1
|
Zhao Y, Zeng R, Chen P, Huang C, Xu K, Huang X, Wang X. Transcriptomic and Proteomic Insights into the Effect of Sterigmatocystin on Aspergillus flavus. J Fungi (Basel) 2023; 9:1193. [PMID: 38132793 PMCID: PMC10745003 DOI: 10.3390/jof9121193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Aspergillus flavus is an important fungus that produces aflatoxins, among which aflatoxin B1 (AFB1) is the most toxic and contaminates food and poses a high risk to human health. AFB1 interacts with another mycotoxin sterigmatocystin (STC), which is also a precursor of AFB1. Herein, we determined the effect of STC on AFB1 by evaluating A. flavus transcriptomic and proteomic profiles in the presence or absence of STC by RNA-seq and isobaric tagging, respectively. Overall, 3377 differentially expressed genes were identified by RNA-seq. These genes were mainly associated with the cellular component organisation and biosynthesis, the synthesis of valine, leucine, and isoleucine, and the synthesis of aflatoxin. Clustered genes responsible for AFB1 biosynthesis exhibited varying degrees of downregulation, and norB expression was completely suppressed in the experimental group. During proteomic analysis, 331 genes were differentially expressed in response to STC. These differentially expressed proteins were associated with cell parts and catalytic and antioxidant activities. Differentially expressed proteins predominantly participated in metabolic pathways associated with aflatoxin biosynthesis, glycolysis/gluconeogenesis, glutathione metabolism, and carbon metabolism. Notably, the upregulated and downregulated enzymes in carbohydrate and glutathione metabolisms may serve as potential gateways for inhibiting aflatoxin biosynthesis. Moreover, twelve proteins including seven downregulated ones involved in aflatoxin biosynthesis were identified; among them, AflG was the most downregulated, suggesting that it may be the key enzyme responsible for inhibiting aflatoxin synthesis. These findings provide novel insights into A. flavus control and the mechanisms regulating mycotoxin production.
Collapse
Affiliation(s)
- Yarong Zhao
- Institute of Quality Standard and Monitoring Technology for Agro-Product of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (Y.Z.); (R.Z.); (P.C.); (C.H.); (K.X.); (X.H.)
- Laboratory of Quality and Safety Risk Assessment for Agro-Product (Guangzhou), Ministry of Agriculture, Guangzhou 510640, China
| | - Rui Zeng
- Institute of Quality Standard and Monitoring Technology for Agro-Product of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (Y.Z.); (R.Z.); (P.C.); (C.H.); (K.X.); (X.H.)
- Laboratory of Quality and Safety Risk Assessment for Agro-Product (Guangzhou), Ministry of Agriculture, Guangzhou 510640, China
| | - Peirong Chen
- Institute of Quality Standard and Monitoring Technology for Agro-Product of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (Y.Z.); (R.Z.); (P.C.); (C.H.); (K.X.); (X.H.)
- Laboratory of Quality and Safety Risk Assessment for Agro-Product (Guangzhou), Ministry of Agriculture, Guangzhou 510640, China
| | - Chulan Huang
- Institute of Quality Standard and Monitoring Technology for Agro-Product of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (Y.Z.); (R.Z.); (P.C.); (C.H.); (K.X.); (X.H.)
- Laboratory of Quality and Safety Risk Assessment for Agro-Product (Guangzhou), Ministry of Agriculture, Guangzhou 510640, China
| | - Kaihang Xu
- Institute of Quality Standard and Monitoring Technology for Agro-Product of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (Y.Z.); (R.Z.); (P.C.); (C.H.); (K.X.); (X.H.)
- Laboratory of Quality and Safety Risk Assessment for Agro-Product (Guangzhou), Ministry of Agriculture, Guangzhou 510640, China
| | - Xiaomei Huang
- Institute of Quality Standard and Monitoring Technology for Agro-Product of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (Y.Z.); (R.Z.); (P.C.); (C.H.); (K.X.); (X.H.)
- Laboratory of Quality and Safety Risk Assessment for Agro-Product (Guangzhou), Ministry of Agriculture, Guangzhou 510640, China
| | - Xu Wang
- Institute of Quality Standard and Monitoring Technology for Agro-Product of Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (Y.Z.); (R.Z.); (P.C.); (C.H.); (K.X.); (X.H.)
- Laboratory of Quality and Safety Risk Assessment for Agro-Product (Guangzhou), Ministry of Agriculture, Guangzhou 510640, China
| |
Collapse
|
2
|
Zingales V, Fernández-Franzón M, Ruiz MJ. Sterigmatocystin-induced DNA damage triggers cell-cycle arrest via MAPK in human neuroblastoma cells. Toxicol Mech Methods 2021; 31:479-488. [PMID: 34039253 DOI: 10.1080/15376516.2021.1916801] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Sterigmatocystin (STE) is a common mycotoxin found in food and feed. Many studies showed that STE is genotoxic. However, up to now, the potential genotoxicity of STE on human neuronal system remains unknown. In this study, we explored the effect of STE on DNA damage and cell-cycle progression on human neuroblastoma SH-SY5Y cells exposed to various concentrations of STE (0.78, 1.56 and 3.12 µM) for 24 h. The results indicated that STE exposure induced DNA damage, as evidenced by DNA comet tails formation and increased γH2AX foci. Additionally, genotoxicity was confirmed by micronuclei (MN) analysis. Furthermore, we found that STE exposure led to cell-cycle arrest at the S and the G2/M phase. Considering the important role played by MAPK and p53 signaling pathways in cell-cycle arrest, we explored their potential involvement in STE-induced cell-cycle arrest by using specific inhibitors. The inhibition of JNK and ERK resulted to attenuate S and G2/M arrest, whereas the inhibition of p38 and p53 attenuated only STE-induced S phase arrest. In conclusion, the present study demonstrates that STE induced DNA damage and triggered MAPK and p53 pathways activation, resulting in cell-cycle arrest at the S and the G2/M phase.
