1
|
Onisiforou A, Zanos P. One path, two solutions: Network-based analysis identifies targetable pathways for the treatment of comorbid type II diabetes and neuropsychiatric disorders. Comput Struct Biotechnol J 2024; 23:3610-3624. [PMID: 39493502 PMCID: PMC11530817 DOI: 10.1016/j.csbj.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/30/2024] [Accepted: 10/06/2024] [Indexed: 11/05/2024] Open
Abstract
Comorbid diseases complicate patient outcomes and escalate healthcare costs, necessitating the need for a deeper mechanistic understanding. Neuropsychiatric disorders (NPDs) such as Neurotic Disorder, Major Depression, Bipolar Disorder, Anxiety Disorder, and Schizophrenia significantly exacerbate Type 2 Diabetes Mellitus (DM2), often leading to suboptimal treatment outcomes. The neurobiological mechanisms underlying this comorbidity remain poorly understood. To address this gap, we developed a novel pathway-based network computational framework to identify critical shared disease mechanisms between DM2 and these five prevalent comorbid NPDs. Our approach involves reconstructing an integrated DM2 ∩ NPDs KEGG pathway-pathway network and employs two complementary analytical methods, including the "minimum path to comorbidity" method to identify the shortest path fostering comorbid development. This analysis uncovered shared pathways like the PI3K-Akt signaling pathway and highlighted key nodes such as calcium signaling, MAPK, estrogen signaling, and apoptosis pathways. Dysregulation of these pathways likely contributes to the development of DM2-NPDs comorbidity. These findings have significant clinical implications, as they identify promising therapeutic targets that could lead to more effective treatments addressing both DM2 and NPDs simultaneously. Our model not only elucidates the intricate molecular interactions driving this comorbidity but also identifies promising therapeutic targets, paving the way for innovative treatment strategies. Additionally, the framework developed in this study can be adapted to study other complex comorbid conditions, advancing personalized medicine for comorbidities and improving patient care.
Collapse
Affiliation(s)
- Anna Onisiforou
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, Nicosia 2109, Cyprus
| | - Panos Zanos
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, Nicosia 2109, Cyprus
| |
Collapse
|
2
|
Natalucci F, Ceccarelli F, Picciariello L, Olivieri G, Ciancarella C, Alessandri C, Conti F. Are Tattoos Safe in Patients with Systemic Lupus Erythematosus? Results From a Single-Center Study. Dermatol Pract Concept 2024; 14:dpc.1404a230. [PMID: 39652953 PMCID: PMC11619997 DOI: 10.5826/dpc.1404a230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2024] [Indexed: 12/12/2024] Open
Abstract
INTRODUCTION Systemic Lupus Erythematosus is a pleiotropic autoimmune disease with common skin involvement. To date, only one study has investigated tattoos safety in SLE patients. OBJECTIVE We performed a single-center study to evaluate the development of local and systemic complications after tattooing in a cohort of systemic lupus erythematosus (SLE) patients. Furthermore, we tried to identify SLE patients who had expressed the will to get a tattoo and why they decided not to. METHODS Consecutive SLE patients were asked to complete a questionnaire about tattoos, including their number, features, and side effects. Open questions were proposed to non-tattooed patients to describe why they did not have tattoos. RESULTS One hundred ninety-two SLE patients were enrolled [M/F 21/171; median age 41 years (IQR 18)]. Almost 50% of them had at least one tattoo. Seven patients (7.4%) referred adverse reactions to tattoos; interestingly, only one patient experienced a systemic reaction, specifically the occurrence of self-limiting lymphadenopathy. The main reason for not getting a tattoo was the diagnosis of SLE. CONCLUSIONS Our results suggest the safety of tattoos in SLE patients, as demonstrated by a low prevalence of mild adverse events.
Collapse
Affiliation(s)
- Francesco Natalucci
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Roma, Italy
| | - Fulvia Ceccarelli
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Roma, Italy
| | - Licia Picciariello
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Roma, Italy
| | - Giulio Olivieri
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Roma, Italy
| | - Claudia Ciancarella
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Roma, Italy
| | - Cristiano Alessandri
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Roma, Italy
| | - Fabrizio Conti
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Roma, Italy
| |
Collapse
|
3
|
Buranasudja V, Sanookpan K, Vimolmangkang S, Binalee A, Mika K, Krobthong S, Kerdsomboon K, Kumkate S, Poolpak T, Kidhakarn S, Yang KM, Limcharoensuk T, Auesukaree C. Pretreatment with aqueous Moringa oleifera Lam. leaf extract prevents cadmium-induced hepatotoxicity by improving cellular antioxidant machinery and reducing cadmium accumulation. Heliyon 2024; 10:e37424. [PMID: 39309955 PMCID: PMC11416483 DOI: 10.1016/j.heliyon.2024.e37424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/25/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024] Open
Abstract
Cadmium (Cd) is a highly harmful pollutant that poses a serious threat to human health. The liver is the primary organ for Cd accumulation, and Cd-induced hepatotoxicity has been shown to be strongly correlated with an oxidative imbalance in hepatocytes. Our previous studies in the eukaryotic model organism Saccharomyces cerevisiae revealed that not only co-treatment but also pretreatment with aqueous Moringa oleifera Lam. leaf extract (AMOLE) effectively mitigated Cd toxicity by reducing intracellular Cd accumulation and Cd-mediated oxidative stress. In this study, we therefore investigated the preventive effect of AMOLE against Cd toxicity in human HepG2 hepatocytes. The results showed that, similar to the case of the yeast model, pretreatment with AMOLE prior to Cd exposure also significantly inhibited Cd-induced oxidative stress in HepG2 cells. Untargeted LC-MS/MS-based metabolomic analysis of AMOLE revealed that its major phytochemical constituents were organic acids, particularly phenolic acids and carboxylic acids. Additionally, DPPH-HPTLC fingerprints suggested that quercetin and other flavonoids possibly contribute to the antioxidant activities of AMOLE. Based on our findings, it appears that pretreatment with AMOLE prevented Cd-induced hepatotoxicity via three possible mechanisms: i) direct elimination of free radicals by AMOLE antioxidant compounds; ii) upregulation of antioxidant defensive machinery (GPx1, and HO-1) via Nrf2 signaling cascade to improve cellular antioxidant capacity; and iii) reduction of intracellular Cd accumulation, probably by suppressing Cd uptake. These data strongly suggest the high potential of AMOLE for clinical utility in the prevention of Cd toxicity.
Collapse
Affiliation(s)
- Visarut Buranasudja
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kittipong Sanookpan
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Nabsolute Co., Ltd., Bangkok, 10330, Thailand
| | - Sornkanok Vimolmangkang
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Plant-Produced Pharmaceuticals, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Asma Binalee
- HPTLC Center, Chula PharTech Co., Ltd., Bangkok, 10330, Thailand
| | - Kamil Mika
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Pharmacological Screening, Jagiellonian University Medical College, 9 Medyczna Street, Krakow, PL, 30-688, Poland
| | - Sucheewin Krobthong
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kittikhun Kerdsomboon
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, 12120, Thailand
- Mahidol University-Osaka University Collaborative Research Center for Bioscience and Biotechnology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Supeecha Kumkate
- Department of Biology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Toemthip Poolpak
- Department of Biology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), CHE, OPS, MHESI, Bangkok, 10400, Thailand
| | - Siraprapa Kidhakarn
- Department of Biology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Kwang Mo Yang
- Department of Biology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), CHE, OPS, MHESI, Bangkok, 10400, Thailand
| | - Tossapol Limcharoensuk
- Mahidol University-Osaka University Collaborative Research Center for Bioscience and Biotechnology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Choowong Auesukaree
- Mahidol University-Osaka University Collaborative Research Center for Bioscience and Biotechnology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| |
Collapse
|
4
|
Li J, Fang X, Cui D, Ma Z, Yang J, Niu Y, Liu H, Xiang P. Mechanistic insights into cadmium exacerbating 2-Ethylhexyl diphenyl phosphate-induced human keratinocyte toxicity: Oxidative damage, cell apoptosis, and tight junction disruption. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116858. [PMID: 39137464 DOI: 10.1016/j.ecoenv.2024.116858] [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: 05/25/2024] [Revised: 07/19/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
Organophosphate flame retardants 2-ethylhexyldiphenyl phosphate (EHDPP) and cadmium (Cd) are ubiquitous in environmental matrices, and dermal absorption is a major human exposure pathway. However, their detrimental effects on the human epidermis remain largely unknown. In this study, human keratinocytes (HaCaT cells) were employed to examine the toxicity and underlying mechanisms of co-exposure to EHDPP and Cd. Their influence on cell morphology and viability, oxidative damage, apoptosis, and tight junction were determined. The results showed that co-exposure decreased cell viability by >40 %, induced a higher level of oxidative damage by increasing the generation of reactive oxygen species (1.3 folds) and inhibited CAT (79 %) and GPX (90 %) activities. Moreover, Cd exacerbated EHDPP-induced mitochondrial disorder and cellular apoptosis, which was evidenced by a reduction in mitochondrial membrane potential and an elevation of cyt-c and Caspase-3 mRNA expression. In addition, greater loss of ZO-1 immunoreactivity at cellular boundaries was observed after co-exposure, indicating skin epithelial barrier function disruption, which may increase the human bioavailability of contaminants via the dermal absorption pathway. Taken together, oxidative damage, cell apoptosis, and tight junction disruption played a crucial role in EHDPP + Cd triggered cytotoxicity in HaCaT cells. The detrimental effects of EHDPP + Cd co-exposure were greater than individual exposure, suggesting the current health risk assessment or adverse effects evaluation of individual exposure may underestimate their perniciousness. Our data imply the importance of considering the combined exposure to accurately assess their health implication.
Collapse
Affiliation(s)
- Jingya Li
- Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming 650224, China
| | - Xianlei Fang
- School of Basic Medical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Daolei Cui
- Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming 650224, China
| | - Ziya Ma
- Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming 650224, China
| | - Ji Yang
- Affiliated Hospital of Yunnan University, Eye Hospital of Yunnan Province, Kunming 650224, China
| | - Youya Niu
- School of Basic Medical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Hai Liu
- Affiliated Hospital of Yunnan University, Eye Hospital of Yunnan Province, Kunming 650224, China.
| | - Ping Xiang
- Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming 650224, China.
| |
Collapse
|
5
|
Maliyam P, Laphookhieo S, Koedrith P, Puttarak P. Antioxidative and anti-cytogenotoxic potential of Lysiphyllum strychnifolium (Craib) A. Schmitz extracts against cadmium-induced toxicity in human embryonic kidney (HEK293) and dermal fibroblast (HDF) cells. Heliyon 2024; 10:e34480. [PMID: 39130464 PMCID: PMC11315074 DOI: 10.1016/j.heliyon.2024.e34480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/12/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
Exposure to cadmium (Cd) results in bioaccumulation and irreversible damage; this encourages an investigation of alternatives to address Cd toxicity, using natural compounds. Lysiphyllum strychnifolium, a well-known Thai medicinal plant, was investigated for its phytochemical compounds and corresponding bioactivities, including antioxidant and anti-cytogenotoxic effects against Cd toxicity in HEK293 renal and HDF dermal cell models. The crude extract of L. strychnifolium (LsCrude) was partitioned into four fractions, using sequential polarity solvents (hexane, dichloromethane, ethyl acetate, and water, denoted as LsH, LsD, LsE, and LsW, respectively). The extraction yields were 1.79 %, 5.08 %, 8.53 %, and 70.25 % (w/w), respectively. Phytochemical screening revealed the presence of tannins, alkaloids, and flavonoids in LsCrude and its fractions, except for LsH. LsE exhibited the highest concentrations of phenolics (286.83 ± 6.83 mg GAE/g extract) and flavonoids (86.36 ± 1.29 mg QE/g extract). Subsequent 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical-scavenging and ferric-reducing ability of plasma (FRAP) reducing powder assays demonstrated the high antioxidant capacity of LsCrude and its fractions. The lowest IC50 value (9.11 ± 0.43 μg/mL) in the DPPH assay corresponded to LsW, whereas the highest total FRAP value (6.06 ± 0.70 mg QE Eq./g dry mass) corresponded to LsE. MTT and alkaline comet assays revealed the lack of toxicity of the extracts, which were considered safe. Upon exposure to Cd at the CC50 level, HEK293 cells treated with LsE suppressed Cd-induced damage. HDF cells treated with LsCrude, LsD, or LsE attenuated Cd-induced damage. In the pre-treatment, LsD protected the HDF cells against Cd-mediated cytogenotoxicity. These anti-cytogenotoxic potentials are likely due to the antioxidant properties of the phytochemicals. Our findings highlight the cyto-geno-protective properties of L. strychnifolium stem extracts against Cd toxicity in HEK293 and HDF cells, and provide a novel approach for combating oxidative stress and DNA damage caused by environmental pollutants.
