase-3 mRNA expression level and Bax protein mRNA expression level were significantly increased, while Bcl-2 mRNA expression level was significantly decreased. While Caspase-3 mRNA expression level and Bax protein mRNA expression level decreased with morin treatment, Bcl-2 mRNA expression level increased significantly. Molecular docking study results showed good binding affinity of morin in SOD, GSH-Px, Bax, Bcl-2, Caspase-3, TNF-α, and IL-1β structures. Morin showed antioxidant, anti-inflammatory, and anti-apoptotic effects against Arsenic-induced cellular toxicity.
Collapse
Affiliation(s)
- Velid Unsal
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Mardin Artuklu University, Mardin, 47200, Türkiye
| | - Mustafa Cicek
- Department of Medical Biology, Faculty of Medicine, Kahramanmaras Sütcü Imam University, Kahramanmaras, 46050, Türkiye
| | - Necmettin Aktepe
- Department of Nursing, Faculty of Health Sciences Mardin Artuklu University, Mardin, 47200, Türkiye
| | - Erkan Oner
- Department of Biochemistry, Faculty of Pharmacy, Adıyaman University, Adıyaman, 02000, Türkiye
| |
Collapse
|
4
|
Tasci T, Orta-Yilmaz B, Aydin Y, Caliskan M. N-acetylcysteine attenuates sodium arsenite-induced oxidative stress and apoptosis in embryonic fibroblast cells. Toxicol Res (Camb) 2024; 13:tfae128. [PMID: 39139367 PMCID: PMC11319482 DOI: 10.1093/toxres/tfae128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 07/05/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
In recent years, the increase in environmental pollutants has been one of the most important factors threatening human and environmental health. Arsenic, a naturally occurring element found in soil, water, and air, easily enters the human body and leads to many metabolic disorders. In this study, we focused on the possible protective effects of N-acetylcysteine (NAC) against sodium arsenite (As)-induced toxic effects on embryonic fibroblast cells. The effects of As and NAC treatment on cells were evaluated, including cytotoxicity, oxidative stress, and apoptosis. Embryonic fibroblast cells were exposed to As (ranging from 0.01 μM to 10 μM) and NAC (at a concentration of 2 mM) for 24 h. The assessment of cytotoxicity markers, such as cell viability and lactate dehydrogenase (LDH), showed that As significantly reduced cell viability and increased LDH levels. Furthermore, we observed that As increased the amount of reactive oxygen species (ROS) in the cell, decreased the activity of antioxidant enzymes, and triggered apoptosis in cells. Additionally, our research revealed that the administration of NAC mitigates the detrimental effects of As. The results showed that As exerted hazardous effects on embryonic fibroblast cells through the induction of oxidative stress and apoptosis. In this context, our study provides evidence that NAC may have a protective effect against the toxicity of As in embryonic fibroblast cells.
Collapse
Affiliation(s)
- Tunahan Tasci
- Department of Biology, Institute of Graduate Studies in Sciences, Istanbul University, Istanbul 34126, Turkey
- Department of Medical Services and Techniques, Vocational School of Health Services, Istanbul Bilgi University, Istanbul 34387, Turkey
| | - Banu Orta-Yilmaz
- Department of Biology, Faculty of Sciences, Istanbul University, Istanbul 34126, Turkey
| | - Yasemin Aydin
- Department of Biology, Faculty of Sciences, Istanbul University, Istanbul 34126, Turkey
| | - Mahmut Caliskan
- Department of Biology, Faculty of Sciences, Istanbul University, Istanbul 34126, Turkey
| |
Collapse
|
5
|
Shariat Razavi F, Kouchak M, Sistani Karampour N, Mahdavinia M, Nazari Khorasgani Z, Rezaie A, Rahbar N. AS1411aptamer conjugated liposomes for targeted delivery of arsenic trioxide in mouse xenograft model of melanoma cancer. J Liposome Res 2024; 34:288-302. [PMID: 37843918 DOI: 10.1080/08982104.2023.2271046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
Development of AS1411aptamer-conjugated liposomes for targeted delivery of arsenic trioxide is the primary goal of this study. AS1411aptamer was used as ligand to target nucleolin, which is highly expressed on the surface of melanoma cancer cells. The targeted liposomes were constructed by the thin film method, and arsenic trioxide was loaded as cobalt (II) hydrogen arsenite (CHA) to increase the loading efficiency and stability of the liposomes. The liposomal structure was characterized by Fourier Transform Infrared Spectroscopy (FT-IR) and field emission scanning electron microscopy (FESEM). In addition, particle sizes and zeta potential of the CHA-loaded liposomes (CHAL) and aptamer-functionalized CHA-loaded liposomes (AP-CHAL) were determined. In vitro cytotoxicity of CHAL and AP-CHAL were evaluated using MTT assay in murine melanoma (B16) and mouse embryonic fibroblast (MEF) cell lines. The encapsulation efficiency of CHAL and AP-CHAL was reported as 60.2 ± 6.5% and 58.7 ± 4.2%, respectively. In vivo antitumor activity of CHAL and AP-CHAL in the B16 tumor-xenograft mouse model was dramatically observed. All mice of both groups survived until the end of treatment and showed body weight gain. The tumor protrusion completely disappeared in 50% of the mice in these groups. Furthermore, histopathology studies demonstrated that CHAL and AP-CHAL did not induce significant toxicity in healthy mice tissues. However, unlike the CHAL group, which showed an initial increase in tumor volume, a specific antitumor effect was observed in the AP-CHAL group from the beginning of treatment. The results showed that AP-CHAL can be used as an effective drug delivery system with high potential in the treatment of solid tumors.
Collapse
Affiliation(s)
- Fatemeh Shariat Razavi
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmaceutics, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Kouchak
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmaceutics, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Neda Sistani Karampour
- Marine Pharmaceutical Science Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmacology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoud Mahdavinia
- Toxicology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Nazari Khorasgani
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Annahita Rezaie
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Nadereh Rahbar
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
6
|
Cruz DRD, Zheng A, Debele T, Larson P, Dion GR, Park YC. Drug delivery systems for wound healing treatment of upper airway injury. Expert Opin Drug Deliv 2024; 21:573-591. [PMID: 38588553 PMCID: PMC11208077 DOI: 10.1080/17425247.2024.2340653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 04/04/2024] [Indexed: 04/10/2024]
Abstract
INTRODUCTION Endotracheal intubation is a common procedure to maintain an open airway with risks for traumatic injury. Pathological changes resulting from intubation can cause upper airway complications, including vocal fold scarring, laryngotracheal stenosis, and granulomas and present with symptoms such as dysphonia, dysphagia, and dyspnea. Current intubation-related laryngotracheal injury treatment approaches lack standardized guidelines, relying on individual clinician experience, and surgical and medical interventions have limitations and carry risks. AREAS COVERED The clinical and preclinical therapeutics for wound healing in the upper airway are described. This review discusses the current developments on local drug delivery systems in the upper airway utilizing particle-based delivery systems, including nanoparticles and microparticles, and bulk-based delivery systems, encompassing hydrogels and polymer-based approaches. EXPERT OPINION Complex laryngotracheal diseases pose challenges for effective treatment, struggling due to the intricate anatomy, limited access, and recurrence. Symptomatic management often requires invasive surgical procedures or medications that are unable to achieve lasting effects. Recent advances in nanotechnology and biocompatible materials provide potential solutions, enabling precise drug delivery, personalization, and extended treatment efficacy. Combining these technologies could lead to groundbreaking treatments for upper airways diseases, significantly improving patients' quality of life. Research and innovation in this field are crucial for further advancements.
Collapse
Affiliation(s)
- Denzel Ryan D. Cruz
- Medical Scientist Training Program, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Avery Zheng
- Chemical Engineering Program, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Tilahun Debele
- Chemical Engineering Program, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Peter Larson
- Department of Otolaryngology – Head and Neck Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Gregory R. Dion
- Department of Otolaryngology – Head and Neck Surgery, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Yoonjee C. Park
- Chemical Engineering Program, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
7
|
Park WH. Propyl gallate induces cell death in human pulmonary fibroblast through increasing reactive oxygen species levels and depleting glutathione. Sci Rep 2024; 14:5375. [PMID: 38438412 PMCID: PMC10912098 DOI: 10.1038/s41598-024-52849-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 01/24/2024] [Indexed: 03/06/2024] Open
Abstract
Propyl gallate (PG) exhibits an anti-growth effect on various cell types. The present study investigated the impact of PG on the levels of reactive oxygen species (ROS) and glutathione (GSH) in primary human pulmonary fibroblast (HPF) cells. Moreover, the effects of N-acetyl cysteine (NAC, an antioxidant), L-buthionine sulfoximine (BSO, a GSH synthesis inhibitor), and small interfering RNA (siRNAs) against various antioxidant genes on ROS and GSH levels and cell death were examined in PG-treated HPF cells. PG (100-800 μM) increased the levels of total ROS and O2·- at early time points of 30-180 min and 24 h, whereas PG (800-1600 μM) increased GSH-depleted cell number at 24 h and reduced GSH levels at 30-180 min. PG downregulated the activity of superoxide dismutase (SOD) and upregulated the activity of catalase in HPF cells. Treatment with 800 μM PG increased the number of apoptotic cells and cells that lost mitochondrial membrane potential (MMP; ΔΨm). NAC treatment attenuated HPF cell death and MMP (ΔΨm) loss induced by PG, accompanied by a decrease in GSH depletion, whereas BSO exacerbated the cell death and MMP (ΔΨm) loss without altering ROS and GSH depletion levels. Furthermore, siRNA against SOD1, SOD2, or catalase attenuated cell death in PG-treated HPF cells, whereas siRNA against GSH peroxidase enhanced cell death. In conclusion, PG induced cell death in HPF cells by increasing ROS levels and depleting GSH. NAC was found to decrease HPF cell death induced by PG, while BSO enhanced cell death. The findings shed light on how manipulating the antioxidant system influence the cytotoxic effects of PG in HPF cells.
Collapse
Affiliation(s)
- Woo Hyun Park
- Department of Physiology, Medical School, Jeonbuk National University, 20 Geonji-Ro, Deokjin, Jeonju, Jeollabuk, 54907, Republic of Korea.
| |
Collapse
|
8
|
Yan M, Wang H, Wei R, Li W. Arsenic trioxide: applications, mechanisms of action, toxicity and rescue strategies to date. Arch Pharm Res 2024; 47:249-271. [PMID: 38147202 DOI: 10.1007/s12272-023-01481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 12/15/2023] [Indexed: 12/27/2023]
Abstract
Arsenical medicine has obtained its status in traditional Chinese medicine for more than 2,000 years. In the 1970s, arsenic trioxide was identified to have high efficacy and potency for the treatment of acute promyelocytic leukemia, which promoted many studies on the therapeutic effects of arsenic trioxide. Currently, arsenic trioxide is widely used to treat acute promyelocytic leukemia and various solid tumors through various mechanisms of action in clinical practice; however, it is accompanied by a series of adverse reactions, especially cardiac toxicity. This review presents a comprehensive overview of arsenic trioxide from preclinical and clinical efficacy, potential mechanisms of action, toxicities, and rescue strategies for toxicities to provide guidance or assistance for the clinical application of arsenic trioxide.