Collapse
Affiliation(s)
- Veronica Zingales
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Mónica Fernández-Franzón
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Maria-José Ruiz
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| |
Collapse
|
3
|
Paving the Road Toward Exploiting the Therapeutic Effects of Ginsenosides: An Emphasis on Autophagy and Endoplasmic Reticulum Stress. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1308:137-160. [PMID: 33861443 DOI: 10.1007/978-3-030-64872-5_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Programmed cell death processes such as apoptosis and autophagy strongly contribute to the onset and progression of cancer. Along with these lines, modulation of cell death mechanisms to combat cancer cells and elimination of resistance to apoptosis is of great interest. It appears that modulation of autophagy and endoplasmic reticulum (ER) stress with specific agents would be beneficial in the treatment of several disorders. Interestingly, it has been suggested that herbal natural products may be suitable candidates for the modulation of these processes due to few side effects and significant therapeutic potential. Ginsenosides are derivatives of ginseng and exert modulatory effects on the molecular mechanisms associated with autophagy and ER stress. Ginsenosides act as smart phytochemicals that confer their effects by up-regulating ATG proteins and converting LC3-I to -II, which results in maturation of autophagosomes. Not only do ginsenosides promote autophagy but they also possess protective and therapeutic properties due to their capacity to modulate ER stress and up- and down-regulate and/or dephosphorylate UPR transducers such as IRE1, PERK, and ATF6. Thus, it would appear that ginsenosides are promising agents to potentially restore tissue malfunction and possibly eliminate cancer.
Collapse
|
4
|
Kövesi B, Kulcsár S, Ancsin Z, Zándoki E, Erdélyi M, Mézes M, Balogh K. Individual and Combined Effects of Aflatoxin B1 and Sterigmatocystin on Lipid Peroxidation and Glutathione Redox System of Common Carp Liver. Toxins (Basel) 2021; 13:toxins13020109. [PMID: 33540648 PMCID: PMC7912975 DOI: 10.3390/toxins13020109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/20/2021] [Accepted: 01/29/2021] [Indexed: 02/03/2023] Open
Abstract
The purpose of the study was to evaluate the short-term effects of aflatoxin B1 (AFB1 100 µg/kg feed) and sterigmatocystin (STC 1000 μg/kg feed) exposure individually and in combination (100 μg AFB1 + 1000 μg STC/kg feed) on the parameters of lipid peroxidation and glutathione redox system both in biochemical and gene expression levels in one-year-old common carp. Lipid peroxidation parameters were slightly affected, as significant differences were observed only in conjugated diene and triene concentrations. Reduced glutathione content decreased more markedly by STC than AFB1 or AFB1+STC, but glutathione peroxidase activity did not change. Expression of gpx4a, gpx4b, gss, and gsr genes was down-regulated due to STC compared to AFB1 or AFB1+STC, while an induction was found as effect of AFB1+STC in the case of gpx4a, but down-regulation for gpx4b as compared to AFB1. Expression of the glutathione biosynthesis regulatory gene, gss, was higher, but glutathione recycling enzyme encoding gene, gsr, was lower as an effect of AFB1+STC compared to AFB1. These results are supported by the changes in the expression of transcription factors encoding genes, nrf2, and keap1. The results revealed that individual effects of AFB1 and STC on different parameters are synergistic or antagonistic in multi-toxin treatment.
Collapse
Affiliation(s)
- Benjamin Kövesi
- Department of Nutrition, Szent István University, H-2100 Gödöllő, Hungary; (B.K.); (S.K.); (Z.A.); (M.E.); (K.B.)
| | - Szabina Kulcsár
- Department of Nutrition, Szent István University, H-2100 Gödöllő, Hungary; (B.K.); (S.K.); (Z.A.); (M.E.); (K.B.)
- Mycotoxins in the Food Chain Research Group, Hungarian Academy of Sciences, Kaposvár Campus, Szent István University, H-7400 Kaposvár, Hungary;
| | - Zsolt Ancsin
- Department of Nutrition, Szent István University, H-2100 Gödöllő, Hungary; (B.K.); (S.K.); (Z.A.); (M.E.); (K.B.)
| | - Erika Zándoki
- Mycotoxins in the Food Chain Research Group, Hungarian Academy of Sciences, Kaposvár Campus, Szent István University, H-7400 Kaposvár, Hungary;
| | - Márta Erdélyi
- Department of Nutrition, Szent István University, H-2100 Gödöllő, Hungary; (B.K.); (S.K.); (Z.A.); (M.E.); (K.B.)
| | - Miklós Mézes
- Department of Nutrition, Szent István University, H-2100 Gödöllő, Hungary; (B.K.); (S.K.); (Z.A.); (M.E.); (K.B.)
- Mycotoxins in the Food Chain Research Group, Hungarian Academy of Sciences, Kaposvár Campus, Szent István University, H-7400 Kaposvár, Hungary;
- Correspondence:
| | - Krisztián Balogh
- Department of Nutrition, Szent István University, H-2100 Gödöllő, Hungary; (B.K.); (S.K.); (Z.A.); (M.E.); (K.B.)