Collapse
Affiliation(s)
- Pattaravan Maliyam
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai District, Songkhla, 90112, Thailand
| | - Surat Laphookhieo
- Center of Chemical Innovation for Sustainability (CIS) and School of Science, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Preeyaporn Koedrith
- Faculty of Environment and Resource Studies, Mahidol University, Phuttamonthon District, NakhonPathom, 73170, Thailand
| | - Panupong Puttarak
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai District, Songkhla, 90112, Thailand
- Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai District, Songkhla, 90112, Thailand
| |
Collapse
|
6
|
Chen S, Fan H, Pei Y, Zhang K, Zhang F, Hu Q, Jin E, Li S. MAPK Signaling Pathway Plays Different Regulatory Roles in the Effects of Boric Acid on Proliferation, Apoptosis, and Immune Function of Splenic Lymphocytes in Rats. Biol Trace Elem Res 2024; 202:2688-2701. [PMID: 37737440 DOI: 10.1007/s12011-023-03862-2] [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: 05/06/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023]
Abstract
Boron is one of the essential trace elements in animals. Although boron supplementation can enhance immune function and promote cell proliferation, high-dose boron supplementation can negatively affect immune function and inhibit cell proliferation. Furthermore, its action pathway is unknown. In this study, ERK1/2, JNK, and p38MAPK signaling pathways were blocked using specific blockers to investigate the impact of low-dose and high-dose boron on proliferation, apoptosis, and immune function of lymphocytes, and the expression of genes related to cell proliferation and apoptosis in rats. The addition of 0.4 mmol/L boron did not affect the ratio of CD4+/CD8+ T cells (P>0.05), IgG and IFN-γ contents (P>0.05), the proliferation rate of lymphocytes (P>0.05), and mRNA and protein expressions of PCNA (P>0.05) in the spleen after ERK1/2 signal pathway was selectively inhibited. Moreover, the addition of 40 mmol/L boron did not affect the proportion of CD4+ T cells, contents of IgG and cytokines (IL-2 and IL-4), proliferation and apoptosis rates of lymphocytes, and expression of proliferation- and apoptosis-related genes in the spleen. Meanwhile, the addition of 0.4 mmol/l boron increased the ratio of CD4+/CD8+ T cells (P<0.05 or P<0.01), IFN-γ or IgG contents (P<0.05), and the proliferation rate of lymphocytes (P<0.05) in spleen after selective inhibition of JNK or p38MAPK signaling pathways, while the protein expression of Caspase-3 decreased (P<0.05 or P<0.01). Furthermore, 40 mmol/L boron decreased the proportion of lymphocyte subsets, cytokine contents, proliferation rate of lymphocytes, and mRNA and protein expressions of PCNA. In contrast, the mRNA and protein expressions of Caspase-3 and protein expression of Bax were increased. These results indicate that ERK1/2 signaling pathway mainly regulates the effects of low-dose and high-dose boron on proliferation, apoptosis, and immune function of splenic lymphocytes.
Collapse
Affiliation(s)
- Shuqin Chen
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, 233100, People's Republic of China
| | - Haoran Fan
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, 233100, People's Republic of China
| | - Yaqiong Pei
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, 233100, People's Republic of China
| | - Kaihuan Zhang
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, 233100, People's Republic of China
| | - Feng Zhang
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, 233100, People's Republic of China
| | - Qianqian Hu
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, 233100, People's Republic of China
| | - Erhui Jin
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, 233100, People's Republic of China.
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, 233100, People's Republic of China.
| | - Shenghe Li
- College of Animal Science, Anhui Science and Technology University, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, 233100, People's Republic of China
- Anhui Province Key Laboratory of Animal Nutritional Regulation and Health, No.9 Donghua Road, Fengyang County, Chuzhou, Anhui Province, 233100, People's Republic of China
| |
Collapse
|
7
|
Sozer Karadagli S, Kaftan G, Cansever I, Armagan G, Sogut O. Tattoo inks: evaluation of cellular responses and analysis of some trace metals. Biometals 2024; 37:495-505. [PMID: 38038794 DOI: 10.1007/s10534-023-00564-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: 08/11/2023] [Accepted: 11/12/2023] [Indexed: 12/02/2023]
Abstract
After tattoo application, inks remain in the skin, mostly in the dermal layer, and manufacturers use inks that have not been adequately evaluated for safety in tattoo production. In this study, the metal contents (Cd, Hg, Pb, and Cr) of tattoo inks available in the Turkish market were determined and the relationship between cell viability and inflammatory response of the detected metal levels was investigated. Nine tattoo inks (3 colors) from 3 different brands abbreviated as E, I, and W were examined. ICP-MS was used for element analysis. The viability of human keratinocyte cells was determined by the WST-1 assay following ink exposures at various dilutions. IL-18 levels were measured in cell culture supernatant by ELISA method following ink or metal (Cd, Cr, Hg, and Pb) exposures. The concentrations of trace elements were found in inks as follows: Cd, 0.0641-1.3857; Hg, 0.0204-0.2675; Pb, 0.8527-6.5981; Cr, 0.1731-45.3962 µg mL-1. It was observed that the levels of Pb and especially Cr in the samples exceeded the limit values. Tattoo inks reduced the cell viability in a dose- and color-dependent manner. IL-18 release was significantly increased in all groups except Cr and black ink of brand I treated cells (p < 0.05). Our results show that the metal contents of tattoo inks exceed Council of Europe Resolution values in some samples and some inks induce immune system activation (IL-18 secretion) and cytotoxic effects. It is thought that these findings may contribute to the toxic/adverse effects of tattoo inks commonly used.
Collapse
Affiliation(s)
- Sumru Sozer Karadagli
- Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Ege University, Izmir, Turkey.
| | - Gizem Kaftan
- Faculty of Pharmacy, Department of Biochemistry, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Islam Cansever
- Central Research Laboratories, Katip Celebi University, Izmir, Turkey
| | - Guliz Armagan
- Faculty of Pharmacy, Department of Biochemistry, Ege University, Izmir, Turkey
| | - Ozlem Sogut
- Faculty of Pharmacy, Department of Analytical Chemistry, Ege University, Izmir, Turkey
| |
Collapse
|
8
|
Korotkov SM. Mitochondrial Oxidative Stress Is the General Reason for Apoptosis Induced by Different-Valence Heavy Metals in Cells and Mitochondria. Int J Mol Sci 2023; 24:14459. [PMID: 37833908 PMCID: PMC10572412 DOI: 10.3390/ijms241914459] [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: 08/09/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023] Open
Abstract
This review analyzes the causes and consequences of apoptosis resulting from oxidative stress that occurs in mitochondria and cells exposed to the toxic effects of different-valence heavy metals (Ag+, Tl+, Hg2+, Cd2+, Pb2+, Al3+, Ga3+, In3+, As3+, Sb3+, Cr6+, and U6+). The problems of the relationship between the integration of these toxic metals into molecular mechanisms with the subsequent development of pathophysiological processes and the appearance of diseases caused by the accumulation of these metals in the body are also addressed in this review. Such apoptosis is characterized by a reduction in cell viability, the activation of caspase-3 and caspase-9, the expression of pro-apoptotic genes (Bax and Bcl-2), and the activation of protein kinases (ERK, JNK, p53, and p38) by mitogens. Moreover, the oxidative stress manifests as the mitochondrial permeability transition pore (MPTP) opening, mitochondrial swelling, an increase in the production of reactive oxygen species (ROS) and H2O2, lipid peroxidation, cytochrome c release, a decline in the inner mitochondrial membrane potential (ΔΨmito), a decrease in ATP synthesis, and reduced glutathione and oxygen consumption as well as cytoplasm and matrix calcium overload due to Ca2+ release from the endoplasmic reticulum (ER). The apoptosis and respiratory dysfunction induced by these metals are discussed regarding their interaction with cellular and mitochondrial thiol groups and Fe2+ metabolism disturbance. Similarities and differences in the toxic effects of Tl+ from those of other heavy metals under review are discussed. Similarities may be due to the increase in the cytoplasmic calcium concentration induced by Tl+ and these metals. One difference discussed is the failure to decrease Tl+ toxicity through metallothionein-dependent mechanisms. Another difference could be the decrease in reduced glutathione in the matrix due to the reversible oxidation of Tl+ to Tl3+ near the centers of ROS generation in the respiratory chain. The latter may explain why thallium toxicity to humans turned out to be higher than the toxicity of mercury, lead, cadmium, copper, and zinc.
Collapse
Affiliation(s)
- Sergey M Korotkov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Thorez pr. 44, 194223 St. Petersburg, Russia
| |
Collapse
|
9
|
Hong W, Liu Y, Liang J, Jiang C, Yu M, Sun W, Huang B, Dong N, Kang L, Tang Y. Molecular Mechanisms of Selenium Mitigating Lead Toxicity in Chickens via Mitochondrial Pathway: Selenoproteins, Oxidative Stress, HSPs, and Apoptosis. TOXICS 2023; 11:734. [PMID: 37755744 PMCID: PMC10536545 DOI: 10.3390/toxics11090734] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/19/2023] [Accepted: 08/24/2023] [Indexed: 09/28/2023]
Abstract
Lead (Pb), a hazardous heavy metal, can damage the health of organisms. However, it is not clear whether Pb can damage chicken cerebellums and thalami. Selenium (Se), an essential nutrient for organisms, has a palliative effect on Pb poisoning in chickens. In our experiment, a model of chickens treated with Pb and Se alone and in combination was established to investigate the molecular mechanism of Se alleviating Pb-caused damage in both chicken cerebellums and thalami. Our morphological results indicated that Pb caused apoptotic lesions, such as mitochondrial and nuclear damage. Further, the anti-apoptotic gene Bcl-2 decreased; on the contrary, four pro-apoptotic genes (p53, Bax, Cyt c, and Caspase-3) increased under Pb treatment, meaning that Pb caused apoptosis via the p53-Cyt c-Caspase-3 pathway. Furthermore, we further demonstrated that Pb elevated four HSPs (HSP27, HSP40, HSP70, and HSP90), as well as HSP70 took part in the molecular mechanism of Pb-caused apoptosis. In addition, we found that Pb exposure led to oxidative stress via up-regulating the oxidant H2O2 and down-regulating four antioxidants (CAT, SOD, GST, and GPx). Moreover, Pb decreased three Se-containing factors (Txnrd1, Txnrd2, and Txnrd3), further confirming that Pb caused oxidative stress. Interestingly, Se supplementation reversed the above changes caused by Pb and alleviated Pb-induced oxidative stress and apoptosis. A time dependency was demonstrated for Bcl-2, Bax, and Cyt c in the cerebellums, as well as CAT, GPx, and p53 in the thalami of Pb-exposed chickens. HSP70 in cerebellums and HSP27 in thalami were more sensitive than those in thalami and cerebellums, respectively, under Pb exposure. Pb-induced apoptosis of thalami was more severe than cerebellums. In conclusion, after Pb treatment, Txnrds mediated oxidative stress, oxidative stress up-regulated HSPs, and finally, HSP70 triggered apoptosis. Se supplementation antagonized Pb-induced oxidative stress and apoptosis via the mitochondrial pathway and selenoproteins in chicken cerebellums and thalami. This study provides new information for the mechanism of environmental pollutant poisoning and the detoxification of Se on abiotic stress.
Collapse
Affiliation(s)
- Weichen Hong
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yuhao Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Jiatian Liang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Chunyu Jiang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Meijin Yu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Wei Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Bin Huang
- Electrical and Information Engineering College, Jilin Agricultural Science and Technology University, Jilin City 132101, China
| | - Na Dong
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Lu Kang
- Institute of Agricultural Quality Standards and Testing Technology, Xinjiang Academy of Agricultural Sciences, Urumqi 830091, China
| | - You Tang
- Electrical and Information Engineering College, Jilin Agricultural Science and Technology University, Jilin City 132101, China
| |
Collapse
|
10
|
Stelmashook EV, Alexandrova OP, Genrikhs EE, Novikova SV, Salmina AB, Isaev NK. Effect of zinc and copper ions on cadmium-induced toxicity in rat cultured cortical neurons. J Trace Elem Med Biol 2022; 73:127012. [PMID: 35679765 DOI: 10.1016/j.jtemb.2022.127012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 05/18/2022] [Accepted: 05/27/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Cadmium is a highly toxic heavy metal that is capable of accumulating in the body and causing neurodegeneration. However, the effect of other trace elements on Cd2+ toxicity is currently poorly understood. The aim of this work was to study the effect of Zn2+ and Cu2+ ions on cadmium-induced death of neurons in the cerebral cortex. METHODS The work was performed on rat cortical primary cultures. The MTT test was used to determine the cytotoxicity effects. Analysis of intracellular Ca2+ concentration was assessed by the Fluo-4 AM calcium indicator that exhibit an increase in fluorescence upon binding Ca2+. MitoSOX Red (mitochondrial superoxide indicator) was used to measuring mitochondrial ROS content in live cells. RESULTS In this article, we show that the administration of CdCl2 (0.005-0.02 mM) for 48 h induced an increase in dose-dependent death rate of cultured cortical neurons. Mature neurons were more sensitive to the damaging effects of Cd2+ than immature ones. ZnCl2 (0.01-0.03 mM) significantly protected neurons from this toxic effect. In contrast to ZnCl2, CuCl2 (0.01 mM) increased cadmium neurotoxicity. Using Fluo-4 AM, measurements of intracellular calcium ions demonstrated that 24 h-exposure to Cd2+ induced intensive increase in Fluo-4 fluorescence in neurons, which was significantly reduced by zinc ions. CuCl2 increased the cadmium-induced Fluo-4 and MitoSOX Red fluorescence in neurons. The chelator of intracellular Ca2+ BAPTA significantly decreased Cd2+-induced intensive increase in Fluo-4 fluorescence in cells. CONCLUSION The data obtained by us indicate that Zn2+ and Cu2+ can affect the neurotoxicity of cadmium in different directions: Zn2+ weaken the violation of intracellular calcium homeostasis caused by cadmium, preventing cell death, while Cu2+ potentiate the increase in the level of free intracellular calcium induced by cadmium and the development of mitochondrial dysfunction with an increase in the production of free radicals in differentiated cultured neurons of the cerebral cortex, which ultimately stimulates cytotoxicity.