Collapse
Affiliation(s)
- Meng Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| | - Hao Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Rui Wei
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Pharmacy Department, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenwen Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
9
|
Ariafar S, Makhdoomi S, Mohammadi M. Arsenic and Tau Phosphorylation: a Mechanistic Review. Biol Trace Elem Res 2023; 201:5708-5720. [PMID: 37211576 DOI: 10.1007/s12011-023-03634-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/14/2023] [Indexed: 05/23/2023]
Abstract
Arsenic poisoning can affect the peripheral nervous system and cause peripheral neuropathy. Despite different studies on the mechanism of intoxication, the complete process is not explained yet, which can prevent further intoxication and produce effective treatment. In the following paper, we would like to consider the idea that arsenic might cause some diseases via inflammation induction, and tauopathy in neurons. Tau protein, one of the microtubule-associated proteins expressed in neurons, contributes to neuronal microtubules structure. Arsenic may be involved in cellular cascades involved in modulating tau function or hyperphosphorylation of tau protein, which ultimately leads to nerve destruction. For proof of this assumption, some investigations have been planned to measure the association between arsenic and quantities of phosphorylation of tau protein. Additionally, some researchers have investigated the association between microtubule trafficking in neurons and the levels of tau protein phosphorylation. It should be noticed that changing tau phosphorylation in arsenic toxicity may add a new feature to understanding the mechanism of poisonousness and aid in discovering novel therapeutic candidates such as tau phosphorylation inhibitors for drug development.
Collapse
Affiliation(s)
- Saba Ariafar
- Department of Pharmacology & Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sajjad Makhdoomi
- Department of Pharmacology & Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mojdeh Mohammadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
10
|
Liu X, Zhong S, Qiu K, Chen X, Wu W, Zheng J, Liu Y, Wu H, Fan S, Nie D, Wang X, Yu Z, Liao Z, Zhong M, Li Y, Zeng C. Targeting NRF2 uncovered an intrinsic susceptibility of acute myeloid leukemia cells to ferroptosis. Exp Hematol Oncol 2023; 12:47. [PMID: 37198609 DOI: 10.1186/s40164-023-00411-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 05/05/2023] [Indexed: 05/19/2023] Open
Abstract
Drug resistance and poor treatment response are major obstacles to the effective treatment of acute myeloid leukemia (AML). A deeper understanding of the mechanisms regulating drug resistance and response genes in AML is therefore urgently needed. Our previous research has highlighted the important role of nuclear factor E2-related factor 2 (NRF2) in AML, where it plays a critical role in detoxifying reactive oxygen species and influencing sensitivity to chemotherapy. In this study, we identify a core set of direct NRF2 targets that are involved in ferroptosis, a novel form of cell death. Of particular interest, we find that glutathione peroxidase 4 (GPX4) is a key ferroptosis gene that is consistently upregulated in AML, and high expression of GPX4 is associated with poor prognosis for AML patients. Importantly, simultaneous inhibition of NRF2 with ML385 and GPX4 with FIN56 or RSL3 synergistically targets AML cells, triggering ferroptosis. Treatment with ML385 + FIN56/RSL3 resulted in a marked reduction in NRF2 and GPX4 expression. Furthermore, NRF2 knockdown enhanced the sensitivity of AML cells to the ferroptosis inducers. Taken together, our results suggest that combination therapy targeting both NRF2 and GPX4 may represent a promising approach for the treatment of AML.
Collapse
Affiliation(s)
- Xin Liu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, P.R. China
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Shuxin Zhong
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, P.R. China
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Kangjie Qiu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, P.R. China
| | - Xi Chen
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, P.R. China
| | - Weiyue Wu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, P.R. China
| | - Jiamian Zheng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, P.R. China
| | - Yanwen Liu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, P.R. China
| | - Haolong Wu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, P.R. China
| | - Shiyun Fan
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, P.R. China
| | - Dingrui Nie
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, P.R. China
| | - Xianfeng Wang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, P.R. China
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, P.R. China
| | - Zhi Yu
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, P.R. China
| | - Ziwei Liao
- Department of Hematology, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, P.R. China
| | - Mengjun Zhong
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, P.R. China.
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, 510632, P.R. China.
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, P.R. China.
| | - Chengwu Zeng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of medicine, Jinan University, No.601, West Huangpu Avenue, Guangzhou, 510632, P.R. China.
| |
Collapse
|
11
|
Dadashi M, Pourbagheri-Sigaroodi A, Anjam-Najmedini A, Bashash D. Synergistic Effects of PI3K Inhibition on Arsenic Trioxide Cytotoxicity in Acute Promyelocytic Leukemia Cells: A New Portrait of Idelalisib as an Adjuvant Therapy. Indian J Hematol Blood Transfus 2023; 39:208-219. [PMID: 37006983 PMCID: PMC10064366 DOI: 10.1007/s12288-022-01573-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/25/2022] [Indexed: 11/06/2022] Open
Abstract
The advent of small-molecule inhibitors targeting the components of oncogenic signaling pathways has revolutionized cancer treatment, where the pharmacological approaches have gone from an era of non-specific chemotherapeutic drugs to the golden age of targeted therapies. In the present study, we evaluated the therapeutic value of an isoform-specific inhibitor of PI3K (Idelalisib) in potentiating the anti-leukemic effects of arsenic trioxide (ATO), an eminent drug used in the treatment of acute promyelocytic leukemia (APL). We found that the abrogation of the PI3K axis profoundly reinforced the anti-leukemic effects of the lower concentrations of ATO, as revealed by the superior reduction in the viability, cell number, and metabolic activity of APL-derived NB4 cells as compared to either agent alone. The cytotoxic effect of Idelalisib in combination with ATO was probably mediated through suppression of c-Myc that was coupled with the elevation in the intracellular level of reactive oxygen species and induction of caspase-3-dependent apoptosis. Notably, our results showed that the suppression of autophagy reinforced the ability of the drugs in eradicating the leukemic cells, suggesting that the compensatory activation of this system may probably overshadow the success of Idelalisib-plus-ATO in APL cells. All in all and given the significant efficacy of Idelalisib against NB4 cells, we proposed the application of this PI3K inhibitor as a foreseeable approach with a safe profile in the treatment of APL.
Collapse
Affiliation(s)
- Maryam Dadashi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Anjam-Najmedini
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
ArulJothi KN, Kumaran K, Senthil S, Nidhu AB, Munaff N, Janitri VB, Kirubakaran R, Singh SK, Gupt G, Dua K, Krishnan A. Implications of reactive oxygen species in lung cancer and exploiting it for therapeutic interventions. Med Oncol 2023; 40:43. [PMID: 36472716 PMCID: PMC9734980 DOI: 10.1007/s12032-022-01900-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022]
Abstract
Lung cancer is the second (11.4%) most commonly diagnosed cancer and the first (18%) to cause cancer-related deaths worldwide. The incidence of lung cancer varies significantly among men, women, and high and low-middle-income countries. Air pollution, inhalable agents, and tobacco smoking are a few of the critical factors that determine lung cancer incidence and mortality worldwide. Reactive oxygen species are known factors of lung carcinogenesis resulting from the xenobiotics and their mechanistic paths are under critical investigation. Reactive oxygen species exhibit dual roles in cells, as a tumorigenic and anti-proliferative factor, depending on spatiotemporal context. During the precancerous state, ROS promotes cancer origination through oxidative stress and base-pair substitution mutations in pro-oncogenes and tumor suppressor genes. At later stages of tumor progression, they help the cancer cells in invasion, and metastases by activating the NF-kB and MAPK pathways. However, at advanced stages, when ROS exceeds the threshold, it promotes cell cycle arrest and induces apoptosis in cancer cells. ROS activates extrinsic apoptosis through death receptors and intrinsic apoptosis through mitochondrial pathways. Moreover, ROS upregulates the expression of beclin-1 which is a critical component to initiate autophagy, another form of programmed cell death. ROS is additionally involved in an intermediatory step in necroptosis, which catalyzes and accelerates this form of cell death. Various therapeutic interventions have been attempted to exploit this cytotoxic potential of ROS to treat different cancers. Growing body of evidence suggests that ROS is also associated with chemoresistance and cancer cell immunity. Considering the multiple roles of ROS, this review highlights the exploitation of ROS for various therapeutic interventions. However, there are still gaps in the literature on the dual roles of ROS and the involvement of ROS in cancer cell immunity and therapy resistance.
Collapse
Affiliation(s)
- K. N. ArulJothi
- grid.412742.60000 0004 0635 5080Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Chennai, 603203 India
| | - K. Kumaran
- grid.412742.60000 0004 0635 5080Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Chennai, 603203 India
| | - Sowmya Senthil
- grid.412742.60000 0004 0635 5080Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Chennai, 603203 India
| | - A. B. Nidhu
- grid.412742.60000 0004 0635 5080Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Chennai, 603203 India
| | - Nashita Munaff
- grid.412742.60000 0004 0635 5080Department of Biotechnology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Chennai, 603203 India
| | - V. B. Janitri
- grid.262613.20000 0001 2323 3518Rochester Institute of Technology, Rochester, NY USA
| | - Rangasamy Kirubakaran
- grid.444708.b0000 0004 1799 6895Department of Biotechnology, Vinayaka Mission’s Kirupananda Variyar Engineering College, Vinayaka Missions Research Foundation, Salem, Tamil Nadu India
| | - Sachin Kumar Singh
- grid.449005.cSchool of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab India ,grid.117476.20000 0004 1936 7611Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007 Australia
| | - Gaurav Gupt
- grid.448952.60000 0004 1767 7579School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, 302017 India ,grid.412431.10000 0004 0444 045XDepartment of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India ,grid.449906.60000 0004 4659 5193Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Kamal Dua
- grid.117476.20000 0004 1936 7611Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007 Australia ,grid.117476.20000 0004 1936 7611Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007 Australia
| | - Anand Krishnan
- grid.412219.d0000 0001 2284 638XDepartment of Chemical Pathology, School of Pathology, Faculty of Health Sciences, University of the Free State, Bloemfontein, 9300 South Africa
| |
Collapse
|
13
|
Burska AN, Ilyassova B, Dildabek A, Khamijan M, Begimbetova D, Molnár F, Sarbassov DD. Enhancing an Oxidative "Trojan Horse" Action of Vitamin C with Arsenic Trioxide for Effective Suppression of KRAS-Mutant Cancers: A Promising Path at the Bedside. Cells 2022; 11:3454. [PMID: 36359850 PMCID: PMC9657932 DOI: 10.3390/cells11213454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
The turn-on mutations of the KRAS gene, coding a small GTPase coupling growth factor signaling, are contributing to nearly 25% of all human cancers, leading to highly malignant tumors with poor outcomes. Targeting of oncogenic KRAS remains a most challenging task in oncology. Recently, the specific G12C mutant KRAS inhibitors have been developed but with a limited clinical outcome because they acquire drug resistance. Alternatively, exploiting a metabolic breach of KRAS-mutant cancer cells related to a glucose-dependent sensitivity to oxidative stress is becoming a promising indirect cancer targeting approach. Here, we discuss the use of a vitamin C (VC) acting in high dose as an oxidative "Trojan horse" agent for KRAS-mutant cancer cells that can be potentiated with another oxidizing drug arsenic trioxide (ATO) to obtain a potent and selective cytotoxic impact. Moreover, we outline the advantages of VC's non-natural enantiomer, D-VC, because of its distinctive pharmacokinetics and lower toxicity. Thus, the D-VC and ATO combination shows a promising path to treat KRAS-mutant cancers in clinical settings.