- Mycotoxins in the Food Chain Research Group, Hungarian Academy of Sciences, Kaposvár Campus, Szent István University, H-7400 Kaposvár, Hungary;
| |
Collapse
|
5
|
Zingales V, Fernández-Franzón M, Ruiz MJ. Sterigmatocystin: Occurrence, toxicity and molecular mechanisms of action – A review. Food Chem Toxicol 2020; 146:111802. [PMID: 33035632 DOI: 10.1016/j.fct.2020.111802] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 12/22/2022]
|
6
|
The role of mitochondria in sterigmatocystin-induced apoptosis on SH-SY5Y cells. Food Chem Toxicol 2020; 142:111493. [PMID: 32553934 DOI: 10.1016/j.fct.2020.111493] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/31/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023]
Abstract
Mitochondria are cellular organelles involved in many crucial functions, such as generation of energy (ATP) and initiation of apoptosis. The aim of the present study was to evaluate the role of mitochondria in the toxicity induced by sterigmatocystin (STE), a mycotoxin produced by fungi of the genus Aspergillus, on SH-SY5Y cells. Our results showed that STE exposure decreased cell viability in a time- and concentration-dependent manner by MTT assay and caused mitochondrial dysfunction, as highlighted by the increase of STE cytotoxicity in cells forced to rely on mitochondrial oxidative phosphorylation. Furthermore, intracellular ATP depletion and increased mitochondrial reactive oxygen species were also observed. Since mitochondria play a pivotal role in apoptosis, the induction of this process in response to STE exposure was decided to study. Our results showed an increase in apoptotic cell population by flow cytometry, further confirmed by the up-regulation of the expression levels of the pro-apoptotic genes Bax and Casp-3 and the down-regulation of the anti-apoptotic gene Bcl-2 by qPCR technique. Taken together, our results provide novel insights in the signalling pathways of the cell death process induced by STE in SH-SY5Y cells, highlighting the key role played by mitochondria in STE toxicity.
Collapse
|
7
|
Rossi F, Gallo A, Bertuzzi T. Emerging mycotoxins in the food chain. MEDITERRANEAN JOURNAL OF NUTRITION AND METABOLISM 2020. [DOI: 10.3233/mnm-190345] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Filippo Rossi
- Università Cattolica del Sacro Cuore, Facoltà di Scienze Agrarie, Alimentari e Ambientali, Dipartimento DiANA, Sezione Scienze degli Alimenti e della Nutrizione, Via Emilia Parmense 84, 29122 Piacenza, Italy
| | - Antonio Gallo
- Università Cattolica del Sacro Cuore, Facoltà di Scienze Agrarie, Alimentari e Ambientali, Dipartimento DiANA, Sezione Scienze degli Alimenti e della Nutrizione, Via Emilia Parmense 84, 29122 Piacenza, Italy
| | - Terenzio Bertuzzi
- Università Cattolica del Sacro Cuore, Facoltà di Scienze Agrarie, Alimentari e Ambientali, Dipartimento DiANA, Sezione Scienze degli Alimenti e della Nutrizione, Via Emilia Parmense 84, 29122 Piacenza, Italy
| |
Collapse
|
8
|
Yin H, Han S, Chen Y, Wang Y, Li D, Zhu Q. T-2 Toxin Induces Oxidative Stress, Apoptosis and Cytoprotective Autophagy in Chicken Hepatocytes. Toxins (Basel) 2020; 12:toxins12020090. [PMID: 32013230 PMCID: PMC7076762 DOI: 10.3390/toxins12020090] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/19/2020] [Accepted: 01/27/2020] [Indexed: 01/08/2023] Open
Abstract
T-2 toxin is type A trichothecenes mycotoxin, which produced by fusarium species in cereal grains. T-2 toxin has been shown to induce a series of toxic effects on the health of human and animal, such as immunosuppression and carcinogenesis. Previous study has proven that T-2 toxin caused hepatotoxicity in chicken, but the regulatory mechanism is unclear. In the present study, we assessed the toxicological effect of T-2 toxin on apoptosis and autophagy in hepatocytes. The total of 120 1-day-old healthy broilers were allocated randomly into four groups and reared for 21 day with complete feed containing 0 mg/kg, 0.5 mg/kg, 1 mg/kg or 2 mg/kg T-2 toxin, respectively. The results showed that the apoptosis rate and pathological changes degree hepatocytes were aggravated with the increase of T-2 toxin. At the molecular mechanism level, T-2 toxin induced mitochondria-mediated apoptosis by producing reactive oxygen species, promoting cytochrome c translocation between the mitochondria and cytoplasm, and thus promoting apoptosomes formation. Meanwhile, the expression of the autophagy-related protein, ATG5, ATG7 and Beclin-1, and the LC3-II/LC3-I ratio were increased, while p62 was downregulated, suggesting T-2 toxin caused autophagy in hepatocytes. Further experiments demonstrated that the PI3K/AKT/mTOR signal may be participated in autophagy induced by T-2 toxin in chicken hepatocytes. These data suggest a possible underlying molecular mechanism for T-2 toxin that induces apoptosis and autophagy in chicken hepatocytes.