Collapse
Affiliation(s)
| | | | | | | | | | - Nickolay K Isaev
- Research Center of Neurology, Moscow, Russia; M.V. Lomonosov Moscow State University, Moscow, Russia.
| |
Collapse
|
11
|
Chen Z, Yang Y, Han Y, Wang X. Neuroprotective Effects and Mechanisms of Senegenin, an Effective Compound Originated From the Roots of Polygala Tenuifolia. Front Pharmacol 2022; 13:937333. [PMID: 35924058 PMCID: PMC9341472 DOI: 10.3389/fphar.2022.937333] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Senegenin is the main bioactive ingredient isolated from the dried roots of Polygala tenuifolia Willd. In recent years, senegenin has been proved to possess a variety of pharmacological activities, such as anti-oxidation, anti-inflammation, anti-apoptosis, enhancement of cognitive function. Besides, it has a good development prospect for the treatment of neurodegenerative diseases, depression, osteoporosis, cognitive dysfunction, ischemia-reperfusion injury and other diseases. However, there is no systematic literature that fully demonstrates the pharmacological effects of senegenin. In order to meet the needs of new drug research and precise medication, this review summarized the neuroprotective effects, mechanisms and gastrointestinal toxicity of senegenin based on the literatures published from the past 2 decades. In addition, an in-depth analysis of the existing problems in the current research as well as the future research directions have been conducted in order to provide a basis for the clinical application of this important plant extract.
Collapse
|
12
|
Cao W, Liu X, Huang X, Liu Z, Cao X, Gao W, Tang B. Hepatotoxicity-Related Oxidative Modifications of Thioredoxin 1/Peroxiredoxin 1 Induced by Different Cadmium-Based Quantum Dots. Anal Chem 2022; 94:3608-3616. [PMID: 35179864 DOI: 10.1021/acs.analchem.1c05181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The hepatotoxicity of cadmium-based quantum dots (Cd-QDs) has become the focus with their extensive applications in biomedicine. Previous reports have demonstrated that high oxidative stress and consequent redox imbalance play critical roles in their toxicity mechanisms. Intracellular antioxidant proteins, such as thioredoxin 1 (Trx1) and peroxiredoxin 1 (Prx1), could regulate redox homeostasis through thiol-disulfide exchange. Herein, we hypothesized that the excessive reactive oxygen species (ROS) induced by Cd-QD exposure affects the functions of Trx1 or Prx1, which further causes abnormal apoptosis of liver cells and hepatotoxicity. Thereby, three types of Cd-QDs, CdS, CdSe, and CdTe QDs, were selected for conducting an intensive study. Under the same conditions, the H2O2 level in the CdTe QD group was much higher than that of CdS or CdSe QDs, and it also corresponded to the higher hepatotoxicity. Mass spectrometry (MS) results show that excessive H2O2 leads to sulfonation modification (-SO3H) at the active sites of Trx1 (Cys32 and Cys35) and Prx1 (Cys52 and Cys173). The irreversible oxidative modifications broke their cross-linking with the apoptosis signal-regulating kinase 1 (ASK1), resulting in the release and activation of ASK1, and activation of the downstream JNK/p38 signaling finally promoted liver cell apoptosis. These results highlight the key effect of the high oxidative stress, which caused irreversible oxidative modifications of Trx1 and Prx1 in the mechanisms involved in Cd-QD-induced hepatotoxicity. This work provides a new perspective on the hepatotoxicity mechanisms of Cd-QDs and helps design safe and reliable Cd-containing nanoplatforms.
Collapse
Affiliation(s)
- Wenhua Cao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, P. R. China
| | - Xiaoqian Liu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, P. R. China
| | - Xiaoqing Huang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, P. R. China
| | - Zhenhua Liu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, P. R. China
| | - Xinyi Cao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, P. R. China
| | - Wen Gao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, P. R. China
| |
Collapse
|
13
|
Xu C, Chen S, Xu M, Chen X, Wang X, Zhang H, Dong X, Zhang R, Chen X, Gao W, Huang S, Chen L. Cadmium Impairs Autophagy Leading to Apoptosis by Ca 2+-Dependent Activation of JNK Signaling Pathway in Neuronal Cells. Neurochem Res 2021; 46:2033-2045. [PMID: 34021889 DOI: 10.1007/s11064-021-03341-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 01/04/2023]
Abstract
Autophagy, a process for self-degradation of intracellular components and dysfunctional organelles, is closely related with neurodegenerative diseases. It has been shown that cadmium (Cd) induces neurotoxicity partly by impairing autophagy. However, the underlying mechanism is not fully elucidated. In this study, we show that Cd induced expansion of autophagosomes with a concomitant abnormal expression of autophagy-related (Atg) proteins in PC12 cells and primary murine neurons. 3-MA, a classical inhibitor of autophagy, attenuated Cd-induced expansion of autophagosomes and apoptosis in the cells. Further investigation demonstrated that Cd activated JNK pathway contributing to autophagosome expansion-dependent neuronal apoptosis. This is supported by the findings that pharmacological inhibition of JNK with SP600125 or expression of dominant negative c-Jun markedly attenuated Cd-induced expansion of autophagosomes and abnormal expression of Atg proteins, as well as apoptosis in PC12 cells and/or primary neurons. Furthermore, we noticed that chelating intracellular free Ca2+ ([Ca2+]i) with BAPTA/AM profoundly blocked Cd-elicited activation of JNK pathway and consequential expansion of autophagosomes, abnormal expression of Atg proteins, and apoptosis in the neuronal cells. Similar events were also seen following prevention of [Ca2+]i elevation with EGTA or 2-APB, implying a Ca2+-dependent mechanism involved. Taken together, the results indicate that Cd impairs autophagy leading to apoptosis by Ca2+-dependent activation of JNK signaling pathway in neuronal cells. Our findings highlight that manipulation of intracellular Ca2+ level and/or JNK activity to ameliorate autophagy may be a promising intervention against Cd-induced neurotoxicity and neurodegeneration.
Collapse
Affiliation(s)
- Chong Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Sujuan Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, 453003, People's Republic of China
| | - Ming Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Xiaoling Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Xiaoxue Wang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Hai Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Xiaoqing Dong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Ruijie Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Xin Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Wei Gao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA.
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA.
| | - Long Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, 1 Wenyuan Road, Chixia District, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
14
|
Cui ZG, Ahmed K, Zaidi SF, Muhammad JS. Ins and outs of cadmium-induced carcinogenesis: Mechanism and prevention. Cancer Treat Res Commun 2021; 27:100372. [PMID: 33865114 DOI: 10.1016/j.ctarc.2021.100372] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 04/03/2021] [Accepted: 04/06/2021] [Indexed: 11/16/2022]
Abstract
Cadmium (Cd) is a heavy metal and a highly toxic pollutant that is released into the environment as a byproduct of most modern factories and industries. Cd enters our body in significant quantities from contaminated water, cigarette smoke, or food product to many detrimental health hazards. Based on causal association all the Cd-related or derived compounds have been classified as carcinogens. In this study, we present an overview of the published literature to understand the molecular mechanisms for Cd-induced carcinogenesis and its prevention. In acute Cd poisoning production of reactive oxygen species is a key factor. However, chronic Cd exposure can transform cells to become more resistant to oxidative stress. Also, as an epigenetic mechanism Cd acts indirectly on DNA repair mechanisms via alteration of reactions upstream. Those transformed cells acquire resistance to apoptosis and deregulation of calcium homeostasis. Leading to uncontrolled carcinogenic cell proliferation and inherent DNA lesions. Flavonoids commonly found in plant foods have been shown to have a protective effect against Cd-induced carcinogenicity. A wide variety of tumorigenic mechanisms involved in chronic Cd exposure and the beneficial effects of flavonoids against Cd-induced carcinogenicity necessitate further investigations.
Collapse
Affiliation(s)
- Zheng-Guo Cui
- Graduate School of Medicine, Henan Polytechnic University, Jiaozuo 454000, China; Department of Environmental Health, University of Fukui School of Medical Science, 23-3 Matsuoka Shimoaizuki, Eiheiji, Fukui 910-1193 Japan
| | - Kanwal Ahmed
- Department of Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University of Health Sciences, Jeddah, Saudi Arabia
| | - Syed Faisal Zaidi
- Department of Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University of Health Sciences, Jeddah, Saudi Arabia
| | - Jibran Sualeh Muhammad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
15
|
Combining Patulin with Cadmium Induces Enhanced Hepatotoxicity and Nephrotoxicity In Vitro and In Vivo. Toxins (Basel) 2021; 13:toxins13030221. [PMID: 33803748 PMCID: PMC8003173 DOI: 10.3390/toxins13030221] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/13/2021] [Accepted: 03/16/2021] [Indexed: 11/29/2022] Open
Abstract
Food can be contaminated by various types of contaminants such as mycotoxins and toxic heavy metals. Therefore, it is very likely that simultaneous intake of more than one type of food contaminant by consumers may take place, which provides a strong rationale for investigating the combined toxicities of these food contaminants. Patulin is one of the most common food-borne mycotoxins, whereas cadmium is a representative of toxic heavy metals found in food. The liver and kidneys are the main target organ sites for both patulin and cadmium. We hypothesized that simultaneous exposure to patulin and cadmium could produce synergistic hepatotoxicity and nephrotoxicity. Alpha mouse liver 12 (AML12) and Human embryonic kidney (HEK) 293 (HEK293) cell lines together with a mouse model were used to explore the combination effect and mechanism. The results demonstrated, for the first time, that the co-exposure of liver or renal cells to patulin and cadmium caused synergistic cytotoxicity in vitro and enhanced liver toxicity in vivo. The synergistic toxicity caused by the co-administration of patulin and cadmium was attributed to the boosted reactive oxygen species (ROS) generation. c-Jun N-terminal kinase 1 (JNK1) and p53 as downstream mediators of oxidative stress contributed to the synergistic toxicity by co-exposure of patulin and cadmium, while p53/JNK1 activation promoted the second-round ROS production through a positive feedback loop. The findings of the present study extend the toxicological knowledge about patulin and cadmium, which could be beneficial to more precisely perform risk assessments on these food contaminants.
Collapse
|
16
|
Souza SO, Lira RB, Cunha CRA, Santos BS, Fontes A, Pereira G. Methods for Intracellular Delivery of Quantum Dots. Top Curr Chem (Cham) 2021; 379:1. [PMID: 33398442 DOI: 10.1007/s41061-020-00313-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023]
Abstract
Quantum dots (QDs) have attracted considerable attention as fluorescent probes for life sciences. The advantages of using QDs in fluorescence-based studies include high brilliance, a narrow emission band allowing multicolor labeling, a chemically active surface for conjugation, and especially, high photostability. Despite these advantageous features, the size of the QDs prevents their free transport across the plasma membrane, limiting their use for specific labeling of intracellular structures. Over the years, various methods have been evaluated to overcome this issue to explore the full potential of the QDs. Thus, in this review, we focused our attention on physical and biochemical QD delivery methods-electroporation, microinjection, cell-penetrating peptides, molecular coatings, and liposomes-discussing the benefits and drawbacks of each strategy, as well as presenting recent studies in the field. We hope that this review can be a useful reference source for researches that already work or intend to work in this area. Strategies for the intracellular delivery of quantum dots discussed in this review (electroporation, microinjection, cell-penetrating peptides, molecular coatings, and liposomes).
Collapse
Affiliation(s)
- Sueden O Souza
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, CB, UFPE, Av. Prof. Moraes Rego, S/N, Recife, PE, 50670-901, Brazil
| | - Rafael B Lira
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, The Netherlands
| | - Cássia R A Cunha
- Laboratório Federal de Defesa Agropecuária em Pernambuco, Recife, Brazil
| | - Beate S Santos
- Departamento de Ciências Farmacêuticas, Universidade Federal de Pernambuco, Recife, Brazil
| | - Adriana Fontes
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, CB, UFPE, Av. Prof. Moraes Rego, S/N, Recife, PE, 50670-901, Brazil.
| | - Goreti Pereira
- Departamento de Química Fundamental, Universidade Federal de Pernambuco, CCEN, UFPE, Av. Jornalista Anibal Fernandes, S/N, Recife, 50740-560, PE, Brazil.
| |
Collapse
|
17
|
Cadmium induces apoptosis via generating reactive oxygen species to activate mitochondrial p53 pathway in primary rat osteoblasts. Toxicology 2020; 446:152611. [PMID: 33031904 DOI: 10.1016/j.tox.2020.152611] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/27/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Cadmium (Cd), a heavy metal produced by various industries, contaminates the environment and seriously damages the skeletal system of humans and animals. Recent studies have reported that Cd can affect the viability of cells, including osteoblasts, both in vivo and in vitro. However, the mechanism of Cd-induced apoptosis remains unclear. In the present study, primary rat osteoblasts were used to investigate the Cd-induced apoptotic mechanism. We found that treatment with 2 and 5 μM Cd for 12 h decreased osteoblast viability and increased apoptosis. Furthermore, Cd increased the generation of reactive oxygen species (ROS), and, thus, DNA damage measured via p-H2AX. The level of the nuclear transcription factor p53 was significantly increased, which upregulated the expression of PUMA, Noxa, Bax, and mitochondrial cytochrome c, downregulated the expression of Bcl-2, and increased the level of cleaved caspase-3. However, pretreatment with the ROS scavenger N-acetyl-l-cysteine (NAC) or the p53 transcription specific inhibitor PFT-α suppressed Cd-induced apoptosis. Our results indicate that Cd can induce apoptosis in osteoblasts by increasing the generation of ROS and activating the mitochondrial p53 signaling pathway, and this mechanism requires the transcriptional activation of p53.