Collapse
Affiliation(s)
- Agata N. Burska
- Department of Biology, Nazarbayev University, Astana 010000, Kazakhstan
| | | | - Aruzhan Dildabek
- Department of Biology, Nazarbayev University, Astana 010000, Kazakhstan
| | - Medina Khamijan
- Department of Biology, Nazarbayev University, Astana 010000, Kazakhstan
| | - Dinara Begimbetova
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
| | - Ferdinand Molnár
- Department of Biology, Nazarbayev University, Astana 010000, Kazakhstan
| | - Dos D. Sarbassov
- Department of Biology, Nazarbayev University, Astana 010000, Kazakhstan
- National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan
| |
Collapse
|
14
|
Li C, Wu X, Zheng C, Xu S, Liu Y, Qin J, Fan X, Ye Y, Fei W. Nanotechnology-integrated ferroptosis inducers: a sharp sword against tumor drug resistance. J Mater Chem B 2022; 10:7671-7693. [PMID: 36043505 DOI: 10.1039/d2tb01350a] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Presently, the biggest hurdle to cancer therapy is the inevitable emergence of drug resistance. Since conventional therapeutic schedules fall short of the expectations in curbing drug resistance, the development of novel drug resistance management strategies is critical. Extensive research over the last decade has revealed that the process of ferroptosis is correlated with cancer resistance; moreover, it has been demonstrated that ferroptosis inducers reverse drug resistance. To elucidate the development and promote the clinical transformation of ferroptosis strategies in cancer therapy, we first analyzed the roles of key ferroptosis-regulating molecules in the progression of drug resistance in-depth and then reviewed the design of ferroptosis-inducing strategies based on nanotechnology for overcoming drug resistance, including glutathione depletion, reactive oxygen species generation, iron donation, lipid peroxidation aggregation, and multiple-drug resistance-associated tumor cell destruction. Finally, the prospects and challenges of regulating ferroptosis as a therapeutic strategy for reversing cancer therapy resistance were evaluated. This review aimed to provide a comprehensive understanding for researchers to develop ferroptosis-inducing nanoplatforms that can overcome drug resistance.
Collapse
Affiliation(s)
- Chaoqun Li
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Xiaodong Wu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Shanshan Xu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yunxi Liu
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Jiale Qin
- Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiaoyu Fan
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
| | - Yiqing Ye
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| |
Collapse
|
15
|
Deng H, Li Y, Li J, Shen W, Chen Q, Weng S, He J, Xu X. Neomycin inhibits Megalocytivirus infection in fish by antagonizing the increase of intracellular reduced glutathione. FISH & SHELLFISH IMMUNOLOGY 2022; 127:148-154. [PMID: 35714896 DOI: 10.1016/j.fsi.2022.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/04/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
Infectious spleen and kidney necrosis virus (ISKNV) is the type species of the Megalocytivirus genus that infects a number of marine and freshwater fishes, causing huge economic losses in aquaculture. The ISKNV infection leads to increase of reducing power in cells. As the antibiotic neomycin can promote the production of reactive oxygen species (ROS) in animal cells, in the current study, the potential therapeutic effect of neomycin on ISKNV infection was explored. We showed that neomycin could decrease the reducing power in cultured MFF-1 cells and inhibit ISKNV infection by antagonizing the shift of the cellular redox balance toward reduction. In vivo experiments further demonstrated that neomycin treatment significantly suppresses ISKNV infection in mandarin fish. Expression of the major capsid protein (MCP) and the proportion of infected cells in tissues were down-regulated after neomycin treatment. Furthermore, neomycin showed complex effects on expression of a set of antiviral related genes of the host. Taking together, the current study suggested that the viral-induced redox imbalance in the infected cells could be used as a target for suppressing ISKNV infection. Neomycin can be potentially utilized for therapeutic treatment of Megalocytivirus diseases by antagonizing intracellular redox changes.
Collapse
Affiliation(s)
- Hengwei Deng
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China; State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, China
| | - Yeyu Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Jinling Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Wenjie Shen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Qiankang Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China
| | - Shaoping Weng
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Jianguo He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Xiaopeng Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Provice Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China.
| |
Collapse
|
16
|
Gandhi D, Bhandari S, Mishra S, Tiwari RR, Rajasekaran S. Non-malignant respiratory illness associated with exposure to arsenic compounds in the environment. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 94:103922. [PMID: 35779705 DOI: 10.1016/j.etap.2022.103922] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 06/22/2022] [Accepted: 06/25/2022] [Indexed: 06/15/2023]
Abstract
Arsenic (As), a toxic metalloid, primarily originates from both natural and anthropogenic activities. Reports suggested that millions of people globally exposed to high levels of naturally occurring As compounds via inhalation and ingestion. There is evidence that As is a well-known lung carcinogen. However, there has been relatively little evidence suggesting its non-malignant lung effects. This review comprehensively summarises current experimental and clinical studies implicating the association of As exposure and the development of several non-malignant lung diseases. Experimental studies provided evidence that As exposure induces redox imbalance, apoptosis, inflammatory response, epithelial-to-mesenchymal transition (EMT), and affected normal lung development through alteration of the components of intracellular signaling cascades. In addition, we also discuss the sources and possible mechanisms of As influx and efflux in the lung. Finally, current experimental studies on treatment strategies using phytochemicals and our perspective on future research with As are also discussed.
Collapse
Affiliation(s)
- Deepa Gandhi
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India
| | - Sneha Bhandari
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India
| | - Sehal Mishra
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India
| | - Rajnarayan R Tiwari
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India
| | - Subbiah Rajasekaran
- Department of Biochemistry, ICMR-National Institute for Research in Environmental, Health, Bhopal, Madhya Pradesh, India.
| |
Collapse
|
17
|
Feng C, Wu Y, Chen Y, Xiong X, Li P, Peng X, Li C, Weng W, Zhu Y, Zhou D, Li Y. Arsenic trioxide increases apoptosis of SK-N-BE (2) cells partially by inducing GPX4-mediated ferroptosis. Mol Biol Rep 2022; 49:6573-6580. [PMID: 35598199 DOI: 10.1007/s11033-022-07497-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/21/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND Neuroblastoma (NB) is the most common extracranial tumor in central nervous system threatening children's health with limited therapeutic options. Arsenic trioxide (ATO) has been identified the cytotoxicity in NB cells but the potential mechanism remains unclear. In this study, we attempted to obtain some insight into the mechanisms of cell death induced by ATO in NB cells. METHODS AND RESULTS Proteomic analyses found that ATO can affect the signaling pathway associated with ferroptosis, including the upregulation of iron absorption (FTL, FTH1, HO-1), ferritinophagy (LC3, P62, ATG7, NCOA4) and modifier of glutathione synthesis (GCLM); downregulation of glutamine synthetase (GS) and GPX4, which was the critical inhibitor of ferroptosis. Western blot analysis revealing GPX4 expression in SK-N-BE (2) cells decreased after treatment with ATO (7.3 µM), resulting in a loss of GPX4 activity. Furthermore, Ferroptosis inhibitor ferrostatin-1 partially blocked ATO-induced cell death. CONCLUSIONS Our study revealed that ATO may induce ferroptosis in neuroblastoma cell SK-N-BE (2) by facilitating the downregulation of GPX4, ultimately resulting in iron-dependent oxidative death.
Collapse
Affiliation(s)
- Chuchu Feng
- Department of Pediatric Hematology/Oncology, Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Yan Jiang Xi Road, No. 107, Guangzhou, 510120, China
| | - Yu Wu
- Department of Pediatric Hematology/Oncology, Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Yan Jiang Xi Road, No. 107, Guangzhou, 510120, China
| | - Yantao Chen
- Department of Orthopaedics, Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Yan Jiang Xi Road, No. 107, Guangzhou, 510120, China
| | - Xilin Xiong
- Department of Pediatric Hematology/Oncology, Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Yan Jiang Xi Road, No. 107, Guangzhou, 510120, China
| | - Peng Li
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Kaiyuan Avenue, No. 190, Guangzhou, 510530, China
| | - Xiaomin Peng
- Department of Pediatric Hematology/Oncology, Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Yan Jiang Xi Road, No. 107, Guangzhou, 510120, China
| | - Chunmou Li
- Department of Pediatric Hematology/Oncology, Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Yan Jiang Xi Road, No. 107, Guangzhou, 510120, China
| | - Wenjun Weng
- Department of Pediatric Hematology/Oncology, Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Yan Jiang Xi Road, No. 107, Guangzhou, 510120, China
| | - Yafeng Zhu
- Medical Researcher Center, Sun Yat-Sen University Memorial Hospital, Sun Yet-Sen University, Yan Jiang Xi Road, No. 107, Guangzhou, 510120, China
| | - Dunhua Zhou
- Department of Pediatric Hematology/Oncology, Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Yan Jiang Xi Road, No. 107, Guangzhou, 510120, China.
| | - Yang Li
- Department of Pediatric Hematology/Oncology, Sun Yet-Sen Memorial Hospital, Sun Yet-Sen University, Yan Jiang Xi Road, No. 107, Guangzhou, 510120, China.
| |
Collapse
|
18
|
Ferroptosis as a mechanism of non-ferrous metal toxicity. Arch Toxicol 2022; 96:2391-2417. [PMID: 35727353 DOI: 10.1007/s00204-022-03317-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/11/2022] [Indexed: 11/02/2022]
Abstract
Ferroptosis is a recently discovered form of regulated cell death, implicated in multiple pathologies. Given that the toxicity elicited by some metals is linked to alterations in iron metabolism and induction of oxidative stress and lipid peroxidation, ferroptosis might be involved in such toxicity. Although direct evidence is insufficient, certain pioneering studies have demonstrated a crosstalk between metal toxicity and ferroptosis. Specifically, the mechanisms underlying metal-induced ferroptosis include induction of ferritinophagy, increased DMT-1 and TfR cellular iron uptake, mitochondrial dysfunction and mitochondrial reactive oxygen species (mitoROS) generation, inhibition of Xc-system and glutathione peroxidase 4 (GPX4) activity, altogether resulting in oxidative stress and lipid peroxidation. In addition, there is direct evidence of the role of ferroptosis in the toxicity of arsenic, cadmium, zinc, manganese, copper, and aluminum exposure. In contrast, findings on the impact of cobalt and nickel on ferroptosis are scant and nearly lacking altogether for mercury and especially lead. Other gaps in the field include limited studies on the role of metal speciation in ferroptosis and the critical cellular targets. Although further detailed studies are required, it seems reasonable to propose even at this early stage that ferroptosis may play a significant role in metal toxicity, and its modulation may be considered as a potential therapeutic tool for the amelioration of metal toxicity.
Collapse
|
19
|
Wei Z, Sun X, He Q, Zhao Y, Wu Y, Han X, Wu Z, Chu X, Guan S. Nephroprotective effect of magnesium isoglycyrrhizinate against arsenic trioxide‑induced acute kidney damage in mice. Exp Ther Med 2022; 23:276. [PMID: 35317438 PMCID: PMC8908469 DOI: 10.3892/etm.2022.11202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/17/2022] [Indexed: 11/15/2022] Open
Abstract
Magnesium isoglycyrrhizinate (MgIG) has anti-inflammatory, antioxidative, antiviral and anti-hepatotoxic effects. However, protective effects of MgIG against renal damage caused by arsenic trioxide (ATO) have not been reported. The present study aimed to clarify the protective function of MgIG on kidney damaged induced by ATO. Other than the control group and the group treated with MgIG alone, mice were injected intraperitoneally with ATO (5 mg/kg/day) for 7 days to establish a mouse model of kidney damage. On the 8th day, blood and kidney tissue were collected and the inflammatory factors and antioxidants levels in the kidney tissue and serum were measured. The expression of protein levels of caspase-3, Bcl-2, Bax, Toll-like receptor-4 (TLR4) and nuclear factor-κB (NF-κB) were determined via western blot analysis. In the renal tissue of mice, ATO exposure dramatically elevated markers of oxidative stress, apoptosis and inflammation. However, MgIG could also restore the activities of urea nitrogen and creatinine to normal levels, decrease the malondialdehyde level and reactive oxygen species formation and increase superoxide dismutase, catalase and glutathione activities. MgIG also ameliorated the morphological abnormalities generated by ATO, reduced inflammation and apoptosis and inhibited the TLR4/NF-κB signaling pathway. In conclusion, MgIG may mitigate ATO-induced kidney damage by decreasing apoptosis, oxidative stress and inflammation and its mechanism may be connected to the inhibition of TLR4/NF-κB signaling.