Collapse
Affiliation(s)
| | | | | | | | | | - Qing Zhu
- Correspondence: ; Tel.: +86-028-8629-0991
| |
Collapse
|
9
|
Dubravka R, Daniela J, Andrea HT, Domagoj K, Nevenka K, Lada R, Davor Ž, Maja P, Maja ŠK. Sterigmatocystin moderately induces oxidative stress in male Wistar rats after short-term oral treatment. Mycotoxin Res 2019; 36:181-191. [DOI: 10.1007/s12550-019-00382-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/28/2019] [Accepted: 12/02/2019] [Indexed: 12/19/2022]
|
10
|
Wang C, Wang X, Liu J, Huang Z, Li C, Liu Y, Sang X, Yang L, Wang S, Su Y, Liu C, Liu Y, Wang Z. Embryonic stem cell microenvironment suppresses the malignancy of cutaneous melanoma cells by down-regulating PI3K/AKT pathway. Cancer Med 2019; 8:4265-4277. [PMID: 31173492 PMCID: PMC6675703 DOI: 10.1002/cam4.2207] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/31/2019] [Accepted: 04/12/2019] [Indexed: 12/18/2022] Open
Abstract
Malignant cancer cells engage in a dynamic reciprocity with the tumor microenvironment (TME) that promotes tumor growth, development, and resistance to therapy. Early embryonic blastocyst microenvironments can reverse the tumorigenic phenotype of malignant cancer cells via ameliorating of TME. It is potential to apply embryonic stem cell (ESC) microenvironment to suppress the malignant behaviors of cancer cells. This study aimed to investigate a better method and the mechanism of ESC microenvironment supplied by ESCs on suppressing the malignancy of cutaneous melanoma cells. Cutaneous melanoma cell line A2058 were cultured and divided into four groups: (a) A2058-only (Control); (b) A2058 and ESCs continuously co-cultured (Group One); (c) A2058 co-cultured with daily refreshed ESCs (Group two); (d) Group one with VO-Ohpic, inhibitor of PTEN (VO-Ohpic Group). The results showed that, compared to control group, A2058 cells in group one exhibited decreased cellular proliferation, migration, invasiveness and vasculogenic mimicry concomitant with an increase in cell apoptosis, accompanied by down-regulation of PI3K/AKT pathway. Besides, the above mentioned anti-tumor effects on A2058 cells were significantly enhanced in group two but statistically weakened after administration of VO-Ohpic compared to group one. We demonstrate that ESC microenvironment reduces the malignancy of A2058 by down-regulating PI3K/AKT pathway. Notably, such anti-tumor effects can be enhanced by appropriately increasing the quality and quantity of ESCs in co-culture system. Our results suggest that ESC microenvironment could be an effective and safe approach to treating cancer.
Collapse
Affiliation(s)
- Chenjie Wang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen UniversityGuangzhou 510060China
| | - Xiaoran Wang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen UniversityGuangzhou 510060China
| | - Jiahui Liu
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen UniversityGuangzhou 510060China
| | - Zheqian Huang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen UniversityGuangzhou 510060China
| | - Chaoyang Li
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen UniversityGuangzhou 510060China
| | - Ying Liu
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen UniversityGuangzhou 510060China
| | - Xuan Sang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen UniversityGuangzhou 510060China
| | - Liu Yang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen UniversityGuangzhou 510060China
| | - Shoubi Wang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen UniversityGuangzhou 510060China
| | - Yaru Su
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen UniversityGuangzhou 510060China
| | - Chengxiu Liu
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen UniversityGuangzhou 510060China
| | - Yizhi Liu
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen UniversityGuangzhou 510060China
| | - Zhichong Wang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen UniversityGuangzhou 510060China
| |
Collapse
|
11
|
Liu Y, Fan D. Ginsenoside Rg5 induces G2/M phase arrest, apoptosis and autophagy via regulating ROS-mediated MAPK pathways against human gastric cancer. Biochem Pharmacol 2019; 168:285-304. [PMID: 31301277 DOI: 10.1016/j.bcp.2019.07.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/08/2019] [Indexed: 12/20/2022]
Abstract
Ginsenoside Rg5, a rare saponin belonging to the family of protopanaxadiol ginsenosides, has been demonstrated to have potential anti-tumor effects in various cancers. However, the effect of Rg5 on human gastric cancer and the underlying molecular mechanisms remain to be elucidated. In this study, Rg5 could suppress cell proliferation by causing G2/M phase arrest. Treatment with Rg5 could induce apoptosis through the extrinsic death receptor and intrinsic mitochondrial pathways. Autophagy induction was demonstrated by the formation of autophagosomes and autophagy-related proteins. Rg5-induced cell death was inhibited by the autophagy inhibitor 3-MA and apoptosis inhibitor Z-VAD-FMK. Moreover, the suppression of apoptosis weakened Rg5-induced autophagy, while the inhibition of autophagy attenuated Rg5-induced apoptosis. Further studies revealed that Rg5 induced ROS production and activated MAPK signaling pathways. The ROS scavenger NAC markedly diminished G2/M arrest, apoptosis, autophagy and activation of MAPK pathways induced by Rg5. The p38 inhibitor SB203580 or knockdown of p38 by siRNA clearly reversed Rg5-induced apoptosis and G2/M arrest. The JNK inhibitor SP600125 or knockdown of JNK by siRNA markedly attenuated Rg5-induced G2/M arrest, apoptosis and autophagy. The inhibition of ERK inhibitor U0126 or knockdown of ERK by siRNA clearly restored Rg5-induced apoptosis and autophagy. Finally, Rg5 significantly suppressed the growth of xenograft gastric tumors with fewer side effects. Overall, the evidence suggested that Rg5 is a novel and promising strategy for the treatment of gastric cancer owing to its high efficacy, multiple mechanisms and fewer side effects.
Collapse
Affiliation(s)
- Yannan Liu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Reserch Institute, Northwest University, Taibai North Road 229, Xi'an 710069 Shaanxi, China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Reserch Institute, Northwest University, Taibai North Road 229, Xi'an 710069 Shaanxi, China.