Collapse
|
18
|
Roy A, Nethi SK, Suganya N, Raval M, Chatterjee S, Patra CR. Attenuation of cadmium-induced vascular toxicity by pro-angiogenic nanorods. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 115:111108. [DOI: 10.1016/j.msec.2020.111108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/07/2020] [Accepted: 05/19/2020] [Indexed: 01/03/2023]
|
19
|
Fang Y, Zhang L, Dong X, Wang H, He L, Zhong S. Downregulation of vdac2 inhibits spermatogenesis via JNK and P53 signalling in mice exposed to cadmium. Toxicol Lett 2020; 326:114-122. [PMID: 32199951 DOI: 10.1016/j.toxlet.2020.03.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 03/05/2020] [Accepted: 03/17/2020] [Indexed: 10/24/2022]
Abstract
Previous studies have reported the reproductive toxicity of cadmium (Cd); however, the effect of Cd on spermatogenesis and the underlying mechanism remain to be elucidated. In this study, mouse Leydig TM3 cells were treated with CdCl2 (0, 5, 10 and 50 μM) for 24 h to evaluate cytotoxicity, and C57BL/6 mice were treated intragastrically with 0.4 mL CdCl2 (0, 0.01, 0.05 and 0.1 g/L) for 2 months to investigate changes in spermatogenesis. The results showed that Cd aggravated apoptosis and proliferation in a dose-dependent manner, concomitant with deteriorated spermatogenesis and testosterone synthesis. For mechanism exploration, RNA-seq was used to profile alterations in gene expression in response to Cd, and the results indicated focus on P53/JNK signalling pathways and membrane proteins. We found that P53/JNK signalling pathways were activated upon Cd treatment, with the Cd-triggered downregulation of the vdac2 gene. P53/JNK pathway blockade ameliorated the Cd-induced inhibition of steroidogenic acute regulatory protein (STAR) expression and testosterone synthesis. Additionally, vdac2 knockdown in TM3 cells contributed to the phosphorylation of JNK/P53 and reduced the testosterone content. Vdac2 overexpression rescued the aforementioned Cd-induced events. Collectively, our study identified an innovative biomarker of Cd exposure in mice. The results demonstrated that vdac2 downregulation inhibits spermatogenesis via the JNK/P53 cascade. This finding may contribute to our understanding of the regulatory mechanism of Cd reproductive toxicity and provide a candidate list for sperm abnormality factors and pathways.
Collapse
Affiliation(s)
- Yu Fang
- Department of Medical Genetics, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Lang Zhang
- Department of Medical Genetics, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Xin Dong
- Department of Medical Genetics, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Hui Wang
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Li He
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China
| | - Shan Zhong
- Department of Medical Genetics, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
20
|
Tang J, Chen L, Yan D, Shen Z, Wang B, Weng S, Wu Z, Xie Z, Shao J, Yang L, Shen L. Surface Functionalization with Proanthocyanidins Provides an Anti-Oxidant Defense Mechanism That Improves the Long-Term Stability and Osteogenesis of Titanium Implants. Int J Nanomedicine 2020; 15:1643-1659. [PMID: 32210558 PMCID: PMC7073973 DOI: 10.2147/ijn.s231339] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/17/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Aseptic loosening is a major complication after total joint replacement. Reactive oxygen species generated by local tissue cells and liberated from implant surfaces have been suggested to cause implant failures. Surface modification of titanium (Ti)-based implants with proanthocyanidins (PAC) is a promising approach for the development of anti-oxidant defense mechanism to supplement the mechanical functions of Ti implants. In this study, a controlled PAC release system was fabricated on the surface of Ti substrates using the layer-by-layer (LBL) assembly. MATERIALS AND METHODS Polyethyleneimine (PEI) base layer was fabricated to enable layer-by-layer (LBL) deposition of hyaluronic acid/chitosan (HA/CS) multi-layers without or with the PAC. Surface topography and wettability of the fabricated HA/CS-PAC substrates were characterized by scanning electron microscopy (SEM), atomic force microscopy (AFM), Fourier-transform infrared spectroscopy (FTIR) and contact angle measurement. PAC release profiles were investigated using drug release assays. MC3T3-E1 pre-osteoblast cells were used to assess the osteo-inductive effects of HA/CS-PAC substrates under conditions H2O2-induced oxidative stress in vitro. A rat model of femoral intramedullary implantation evaluated the osseo-integration and osteo-inductive potential of the HA/CS-PAC coated Ti implants in vivo. RESULTS SEM, AFM, FTIR and contact angle measurements verified the successful fabrication of Ti surfaces with multi-layered HA/CS-PAC coating. Drug release assays revealed controlled and sustained release of PAC over 14 days. In vitro, cell-based assays showed high tolerability and enhanced the osteogenic potential of MC3T3-E1 cells on HA/CS-PAC substrates when under conditions of H2O2-induced oxidative stress. In vivo evaluation of femoral bone 14 days after femoral intramedullary implantation confirmed the enhanced osteo-inductive potential of the HA/CS-PAC coated Ti implants. CONCLUSION Multi-layering of HA/CS-PAC coating onto Ti-based surfaces by the LBL deposition significantly enhances implant osseo-integration and promotes osteogenesis under conditions of oxidative stress. This study provides new insights for future applications in the field of joint arthroplasty.
Collapse
Affiliation(s)
- Jiahao Tang
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Liang Chen
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Deyi Yan
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Zijian Shen
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Bingzhang Wang
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Sheji Weng
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Zongyi Wu
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Zhongjie Xie
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Jiancan Shao
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Lei Yang
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Liyan Shen
- The Second School of Medicine Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| |
Collapse
|
21
|
El-Boshy M, Refaat B, Almaimani RA, Abdelghany AH, Ahmad J, Idris S, Almasmoum H, Mahbub AA, Ghaith MM, BaSalamah MA. Vitamin D 3 and calcium cosupplementation alleviates cadmium hepatotoxicity in the rat: Enhanced antioxidative and anti-inflammatory actions by remodeling cellular calcium pathways. J Biochem Mol Toxicol 2020; 34:e22440. [PMID: 31926057 DOI: 10.1002/jbt.22440] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 12/10/2019] [Accepted: 12/17/2019] [Indexed: 12/13/2022]
Abstract
Although vitamin D (VD) and calcium (Ca) attenuate cadmium (Cd) metabolism, their combined antioxidant and anti-inflammatory actions against Cd toxicity have not been previously explored. Hence, this study measured the protective effects of VD ± Ca supplements against Cd hepatotoxicity. Forty adult male rats were distributed to: negative controls (NCs), positive controls (PCs), VD, Ca, and VD3 and Ca (VDC) groups. All groups, except NC, received CdCl2 in drinking water (44 mg/L) for 4 weeks individually or concurrently with intramuscular VD3 (600 IU/kg; three times per week) and/or oral Ca (100 mg/kg; five times per week). The PC group showed abnormal hepatic biochemical parameters and increase in cellular cytochrome C, caspase-9, and caspase-3 alongside the apoptotic/necrotic cell numbers by terminal deoxynucleotidyl transferase dUTP nick end labeling technique. The PC hepatic tissue also had substantially elevated pro-oxidants (malondialdehyde [MDA]/H2 O2 /protein carbonyls) and inflammatory cytokines (interleukin 1β [IL-1β]/IL-6/IL17A/tumor necrosis factor-α), whereas the anti-inflammatory (IL-10/IL-22) and antioxidants (glutathione [GSH]/GPx/catalase enzyme [CAT]) markers declined. Hypovitaminosis D, low hepatic tissue Ca, aberrant hepatic expression of VD-metabolizing enzymes (Cyp2R1/Cyp27a1/cyp24a1), receptor and binding protein alongside Ca-membrane (CaV 1.1/CaV 3.1), and store-operated (RyR1/ITPR1) channels, and Ca-binding proteins (CAM/CAMKIIA/S100A1/S100B) were observed in the PC group. Both monotherapies decreased serum, but not tissue Cd levels, restored the targeted hepatic VD/Ca molecules' expression. However, these effects were more prominent in the VD group than the Ca group. The VDC group, contrariwise, disclosed the greatest alleviations on serum and tissue Cd, inflammatory and oxidative markers, the VD/Ca molecules and tissue integrity. In conclusion, this report is the first to reveal boosted protection for cosupplementing VD and Ca against Cd hepatotoxicity that could be due to enhanced antioxidative, anti-inflammatory, and modulation of the Ca pathways.
Collapse
Affiliation(s)
- Mohamed El-Boshy
- Faculty of Applied Medical Sciences, Department of Laboratory Medicine, Umm Al-Qura University, Makkah, Saudi Arabia.,Faculty of Veterinary Medicine, Department of Clinical Pathology, Mansoura University, Mansoura, Egypt
| | - Bassem Refaat
- Faculty of Applied Medical Sciences, Department of Laboratory Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Riyad A Almaimani
- Faculty of Medicine, Department of Biochemistry, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abdelghany H Abdelghany
- Faculty of Applied Medical Sciences, Department of Laboratory Medicine, Umm Al-Qura University, Makkah, Saudi Arabia.,Faculty of Medicine, Department of Anatomy, Alexandria University, Alexandria, Egypt
| | - Jawwad Ahmad
- Faculty of Applied Medical Sciences, Department of Laboratory Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Shakir Idris
- Faculty of Applied Medical Sciences, Department of Laboratory Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Hussain Almasmoum
- Faculty of Applied Medical Sciences, Department of Laboratory Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Amani A Mahbub
- Faculty of Applied Medical Sciences, Department of Laboratory Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mazen M Ghaith
- Faculty of Applied Medical Sciences, Department of Laboratory Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mohammad A BaSalamah
- Faculty of Medicine, Department of Pathology, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
22
|
Lacave JM, Bilbao E, Gilliland D, Mura F, Dini L, Cajaraville MP, Orbea A. Bioaccumulation, cellular and molecular effects in adult zebrafish after exposure to cadmium sulphide nanoparticles and to ionic cadmium. CHEMOSPHERE 2020; 238:124588. [PMID: 31545210 DOI: 10.1016/j.chemosphere.2019.124588] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/06/2019] [Accepted: 08/13/2019] [Indexed: 06/10/2023]
Abstract
Few works have addressed the effects provoked by the exposure to cadmium containing nanoparticles (NPs) on adult zebrafish (Danio rerio). We studied the effects of CdS NPs (5 nm) or ionic cadmium (10 μg Cd/L) after 3 and 21 d of exposure and at 6 months post-exposure (mpe). Acute toxicity was recorded after exposure to both forms of cadmium. Significant cadmium accumulation was measured in the whole fish after both treatments and autometallography showed a higher accumulation of metal in the intestine than that in the liver. Histopathological alterations, such as inflammation in gills and vacuolization in the liver, were detected after the exposure to both cadmium forms and, in a lower extent, at 6 mpe. X-ray analysis proved the presence of CdS NPs in these organs. The hepatic transcriptome analysis revealed that gene ontology terms such as "immune response" or "actin binding" were over-represented after 21 d of exposure to ionic cadmium respect to CdS NPs treatment. Exposure to CdS NPs caused a significant effect on pathways involved in the immune response and oxidative stress, while the exposure to ionic cadmium affected significantly pathways involved in DNA damage and repair and in the energetic metabolism. Oxidative damage to liver proteins was detected after the exposure to ionic cadmium, while a stronger destabilization of the hepatocyte lysosomal membrane was recorded under exposure to CdS NPs. In summary, although ionic cadmium provoked stronger effects than CdS NPs, both cadmium forms exerted an array of lethal and sublethal effects to zebrafish.
Collapse
Affiliation(s)
- José María Lacave
- CBET Research Group, Dept. of Zoology and Animal Cell Biology, Research Centre for Experimental Marine Biology and Biotechnology PiE and Science and Technology Faculty, University of the Basque Country (UPV/EHU), Sarriena z/g, E-48940, Leioa, Basque Country, Spain
| | - Eider Bilbao
- CBET Research Group, Dept. of Zoology and Animal Cell Biology, Research Centre for Experimental Marine Biology and Biotechnology PiE and Science and Technology Faculty, University of the Basque Country (UPV/EHU), Sarriena z/g, E-48940, Leioa, Basque Country, Spain
| | - Douglas Gilliland
- European Commission, JRC Directorate F, Health, Consumers and Reference Materials, Via E. Fermi, 2749, I-21027, Ispra, VA, Italy
| | - Francesco Mura
- Dept. of Basic and Applied Sciences for Engineering and Center for Nanotechnologies Applied to Engineering, Sapienza University of Rome Via A. Scarpa 16, Rome, 00161, Italy
| | - Luciana Dini
- Dept. of Biological and Environmental Science and Technology (Di.S.Te.B.A), University of Salento & CNR, Nanotec, 73100, Lecce, Italy
| | - Miren P Cajaraville
- CBET Research Group, Dept. of Zoology and Animal Cell Biology, Research Centre for Experimental Marine Biology and Biotechnology PiE and Science and Technology Faculty, University of the Basque Country (UPV/EHU), Sarriena z/g, E-48940, Leioa, Basque Country, Spain
| | - Amaia Orbea
- CBET Research Group, Dept. of Zoology and Animal Cell Biology, Research Centre for Experimental Marine Biology and Biotechnology PiE and Science and Technology Faculty, University of the Basque Country (UPV/EHU), Sarriena z/g, E-48940, Leioa, Basque Country, Spain.
| |
Collapse
|
23
|
Attafi IM, Bakheet SA, Korashy HM. The role of NF-κB and AhR transcription factors in lead-induced lung toxicity in human lung cancer A549 cells. Toxicol Mech Methods 2019; 30:197-207. [PMID: 31682781 DOI: 10.1080/15376516.2019.1687629] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lead (Pb) is recognized as the first heavy metal of the top six toxic air pollutants threatening human health and the second hazardous substance. Pb exposure is associated with lung impairment and high incidences of lung cancer. Nuclear factor kappa B (NF-κB) and aryl hydrocarbon receptor (AhR) signaling pathways are known to be expressed and play an important role in the lung. However, the link between Pb lung toxicity and NF-κB and/or AhR pathways remains unclear. This study was established to explore the role of NF-κB and AhR modulation in Pb-induced lung toxicity in human lung cancer A549 cells. In the current study, treatment of A549 cells with Pb significantly induced cell apoptosis as evidenced by increasing a) the percentage of cells underwent apoptosis determined by flow cytometry and b) p53 mRNA level. Pb treatment induced oxidative stress by a) increasing the formation of reactive oxygen species and b) decreasing GSTA1 mRNA levels. The toxic effects of Pb on the lung was associated with significant increases in NF-κB and AhR levels which was accompanied with increases in downstream targets genes, iNOS and CYP1A1, respectively. Inhibition of NF-κB or AhR either chemically using resveratrol or genetically using small interfering RNA (siRNA) significantly rescued A549 cells from Pb-mediated lung toxicity. The results clearly indicate that Pb-mediated lung toxicities are NF-κB and AhR-dependent mechanism.