Collapse
Affiliation(s)
- Ziheng Wei
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xiaoqi Sun
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Qianqian He
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Yang Zhao
- Department of Academic Research, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yongchao Wu
- Department of Radiological Intervention, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Xue Han
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Zhonglin Wu
- Department of Radiological Intervention, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Xi Chu
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Shengjiang Guan
- School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| |
Collapse
|
20
|
Xie Q, Shen Y, Liang J, Zhang C, Ling X, Gu L, Wang Y, Wang Y, Liu X, Hu C. Design and synthesis of 1,3-diphenylpyrimidine-2,4(1 H,3 H)-dione derivatives as antitumor agents via elevating ROS production to induce apoptosis. NEW J CHEM 2022. [DOI: 10.1039/d2nj01922a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The target compound XS23 have exhibited excellent antitumor cell proliferative activity against multiple tumor cell lines and can induce cancer cell apoptosis by elevating ROS production.
Collapse
Affiliation(s)
- Qian Xie
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 110016, China
| | - Yanni Shen
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 110016, China
| | - Jianhui Liang
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 110016, China
| | - Chao Zhang
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 110016, China
| | - Xianwu Ling
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 110016, China
| | - Liangxiao Gu
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 110016, China
| | - Yiling Wang
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 110016, China
| | - Yan Wang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaoping Liu
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 110016, China
| | - Chun Hu
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 110016, China
| |
Collapse
|
21
|
Liu T, Sun L, Zhang Y, Wang Y, Zheng J. Imbalanced GSH/ROS and sequential cell death. J Biochem Mol Toxicol 2021; 36:e22942. [PMID: 34725879 DOI: 10.1002/jbt.22942] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/31/2021] [Accepted: 10/18/2021] [Indexed: 12/17/2022]
Abstract
Reactive oxygen species (ROS) are produced in cells during metabolic processes. Excessive intracellular ROS may react with large biomolecules, such as DNA, RNA, proteins, and small biomolecules, that is, glutathione (GSH) and unsaturated fatty acids. GSH has physiological functions, including free radical scavenging, anti-oxidation, and electrophile elimination. The disruption of ROS/GSH balance results in the deleterious oxidation and chemical modification of biomacromolecules, which eventually leads to cell-cycle arrest and proliferation inhibition, and even induces cell death. Imbalanced ROS/GSH may result from a direct increase of ROS, consumption of GSH, intracellular oxidoreductase interference, or thioredoxin activity reduction. Some chemicals including arsenic trioxide (ATO), pyrogallol (PG), and carbobenzoxy-Leu-Leu-leucinal (MG132) could also disrupt the balance of GSH and ROS. This article reviews the occurrence and consequences of the imbalance between GSH and ROS and introduces factors responsible for the disruption of cellular ROS and GSH balance, resulting in cell death. "GSH" and "ROS" were used as keywords to search the relevant literaturess.
Collapse
Affiliation(s)
- Ting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China
| | - Li Sun
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China.,School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Yubin Zhang
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China.,School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Yonglin Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China
| | - Jiang Zheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China
| |
Collapse
|
22
|
Su Y, Li L, Farooq MU, Huang X, Zheng T, Zhang YJ, Ei HH, Panhwar FH, Tang Z, Zeng R, Liang Y, Ye X, Jia X, Zhu J. Rescue effects of Se-enriched rice on physiological and biochemical characteristics in cadmium poisoning mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:20023-20033. [PMID: 33409993 DOI: 10.1007/s11356-020-11854-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 11/26/2020] [Indexed: 06/12/2023]
Abstract
Cadmium (Cd) is an element injurious for human health and is possibly toxic to organisms at minor concentrations. While some of other trace metallic elements have antagonistic features to it. One of them is the interaction between selenium (Se) and Cd in plant different organs. Literature review disclosed that the intake of Se to some extent can reduce the accumulation of Cd in plants, while the research on of trace metallic elements (Cd) and Se-enriched food (rice) in the living body has rarely been reported. This study intended to explore whether there was a mitigating effect of Se-enriched rice on mice poisoned with Cd. A mouse model of low-dose and high-dose Cd poisoning was established (supplemented with cadmium chloride(CdCl2·2½H20)), followed by feeding two groups (1) Se-enriched rice and (2) setting an equal amount of inorganic Se group. After that, the impact of Se-enriched rice on the antioxidant activity was evaluated. The Se-enriched diet enhanced the total antioxidant capacity (T-AOC), superoxide dismutase (SOD), and enzyme activities of GSH peroxidase (GSH-Px) in mice livers and kidney whereas significantly decreased the malondialdehyde (MDA) contents. Moreover, the degree of physiological damage in mice with low cadmium poisoning was significantly alleviated, and the expression of antioxidant genes (Nrf-2, GPX1, TrxR2, TNF-2) was increased. In conclusion, the Se-enriched diet has a positive effect on the biological effects in mice, and it can be used as a daily diet to resist damage to the body's low Cd state and support enzymatic antioxidant systems by eliminating oxidative injury.
Collapse
Affiliation(s)
- Yang Su
- Rice Research Institute, Sichuan Agricultural University, 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Ling Li
- Rice Research Institute, Sichuan Agricultural University, 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Muhammad Umer Farooq
- Rice Research Institute, Sichuan Agricultural University, 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
- Department of Botany, Faculty of Life Sciences, University of Okara, Okara, 56130, Pakistan
| | - Xin Huang
- Rice Research Institute, Sichuan Agricultural University, 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Tengda Zheng
- Rice Research Institute, Sichuan Agricultural University, 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Yu Jie Zhang
- Rice Research Institute, Sichuan Agricultural University, 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Hla Hla Ei
- Rice Research Institute, Sichuan Agricultural University, 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Faiz Hussain Panhwar
- Rice Research Institute, Sichuan Agricultural University, 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Zhichen Tang
- Rice Research Institute, Sichuan Agricultural University, 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Rui Zeng
- Rice Research Institute, Sichuan Agricultural University, 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
- College of Chemistry and Life Science, Chengdu Normal University, Chengdu, 611130, China
| | - Yuanke Liang
- Rice Research Institute, Sichuan Agricultural University, 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Xiaoying Ye
- Rice Research Institute, Sichuan Agricultural University, 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Xiaomei Jia
- Rice Research Institute, Sichuan Agricultural University, 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Jianqing Zhu
- Rice Research Institute, Sichuan Agricultural University, 211, Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
23
|
Li Y, Li M, Wang X, Wang Y, Li C, Zhao Y, Li Z, Chen J, Li J, Ren K, Duan X, Ren J, Han X, Li Q. Arsenic trioxide-eluting electrospun nanofiber-covered self-expandable metallic stent reduces granulation tissue hyperplasia in rabbit trachea. ACTA ACUST UNITED AC 2020; 16:015013. [PMID: 33325379 DOI: 10.1088/1748-605x/abb25a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Stent-related granulation tissue hyperplasia is a major complication that limits the application of stents in airways. In this study, an arsenic trioxide-eluting electrospun nanofiber-covered self-expandable metallic stent (ATO-NFCS) was developed. Poly-L-lactide-caprolactone (PLCL) was selected as the drug-carrying polymer. Stents with two different ATO contents (0.4% ATO/PLCL and 1.2% ATO/PLCL) were fabricated. The in vitro release in simulated airway fluid suggested that the total ATO release time was 1 d. The growth of human embryonic pulmonary fibroblasts (CCC-HPF-1), normal human bronchial epithelial cells and airway smooth muscle cells was inhibited by ATO. When embedded in paravertebral muscle, the nanofiber membrane showed good short-term and long-term biological effects. In an animal study, placement of the ATO-NFCS in the trachea through a delivery system under fluoroscopy was feasible. The changes in liver and kidney function 1 and 7 d after ATO-NFCS placement were within the normal range. On pathological examination, the heart, liver, spleen, lungs and kidneys were normal. The effectiveness of the ATO-NFCS in reducing granulation tissue hyperplasia and collagen deposition was demonstrated in the rabbit airway (n = 18) at 4 weeks. The present study preliminarily investigated the efficacy of the ATO-NFCS in reducing granulation tissue formation in the trachea of rabbits. The results suggest that the ATO-NFCS is safe in vivo, easy to place, and effective for the suppression of granulation tissue formation.
Collapse
Affiliation(s)
- Yahua Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China. Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, People's Republic of China. These authors contributed equally to this work
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Park HK, Han BR, Park WH. Combination of Arsenic Trioxide and Valproic Acid Efficiently Inhibits Growth of Lung Cancer Cells via G2/M-Phase Arrest and Apoptotic Cell Death. Int J Mol Sci 2020; 21:ijms21072649. [PMID: 32290325 PMCID: PMC7177455 DOI: 10.3390/ijms21072649] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/03/2020] [Accepted: 04/09/2020] [Indexed: 12/15/2022] Open
Abstract
Arsenic trioxide (ATO; As2O3) has anti-cancer effects in various solid tumors as well as hematological malignancy. Valproic acid (VPA), which is known to be a histone deacetylase inhibitor, has also anti-cancer properties in several cancer cells including lung cancer cells. Combined treatment of ATO and VPA (ATO/VPA) could synergistically enhance anti-cancer effects and reduce ATO toxicity ATO. In this study, the combined anti-cancer effects of ATO and VPA (ATO/VPA) was investigated in NCI-H460 and NCI-H1299 lung cancer cells in vitro and in vivo. A combination of 3 μM ATO and 3 mM VPA (ATO/VPA) strongly inhibited the growths of both lung cancer cell types. DNA flow cytometry indicated that ATO/VPA significantly induced G2/M-phase arrest in both cell lines. In addition, ATO/VPA strongly increased the percentages of sub-G1 cells and annexin V-FITC positive cells in both cells. However, lactate dehydrogenase (LDH) release from cells was not increased in ATO/VPA-treated cells. In addition, ATO/VPA increased apoptosis in both cell types, accompanied by loss of mitochondrial membrane potential (MMP, ∆Ψm), activation of caspases, and cleavage of anti-poly ADP ribose polymerase-1. Moreover, a pan-caspase inhibitor, Z-VAD, significantly reduced apoptotic cell death induced by ATO/VPA. In the xenograft model, ATO/VPA synergistically inhibited growth of NCI-H460-derived xenograft tumors. In conclusion, the combination of ATO/VPA effectively inhibited the growth of lung cancer cells through G2/M-phase arrest and apoptotic cell death, and had a synergistic antitumor effect in vivo.