| |
Collapse
|
12
|
Balogh K, Kövesi B, Zándoki E, Kulcsár S, Ancsin Z, Erdélyi M, Dobolyi C, Bata-Vidács I, Inotai K, Szekeres A, Mézes M, Kukolya J. Effect of Sterigmatocystin or Aflatoxin Contaminated Feed on Lipid Peroxidation and Glutathione Redox System and Expression of Glutathione Redox System Regulatory Genes in Broiler Chicken. Antioxidants (Basel) 2019; 8:E201. [PMID: 31261801 PMCID: PMC6680631 DOI: 10.3390/antiox8070201] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/17/2019] [Accepted: 06/26/2019] [Indexed: 12/02/2022] Open
Abstract
Authors studied the effect of sterigmatocystin from infected corn (STC), purified sterigmatocystin (PSTC), and aflatoxin B1 from infected corn (AFB1) on lipid peroxidation and glutathione redox parameters, including the expression of their encoding genes in a sub-chronic (14 days) trial. A total of 144 three-week-old cockerels was divided into four experimental groups (n = 36 in each). Control feed was contaminated with STC or PSTC (1590 µg STC/kg or 1570.5 µg STC/kg feed), or with AFB1 (149.1 µg AFB1/kg feed). Six birds from each group were sampled at day 1, 2, 3, 7 and 14 of mycotoxin exposure. As parameters of lipid peroxidation, conjugated dienes (CD) and trienes (CT) were measured in the liver, while malondialdehyde (MDA) concentration was determined in blood plasma, red blood cell hemolysate and liver. Reduced glutathione (GSH) concentration and glutathione peroxidase (GPx) activity were determined in the same samples, and expression of glutathione peroxidase 4 (GPX4), glutathione synthetase (GSS) and glutathione reductase (GSR) genes was measured by RT-PCR in the liver. STC, PSTC or AFB1 caused a slight, but not significant, increase in CD and CT levels; however, in the case of MDA, no increase was found in the liver. Glutathione redox system was activated in the liver by AFB1, but less markedly by STC/PSTC. PSTC and AFB1 resulted in a higher expression of GPX4, while GSS expression was down-regulated by AFB1 on day 1, but up-regulated by STC on day 2 and by both mycotoxins on day 7. However, on day 14, GSS expression was down-regulated by PSTC. Expression of GSR was low on day 1 in AFB1 and PSTC groups, but later it was up-regulated by AFB1. The observed changes regarding gene expression strengthen the hypothesis that the mild oxidative stress, caused by the applied STC doses, activates the glutathione redox system of broiler chickens.
Collapse
Affiliation(s)
- Krisztián Balogh
- Department of Nutrition, Szent István University, H-2100 Gödöllő, Hungary.
- Mycotoxins in the Food Chain" Research Group, Hungarian Academy of Sciences-Kaposvár University-Szent István University, H-7400 Kaposvár, Hungary.
| | - Benjámin Kövesi
- Department of Nutrition, Szent István University, H-2100 Gödöllő, Hungary.
| | - Erika Zándoki
- Mycotoxins in the Food Chain" Research Group, Hungarian Academy of Sciences-Kaposvár University-Szent István University, H-7400 Kaposvár, Hungary.
| | - Szabina Kulcsár
- Mycotoxins in the Food Chain" Research Group, Hungarian Academy of Sciences-Kaposvár University-Szent István University, H-7400 Kaposvár, Hungary.
| | - Zsolt Ancsin
- Department of Nutrition, Szent István University, H-2100 Gödöllő, Hungary.
| | - Márta Erdélyi
- Department of Nutrition, Szent István University, H-2100 Gödöllő, Hungary.
| | - Csaba Dobolyi
- Department of Environmental and Applied Microbiology, Agro-Environmental Research Institute, National Agricultural Research and Innovation Centre (NARIC) H-1022 Budapest, Hungary.
| | - Ildikó Bata-Vidács
- Department of Environmental and Applied Microbiology, Agro-Environmental Research Institute, National Agricultural Research and Innovation Centre (NARIC) H-1022 Budapest, Hungary.
| | - Katalin Inotai
- Department of Environmental and Applied Microbiology, Agro-Environmental Research Institute, National Agricultural Research and Innovation Centre (NARIC) H-1022 Budapest, Hungary.
| | - András Szekeres
- Department of Microbiology, Faculty of Science and Informatics, University of Szeged, H-6726 Szeged, Hungary.
| | - Miklós Mézes
- Department of Nutrition, Szent István University, H-2100 Gödöllő, Hungary.
- Mycotoxins in the Food Chain" Research Group, Hungarian Academy of Sciences-Kaposvár University-Szent István University, H-7400 Kaposvár, Hungary.
| | - József Kukolya
- Department of Environmental and Applied Microbiology, Agro-Environmental Research Institute, National Agricultural Research and Innovation Centre (NARIC) H-1022 Budapest, Hungary.
| |
Collapse
|
13
|
Kövesi B, Pelyhe C, Zándoki E, Mézes M, Balogh K. Effect of short-term sterigmatocystin exposure on lipid peroxidation and glutathione redox system and expression of glutathione redox system regulatory genes in common carp liver. Toxicon 2019; 161:50-56. [DOI: 10.1016/j.toxicon.2019.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 02/26/2019] [Accepted: 03/03/2019] [Indexed: 11/29/2022]
|
14
|
Sun Z, Li Y, Wang H, Cai M, Gao S, Liu J, Tong L, Hu Z, Wang Y, Wang K, Zhang L, Cao X, Zhang S, Shi F, Zhao J. miR-181c-5p mediates simulated microgravity-induced impaired osteoblast proliferation by promoting cell cycle arrested in the G 2 phase. J Cell Mol Med 2019; 23:3302-3316. [PMID: 30761733 PMCID: PMC6484313 DOI: 10.1111/jcmm.14220] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 01/06/2019] [Accepted: 01/23/2019] [Indexed: 12/15/2022] Open
Abstract
Impaired osteoblast proliferation plays fundamental roles in microgravity‐induced bone loss, and cell cycle imbalance may result in abnormal osteoblast proliferation. However, whether microgravity exerts an influence on the cell cycle in osteoblasts or what mechanisms may underlie such an effect remains to be fully elucidated. Herein, we confirmed that simulated microgravity inhibits osteoblast proliferation. Then, we investigated the effect of mechanical unloading on the osteoblast cell cycle and found that simulated microgravity arrested the osteoblast cell cycle in the G2 phase. In addition, our data showed that cell cycle arrest in osteoblasts from simulated microgravity was mainly because of decreased cyclin B1 expression. Furthermore, miR‐181c‐5p directly inhibited cyclin B1 protein translation by binding to a target site in the 3′UTR. Lastly, we demonstrated that inhibition of miR‐181c‐5p partially counteracted cell cycle arrest and decreased the osteoblast proliferation induced by simulated microgravity. In conclusion, our study demonstrates that simulated microgravity inhibits cell proliferation and induces cell cycle arrest in the G2 phase in primary mouse osteoblasts partially through the miR‐181c‐5p/cyclin B1 pathway. This work may provide a novel mechanism of microgravity‐induced detrimental effects on osteoblasts and offer a new avenue to further investigate bone loss induced by mechanical unloading.