Collapse
Affiliation(s)
- Ibraheem M Attafi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Poison Control & Medical Forensic Chemistry Center, Jazan Health Affairs, Jazan, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hesham M Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, Qatar University, Doha, Qatar
| |
Collapse
|
24
|
Huang CC, Kuo CY, Yang CY, Liu JM, Hsu RJ, Lee KI, Su CC, Wu CC, Lin CT, Liu SH, Huang CF. Cadmium exposure induces pancreatic β-cell death via a Ca 2+-triggered JNK/CHOP-related apoptotic signaling pathway. Toxicology 2019; 425:152252. [PMID: 31348969 DOI: 10.1016/j.tox.2019.152252] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/07/2019] [Accepted: 07/22/2019] [Indexed: 01/01/2023]
Abstract
Cadmium (Cd) is known to be ranked the 7th hazardous substance in the Substance Priority List by Agency for Toxic Substances and Disease Registry. The experimental and epidemiological data have suggested that Cd is linked to the development of diabetes mellitus (DM). The molecular mechanism of Cd on the pancreatic β-cell cytotoxicity still remains unclear. Evidence has pointed toward that Ca2+ is an important regulator of toxic insult-induced β-cell cytotoxicity. The role of Ca2+ in the Cd-induced β-cell cytotoxicity is still unknown. In this study, we found that Cd exposure significantly inhibited insulin secretion and cell viability in the pancreatic β-cell-derived RIN-m5F cells. Cd exposure induced apoptotic events, including the increased populations of apoptotic cells and sub-G1 hypodiploid cells, and caspase-3/-7/-9 and poly (ADP-ribose) polymerase (PARP) activation, which largely depended on the activation of c-Jun N-terminal kinase (JNK) and C/EBP homologous protein (CHOP). Transfection with siRNAs for JNK and CHOP or pretreatment with specific pharmacological inhibitor of JNK (SP600125) in β-cells effectively prevented the Cd-induced insulin secretion dysfunction and apoptosis. JNK-specific siRNA dramatically suppressed Cd-induced JNK phosphorylation and CHOP protein expression, but JNK phosphorylation could not be inhibited by CHOP-specific siRNA. Furthermore, Cd exposure significantly increased the intracellular calcium ([Ca2+]i) levels. Buffering the Ca2+ response with BAPTA/AM effectively abrogated the Cd-induced [Ca2+]i elevation, insulin secretion dysfunction, apoptosis, and protein expression of JNK phosphorylation and CHOP activation. Taken together, these findings demonstrated that Cd exposure exerts β-cell death via a [Ca2+]i-dependent JNK activation-activated downstream CHOP-related apoptotic signaling pathway.
Collapse
Affiliation(s)
- Cheng-Chin Huang
- Department of Emergency, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, 427, Taiwan
| | - Chun-Ying Kuo
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County, 500, Taiwan
| | - Ching-Yao Yang
- Department of Surgery, National Taiwan University Hospital, and Department of Surgery, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Jui-Ming Liu
- Division of Urology, Department of Surgery, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, 330, Taiwan
| | - Ren-Jun Hsu
- Department of Pathology and Graduate Institute of Pathology and Parasitology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 114, Taiwan; Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, 114, Taiwan
| | - Kuan-I Lee
- Department of Emergency, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, 427, Taiwan
| | - Chin-Chuan Su
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua County, 500, Taiwan
| | - Chin-Ching Wu
- Department of Public Health, China Medical University, Taichung, 404, Taiwan
| | - Ching-Ting Lin
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 404, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.
| | - Chun-Fa Huang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 404, Taiwan; Department of Nursing, College of Medical and Health Science, Asia University, Taichung, 413, Taiwan.
| |
Collapse
|
25
|
Lee JY, Tokumoto M, Satoh M. Novel Mechanisms of Cadmium-Induced Toxicity in Renal Cells. CURRENT TOPICS IN ENVIRONMENTAL HEALTH AND PREVENTIVE MEDICINE 2019. [DOI: 10.1007/978-981-13-3630-0_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
26
|
Tucovic D, Popov Aleksandrov A, Mirkov I, Ninkov M, Kulas J, Zolotarevski L, Vukojevic V, Mutic J, Tatalovic N, Kataranovski M. Oral cadmium exposure affects skin immune reactivity in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 164:12-20. [PMID: 30092388 DOI: 10.1016/j.ecoenv.2018.07.117] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/11/2018] [Accepted: 07/28/2018] [Indexed: 06/08/2023]
Abstract
Skin can acquire cadmium (Cd) by oral route, but there is paucity of data concerning cutaneous effects of this metal. Cd acquired by oral route can affect skin wound healing, but the effect of Cd on other activities involved in skin homeostasis, including skin immunity, are not explored. Using the rat model of 30-day oral administration of Cd (5 ppm and 50 ppm) in drinking water, basic aspects of immune-relevant activity of epidermal cells were examined. Dose-dependent Cd deposition in the the skin was observed (0.035 ± 0.02 µg/g and 0.127 ± 0.04 µg/g at 5 ppm and 50 ppm, respectively, compared to 0.012 ± 0.009 µg/g at 0 ppm of Cd). This resulted in skin inflammation (oxidative stress at both Cd doses and dose-dependent structural changes in the skin and the presence/activation of innate immunity cells). At low Cd dose inflammatory response (nitric oxide and IL-1β) was observed. Other inflammatory cytokines (IL-6 and TNF) response occurred at 50 ppm, which was increased further following skin sensitization with contact allergen dinitro-chlorobenzene (DNCB). Epidermal cells exposed to both Cd doses enhanced concanavalin A (ConA)-stimulated lymphocyte production of IL-17. This study showed for the first time the effect of the metal which gained access to the skin via gut on immune reactivity of epidermal cells. Presented data might be relevant for the link between dietary Cd and the risk of skin pathologies.
Collapse
Affiliation(s)
- Dina Tucovic
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, 142 Bulevar despota Stefana, 11000 Belgrade, Serbia
| | - Aleksandra Popov Aleksandrov
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, 142 Bulevar despota Stefana, 11000 Belgrade, Serbia
| | - Ivana Mirkov
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, 142 Bulevar despota Stefana, 11000 Belgrade, Serbia
| | - Marina Ninkov
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, 142 Bulevar despota Stefana, 11000 Belgrade, Serbia
| | - Jelena Kulas
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, 142 Bulevar despota Stefana, 11000 Belgrade, Serbia
| | - Lidija Zolotarevski
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, 142 Bulevar despota Stefana, 11000 Belgrade, Serbia
| | - Vesna Vukojevic
- Innovation Center of the Faculty of Chemistry, University of Belgrade, 12-16 Studentski trg, 11000 Belgrade, Serbia
| | - Jelena Mutic
- Innovation Center of the Faculty of Chemistry, University of Belgrade, 12-16 Studentski trg, 11000 Belgrade, Serbia
| | - Nikola Tatalovic
- Department of Physiology, Institute for Biological Research "Siniša Stanković", University of Belgrade, 142 Bulevar despota Stefana, 11000 Belgrade, Serbia
| | - Milena Kataranovski
- Immunotoxicology Group, Department of Ecology, Institute for Biological Research "Sinisa Stankovic", University of Belgrade, 142 Bulevar despota Stefana, 11000 Belgrade, Serbia; Institute of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, 16 Studentski trg, 11000 Belgrade, Serbia.
| |
Collapse
|
27
|
Protective effects of Fragaria ananassa methanolic extract in a rat model of cadmium chloride-induced neurotoxicity. Biosci Rep 2018; 38:BSR20180861. [PMID: 30291211 PMCID: PMC6240722 DOI: 10.1042/bsr20180861] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 09/14/2018] [Accepted: 09/19/2018] [Indexed: 01/05/2023] Open
Abstract
Cadmium (Cd) is a common environmental toxicant that has harmful effects on plants, animals, and humans. The present study evaluated the protective effects of Fragaria ananassa methanolic extract (SME) on cadmium chloride (CdCl2)-induced neuronal toxicity in rats. Male albino rats were intraperitoneally (i.p) injected with CdCl2 (6.5 mg/kg) for 5 days with or without the SME (250 mg/kg). We measured the levels of Cd, lipid peroxidation (LPO), nitric oxide, glutathione (GSH), and oxidative enzymes such as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase, and glutathione reductase (GR) in the whole brain homogenate. Compared with the control group, the Cd-intoxicated group showed a marked increase in the brain levels of Cd, LPO, and nitric oxide and a decrease in the levels of GSH and all tested antioxidant enzymes. Compared with Cd-intoxicated rats, the rats pretreated with SME showed restoration of oxidative balance in the brain tissue. While the expression of brain SOD2, CAT, glutathione peroxidase 1, and GR was down-regulated in the Cd-treated group, the expression of these enzymes was up-regulated in rats pretreated with SME. In addition, administration of SME before CdCl2 increased the Bcl-2 expression, but significantly decreased the expression of Bax. Immunohistochemical analysis showed that compared with Cd-intoxicated rats, rats pretreated with SME showed a decrease in the protein expression of tumor necrosis factor α (TNF-α). Our findings indicate that SME protects the brain tissue from Cd-induced neuronal toxicity by improving the antioxidant system and increasing antiapoptotic and anti-inflammatory activities.
Collapse
|
28
|
Wei Y, Wang L, Wang D, Wang D, Wen C, Han B, Ouyang Z. Characterization and anti-tumor activity of a polysaccharide isolated from Dendrobium officinale grown in the Huoshan County. Chin Med 2018; 13:47. [PMID: 30214471 PMCID: PMC6131812 DOI: 10.1186/s13020-018-0205-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/03/2018] [Indexed: 12/13/2022] Open
Abstract
Background Polysaccharides are carbohydrate chains composed of linked monosaccharide units. Accumulating studies report that polysaccharides isolated from Dendrobium officinale have a variety of functions. However, the composition and anti-tumor activity of D. officinale grown in the Huoshan area are largely unknown. Methods A polysaccharide (DOPA-1) was isolated from D. officinale by hot water extraction and ethanol precipitation, followed by purification via DEAE-cellulose and Sephadex G-100 chromatography. DOPA-1 was analyzed by infrared and nuclear magnetic resonance and then characterized by periodate oxidation and Smith degradation. The anti-tumor activity of DOPA-1 was then tested in HepG-2 cells. Results Our results show that DOPA-1 is mainly comprised of mannose, glucose, and galactose at a molar ratio of 1:0.42:0.27 and has an average molecular weight of 2.29 × 105 Da. Additionally, DOPA-1 inhibited HepG-2 cell growth in a dose-dependent manner. DOPA-1-treated HepG-2 cells also had increased reactive oxygen species (ROS) levels and decreased mitochondrial membrane potential. Furthermore, apoptosis was observed in DOPA-1-treated HepG-2 cells along with Bcl-2 downregulation and Bax upregulation at the protein level. Conclusions Our findings suggest that DOPA-1 induces apoptosis in tumor cells via altered mitochondrial function, ROS production, and altered apoptosis-related protein expression. This bioactive polysaccharide could, therefore, potentially be further developed as an anti-tumor adjuvant drug.