Collapse
Affiliation(s)
| | | | - Woo Hyun Park
- Correspondence: ; Tel.: +82-63-270-3079; Fax: +82-63-274-9892
| |
Collapse
|
25
|
Wu Y, Du D, Chen J, Liu C. Preparation of PLGA microspheres loaded with 10-hydroxycamptothecin and arsenic trioxide and their treatment for rabbit hepatocellular carcinoma. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2020; 165:57-63. [PMID: 31907490 DOI: 10.5507/bp.2019.063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/17/2019] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE This study aims to study the preparation method of arsenic trioxide (As2O3) polylactic-co-glyconlic acid (PLGA) microspheres and 10-hydroxycamptothecin (HCPT) PLGA microspheres and explore their therapeutic effects as embolic agents for VX2 hepatocellular carcinoma in rabbits. METHODS As2O3 and HCPT PLGA microspheres were prepared by multiple emulsion solvent evaporation method. Scanning electron microscopy (SEM) and particle size distribution were used to analyze the morphology, the drug sustained release ability was observed by the release of microspheres in vitro. The rabbit model of VX2 hepatocellular carcinoma was established and the hepatocellular carcinoma was treated with combined microspheres. The therapeutic effects were detected by qPCR, western blotting, HE staining and immunohistochemical methods. RESULTS The PLGA microspheres loaded with As2O3 and HCPT were successfully prepared by optimizing the ratio. The particle size was between 30 and 50 μm. In vitro release results showed that PLGA microspheres loaded with As2O3 released completely in 10 days and PLGA microspheres loaded with HCPT released completely in 12 days. Western blotting and qPCR results showed that the expression of ALDH1A1 and Nanog decreased significantly in treatment group. HE staining and immunohistochemical analysis showed that the expression of CD31, HIF and VEGF decreased significantly and the apoptosis of tissues was obvious. CONCLUSION The combination of As2O3 and HCPT PLGA microspheres as embolization for VX2 hepatocellular carcinoma in rabbits has significant therapeutic effect.
Collapse
Affiliation(s)
- Yumin Wu
- Department of Interventional Therapy, Shenzhen Second People's Hospital. No. 3002 Sungang West Road, Futian District, Shenzhen, Guangdong 518035, P.R.China
| | - Duanming Du
- Department of Interventional Therapy, Shenzhen Second People's Hospital. No. 3002 Sungang West Road, Futian District, Shenzhen, Guangdong 518035, P.R.China
| | - Juanping Chen
- Department of Interventional Therapy, Shenzhen Second People's Hospital. No. 3002 Sungang West Road, Futian District, Shenzhen, Guangdong 518035, P.R.China
| | - Chunlin Liu
- Department of Interventional Therapy, Shenzhen Second People's Hospital. No. 3002 Sungang West Road, Futian District, Shenzhen, Guangdong 518035, P.R.China
| |
Collapse
|
26
|
Lee HM, Kim DH, Lee HL, Cha B, Kang DH, Jeong YIL. Synergistic effect of buthionine sulfoximine on the chlorin e6-based photodynamic treatment of cancer cells. Arch Pharm Res 2019; 42:990-999. [DOI: 10.1007/s12272-019-01179-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/10/2019] [Indexed: 11/28/2022]
|
27
|
Wang C, Ning Z, Wan F, Huang R, Chao L, Kang Z, Yang F, Zhong G, Li Y, Pan J, Tang Z, Hu L. Characterization of the cellular effects and mechanism of arsenic trioxide-induced hepatotoxicity in broiler chickens. Toxicol In Vitro 2019; 61:104629. [PMID: 31442540 DOI: 10.1016/j.tiv.2019.104629] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 08/15/2019] [Accepted: 08/17/2019] [Indexed: 11/25/2022]
Abstract
To characterize the cellular effects and mechanism of arsenic trioxide (ATO)-induced hepatotoxicity in broiler chickens, increasing concentrations of ATO (0, 0.6, 1.2, 2.4, and 4.8 μM) were added to chicken hepatocyte cultures in vitro. The changes in hepatocyte morphology, oxidative stress and apoptosis were evaluated using fluorescence microscopy and flow cytometry. The effects of ATO on mRNA or protein expression of antioxidant enzymes, especially methionine sulfoxide reductase (Msr), were analyzed using qRT-PCR and western blotting assays. Increased apoptosis were concomitant with increased reactive oxygen species (ROS) accumulation and upregulation of antioxidant enzymes such as catalase (CAT) and superoxide dismutase (SOD) with increasing ATO concentrations. Moreover, G1 phase arrest and dysregulation of the balance between antiapoptotic versus proapoptotic factors were noted. Furthermore, upregulation of HO-1, SOD-1, and TRX in the ATO groups were consistent with ATO-induced oxidative damage. High Msr, SOD-1, TRX, Bak1, Bax, and p53 protein levels in the ATO groups indicate that these proteins may have accumulated to counter ATO-induced oxidative stress. ROS scavenger N-acetyl-l-cysteine (NAC) could reverse ATO-induced oxidative damage and restore hepatocyte viability, even with compromised Msr function. Our findings suggest that Msr can protect broiler hepatocytes against ATO-induced oxidative stress. Furthermore, NAC-mediated reversal of oxidative damage may represent a strategy to mitigate potential economic losses associated with arsenic poisoning in the poultry industry.
Collapse
Affiliation(s)
- Congcong Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhijun Ning
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Fang Wan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Riming Huang
- Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Limin Chao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhenlong Kang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Fan Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Gaolong Zhong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jiaqiang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
28
|
Zeng R, Liang Y, Farooq MU, Zhang Y, Ei HH, Tang Z, Zheng T, Su Y, Ye X, Jia X, Zhu J. Alterations in transcriptome and antioxidant activity of naturally aged mice exposed to selenium-rich rice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:17834-17844. [PMID: 31037530 DOI: 10.1007/s11356-019-05226-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/17/2019] [Indexed: 06/09/2023]
Abstract
Selenium (Se) is a vital element which leads to strong antioxidation in animals and humans. However, the mechanism underlying natural cereal Se-induced biological changes is not well understood. This study intended to explore the gene differential expression in naturally aged mice exposed to selenium by RNA-Seq technique. A total spectrum of 53 differentially expressed genes was quantified in mice heart tissues treated with Se-rich and general rice. The GO functional annotation of differentially expressed genes disclosed the enrichment of cellular process, ionic binding, biological regulation, and catalytic activity. One hundred twenty-three differential pathways (cardiovascular diseases, immune system, transport and catabolism, longevity regulating, and PI3K-AKT signaling) were identified according to KEGG metabolic terms. Afterwards, the effect of Se-rich rice on the antioxidant activity was assessed. The selenium-rich diet increased the total antioxidant capacity (T-AOC), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) in mice serum and livers while significantly reduces methane dicarboxylic aldehyde (MDA) contents. FOXO1 and FOXO3 genes, which acted as the regulators of apoptosis and the antioxidant enzyme, were significantly enhanced in mice when fed with Se-rich rice. In short, the present findings disclosed the alluring insights of organic and inorganic selenium sources on certain biological processes and antioxidant activity of living bodies. However, long-term trials are still required to draw a definitive conclusion, including risks and benefit analysis for various management strategies.
Collapse
Affiliation(s)
- Rui Zeng
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yuanke Liang
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Muhammad Umer Farooq
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yujie Zhang
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Hla Hla Ei
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Zhichen Tang
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Tengda Zheng
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yang Su
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Xiaoying Ye
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Xiaomei Jia
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Jianqing Zhu
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
29
|
The Antioxidant from Ethanolic Extract of Rosa cymosa Fruits Activates Phosphatase and Tensin Homolog In Vitro and In Vivo: A New Insight on Its Antileukemic Effect. Int J Mol Sci 2019; 20:ijms20081935. [PMID: 31010164 PMCID: PMC6514837 DOI: 10.3390/ijms20081935] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/13/2019] [Accepted: 04/17/2019] [Indexed: 12/18/2022] Open
Abstract
Rosa cymosa Tratt is a Chinese herbal remedy that is used in the treatment of diarrhea, burns, rheumatoid arthritis, and hemorrhage. Despite its use in Asian folk medicine, there are limited reports on the biological activity of R. cymosa fruits. This study focused on the investigation of the antitumor effect of the antioxidative ethanolic extract of R. cymosa fruits (RCE) along with its underlying mechanism of action. RCE showed a potent cytotoxic effect against Sup-T1 and Molt-4 lymphoblastic leukemia cells. In the xenograft animal model, the tumor size was significantly reduced to about 59.42% in the RCE-treated group in comparison with the control group. The use of RCE (37.5, 75, or 150 μg/mL) triggered apoptosis by 26.52–83.49%, disrupted mitochondrial membrane potential (MMP) by 10.44–58.60%, and promoted calcium release by 1.29-, 1.44-, and 1.71-fold compared with the control group. The extract induced redox oxygen species (ROS) generation through the elimination of Nrf2/Keap1/P62-mediated oxidative stress response. The loss of phosphatase and tensin homolog (PTEN) activation by RCE impaired PI3K/Akt/Foxo and Jak/Stat activation pathways, which contributed to tumorigenesis. These multiple targets of R. cymosa against hematologic cancer cells suggested its potential application as an antileukemic dietary supplement.
Collapse
|
30
|
Pre-application of arsenic trioxide may potentiate cytotoxic effects of vinorelbine/docetaxel on neuroblastoma SK-N-SH cells. Biomed Pharmacother 2019; 113:108665. [PMID: 30889490 DOI: 10.1016/j.biopha.2019.108665] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/25/2019] [Accepted: 02/04/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Arsenic trioxide is effective in the treatment of acute promyelocytic leukemia and is currently in use in clinical trials for the treatment of solid tumor types. Given that arsenic trioxide is able to arrest neuroblastoma cell cycle in the G2/M phase, the present study is, to the best of our knowledge, the first to investigate whether the combination of arsenic trioxide with mitosis-phase-specific antineoplastic agents (vinorelbine or docetaxel) or non-mitosis-phase-specific antineoplastic agents (etoposide or cisplatin) exert synergistic effects in cytotoxicity on the human SK-N-SH neuroblastoma cell line. METHODS Neuroblastoma cells were either incubated with one of the four drugs individually, or preincubated with arsenic trioxide and then followed by another drug when cell cycle was arrested at the G2/M phase with the highest proportion. RESULTS The results of the present study revealed that arsenic trioxide potentiated the apoptotic rate of neuroblastoma cells induced by chemotherapeutic drugs. The present study further demonstrated that preincubation with arsenic trioxide followed by a mitosis-phase-specific antineoplastic agent result in a higher cytotoxicity effect compared with a non mitosis-phase-specific antineoplastic agent. Along with the enhanced cytotoxicity in combination group, the cell cycle distribution demonstrated a decreased proportion of G2/M phase in the combination group. CONCLUSION The in vitro study revealed that the pre-application of arsenic trioxide followed by mitosis-phase-specific antineoplastic agents potentiate the cytotoxic effects on neuroblastoma cells, therefore arsenic trioxide may be a promising therapeutic option for treating neuroblastoma.
Collapse
|
31
|
Pradhan J, Mohanty C, Sahoo SK. Protective efficacy of crocetin and its nanoformulation against cyclosporine A-mediated toxicity in human embryonic kidney cells. Life Sci 2018; 216:39-48. [PMID: 30444987 DOI: 10.1016/j.lfs.2018.11.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 10/23/2018] [Accepted: 11/12/2018] [Indexed: 01/15/2023]
Abstract
AIM This study is aimed to formulate crocetin-loaded lipid Nanoparticles (NPs) and to evaluate its antioxidant properties in a cyclosporine A-mediated toxicity in Human Embryonic Kidney (HEK-293) cells in vitro. MAIN METHODS Crocetin-loaded NPs were prepared followed by physicochemical characterization. In vitro protective efficacy of crocetin and crocetin loaded NPs was investigated in cyclosporine A-mediated toxicity in HEK-293 cells by assessing free radical scavenging, DNA Nicking, cytotoxicity, intracellular Reactive oxygen species (ROS) inhibition, Mitochondrial membrane potential (MMPs) loss and evaluating the activity and expression of antioxidant enzymes and non-enzyme level. Further, we have studied the mechanism of protective activity of crocetin either native or in NPs by studying the expression of phase II detoxifying proteins (HO-1) via Nrf2 mediated regulation. KEY FINDINGS Our results showed that pretreatment with crocetin and crocetin-loaded NPs attenuated the cyclosporine A-mediated toxicity, ROS production and exhibited enhance free radical scavenging ability and cytoprotective activity. Further, the treatment prevented MMPs loss by directly scavenging the ROS and restored the antioxidant enzyme network with normalization of heme oxygenase-1 (HO-1) expression by inhibiting nuclear translocation of Nrf2. SIGNIFICANCE Pretreatment of crocetin and crocetin-loaded NPs provided pronounce protective effect against cyclosporine A-mediated toxicity in HEK-293 cells by nullifying the ROS formation and restored antioxidant network through inhibition of Nrf2 translocation and followed by expression of HO-1. Such an approach may be anticipated to be beneficial for antioxidant therapy.