Collapse
Affiliation(s)
- Zhongyang Sun
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Orthopedics, Junxie Hospital, Anhui Medical University, Nanjing, China.,The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Ying Li
- Department of Orthopedics, Junxie Hospital, Anhui Medical University, Nanjing, China
| | - Han Wang
- Department of Orthopedics, Affiliated Hospital of Air Force Aviation Medicine Research Institute, Fourth Military Medical University, Beijing, China
| | - Min Cai
- Department of Orthopedics, Junxie Hospital, Anhui Medical University, Nanjing, China.,Medical Services Section, Junxie Hospital, Anhui Medical University, Nanjing, China
| | - Shanshan Gao
- Medical Services Section, Junxie Hospital, Anhui Medical University, Nanjing, China
| | - Jing Liu
- Department of Pharmacy, Junxie Hospital, Anhui Medical University, Nanjing, China
| | - Liangcheng Tong
- Department of Orthopedics, Junxie Hospital, Anhui Medical University, Nanjing, China
| | - Zebing Hu
- The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Yixuan Wang
- The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Ke Wang
- The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Lijun Zhang
- The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Xinsheng Cao
- The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Shu Zhang
- The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Fei Shi
- The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Jianning Zhao
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
15
|
Up-regulation of miR-383-5p suppresses proliferation and enhances chemosensitivity in ovarian cancer cells by targeting TRIM27. Biomed Pharmacother 2019; 109:595-601. [DOI: 10.1016/j.biopha.2018.10.148] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 09/03/2018] [Accepted: 10/24/2018] [Indexed: 11/21/2022] Open
|
16
|
Ghufran MS, Soni P, Kanade SR. Aflatoxin-induced upregulation of protein arginine methyltransferase 5 is mediated by protein kinase C and extracellular signal-regulated kinase. Cell Biol Toxicol 2018; 35:67-80. [DOI: 10.1007/s10565-018-9439-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/27/2018] [Indexed: 12/24/2022]
|
17
|
Cui J, Wang J, Huang S, Jiang X, Li Y, Wu W, Zhang X. The G2 phase arrest induced by sterigmatocystin is dependent on hMLH1- ERK/p38-p53 pathway in human esophageal epithelium cells in vitro. Food Chem Toxicol 2018; 115:205-211. [DOI: 10.1016/j.fct.2018.03.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 03/06/2018] [Accepted: 03/09/2018] [Indexed: 01/12/2023]
|
18
|
Jiang X, Wang J, Xing L, Shen H, Lian W, Yi L, Zhang D, Yang H, Liu J, Zhang X. Sterigmatocystin-induced checkpoint adaptation depends on Chk1 in immortalized human gastric epithelial cells in vitro. Arch Toxicol 2016; 91:259-270. [PMID: 26914363 DOI: 10.1007/s00204-016-1682-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/15/2016] [Indexed: 12/29/2022]
Abstract
Sterigmatocystin (ST) is a common contaminant detected in food and animal feed that has been recognized as a possible human carcinogen. Our previous studies demonstrate that ST causes DNA damage and subsequently triggers cell cycle arrest in G2 and apoptosis in immortalized human gastric epithelial cells (GES-1). Recently, studies have shown that in certain contexts, cells with DNA damage may escape checkpoint arrest and enter mitosis without repairing the damage. The term for this process is "checkpoint adaptation," and it increases the risk of unstable genome propagation, which may contribute to carcinogenesis. Thus, we aimed to investigate whether checkpoint adaptation occurs in GES-1 cells treated with ST and explored the underlying molecular mechanisms that contribute to this phenotype. In this study, we found that ST treatment for 24 h in GES-1 cells led to an initial G2 arrest; however, a fraction of GES-1 cells became large and rounded, and the number of p-H3-positive cells increased sharply after ST treatment for 48 h. Moreover, collection of the large and rounded cells by mechanical shake-off revealed that the majority of these large cells were found in the mitotic phase of the cell cycle. Importantly, we found that these rounded cells entered mitosis despite damaged DNA and that a small subset of this cell population survived and continued to propagate. These results suggest that ST induces an initial G2 arrest that is subsequently followed by G2 phase checkpoint adaptation, which may potentially promote genomic instability and result in tumorigenesis. Furthermore, we showed that activation of Chk1 contributes to the G2 arrest in GES-1 cells that are treated with ST for 24 h and that prolonged treatment of cells with ST for 48 h led to a decrease in the total protein and phosphorylation levels of Chk1 in mitotic cells, indicating that checkpoint adaptation may be driven by inactivation of Chk1. Knockdown studies confirmed that cells entered mitosis following inactivation of Chk1. Taken together, we show that ST treatment for 24 h activates Chk1 and induces a G2 arrest in GES-1 cells. However, prolonged ST treatment for 48 h led to Chk1 inactivation in GES-1 cells, which promotes checkpoint adaptation and entry of cells into mitosis despite damaged DNA. Importantly, checkpoint adaptation in GES-1 cells treated with ST may potentially promote genomic instability and drive tumorigenesis.