Collapse
Affiliation(s)
- Yuan Wei
- 1School of Pharmacy, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013 Jiangsu China
| | - Linwei Wang
- 1School of Pharmacy, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013 Jiangsu China
| | - Dujun Wang
- 1School of Pharmacy, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013 Jiangsu China
| | - Dan Wang
- 1School of Pharmacy, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013 Jiangsu China
| | - Chongwei Wen
- 1School of Pharmacy, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013 Jiangsu China
| | - Bangxin Han
- 2School of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, 237012 China
| | - Zhen Ouyang
- 1School of Pharmacy, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013 Jiangsu China
| |
Collapse
|
29
|
Wang Y, Mandal AK, Son YO, Pratheeshkumar P, Wise JTF, Wang L, Zhang Z, Shi X, Chen Z. Roles of ROS, Nrf2, and autophagy in cadmium-carcinogenesis and its prevention by sulforaphane. Toxicol Appl Pharmacol 2018; 353:23-30. [PMID: 29885333 PMCID: PMC6281793 DOI: 10.1016/j.taap.2018.06.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 12/14/2022]
Abstract
Environmental and occupational exposures to cadmium increase the risk of various cancers, including lung cancer. The carcinogenic mechanism of cadmium, including its prevention remains to be investigated. Using fluorescence and electron spin resonance spin trapping, the present study shows that in immortalized lung cells (BEAS-2BR cells), exposure cadmium generated reactive oxygen species (ROS). Through ROS generation, cadmium increased the protein level of TNF-α, which activated NF-κB and its target protein COX-2, creating an inflammatory microenvironment. As measured by anchorage-independent colony formation assay, cadmium induced malignant cell transformation. Inhibition of ROS by antioxidants inhibited transformation, showing that ROS were important in the mechanism of this process. The inflammatory microenvironment created by cadmium may also contribute to the mechanism of the transformation. Using tandem fluorescence protein mCherry-GFP-LC3 construct, the present study shows that cadmium-transformed cells had a property of autophagy deficiency, resulting in accumulation of autophagosomes and increased p62. This protein upregulated Nrf2, which also upregulated p62 through positive feed-back mechanism. Constitutive Nrf2 activation increased its downstream anti-apoptotic proteins, Bcl-2 and Bcl-xl, resulting in apoptosis resistance. In untransformed BEAS-2BR cells, sulforaphane, a natural compound, increased autophagy, activated Nrf2, and decreased ROS. In cadmium-transformed BEAS-2BR cells, sulforaphane restored autophagy, decreased Nrf2, and decreased apoptosis resistance. In untransformed cells, this sulforaphane induced inducible Nrf2 to decrease ROS and possibly malignant cell transformation. In cadmium-transformed cells, it decreased constitutive Nrf2 and reduced apoptosis resistance. The dual roles of sulforaphane make this natural compound a valuable agent for prevention against cadmium-induced carcinogenesis.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, People's Republic of China; Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Ardhendu Kumar Mandal
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Young-Ok Son
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Poyil Pratheeshkumar
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - James T F Wise
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Division of Nutritional Sciences, Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536-0305,USA
| | - Lei Wang
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Zhuo Zhang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Xianglin Shi
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA.
| | - Zhimin Chen
- Department of Pulmonology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, People's Republic of China.
| |
Collapse
|
30
|
Shen J, Wang X, Zhou D, Li T, Tang L, Gong T, Su J, Liang P. Modelling cadmium-induced cardiotoxicity using human pluripotent stem cell-derived cardiomyocytes. J Cell Mol Med 2018; 22:4221-4235. [PMID: 29993192 PMCID: PMC6111808 DOI: 10.1111/jcmm.13702] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/27/2018] [Indexed: 12/29/2022] Open
Abstract
Cadmium, a highly ubiquitous toxic heavy metal, has been widely recognized as an environmental and industrial pollutant, which confers serious threats to human health. The molecular mechanisms of the cadmium-induced cardiotoxicity (CIC) have not been studied in human cardiomyocytes at the cellular level. Here we showed that human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) can recapitulate the CIC at the cellular level. The cadmium-treated hPSC-CMs exhibited cellular phenotype including reduced cell viability, increased apoptosis, cardiac sarcomeric disorganization, elevated reactive oxygen species, altered action potential profile and cardiac arrhythmias. RNA-sequencing analysis revealed a differential transcriptome profile and activated MAPK signalling pathway in cadmium-treated hPSC-CMs, and suppression of P38 MAPK but not ERK MAPK or JNK MAPK rescued CIC phenotype. We further identified that suppression of PI3K/Akt signalling pathway is sufficient to reverse the CIC phenotype, which may play an important role in CIC. Taken together, our data indicate that hPSC-CMs can serve as a suitable model for the exploration of molecular mechanisms underlying CIC and for the discovery of CIC cardioprotective drugs.
Collapse
Affiliation(s)
- Jiaxi Shen
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Xiaochen Wang
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Danni Zhou
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Tongyu Li
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Ling Tang
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Tingyu Gong
- The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China
| | - Jun Su
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Ping Liang
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
31
|
Qi J, Qian K, Tian L, Cheng Z, Wang Y. Gallium(iii)–2-benzoylpyridine-thiosemicarbazone complexes promote apoptosis through Ca2+ signaling and ROS-mediated mitochondrial pathways. NEW J CHEM 2018. [DOI: 10.1039/c8nj00697k] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ga(iii) complexes promoted apoptosis as a result of a combination of multiple apoptotic pathways.
Collapse
Affiliation(s)
- Jinxu Qi
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- China
| | - Kun Qian
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- China
| | - Liang Tian
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- China
| | - Zhen Cheng
- Stanford Cancer Institute
- Member of Academic Council
- Stanford University
- USA
| | - Yihong Wang
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- China
| |
Collapse
|
32
|
Wu X, Song Y, Li S, Liu X, Hua W, Wang K, Liu W, Li S, Zhang Y, Shao Z, Yang C. Pramlintide regulation of extracellular matrix (ECM) and apoptosis through mitochondrial-dependent pathways in human nucleus pulposus cells. Int J Immunopathol Pharmacol 2017; 31:394632017747500. [PMID: 29256292 PMCID: PMC5849218 DOI: 10.1177/0394632017747500] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Pramlintide, an approved analog of amylin, is responsible for regulating the physiology of energy homeostasis. The goals of this study were to investigate the roles of pramlintide in the regulation of cell survival and matrix metabolism, and further explore their underlying mechanisms, in human nucleus pulposus (NP) cells. NP cells were treated with different concentrations of pramlintide in normoxic or hypoxic conditions. Cell viability, LAC concentration, calcium concentration, mitochondrial membrane potential (ΔΨm), MMPs proteins, and apoptotic related proteins were detected. The results indicate that pramlintide could improve NP cell proliferation, glycolytic activity, and the ECM synthesis under hypoxia, which is evident from the increased precipitation of proteoglycans; increased expression of AGG, Col2, and SOX9 proteins; and decreased expression of MMP3, MMP9, and MMP13 proteins, which are Ca2+-dependent enzymes. And, pramlintide could facilitate the survival of NP cells through mitochondrial-mediated, Bcl-2/caspase-3-dependent apoptosis. In addition, activation of AKT-AMPK/mTOR signaling pathway is also observed by the treatment. These findings demonstrate that pramlintide may play a pivotal role in reversing intervertebral disk degeneration and may relieve the impairment of ECM metabolism and NP cells survival through mitochondrial-dependent apoptotic signaling pathway, thus offering a novel potential pharmacological treatment strategy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Cao Yang
- Cao Yang, Department of Orthopaedic Surgery,
Union Hospital, Tongji Medical College, Huazhong University of Science and
Technology, No. 1277 Jiefang Avenue, Wuhan 430022, Hubei, China.
| |
Collapse
|
33
|
Tang L, Su J, Liang P. Modeling cadmium-induced endothelial toxicity using human pluripotent stem cell-derived endothelial cells. Sci Rep 2017; 7:14811. [PMID: 29093498 PMCID: PMC5665915 DOI: 10.1038/s41598-017-13694-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/29/2017] [Indexed: 11/08/2022] Open
Abstract
Cadmium (Cd) is a harmful heavy metal that results in vascular diseases such as atherosclerosis. Prior evidence revealed that Cd induced endothelial cell (EC) death and dysfunction, supporting that ECs are a primary target of Cd-induced toxicity, and can cause severe pathologies of vascular diseases. However, the underlying mechanisms remain unclear. In this study, we investigated the mechanisms of Cd-induced endothelial toxicity in a human model system of H9 human pluripotent stem cell-derived endothelial cells (H9-ECs). We showed that H9-ECs were susceptible to CdCl2 induction, leading to detrimental changes of cell structure and significantly elevated level of apoptosis. We demonstrated that CdCl2-treated H9-ECs gave rise to a clear EC dysfunction phenotype and significantly differential transcriptomic profile. Signaling pathway analysis revealed that P38 or ERK signaling pathway is critical to cadmium-induced EC apoptosis and dysfunction, and inhibition of P38 or ERK effectively rescued CdCl2-induced endothelial toxicity in H9-ECs. Conclusively, hPSC-ECs can be a reliable model to recapitulate the EC pathological features and transcriptomic profile, which may provide a unique platform for understanding the cellular and molecular mechanisms of Cd-induced endothelial toxicity and for identifying therapeutic drugs for Cd-induced vascular diseases.
Collapse
Affiliation(s)
- Ling Tang
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University, 310003, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, 310029, Hangzhou, China
| | - Jun Su
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University, 310003, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, 310029, Hangzhou, China
| | - Ping Liang
- Key Laboratory of combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University, 310003, Hangzhou, China.
- Institute of Translational Medicine, Zhejiang University, 310029, Hangzhou, China.
| |
Collapse
|
34
|
Rehman K, Fatima F, Waheed I, Akash MSH. Prevalence of exposure of heavy metals and their impact on health consequences. J Cell Biochem 2017. [PMID: 28643849 DOI: 10.1002/jcb.26234] [Citation(s) in RCA: 670] [Impact Index Per Article: 83.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Even in the current era of growing technology, the concentration of heavy metals present in drinking water is still not within the recommended limits as set by the regulatory authorities in different countries of the world. Drinking water contaminated with heavy metals namely; arsenic, cadmium, nickel, mercury, chromium, zinc, and lead is becoming a major health concern for public and health care professionals. Occupational exposure to heavy metals is known to occur by the utilization of these metals in various industrial processes and/or contents including color pigments and alloys. However, the predominant source resulting in measurable human exposure to heavy metals is the consumption of contaminated drinking water and the resulting health issues may include cardiovascular disorders, neuronal damage, renal injuries, and risk of cancer and diabetes. The general mechanism involved in heavy metal-induced toxicity is recognized to be the production of reactive oxygen species resulting oxidative damage and health related adverse effects. Thus utilization of heavy metal-contaminated water is resulting in high morbidity and mortality rates all over the world. Thereby, feeling the need to raise the concerns about contribution of different heavy metals in various health related issues, this article has discussed the global contamination of drinking water with heavy metals to assess the health hazards associated with consumption of heavy metal-contaminated water. A relationship between exposure limits and ultimate responses produced as well as the major organs affected have been reviewed. Acute and chronic poisoning symptoms and mechanisms responsible for such toxicities have also been discussed.
Collapse
Affiliation(s)
- Kanwal Rehman
- Institute of Pharmacy, Physiology, and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | - Fiza Fatima
- Institute of Pharmacy, Physiology, and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | - Iqra Waheed
- Institute of Pharmacy, Physiology, and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | | |
Collapse
|
35
|
Wang H, Zhai N, Chen Y, Xu H, Huang K. Cadmium induces Ca 2+ mediated, calpain-1/caspase-3-dependent apoptosis in primary cultured rat proximal tubular cells. J Inorg Biochem 2017; 172:16-22. [DOI: 10.1016/j.jinorgbio.2017.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 04/06/2017] [Accepted: 04/08/2017] [Indexed: 01/15/2023]
|
36
|
Tang Y, Xie M, Jiang N, Huang F, Zhang X, Li R, Lu J, Liao S, Liu Y. Icarisid II inhibits the proliferation of human osteosarcoma cells by inducing apoptosis and cell cycle arrest. Tumour Biol 2017. [PMID: 28621234 DOI: 10.1177/1010428317705745] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Icarisid II, one of the main active components of Herba Epimedii extracts, shows potent antitumor activity in various cancer cell lines, including osteosarcoma cells. However, the anticancer mechanism of icarisid II against osteosarcoma U2OS needs further exploration. This study aims to investigate further antitumor effects of icarisid II on human osteosarcoma cells and elucidate the underlying mechanism. We cultivated human osteosarcoma USO2 cells in vitro using different concentrations of icarisid II (0-30 µM). Cell viability was detected at 24, 48, and 72 h using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide analysis. Cell cycle was tested by flow cytometry after treatment with icarisid II for 48 h. Annexin V-allophycocyanin and 7-aminoactinomycin D staining were conducted to detect cell apoptosis. Quantitative real-time polymerase chain reaction and Western blot assay were performed to measure the levels of genes and proteins related to cell cycle and apoptosis. Results showed that icarisid II significantly inhibited the proliferation and induced apoptosis of human osteosarcoma U2OS cells. The half maximal inhibitory concentration values were 14.44, 11.02, and 7.37 µM at 24, 48, and 72 h, respectively. Cell cycle was arrested in the G2/M phase in vitro. In addition, icarisid II upregulated the expression levels of P21 and CyclinB1 whereas downregulated the expression levels of CyclinD1, CDC2, and P-Cdc25C, which were related to cell cycle arrest in U2OS cells. The cell apoptotic rate increased in a dose-dependent manner after treatment with icarisid II for 48 h. Icarisid II induced apoptosis by upregulating Bax, downregulating Bcl-2, and activating apoptosis-related proteins, including cleaved caspase-3, caspase-7, caspase-9, and poly (ADP-ribose) polymerase. These data indicate that icarisid II exhibits an antiproliferation effect on human osteosarcoma cells and induces apoptosis by activating the caspase family in a time- and dose-dependent manner in vitro. Therefore, icarisid II may be used as a candidate agent for the clinical treatment of osteosarcoma in the future.