Collapse
Affiliation(s)
- Jyotsnarani Pradhan
- Institute of Life Sciences, Bhubaneswar, Odisha, India; P.G. Department of Biotechnology, Utkal University, Bhubaneswar, Odisha, India
| | | | | |
Collapse
|
32
|
Martin R, Dowling K, Nankervis S, Pearce D, Florentine S, McKnight S. In vitro assessment of arsenic mobility in historical mine waste dust using simulated lung fluid. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2018; 40:1037-1049. [PMID: 28497229 DOI: 10.1007/s10653-017-9974-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 05/08/2017] [Indexed: 06/07/2023]
Abstract
Exposure studies have linked arsenic (As) ingestion with disease in mining-affected populations; however, inhalation of mine waste dust as a pathway for pulmonary toxicity and systemic absorption has received limited attention. A biologically relevant extractant was used to assess the 24-h lung bioaccessibility of As in dust isolated from four distinct types of historical gold mine wastes common to regional Victoria, Australia. Mine waste particles less than 20 µm in size (PM20) were incubated in a simulated lung fluid containing a major surface-active component found in mammalian lungs, dipalmitoylphosphatidylcholine. The supernatants were extracted, and their As contents measured after 1, 2, 4, 8 and 24 h. The resultant As solubility profiles show rapid dissolution followed by a more modest increasing trend, with between 75 and 82% of the total 24-h bioaccessible As released within the first 8 h. These profiles are consistent with the solubility profile of scorodite, a secondary As-bearing phase detected by X-ray diffraction in one of the investigated waste materials. Compared with similar studies, the cumulative As concentrations released at the 24-h time point were extremely low (range 297 ± 6-3983 ± 396 µg L-1), representing between 0.020 ± 0.002 and 0.036 ± 0.003% of the total As in the PM20.
Collapse
Affiliation(s)
- Rachael Martin
- Faculty of Science and Technology, Federation University Australia, Mount Helen, VIC, Australia.
| | - Kim Dowling
- Faculty of Science and Technology, Federation University Australia, Mount Helen, VIC, Australia
| | - Scott Nankervis
- Faculty of Science and Technology, Federation University Australia, Mount Helen, VIC, Australia
| | - Dora Pearce
- Faculty of Science and Technology, Federation University Australia, Mount Helen, VIC, Australia
- Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Singarayer Florentine
- Faculty of Science and Technology, Federation University Australia, Mount Helen, VIC, Australia
| | - Stafford McKnight
- Faculty of Science and Technology, Federation University Australia, Mount Helen, VIC, Australia
| |
Collapse
|
33
|
Park WH, Han BR, Park HK, Kim SZ. Arsenic trioxide induces growth inhibition and death in human pulmonary artery smooth muscle cells accompanied by mitochondrial O2•- increase and GSH depletion. ENVIRONMENTAL TOXICOLOGY 2018; 33:833-840. [PMID: 29708299 DOI: 10.1002/tox.22569] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 04/01/2018] [Accepted: 04/10/2018] [Indexed: 06/08/2023]
Abstract
Arsenic trioxide (ATO; As2 O3 ) induces cell death in various cells via oxidative stress. Expose to chronic arsenic is involved in the development of vascular diseases. However, little is known about the cytotoxic effects of ATO on human normal vascular smooth muscle cells (VSMCs). Thus, in this study, we investigated the effects of ATO on cell growth and death in human pulmonary artery smooth muscle (HPASM) cells in relation to reactive oxygen species (ROS) and glutathione (GSH) levels. ATO treatment decreased the growth of HPASM cells with an IC50 of ∼30-50 μM at 24 h, and ATO induced HPASM cell death via apoptosis or necrosis dependent on the doses of it at this time. Treatment with 50 μM ATO did not increase ROS levels at the early time points, but it significantly increased mitochondrial O2•- levels at 24 h. ATO also induced GSH depletion in HPASM cells. N-acetyl cysteine (NAC; a well-known antioxidant) did not significantly affect apoptotic cell death, ROS levels, or GSH depletion in ATO-treated HPASM cells. However, l-buthionine sulfoximine (BSO; an inhibitor of GSH synthesis) intensified mitochondrial O2•- levels in ATO-treated HPASM cells, and significantly increased cell death and GSH depletion in these cells as well. In summary, we provided the first evidence that ATO inhibited the growth of HPASM cells, and induced apoptotic and/or necrotic cell death in these cells, accompanied by increases in mitochondrial O2•- level and GSH depletion.
Collapse
Affiliation(s)
- Woo Hyun Park
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, 20 Geonji-ro, Deokjin, Jeonju, Jeollabuk, 54907, Republic of Korea
| | - Bo Ran Han
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, 20 Geonji-ro, Deokjin, Jeonju, Jeollabuk, 54907, Republic of Korea
| | - Hyun Kyung Park
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, 20 Geonji-ro, Deokjin, Jeonju, Jeollabuk, 54907, Republic of Korea
| | - Sung Zoo Kim
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, 20 Geonji-ro, Deokjin, Jeonju, Jeollabuk, 54907, Republic of Korea
| |
Collapse
|
34
|
Huang Q, Xi G, Alamdar A, Zhang J, Shen H. Comparative proteomic analysis reveals heart toxicity induced by chronic arsenic exposure in rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 229:210-218. [PMID: 28599205 DOI: 10.1016/j.envpol.2017.05.077] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/12/2017] [Accepted: 05/27/2017] [Indexed: 06/07/2023]
Abstract
Arsenic is a widespread metalloid in the environment, which poses a broad spectrum of adverse effects on human health. However, a global view of arsenic-induced heart toxicity is still lacking, and the underlying molecular mechanisms remain unclear. By performing a comparative quantitative proteomic analysis, the present study aims to investigate the alterations of proteome profile in rat heart after long-term exposure to arsenic. As a result, we found that the abundance of 81 proteins were significantly altered by arsenic treatment (35 up-regulated and 46 down-regulated). Among these, 33 proteins were specifically associated with cardiovascular system development and function, including heart development, heart morphology, cardiac contraction and dilation, and other cardiovascular functions. It is further proposed that the aberrant regulation of 14 proteins induced by arsenic would disturb cardiac contraction and relaxation, impair heart morphogenesis and development, and induce thrombosis in rats, which is mediated by the Akt/p38 MAPK signaling pathway. Overall, these findings will augment our knowledge of the involved mechanisms and develop useful biomarkers for cardiotoxicity induced by environmental arsenic exposure.
Collapse
Affiliation(s)
- Qingyu Huang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | - Guochen Xi
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China
| | - Ambreen Alamdar
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China
| | - Jie Zhang
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China
| | - Heqing Shen
- Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, PR China.
| |
Collapse
|
35
|
Han BR, Park WH. MAPK inhibitors enhance cell death in pyrogallol-treated human pulmonary fibroblast cells via increasing O 2•- levels. Oncol Lett 2017; 14:1179-1185. [PMID: 28693293 DOI: 10.3892/ol.2017.6185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/21/2017] [Indexed: 01/24/2023] Open
Abstract
Pyrogallol (PG) induces apoptosis in lung cancer cells via the overproduction of O2•- and affects mitogen-activated protein kinases (MAPKs) in these cells. The aim of the present study was to elucidate the effect of PG and/or MAPK inhibitors on human pulmonary fibroblast (HPF) cell viability in relation to reactive oxygen species (ROS) and glutathione (GSH). Treatment with 50 or 100 µM PG inhibited the viability of HPF cells, and induced cell death and the loss of mitochondrial membrane potential (MMP; ΔΨm). In particular, treatment with 100 µM PG induced cell death via apoptosis as well as necrosis in HPF cells. PG increased mitochondrial O2•- levels and the number of GSH-depleted HPF cells. All the MAPK (mitogen-activated protein kinase kinase, c-Jun N-terminal kinase and p38) inhibitors enhanced the inhibition of cell viability, cell death and MMP (ΔΨm) loss in 100 µM PG-treated HPF cells. All the inhibitors increased the O2•- levels in 100 µM PG-treated HPF cells, but none of the inhibitors significantly altered the PG-induced GSH depletion. In conclusion, PG treatment induced cell death via apoptosis and necrosis in HPF cells. Treatment with MAPK inhibitors slightly enhanced cell death in PG-treated HPF cells. HPF cell death induced by PG and/or MAPK inhibitors was at least partially associated with changes in O2•- levels and GSH content. The present data provided useful information to understand PG-induced normal lung cell death in association with MAPK signaling pathways and ROS levels.
Collapse
Affiliation(s)
- Bo Ram Han
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, Jeonju, Jeollabuk 54907, Republic of Korea
| | - Woo Hyun Park
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, Jeonju, Jeollabuk 54907, Republic of Korea
| |
Collapse
|
36
|
Galadari S, Rahman A, Pallichankandy S, Thayyullathil F. Reactive oxygen species and cancer paradox: To promote or to suppress? Free Radic Biol Med 2017; 104:144-164. [PMID: 28088622 DOI: 10.1016/j.freeradbiomed.2017.01.004] [Citation(s) in RCA: 626] [Impact Index Per Article: 89.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 12/16/2016] [Accepted: 01/03/2017] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS), a group of highly reactive ions and molecules, are increasingly being appreciated as powerful signaling molecules involved in the regulation of a variety of biological processes. Indeed, their role is continuously being delineated in a variety of pathophysiological conditions. For instance, cancer cells are shown to have increased ROS levels in comparison to their normal counterparts. This is partly due to an enhanced metabolism and mitochondrial dysfunction in cancer cells. The escalated ROS generation in cancer cells contributes to the biochemical and molecular changes necessary for the tumor initiation, promotion and progression, as well as, tumor resistance to chemotherapy. Therefore, increased ROS in cancer cells may provide a unique opportunity to eliminate cancer cells via elevating ROS to highly toxic levels intracellularly, thereby, activating various ROS-induced cell death pathways, or inhibiting cancer cell resistance to chemotherapy. Such results can be achieved by using agents that either increase ROS generation, or inhibit antioxidant defense, or even a combination of both. In fact, a large variety of anticancer drugs, and some of those currently under clinical trials, effectively kill cancer cells and overcome drug resistance via enhancing ROS generation and/or impeding the antioxidant defense mechanism. This review focuses on our current understanding of the tumor promoting (tumorigenesis, angiogenesis, invasion and metastasis, and chemoresistance) and the tumor suppressive (apoptosis, autophagy, and necroptosis) functions of ROS, and highlights the potential mechanism(s) involved. It also sheds light on a very novel and an actively growing field of ROS-dependent cell death mechanism referred to as ferroptosis.