Collapse
Affiliation(s)
- Xiujuan Jiang
- Laboratory of Pathology, Hebei Medical University, No. 361, Zhongshan Eastern Road, Shijiazhuang, Hebei Province, People's Republic of China.,Department of Pathology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, People's Republic of China
| | - Juan Wang
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, Hebei Province, People's Republic of China
| | - Lingxiao Xing
- Laboratory of Pathology, Hebei Medical University, No. 361, Zhongshan Eastern Road, Shijiazhuang, Hebei Province, People's Republic of China
| | - Haitao Shen
- Laboratory of Pathology, Hebei Medical University, No. 361, Zhongshan Eastern Road, Shijiazhuang, Hebei Province, People's Republic of China
| | - Weiguang Lian
- Laboratory of Pathology, Hebei Medical University, No. 361, Zhongshan Eastern Road, Shijiazhuang, Hebei Province, People's Republic of China
| | - Li Yi
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, Hebei Province, People's Republic of China
| | - Donghui Zhang
- Department of Pathology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, People's Republic of China
| | - Haiyan Yang
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, Hebei Province, People's Republic of China
| | - Jianghui Liu
- The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei Province, People's Republic of China
| | - Xianghong Zhang
- Laboratory of Pathology, Hebei Medical University, No. 361, Zhongshan Eastern Road, Shijiazhuang, Hebei Province, People's Republic of China.
| |
Collapse
|
19
|
Downregulation of TRIM27 expression inhibits the proliferation of ovarian cancer cells in vitro and in vivo. J Transl Med 2016; 96:37-48. [PMID: 26568293 DOI: 10.1038/labinvest.2015.132] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 08/22/2015] [Accepted: 08/26/2015] [Indexed: 01/24/2023] Open
Abstract
TRIM27 (tripartite motif-containing 27) was originally identified as a fusion partner with the RET (REarranged during transfection) proto-oncogene and is highly expressed in various tumor cells and tissues. However, the level of expression and function of TRIM27 in ovarian cancer remain unclear. Here we have measured the expression of TRIM27 in normal ovarian and fallopian tube epithelial cells and in ovarian serous carcinoma cells and correlated TRIM27 expression with clinical and pathological parameters. In addition, we detected the effect of TRIM27 knockdown on proliferation of ovarian cancer cells in cell culture and xenografts. The results demonstrated that TRIM27 was highly expressed in ovarian serous carcinoma cells, and TRIM27 expression was significantly correlated with metastasis and FIGO stage in ovarian serous carcinoma patients. Downregulation of TRIM27 expression suppressed the proliferation of ovarian cancer cells in cell culture and inhibited the growth of xenografts in nude mice. TRIM27 knockdown induced cell cycle arrest and apoptosis in ovarian cancer cells by upregulating the expression of p-P38 and downregulating the expression of p-AKT. Thus the present study suggests that TRIM27 could have important roles as an oncogene during the development of ovarian cancer and could serve as a diagnostic and therapeutic target.
Collapse
|
20
|
Sterigmatocystin induces G1 arrest in primary human esophageal epithelial cells but induces G2 arrest in immortalized cells: key mechanistic differences in these two models. Arch Toxicol 2014; 89:2015-25. [DOI: 10.1007/s00204-014-1362-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 09/04/2014] [Indexed: 01/03/2023]
|
21
|
Yang Q, He X, Li X, Xu W, Luo Y, Yang X, Wang Y, Li Y, Huang K. DNA damage and S phase arrest induced by Ochratoxin A in human embryonic kidney cells (HEK 293). Mutat Res 2014; 765:22-31. [PMID: 25847125 DOI: 10.1016/j.mrfmmm.2014.05.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 04/30/2014] [Accepted: 05/03/2014] [Indexed: 12/29/2022]
Abstract
Ochratoxin A (OTA) is a ubiquitous mycotoxin with potential nephrotoxic, hepatotoxic and immunotoxic effects. The mechanisms underlying the nephrotoxicity of OTA remain obscure. To investigate DNA damage and the changes of the cell cycle distribution induced by OTA, human embryonic kidney cells (HEK 293 cells) were incubated with various concentrations of OTA for 24h in vitro. The results indicated that OTA treatment led to the production of reactive oxygen species (ROS) and to a decrease of the mitochondrial membrane potential (ΔΨm). OTA-induced DNA damage in HEK 293 cells was evidenced by DNA comet tails formation and increased expression of γ-H2AX. In addition, OTA could induce cell cycle arrest at the S phase in HEK 293 cells. The expression of key cell cycle regulatory factors that were critical to the S phase, including cyclin A2, cyclin E1, and CDK2, were further detected. The expression of cyclin A2, cyclin E1, and CDK2 were significantly decreased by OTA treatment at both the mRNA and protein levels. The apoptosis of HEK 293 cells after OTA treatment was observed using Hoechst 33342 staining. The results confirmed that OTA did induce apoptosis in HEK 293 cells. In conclusion, our results provided new insights into the molecular mechanisms by which OTA might promote nephrotoxicity.