Collapse
Affiliation(s)
- Yuanyuan Tang
- 1 The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Mao Xie
- 1 The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Neng Jiang
- 2 Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
| | - Feifei Huang
- 1 The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiao Zhang
- 1 The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ruishan Li
- 1 The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jingjing Lu
- 1 The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shijie Liao
- 3 Department of Orthopedics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yun Liu
- 3 Department of Orthopedics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
37
|
Son YO, Pratheeshkumar P, Divya SP, Zhang Z, Shi X. Nuclear factor erythroid 2-related factor 2 enhances carcinogenesis by suppressing apoptosis and promoting autophagy in nickel-transformed cells. J Biol Chem 2017; 292:8315-8330. [PMID: 28330870 DOI: 10.1074/jbc.m116.773986] [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: 12/22/2016] [Revised: 03/20/2017] [Indexed: 12/13/2022] Open
Abstract
Nickel-containing compounds are widely used in industry. Nickel is a known human carcinogen that primarily affects the lungs. Proposed mechanisms of nickel-induced carcinogenesis include disruption of cellular iron homeostasis, generation of reactive oxygen species (ROS), and induction of hypoxia signaling. However, the precise molecular mechanisms of nickel-induced malignant transformation and tumor development remain unclear. This study shows that the transcription factor Nrf2 is highly expressed in lung tumor tissue and in nickel-transformed human lung bronchial epithelial BEAS-2B cells (NiT cells). Additionally, constitutively high levels of Nrf2 play a critical role in apoptosis resistance in NiT cells. Basal ROS levels were extremely low in NiT cells and were correlated with elevated expression levels of both antioxidant enzymes (e.g. catalase and superoxide dismutases) and antiapoptotic proteins (e.g. Bcl-2 and Bcl-xL). These processes are tightly controlled by Nrf2. Autophagy inhibition, induced pharmacologically or genetically, enhanced Ni2+-induced apoptosis, indicating that the induction of autophagy is the cause of apoptosis resistance in NiT cells. Using similar approaches, we show that in NiT cells the inhibition of apoptosis decreases autophagy. We have shown that Stat3, which is up-regulated by Nrf2, controls autophagy induction in NiT cells. Colony formation and tumor growth were significantly attenuated by knockdown of Nrf2 or Bcl-2. Taken together, this study demonstrates that in NiT cells constitutively high Nrf2 expression inhibits apoptosis by up-regulating antioxidant enzymes and antiapoptotic proteins to increase autophagy via Stat3 signaling. These findings indicate that the Nrf2-mediated suppression of apoptosis and promotion of autophagy contribute to nickel-induced transformation and tumorigenesis.
Collapse
Affiliation(s)
- Young-Ok Son
- Center for Research on Environmental Disease; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536-0305; National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, South Korea.
| | - Poyil Pratheeshkumar
- Center for Research on Environmental Disease; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536-0305
| | - Sasidharan Padmaja Divya
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536-0305
| | - Zhuo Zhang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536-0305
| | - Xianglin Shi
- Center for Research on Environmental Disease; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536-0305.
| |
Collapse
|
38
|
Zhang R, Zhu Y, Dong X, Liu B, Zhang N, Wang X, Liu L, Xu C, Huang S, Chen L. Celastrol Attenuates Cadmium-Induced Neuronal Apoptosis via Inhibiting Ca 2+ -CaMKII-Dependent Akt/mTOR Pathway. J Cell Physiol 2017; 232:2145-2157. [PMID: 27891586 DOI: 10.1002/jcp.25703] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 11/22/2016] [Indexed: 11/06/2022]
Abstract
Cadmium (Cd), an environmental and industrial pollutant, affects the nervous system and consequential neurodegenerative disorders. Recently, we have shown that celastrol prevents Cd-induced neuronal cell death partially by suppressing Akt/mTOR pathway. However, the underlying mechanism remains to be elucidated. Here, we show that celastrol attenuated Cd-elevated intracellular-free calcium ([Ca2+ ]i ) level and apoptosis in neuronal cells. Celastrol prevented Cd-induced neuronal apoptosis by inhibiting Akt-mediated mTOR pathway, as inhibition of Akt with Akt inhibitor X or ectopic expression of dominant negative Akt reinforced celastrol's prevention of Cd-induced phosphorylation of S6K1/4E-BP1 and cell apoptosis. Furthermore, chelating intracellular Ca2+ with BAPTA/AM or preventing [Ca2+ ]i elevation using EGTA potentiated celastrol's repression of Cd-induced [Ca2+ ]i elevation and consequential activation of Akt/mTOR pathway and cell apoptosis. Moreover, celastrol blocked Cd-elicited phosphorylation of CaMKII, and pretreatment with BAPTA/AM or EGTA enhanced celastrol's suppression of Cd-increased phosphorylation of CaMKII in neuronal cells, implying that celastrol hinders [Ca2+ ]i -mediated CaMKII phosphorylation. Inhibiting CaMKII with KN93 or silencing CaMKII attenuated Cd activation of Akt/mTOR pathway and cell apoptosis, and this was strengthened by celastrol. Taken together, these data demonstrate that celastrol attenuates Cd-induced neuronal apoptosis via inhibiting Ca2+ -CaMKII-dependent Akt/mTOR pathway. Our findings underscore that celastrol may act as a neuroprotective agent for the prevention of Cd-induced neurodegenerative disorders. J. Cell. Physiol. 232: 2145-2157, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ruijie Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, P. R. China
| | - Yu Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, P. R. China
| | - Xiaoqing Dong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, P. R. China
| | - Beibei Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, P. R. China
| | - Nana Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, P. R. China
| | - Xiaoxue Wang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, P. R. China
| | - Lei Liu
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Chong Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, P. R. China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, Louisiana.,Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Long Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, P. R. China
| |
Collapse
|
39
|
Roy RV, Pratheeshkumar P, Son YO, Wang L, Hitron JA, Divya SP, Zhang Z, Shi X. Different roles of ROS and Nrf2 in Cr(VI)-induced inflammatory responses in normal and Cr(VI)-transformed cells. Toxicol Appl Pharmacol 2016; 307:81-90. [PMID: 27470422 DOI: 10.1016/j.taap.2016.07.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/22/2016] [Accepted: 07/24/2016] [Indexed: 12/11/2022]
Abstract
Hexavalent chromium (Cr(VI)) is classified as a human carcinogen. Cr(VI) has been associated with adenocarcinomas and squamous cell carcinoma of the lung. The present study shows that acute Cr(VI) treatment in human bronchial epithelial cells (BEAS-2B) increased inflammatory responses (TNF-α, COX-2, and NF-кB/p65) and expression of Nrf2. Cr(VI)-induced generation of reactive oxygen species (ROS) are responsible for increased inflammation. Despite the fact that Nrf2 is a master regulator of response to oxidative stress, silencing of Nrf2 in the acute Cr(VI) treatment had no effect on Cr(VI)-induced inflammation. In contrast, in Cr(VI)-transformed (CrT) cells, Nrf2 is constitutively activated. Knock-down of this protein resulted in decreased inflammation, while silencing of SOD2 and CAT had no effect in the expression of these inflammatory proteins. Results obtained from the knock-down of Nrf2 in CrT cells are very different from the results obtained in the acute Cr(VI) treatment. In BEAS-2B cells, knock-down of Nrf2 had no effect in the inflammation levels, while in CrT cells a decrease in the expression of inflammation markers was observed. These results indicate that before transformation, ROS plays a critical role while Nrf2 not in Cr(VI)-induced inflammation, whereas after transformation (CrT cells), Nrf2 is constitutively activated and this protein maintains inflammation while ROS not. Constitutively high levels of Nrf2 in CrT binds to the promoter regions of COX-2 and TNF-α, leading to increased inflammation. Collectively, our results demonstrate that before cell transformation ROS are important in Cr(VI)-induced inflammation and after transformation a constitutively high level of Nrf2 is important.
Collapse
Affiliation(s)
- Ram Vinod Roy
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Poyil Pratheeshkumar
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Yong-Ok Son
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Lei Wang
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - John Andrew Hitron
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Sasidharan Padmaja Divya
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Zhuo Zhang
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Xianglin Shi
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA.
| |
Collapse
|
40
|
Zhu XQ, Li XM, Zhao YD, Ji XL, Wang YP, Fu YM, Wang HD, Lu DX, Qi RB. Effects of Senegenin against hypoxia/reoxygenation-induced injury in PC12 cells. Chin J Integr Med 2016; 22:353-61. [PMID: 26759162 DOI: 10.1007/s11655-015-2091-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To investigate the effect and the potential mechanism of Senegenin (Sen) against injury induced by hypoxia/reoxygenation (H/R) in highly differentiated PC12 cells. METHODS The cultured PC12 cells were treated with H/R in the presence or absence of Sen (60 μmol/L). Four groups were included in the experiment: control group, H/R group, H/R+Sen group and Sen group. Cell viability of each group and the level of lactate dehydrogenase (LDH) in culture medium were detected for the pharmacological effect of Sen. Hoechst 33258 staining and annexin V/propidium iodide double staining were used to analyze the apoptosis rate. Moreover, mitochondrial membrane potential (△Ψm), reactive oxygen species (ROS) and intracellular free calcium ([Ca(2+)]i) were measured by fluorescent staining and flow cytometry. Cleaved caspase-3 and activity of NADPH oxidase (NOX) were determined by colorimetric protease assay and enzyme linked immunosorbent assay, respectively. RESULTS Sen significantly elevated cell viability (P<0.05), decreased the leakage of LDH (P<0.05) and apoptosis rate (P<0.05) in H/R-injured PC12 cells. Sen maintained the value of △Ψm (P<0.05) and suppressed the activity of caspase-3 (P<0.05). Moreover, Sen reduced ROS accumulation P<0.05) and [Ca(2+)]i increment (P<0.05) by inhibiting the activity of NOX (P<0.05). CONCLUSION Sen may exert cytoprotection against H/R injury by decreasing the levels of intracellular ROS and [Ca(2+)]i, thereby suppressing the mitochondrial pathway of cellular apoptosis.
Collapse
Affiliation(s)
- Xiao-Qing Zhu
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, China
- Clifford Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 511496, China
| | - Xue-Min Li
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, China
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, 230031, China
| | - Yan-Dong Zhao
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xi-Luan Ji
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yan-Ping Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yong-Mei Fu
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Hua-Dong Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Da-Xiang Lu
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Ren-Bin Qi
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
41
|
Lee JY, Tokumoto M, Fujiwara Y, Hasegawa T, Seko Y, Shimada A, Satoh M. Accumulation of p53 via down-regulation of UBE2D family genes is a critical pathway for cadmium-induced renal toxicity. Sci Rep 2016; 6:21968. [PMID: 26912277 PMCID: PMC4766413 DOI: 10.1038/srep21968] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 02/03/2016] [Indexed: 12/19/2022] Open
Abstract
Chronic cadmium (Cd) exposure can induce renal toxicity. In Cd renal toxicity, p53 is thought to be involved. Our previous studies showed that Cd down-regulated gene expression of the UBE2D (ubiquitin-conjugating enzyme E2D) family members. Here, we aimed to define the association between UBE2D family members and p53-dependent apoptosis in human proximal tubular cells (HK-2 cells) treated with Cd. Cd increased intracellular p53 protein levels and decreased UBE2D2 and UBE2D4 gene expression via inhibition of YY1 and FOXF1 transcription factor activities. Double knockdown of UBE2D2 and UBE2D4 caused an increase in p53 protein levels, and knockdown of p53 attenuated not only Cd-induced apoptosis, but also Cd-induced apoptosis-related gene expression (BAX and PUMA). Additionally, the mice exposed to Cd for 6 months resulted in increased levels of p53 and induction of apoptosis in proximal tubular cells. These findings suggest that down-regulation of UBE2D family genes followed by accumulation of p53 in proximal tubular cells is an important mechanism for Cd-induced renal toxicity.
Collapse
Affiliation(s)
- Jin-Yong Lee
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| | - Maki Tokumoto
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| | - Yasuyuki Fujiwara
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan.,Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Tatsuya Hasegawa
- Department of Environmental Biochemistry, Mount Fuji Research Institute, 5597-1 Kenmarubi, Kamiyoshida, Fujiyoshida, Yamanashi 403-0005, Japan
| | - Yoshiyuki Seko
- Department of Environmental Biochemistry, Mount Fuji Research Institute, 5597-1 Kenmarubi, Kamiyoshida, Fujiyoshida, Yamanashi 403-0005, Japan
| | - Akinori Shimada
- Laboratory of Pathology, Department of Medical Technology, School of Life and Environmental Science, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5201, Japan
| | - Masahiko Satoh
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi 464-8650, Japan
| |
Collapse
|
42
|
Guo C, Liu S, Dong P, Zhao D, Wang C, Tao Z, Sun MZ. Akbu-LAAO exhibits potent anti-tumor activity to HepG2 cells partially through produced H2O2 via TGF-β signal pathway. Sci Rep 2015; 5:18215. [PMID: 26655928 PMCID: PMC4677388 DOI: 10.1038/srep18215] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/16/2015] [Indexed: 02/07/2023] Open
Abstract
Previously, we characterized the biological properties of Akbu-LAAO, a novel L-amino acid oxidase from Agkistrodon blomhoffii ussurensis snake venom (SV). Current work investigated its in vitro anti-tumor activity and underlying mechanism on HepG2 cells. Akbu-LAAO inhibited HepG2 growth time and dose-dependently with an IC50 of ~38.82 μg/mL. It could induce the apoptosis of HepG2 cells. Akbu-LAAO exhibited cytotoxicity by inhibiting growth and inducing apoptosis of HepG2 as it showed no effect on its cell cycle. The inhibition of Akbu-LAAO to HepG2 growth partially relied on enzymatic-released H2O2 as catalase only partially antagonized this effect. cDNA microarray results indicated TGF-β signaling pathway was linked to the cytotoxicity of Akbu-LAAO on HepG2. TGF-β pathway related molecules CYR61, p53, GDF15, TOB1, BTG2, BMP2, BMP6, SMAD9, JUN, JUNB, LOX, CCND1, CDK6, GADD45A, CDKN1A were deregulated in HepG2 following Akbu-LAAO stimulation. The presence of catalase only slightly restored the mRNA changes induced by Akbu-LAAO for differentially expressed genes. Meanwhile, LDN-193189, a TGF-β pathway inhibitor reduced Akbu-LAAO cytotoxicity on HepG2. Collectively, we reported, for the first time, SV-LAAO showed anti-tumor cell activity via TGF-β pathway. It provides new insight of SV-LAAO exhibiting anti-tumor effect via a novel signaling pathway.