Collapse
Affiliation(s)
- Sehamuddin Galadari
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE; Al Jalila Foundation Research Centre, P.O. Box 300100, Dubai, UAE.
| | - Anees Rahman
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| | - Siraj Pallichankandy
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| | - Faisal Thayyullathil
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| |
Collapse
|
37
|
Chayapong J, Madhyastha H, Madhyastha R, Nurrahmah QI, Nakajima Y, Choijookhuu N, Hishikawa Y, Maruyama M. Arsenic trioxide induces ROS activity and DNA damage, leading to G0/G1 extension in skin fibroblasts through the ATM-ATR-associated Chk pathway. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2017; 24:5316-5325. [PMID: 28013460 DOI: 10.1007/s11356-016-8215-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 12/06/2016] [Indexed: 06/06/2023]
Abstract
Arsenic (As) toxicity is a global health problem, affecting millions of people. Exposure to arsenic, mostly via drinking water, has been associated with cancer of skin, lungs, and blood, in addition to several kinds of skin lesions. The present study focused on the effect of arsenic trioxide (As2O3) on normal skin fibroblast cells. Specifically, the effect of As2O3 on ROS generation and oxidative stress was investigated. Proteins involved in the DNA damage signaling pathway and cell cycle were also studied. As2O3 induced the generation of intracellular ROS. Immunohistochemistry analysis revealed a dose-dependent increase in the number of 8-OHdG-positive cells, an indication of oxidative stress. Cell cycle analysis by flow cytometry demonstrated that As2O3 caused a significant percentage of cells to accumulate in the G0/G1 phase with a concomitant reduction in the S phase. Increases in the activated forms of DNA damage signaling proteins, ATM and ATR, and their effector molecules, Chk2 and p53, were also observed. In addition, expression of oncogene p21 was also increased. The study shows that exposure of normal skin fibroblast cells to As2O3 could lead to cell cycle arrest through ATM/ATR and DNA damage signaling pathways. In conclusion, we report here that arsenic trioxide increases cellular oxidative stress leading to shift in cell cycle and leads to DNA damage through ATM/ATR and the CHK-dependent signaling pathway.
Collapse
Affiliation(s)
- Jutapon Chayapong
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Harishkumar Madhyastha
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Radha Madhyastha
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Queen Intan Nurrahmah
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Yuichi Nakajima
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Narantsog Choijookhuu
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Yoshitaka Hishikawa
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan
| | - Masugi Maruyama
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, 889-1692, Japan.
| |
Collapse
|
38
|
Pan B, Zhong W, Deng Z, Lai C, Chu J, Jiao G, Liu J, Zhou Q. Inhibition of prostate cancer growth by solanine requires the suppression of cell cycle proteins and the activation of ROS/P38 signaling pathway. Cancer Med 2016; 5:3214-3222. [PMID: 27726305 PMCID: PMC5119977 DOI: 10.1002/cam4.916] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/21/2016] [Accepted: 08/23/2016] [Indexed: 12/20/2022] Open
Abstract
Solanine, a naturally steroidal glycoalkaloid in nightshade (Solanum nigrum Linn.), can inhibit proliferation and induce apoptosis of tumor cells. However, the mechanism of solanine-suppressing prostate cancer cell growth remains to be elucidated. This study investigates the inhibition mechanism of solanine on cancer development in vivo and in cultured human prostate cancer cell DU145 in vitro. Results show that solanine injection significantly suppresses the tumor cell growth in xenograft athymic nude mice. Solanine regulates the protein levels of cell cycle proteins, including Cyclin D1, Cyclin E1, CDK2, CDK4, CDK6, and P21 in vivo and in vitro. Also, in cultured DU145 cell, solanine significantly inhibits cell growth. Moreover, the administration of NAC, an active oxygen scavenger, markedly reduces solanine-induced cell death. Blockade of P38 MAPK kinase cannot suppress reactive oxygen species (ROS), but can suppress solanine-induced cell apoptosis. Also, inhibition of ROS by NAC inactivates P38 pathway. Taken together, the data suggest that inhibition of prostate cancer growth by solanine may be through blocking the expression of cell cycle proteins and inducing apoptosis via ROS and activation of P38 pathway. These findings indicate an attractive therapeutic potential of solanine for suppression of prostate cancer.
Collapse
Affiliation(s)
- Bin Pan
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Weifeng Zhong
- Sun Yat-sen Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Zhihai Deng
- Department of Urology, Gao Zhou People's Hospital, Gaozhou, China
| | - Caiyong Lai
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jing Chu
- Department of Urology, Zhuhai People's Hospital, Zhuhai, China
| | - Genlong Jiao
- Department of Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Junfeng Liu
- Department of Urology, Inner Mongolia People's Hospital, Hohhot, China
| | - Qizhao Zhou
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
39
|
Zhang Y, Wu X, Li Y, Zhang H, Li Z, Zhang Y, Zhang L, Ju J, Liu X, Chen X, Glybochko PV, Nikolenko V, Kopylov P, Xu C, Yang B. Endothelial to mesenchymal transition contributes to arsenic-trioxide-induced cardiac fibrosis. Sci Rep 2016; 6:33787. [PMID: 27671604 PMCID: PMC5037371 DOI: 10.1038/srep33787] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 09/01/2016] [Indexed: 12/26/2022] Open
Abstract
Emerging evidence has suggested the critical role of endothelial to mesenchymal transition (EndMT) in fibrotic diseases. The present study was designed to examine whether EndMT is involved in arsenic trioxide (As2O3)-induced cardiac fibrosis and to explore the underlying mechanisms. Cardiac dysfunction was observed in rats after exposure to As2O3 for 15 days using echocardiography, and the deposition of collagen was detected by Masson’s trichrome staining and electron microscope. EndMT was indicated by the loss of endothelial cell markers (VE-cadherin and CD31) and the acquisition of mesenchymal cell markers (α-SMA and FSP1) determined by RT-PCR at the mRNA level and Western blot and immunofluorescence analysis at the protein level. In the in-vitro experiments, endothelial cells acquired a spindle-shaped morphology accompanying downregulation of the endothelial cell markers and upregulation of the mesenchymal cell markers when exposed to As2O3. As2O3 activated the AKT/GSK-3β/Snail signaling pathway, and blocking this pathway with PI3K inhibitor (LY294002) abolished EndMT in As2O3-treated endothelial cells. Our results highlight that As2O3 is an EndMT-promoting factor during cardiac fibrosis, suggesting that targeting EndMT is beneficial for preventing As2O3-induced cardiac toxicity.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.,Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, 150086, China
| | - Xianxian Wu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yang Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province &Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, China
| | - Haiying Zhang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Zhange Li
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ying Zhang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Longyin Zhang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jiaming Ju
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xin Liu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiaohui Chen
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Peter V Glybochko
- The Research Center, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Vladimir Nikolenko
- The Research Center, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Philipp Kopylov
- Department of preventive and emergency cardiology, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Chaoqian Xu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Baofeng Yang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.,Department of Pharmacology and Therapeutics, Melbourne School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, 3010, Australia
| |
Collapse
|
40
|
Yamaguchi Y, Madhyastha H, Madhyastha R, Choijookhuu N, Hishikawa Y, Pengjam Y, Nakajima Y, Maruyama M. Arsenic acid inhibits proliferation of skin fibroblasts, and increases cellular senescence through ROS mediated MST1-FOXO signaling pathway. J Toxicol Sci 2016; 41:105-13. [PMID: 26763397 DOI: 10.2131/jts.41.105] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Arsenic exposure through drinking water is a major public health problem. It causes a number of toxic effects on skin. Arsenic has been reported to inhibit cell proliferation in in vitro conditions. However, reports about the molecular mechanisms are limited. Here, we investigated the mechanism involved in arsenic acid-mediated inhibition of cell proliferation using mouse skin fibroblast cell line. The present study found that 10 ppm arsenic acid inhibited cell proliferation, without any effect on cell death. Arsenic acid induced the generation of reactive oxygen species (ROS), resulting in oxidative stress to DNA. It also activated the mammalian Ste20-like protein kinase 1 (MST1); however the serine/threonine kinase Akt was downregulated. Forkhead box O (FOXO) transcription factors are activated through phosphorylation by MST1 under stress conditions. They are inhibited by phosphorylation by Akt through external and internal stimuli. Activation of FOXOs results in their nuclear localization, followed by an increase in transcriptional activity. Our results showed that arsenic induced the nuclear translocation of FOXO1 and FOXO3a, and altered the cell cycle, with cells accumulating at the G2/M phase. These effects caused cellular senescence. Taken together, our results indicate that arsenic acid inhibited cell proliferation through cellular senescence process regulated by MST1-FOXO signaling pathway.
Collapse
Affiliation(s)
- Yuya Yamaguchi
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Park WH. Exogenous H2O2 induces growth inhibition and cell death of human pulmonary artery smooth muscle cells via glutathione depletion. Mol Med Rep 2016; 14:936-42. [PMID: 27220315 DOI: 10.3892/mmr.2016.5307] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 05/10/2016] [Indexed: 11/05/2022] Open
Abstract
Reactive oxygen species (ROS) are associated with various pathophysiological processes of vascular smooth muscle cells (VSMCs). Pyrogallol (PG) induces the superoxide anion (O2•‑)‑mediated cell death of numerous cell types. The present study aimed to investigate the effects of exogenous hydrogen peroxide (H2O2) and PG treatment on the cell growth and death of human pulmonary artery smooth muscle cells (HPASMCs), with regards to intracellular ROS and glutathione (GSH) levels, as determined by MTT and cell number assays. H2O2 led to reduced growth of HPASMCs, with a half maximal inhibitory concentration of 250‑500 µM at 24 h, and induced apoptosis, as determined by Annexin V‑staining and benzyloxycarbonyl‑Val‑Ala‑Asp‑fluoromethylketone treatment. However, PG did not strongly induce growth inhibition and death of HPASMCs. In addition, H2O2 led to increased ROS levels, including mitochondrial O2•‑, and induced GSH depletion in HPASMCs. Treatment with N‑acetyl cysteine (NAC) attenuated apoptotic cell death and ROS levels in H2O2‑treated HPASMCs, and also prevented GSH depletion. Notably, PG treatment did not increase ROS levels, including mitochondrial O2•‑. Furthermore, NAC induced a significant increase in mitochondrial O2•‑ levels in PG‑treated HPASMCs, and cell death and GSH depletion were significantly increased. L‑buthionine sulfoximine intensified cell death and GSH depletion in PG‑treated HPASMCs. In conclusion, exogenous H2O2 induced growth inhibition and cell death of HPASMCs via GSH depletion.