Collapse
Affiliation(s)
- Chunmei Guo
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Shuqing Liu
- Department of Biochemistry, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Panpan Dong
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China.,Department of Biochemistry, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Dongting Zhao
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China.,Department of Biochemistry, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Chengyi Wang
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China.,Department of Biochemistry, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Zhiwei Tao
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China.,Department of Biochemistry, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Ming-Zhong Sun
- Department of Biotechnology, Dalian Medical University, Dalian, Liaoning, 116044, China
| |
Collapse
|
43
|
2D-DIGE and MALDI TOF/TOF MS analysis reveal that small GTPase signaling pathways may play an important role in cadmium-induced colon cell malignant transformation. Toxicol Appl Pharmacol 2015. [DOI: 10.1016/j.taap.2015.07.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
44
|
He W, Guo W, Qian Y, Zhang S, Ren D, Liu S. Synergistic hepatotoxicity by cadmium and chlorpyrifos: disordered hepatic lipid homeostasis. Mol Med Rep 2015; 12:303-8. [PMID: 25707953 DOI: 10.3892/mmr.2015.3381] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 01/23/2015] [Indexed: 11/06/2022] Open
Abstract
Due to its extensive application, chlorpyrifos (CPF) has contaminated a diverse range of environmental substrates, fruits and vegetables. A number of studies have suggested that CPF may incur adverse effects on human health, including neurotoxicity, hepatotoxicity and endocrine disruption. Additionally, cadmium (Cd) is one of the most prevalent environmental heavy metals, as a result of considerable use in a wide spectrum of industrial fields. Exposure to Cd can cause several lesions in various organs, including the liver, kidneys and lungs. CPF and Cd often co-exist in the environment, food and crops, however, their joint exposure and potential synergistic toxicity are largely neglected and unrecognized. Our previous study characterized an interaction between CPF and Cd, which may occur via bonding between Cd2+ and the nitrogen atom in the pyridine ring of CPF, or the chelation between one Cd2+ and two CPF molecules. Our previous study also identified increased hepatotoxicity induced by CPF and Cd together compared with the individual compounds. In the present study, the effects of the concomitant exposure of CPF and Cd on lipid metabolism in hepatocytes was investigated. The results demonstrated an accumulation of lipids in hepatocytes, induced by the CPF and Cd complex, which was fundamentally distinct from its parental chemicals. Notably, the molecular mechanism by which the CPF-Cd complex significantly induced hepatic lipogenesis was revealed, elevating the concentrations of sterol regulatory element-binding protein-1 and fatty acid synthase. These findings pave the way for future studies in recognizing synergistic biological effects between pollutants.
Collapse
Affiliation(s)
- Wei He
- Beijing Key Laboratory of Forestry Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Wenli Guo
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco‑Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P.R. China
| | - Yi Qian
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco‑Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P.R. China
| | - Shuping Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco‑Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P.R. China
| | - Difeng Ren
- Beijing Key Laboratory of Forestry Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco‑Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, P.R. China
| |
Collapse
|
45
|
Zhou X, Hao W, Shi H, Hou Y, Xu Q. Calcium homeostasis disruption - a bridge connecting cadmium-induced apoptosis, autophagy and tumorigenesis. Oncol Res Treat 2015; 38:311-5. [PMID: 26045029 DOI: 10.1159/000431032] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 04/30/2015] [Indexed: 11/19/2022]
Abstract
Calcium and cadmium are divalent metals and have similar chemical properties. Both can enter cells through, albeit different, channels, or through protein-dependent permeation. However, cadmium disturbs the calcium homeostasis by inhibiting calcium channels and/or related proteins. Cadmium can also alter membrane phospholipid concentrations, and so induce a calcium homeostasis disorder. The altered calcium homeostasis induced by cadmium results in cell apoptosis, autophagy or tumorigenesis. In this review, calcium homeostasis disruption is summarized as a bridge connecting cadmium-induced apoptosis, autophagy, and tumorigenesis.
Collapse
Affiliation(s)
- Xuehai Zhou
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | | | | | | | | |
Collapse
|
46
|
Zhang Z, Zheng L, Zhao Z, Shi J, Wang X, Huang J. Grape seed proanthocyanidins inhibit H2O2-induced osteoblastic MC3T3-E1 cell apoptosis via ameliorating H2O2-induced mitochondrial dysfunction. J Toxicol Sci 2015; 39:803-13. [PMID: 25242411 DOI: 10.2131/jts.39.803] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Oxidative stress represents a major cause of cellular damage and death in pathological conditions including osteoporosis, in which oxidative stress is associated with increased bone resorption and low bone mass. And grape seed proanthocyanidins are a group of polyphenolic bioflavonoids which are known to possess broad pharmacological activity and therapeutic potential, exerting a protective role against oxidant injury. The aim of our study was to investigate whether proanthocyanidins exert an anti-apoptosis effect in osteoblastic MC3T3-E1 cells, via their antioxidant activity. Firstly, we determined the anti-apoptosis effect of proanthocyanidins in osteoblastic MC3T3-E1 cells, which were subject to H2O2 treatment, then we determined the association of the antioxidant activity exerted by proanthocyanidins with their anti-apoptosis effect. Results demonstrated that proanthocyanidins inhibit H2O2-promoted apoptosis in MC3T3-E1 cells, via ameliorating the viability of MC3T3-E1 cells post H2O2 treatment and reducing the apoptotic cell numbers. And the proanthocyanidins treatment also ameliorates the H2O2-induced mitochondrial dysfunction via promoting the mitochondrial membrane potential (MMP) and respiratory chain complex IV, and reducing the mitochondrial free radical production, ROS and mitochondrial superoxide. Moreover, the proanthocyanidins inhibit H2O2-induced apoptosis signaling which is mediated by p53. This study implied a possible anti-osteoporosis effect of proanthocyanidins via their antioxidant and anti-apoptosis activity.
Collapse
Affiliation(s)
- Zhifeng Zhang
- Department of Orthopaedics, the Second Affiliated Hospital of Inner Mongolia Medical University, China
| | | | | | | | | | | |
Collapse
|
47
|
Feng L, Zhang Y, Jiang M, Mo Y, Wan R, Jia Z, Tollerud DJ, Zhang X, Zhang Q. Up-regulation of Gadd45α after exposure to metal nanoparticles: the role of hypoxia inducible factor 1α. ENVIRONMENTAL TOXICOLOGY 2015; 30:490-9. [PMID: 24277352 PMCID: PMC4033704 DOI: 10.1002/tox.21926] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 11/09/2013] [Indexed: 05/05/2023]
Abstract
The increased development and use of nanoparticles in various fields may lead to increased exposure, directly affecting human health. Our current knowledge of the health effects of metal nanoparticles such as cobalt and titanium dioxide (Nano-Co and Nano-TiO2 ) is limited but suggests that some metal nanoparticles may cause genotoxic effects including cell cycle arrest, DNA damage, and apoptosis. The growth arrest and DNA damage-inducible 45α protein (Gadd45α) has been characterized as one of the key players in the cellular responses to a variety of DNA damaging agents. The aim of this study was to investigate the alteration of Gadd45α expression in mouse embryo fibroblasts (PW) exposed to metal nanoparticles and the possible mechanisms. Non-toxic doses of Nano-Co and Nano-TiO2 were selected to treat cells. Our results showed that Nano-Co caused a dose- and time-dependent increase in Gadd45α expression, but Nano-TiO2 did not. To investigate the potential pathways involved in Nano-Co-induced Gadd45α up-regulation, we measured the expression of hypoxia inducible factor 1α (HIF-1α) in PW cells exposed to Nano-Co and Nano-TiO2 . Our results showed that exposure to Nano-Co caused HIF-1α accumulation in the nucleus. In addition, hypoxia inducible factor 1α knock-out cells [HIF-1α (-/-)] and its wild-type cells [HIF-1α (+/+)] were used. Our results demonstrated that Nano-Co caused a dose- and time-dependent increase in Gadd45α expression in wild-type HIF-1α (+/+) cells, but only a slight increase in HIF-1α (-/-) cells. Pre-treatment of PW cells with heat shock protein 90 inhibitor, 17-(Allylamino)-17-demethoxygeldanamycin (17-AAG), prior to exposure to Nano-Co significantly abolished Nano-Co-induced Gadd45α expression. These results suggest that HIF-1α accumulation may be partially involved in the increased Gadd45α expression in cells exposed to Nano-Co. These findings may have important implications for understanding the potential health effects of metal nanoparticle exposure.
Collapse
Affiliation(s)
- Lingfang Feng
- Institute of Occupational Health, Zhejiang Academy of Medical Sciences; Hangzhou, Zhejiang, P. R. of China
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky, USA
| | - Yue Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky, USA
| | - Mizu Jiang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky, USA
- Department of Gastroenterology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P. R. of China
| | - Yiqun Mo
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky, USA
| | - Rong Wan
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky, USA
- Department of Pathology, Fujiang Medical University, Fuzhou, Fujiang, P. R. of China
| | - Zhenyu Jia
- Institute of Occupational Health, Zhejiang Academy of Medical Sciences; Hangzhou, Zhejiang, P. R. of China
| | - David J. Tollerud
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky, USA
| | - Xing Zhang
- Institute of Occupational Health, Zhejiang Academy of Medical Sciences; Hangzhou, Zhejiang, P. R. of China
| | - Qunwei Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
48
|
Zou H, Liu X, Han T, Hu D, Yuan Y, Gu J, Bian J, Liu Z. Alpha-lipoic acid protects against cadmium-induced hepatotoxicity via calcium signalling and gap junctional intercellular communication in rat hepatocytes. J Toxicol Sci 2015; 40:469-77. [DOI: 10.2131/jts.40.469] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Hui Zou
- College of Veterinary Medicine, Yangzhou University, and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, China
| | - Xuezhong Liu
- College of Veterinary Medicine, Yangzhou University, and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, China
| | - Tao Han
- College of Veterinary Medicine, Yangzhou University, and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, China
| | - Di Hu
- College of Veterinary Medicine, Yangzhou University, and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, China
| |
Collapse
|
49
|
Xie G, Tian W, Wei T, Liu F. The neuroprotective effects of β-hydroxybutyrate on Aβ-injected rat hippocampus in vivo and in Aβ-treated PC-12 cells in vitro. Free Radic Res 2014; 49:139-50. [PMID: 25410532 DOI: 10.3109/10715762.2014.987274] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease is a neurodegenerative disorder associated with the deposition of the peptide amyloid-beta (Aβ) in senile plaques and cerebral vasculature. The neurotoxic mechanisms of this condition have been linked to oxidative-stress-induced apoptosis leading to widespread neuronal loss. Herein, we demonstrate the neuroprotective effects of a ketone body D-β-hydroxybutyrate (β-HB) in neural cell lines and an animal model induced by injecting Aβ into the hippocampus. Using histological examination and the TUNEL assay, we show that administration of exogenous β-HB effectively prevents Aβ deposition and neuron apoptosis in this rat model. β-HB pretreatment also relieves the oxidative stress in Aβ-induced PC-12 cells, as shown by decreased intracellular reactive oxygen species and Ca(2+) levels, activated Nrf2 and recovered superoxide dismutase and catalase activities. Consequently, the apoptotic pathway is also inhibited in these cells, with decreased levels of p53, caspase-12, caspase-9, caspase-3; a decreased Bax/Bcl-2 ratio; and decreased cytochrome c release. Taken together, our study provides a molecular basis for the neuroprotective effects of β-HB in line with the suppression of oxidative stress and the inhibition of apoptotic protein activation.
Collapse
Affiliation(s)
- G Xie
- College of Veterinary Medicine, Jilin University , Changchun, Jilin , P. R. China
| | | | | | | |
Collapse
|
50
|
Lee JY, Tokumoto M, Fujiwara Y, Satoh M. Gene expression analysis using DNA microarray in HK-2 human proximal tubular cells treated with cadmium. J Toxicol Sci 2014; 38:959-62. [PMID: 24284285 DOI: 10.2131/jts.38.959] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
We examined the alteration of gene expression in HK-2 human proximal tubular cells exposed to cadmium (Cd) using DNA microarray analysis. Cd increased the expression of 30 genes, including 7 genes coding for heat shock proteins, more than 2.0-fold and decreased the expression of 21 genes, including transcription-related genes, such as AP2B1, HOXA7, HOXA9 and TCEB2, less than 0.5-fold prior to the appearance of cytotoxicity in HK-2 cells.
Collapse
Affiliation(s)
- Jin-Yong Lee
- Laboratory of Pharmaceutical Health Sciences, School of Pharmacy, Aichi Gakuin University
| | | | | | | |
Collapse
|