Collapse
Affiliation(s)
- Woo Hyun Park
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, Jeonju, North Jeolla 561‑180, Republic of Korea
| |
Collapse
|
42
|
Zhang P, Wang W, Wei Z, Xu LI, Yang X, DU Y. xCT expression modulates cisplatin resistance in Tca8113 tongue carcinoma cells. Oncol Lett 2016; 12:307-314. [PMID: 27347143 DOI: 10.3892/ol.2016.4571] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/19/2016] [Indexed: 12/26/2022] Open
Abstract
Tongue squamous cell carcinoma (TSCC), which is a subtype of head and neck cancer, is the most common type of oral cancer. Due to its high recurrence rate and chemoresistance, the average survival rate for patients with TSCC remains unsatisfactory. At present, cisplatin (CDDP) is utilized as the first-line treatment for numerous solid neoplasms, including TSCC. CDDP resistance develops in the majority of patients; however, the mechanism of such resistance remains unknown. Therefore, the present study aimed to clarify the mechanism of CDDP resistance and attempted to reduce chemoresistance. The results indicated that CDDP significantly increased expression of xCT, which is the light chain and functional subunit of the glutamate/cysteine transporter system xc-, and a subsequent increase in glutathione (GSH) levels was observed. The present study demonstrated that the upregulation of xCT expression and intercellular GSH levels contributed to CDDP resistance in TSCC cells. Furthermore, xCT suppression, induced by small interfering RNA or pharmacological inhibitors, sensitized TSCC cells to CDDP treatment. In conclusion, the present study revealed that CDDP-induced xCT expression promotes CDDP chemoresistance, and xCT inhibition sensitizes TSCC cells to CDDP treatment. These results provide a novel insight into the molecular mechanisms involved in TSCC cell chemoresistance.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Stomatology, No. 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Wei Wang
- Department of Stomatology, No. 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Zhenhui Wei
- Department of Stomatology, No. 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - L I Xu
- Department of Stomatology, No. 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Xuanning Yang
- Department of Stomatology, No. 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Yuanhong DU
- Department of Stomatology, No. 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
43
|
Yu H, Liu S, Li M, Wu B. Influence of diet, vitamin, tea, trace elements and exogenous antioxidants on arsenic metabolism and toxicity. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2016; 38:339-351. [PMID: 26169729 DOI: 10.1007/s10653-015-9742-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 07/02/2015] [Indexed: 06/04/2023]
Abstract
Health risk of arsenic (As) has received increasing attention. Acute and chronic exposure to As could cause several detrimental effects on human health. As toxicity is closely related to its bioaccessibility and metabolism. In real environment, many factors, such as diet and nutrition, can influence As bioaccessibility, metabolism and toxicity. This paper mainly reviews the influences of diets and elements on As bioaccessibility, metabolism and toxicity and their underlying mechanisms to provide suggestions for future investigations. Vitamins, jaggery, fruit, tea, glutathione, N-acetylcysteine and zinc could reduce the As-induced toxicity by increasing antioxidative enzymes to antagonize oxidative stress caused by As and/or increasing As methylation. However, bean and betel nut could increase risk of skin lesions caused by As. Interestingly, high-fat diet, selenium and iron have incompatible effects on As bioaccessibility, metabolism and toxicity in different experimental conditions. Based on current literatures, the As methylation and As-induced oxidative damage might be two main ways that the diets and elements influence As toxicity. Combined application of in vitro human cell lines and gastrointestinal models might be useful tools to simultaneously characterize the changes in As bioaccessibility and toxicity in the future research.
Collapse
Affiliation(s)
- Haiyan Yu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, People's Republic of China
| | - Su Liu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, People's Republic of China
| | - Mei Li
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, People's Republic of China
| | - Bing Wu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
44
|
Ochi M, Tanaka Y, Toyoda H. Protective effect of N-acetylcysteine against nicardipine hydrochloride-induced autophagic cell death of human vascular endothelial cells. J Toxicol Sci 2015; 40:551-8. [DOI: 10.2131/jts.40.551] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Masanori Ochi
- Biological Research Department, Sawai Pharmaceutical Co., Ltd
| | | | - Hiromu Toyoda
- Biological Research Department, Sawai Pharmaceutical Co., Ltd
| |
Collapse
|
45
|
You BR, Kim SH, Park WH. Reactive oxygen species, glutathione, and thioredoxin influence suberoyl bishydroxamic acid-induced apoptosis in A549 lung cancer cells. Tumour Biol 2014; 36:3429-39. [PMID: 25537089 DOI: 10.1007/s13277-014-2978-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/11/2014] [Indexed: 12/12/2022] Open
Abstract
Suberoyl bishydroxamic acid (SBHA) as a histone deacetylase (HDAC) inhibitor can induce apoptosis through the formation of reactive oxygen species (ROS). However, there is no report about the regulation of ROS and antioxidant enzymes in SBHA-treated lung cancer cells. Here, we investigated the toxicological effects of SBHA on the regulations of ROS, glutathione (GSH), and antioxidant enzymes, especially thioredoxin (Trx) in A549 lung cancer cells. SBHA inhibited the growth of A549 cells in time- and dose-dependent manners, and it induced apoptosis which accompanied by the loss of mitochondrial membrane potential (MMP; ΔΨm). SBHA significantly increased ROS levels including O2 (•-) level at 72 h whereas it decreased ROS levels at the early time points (30 min to 3 h). SBHA also induced GSH depletion at 24 and 72 h. N-acetyl cysteine (NAC; a well-known antioxidant) prevented apoptotic cell death and GSH depletion via decreasing ROS in SBHA-treated A549 cells. In addition, SBHA changed the levels of antioxidant-related proteins, especially Trx1. The expression and activity of Trx1 in A549 cells were reduced by SBHA. While the downregulation of Trx1 enhanced cell death, ROS level, and GSH depletion in SBHA-treated A549 cells, the overexpression of Trx1 decreased ROS level in these cells without the prevention of cell death and GSH depletion. In conclusion, SBHA-induced A549 cell death was influenced by changes in ROS and GSH levels. The basal status of Trx1 among other antioxidant proteins was closely correlated with the survival of A549 cells.
Collapse
Affiliation(s)
- Bo Ra You
- Department of Physiology, Medical School, Research Institute for Endocrine Sciences, Chonbuk National University, JeonJu, 561-180, Republic of Korea
| | | | | |
Collapse
|
46
|
PX-12 induces apoptosis in Calu-6 cells in an oxidative stress-dependent manner. Tumour Biol 2014; 36:2087-95. [PMID: 25391429 DOI: 10.1007/s13277-014-2816-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 11/04/2014] [Indexed: 02/06/2023] Open
Abstract
PX-12 (1-methylpropyl 2-imidazolyl disulfide) as a thioredoxin (Trx) inhibitor has an anti-tumor effect. However, there is no report about the toxicological effect of PX-12 on lung cancer cells. Here, we investigated the anti-growth effects of PX-12 on Calu-6 lung cancer cells in relation to reactive oxygen species (ROS) and glutathione (GSH) levels. PX-12 induced the growth inhibition of Calu-6 cells with IC50 of nearly 3 μM at 72 h. In contrast, PX-12 did not affect the growth of human small airway epithelial cells (HSAECs). Cell cycle distribution analysis indicated that PX-12 significantly induced a G2/M phase arrest in Calu-6 cells. PX-12 also increased the number of annexin V-FITC-positive cells in Calu-6 cells. All the tested caspase inhibitors markedly prevented Calu-6 cell death induced by PX-12. With regard to ROS and GSH levels, PX-12 increased ROS levels containing O2(·-) in Calu-6 cells and induced the depletion of GSH. N-acetyl cysteine (NAC), which is a well-known antioxidant, significantly reduced O2(·-) level in PX-12-treated Calu-6 cells and prevented apoptosis and GSH depletion in these cells. In conclusion, it is the first report that PX-12 inhibited the growth of Calu-6 cells via a G2/M phase arrest as well as apoptosis, which effect was related to the intracellular increases in ROS levels.
Collapse
|
47
|
Health Effects Associated with Inhalation of Airborne Arsenic Arising from Mining Operations. GEOSCIENCES 2014. [DOI: 10.3390/geosciences4030128] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
48
|
Autophagy interplays with apoptosis and cell cycle regulation in the growth inhibiting effect of Trisenox in HEP-2, a laryngeal squamous cancer. Pathol Oncol Res 2014; 21:103-11. [PMID: 24838151 DOI: 10.1007/s12253-014-9794-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 05/06/2014] [Indexed: 02/02/2023]
Abstract
Laryngeal squamous cell carcinoma (LSCC) is the most common among several types of head and neck cancers. Current treatments have a poor effect on early and advanced cases, and further investigations for novel agents against LSCCs are desirable. In this study, we elucidate the cytotoxic enhancing effect of arsenic trioxide (As2O3) combined with L-buthionine sulfoximine (BSO) in LSCC. The effect of BSO with As2O3 or Cisplatin (CDDP) on cell viability was examined using 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). The reactive oxygen species (ROS) levels, cell cycle, and apoptosis were measured by flow cytometry using 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA), propidium iodide (PI) and annexin V/PI. The acidic vacuolar organelles were visualized by fluorescence microscope and quantified using flow cytometry. Neither CDDP nor As2O3 when used alone reduced the cell viability. BSO was found to enhance only As2O3 sensitivity, leading to G2/M arrest and autophagy with no correlation of ROS induction. This result suggests that modulation of glutathione enhances autophagy, which interplays with apoptosis. In this study, we obtained initial preclinical evidence for the potential efficacy of these drugs in a combined therapy protocol.
Collapse
|
49
|
Luo F, Zhuang Y, Sides MD, Sanchez CG, Shan B, White ES, Lasky JA. Arsenic trioxide inhibits transforming growth factor-β1-induced fibroblast to myofibroblast differentiation in vitro and bleomycin induced lung fibrosis in vivo. Respir Res 2014; 15:51. [PMID: 24762191 PMCID: PMC4113202 DOI: 10.1186/1465-9921-15-51] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 02/10/2014] [Indexed: 01/02/2023] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a progressive disease of insidious onset, and is responsible for up to 30,000 deaths per year in the U.S. Excessive production of extracellular matrix by myofibroblasts has been shown to be an important pathological feature in IPF. TGF-β1 is expressed in fibrotic lung and promotes fibroblast to myofibroblast differentiation (FMD) as well as matrix deposition. Methods To identify the mechanism of Arsenic trioxide’s (ATO)’s anti-fibrotic effect in vitro, normal human lung fibroblasts (NHLFs) were treated with ATO for 24 hours and were then exposed to TGF-β1 (1 ng/ml) before harvesting at multiple time points. To investigate whether ATO is able to alleviate lung fibrosis in vivo, C57BL/6 mice were administered bleomycin by oropharyngeal aspiration and ATO was injected intraperitoneally daily for 14 days. Quantitative real-time PCR, western blotting, and immunofluorescent staining were used to assess the expression of fibrotic markers such as α-smooth muscle actin (α-SMA) and α-1 type I collagen. Results Treatment of NHLFs with ATO at very low concentrations (10-20nM) inhibits TGF-β1-induced α-smooth muscle actin (α-SMA) and α-1 type I collagen mRNA and protein expression. ATO also diminishes the TGF-β1-mediated contractile response in NHLFs. ATO’s down-regulation of profibrotic molecules is associated with inhibition of Akt, as well as Smad2/Smad3 phosphorylation. TGF-β1-induced H2O2 and NOX-4 mRNA expression are also blocked by ATO. ATO-mediated reduction in Smad3 phosphorylation correlated with a reduction of promyelocytic leukemia (PML) nuclear bodies and PML protein expression. PML-/- mouse embryonic fibroblasts (MEFs) showed decreased fibronectin and PAI-1 expression in response to TGF-β1. Daily intraperitoneal injection of ATO (1 mg/kg) in C57BL/6 mice inhibits bleomycin induced lung α-1 type I collagen mRNA and protein expression. Conclusions In summary, these data indicate that low concentrations of ATO inhibit TGF-β1-induced fibroblast to myofibroblast differentiation and decreases bleomycin induced pulmonary fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Joseph A Lasky
- Department of Medicine, Section of Pulmonary Diseases, Critical Care and Environmental Medicine, Tulane University Health Science Center, New Orleans, LA 70112, USA.
| |
Collapse
|
50
|
YOU BORA, PARK WOOHYUN. Suberoylanilide hydroxamic acid-induced HeLa cell death is closely correlated with oxidative stress and thioredoxin 1 levels. Int J Oncol 2014; 44:1745-55. [DOI: 10.3892/ijo.2014.2337] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/06/2014] [Indexed: 11/05/2022] Open
|