Collapse
Affiliation(s)
- Qian Yang
- Laboratory of Food Safety and Molecular Biology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, PR China
| | - Xiaoyun He
- The Supervision, Inspection & Testing Center of Genetically Modified Organisms, Ministry of Agriculture, Beijing 100083, PR China
| | - Xiaohong Li
- Laboratory of Food Safety and Molecular Biology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, PR China
| | - Wentao Xu
- Laboratory of Food Safety and Molecular Biology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, PR China.
| | - Yunbo Luo
- Laboratory of Food Safety and Molecular Biology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, PR China
| | - Xuan Yang
- The Supervision, Inspection & Testing Center of Genetically Modified Organisms, Ministry of Agriculture, Beijing 100083, PR China
| | - Yan Wang
- The Supervision, Inspection & Testing Center of Genetically Modified Organisms, Ministry of Agriculture, Beijing 100083, PR China
| | - Yingcong Li
- Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Kunlun Huang
- Laboratory of Food Safety and Molecular Biology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, PR China
| |
Collapse
|
22
|
Impairment of cell cycle progression by sterigmatocystin in human pulmonary cells in vitro. Food Chem Toxicol 2014; 66:89-95. [DOI: 10.1016/j.fct.2014.01.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 01/11/2014] [Accepted: 01/14/2014] [Indexed: 01/11/2023]
|
23
|
Cheng HC, Liu YP, Shan YS, Huang CY, Lin FC, Lin LC, Lee L, Tsai CH, Hsiao M, Lu PJ. Loss of RUNX3 increases osteopontin expression and promotes cell migration in gastric cancer. Carcinogenesis 2013; 34:2452-2459. [PMID: 23774402 DOI: 10.1093/carcin/bgt218] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Loss of RUNX3 expression is frequently observed in gastric cancer and is highly associated with lymph node metastasis and poor prognosis. However, the underlying molecular mechanisms of gastric cancer remain unknown. In this study, we found that the protein levels of RUNX3 and osteopontin (OPN) are inversely correlated in gastric cancer clinical specimens and cell lines. Furthermore, similar inverse trends between RUNX3 and OPN messenger RNA (mRNA) expression were demonstrated in six out of seven normal-tumor-paired gastric cancer clinical specimens. In addition, low RUNX3 and high OPN expression were associated with poor prognosis in gastric cancer patients. Ectopic expression of green fluorescent protein-RUNX3 reduced OPN protein and mRNA expression in the AGS and SCM-1 gastric cancer cell lines. In contrast, knockdown of RUNX3 in GES-1, a normal gastric epithelial cell line, increased OPN expression. Although three RUNX3-binding sequences have been identified in the OPN promoter region, direct binding of RUNX3 to the specific binding site, -142 to -137bp, was demonstrated by chromatin immunoprecipitation assay. The binding of RUNX3 to the OPN promoter significantly decreased OPN promoter activity. The knockdown of OPN or overexpression of RUNX3 inhibited cell migration in AGS and SCM-1 cells; however, the coexpression of RUNX3 and OPN reversed the RUNX3-reduced migration ability in AGS and SCM-1 cells. In contrast, the knockdown of both RUNX3 and OPN inhibited RUNX3-knockdown-induced migration of GES-1 cells. Together, our data demonstrated that RUNX3 is a transcriptional repressor of OPN and that loss of RUNX3 upregulates OPN, which promotes migration in gastric cancer cells.
Collapse
Affiliation(s)
- Hui-Chuan Cheng
- Institute of Clinical Medicine, National Cheng Kung University Medical College, 138 Sheng-Li Road, Tainan 704, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Scientific Opinion on the risk for public and animal health related to the presence of sterigmatocystin in food and feed. EFSA J 2013. [DOI: 10.2903/j.efsa.2013.3254] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
25
|
Zhang D, Cui Y, Shen H, Xing L, Cui J, Wang J, Zhang X. Sterigmatocystin-induced DNA damage triggers G2 arrest via an ATM/p53-related pathway in human gastric epithelium GES-1 cells in vitro. PLoS One 2013; 8:e65044. [PMID: 23705030 PMCID: PMC3660384 DOI: 10.1371/journal.pone.0065044] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 04/25/2013] [Indexed: 11/18/2022] Open
Abstract
Sterigmatocystin (ST), which is commonly detected in food and feed commodities, is a mutagenic and carcinogenic mycotoxin that has been recognized as a possible human carcinogen. Our previous study showed that ST can induce G2 phase arrest in GES-1 cells in vitro and that the MAPK and PI3K signaling pathways are involved in the ST-induced G2 arrest. It is now widely accepted that DNA damage plays a critical role in the regulation of cell cycle arrest and apoptosis. In response to DNA damage, a complex signaling network is activated in eukaryotic cells to trigger cell cycle arrest and facilitate DNA repair. To further explore the molecular mechanism through which ST induces G2 arrest, the current study was designed to precisely dissect the role of DNA damage and the DNA damage sensor ataxia telangiectasia-mutated (ATM)/p53-dependent pathway in the ST-induced G2 arrest in GES-1 cells. Using the comet assay, we determined that ST induces DNA damage, as evidenced by the formation of DNA comet tails, in GES-1 cells. We also found that ST induces the activation of ATM and its downstream molecules, Chk2 and p53, in GES-1 cells. The ATM pharmacological inhibitor caffeine was found to effectively inhibit the activation of the ATM-dependent pathways and to rescue the ST-induced G2 arrest in GES-1 cells, which indicating its ATM-dependent characteristic. Moreover, the silencing of the p53 expression with siRNA effectively attenuated the ST-induced G2 arrest in GES-1 cells. We also found that ST induces apoptosis in GES-1 cells. Thus, our results show that the ST-induced DNA damage activates the ATM/53-dependent signaling pathway, which contributes to the induction of G2 arrest in GES-1 cells.
Collapse
Affiliation(s)
- Donghui Zhang
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Yu Cui
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Haitao Shen
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Lingxiao Xing
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Jinfeng Cui
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Juan Wang
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Xianghong Zhang
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China
- Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
- * E-mail:
| |
Collapse
|
26
|
Wang J, Huang S, Xing L, Shen H, Yan X, Wang J, Zhang X. Role of hMLH1 in sterigmatocystin-induced G2 phase arrest in human esophageal epithelial Het-1A cells in vitro. Toxicol Lett 2013; 217:226-34. [DOI: 10.1016/j.toxlet.2012.12.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Revised: 12/19/2012] [Accepted: 12/27/2012] [Indexed: 12/12/2022]
|
27
|
Lin FC, Liu YP, Lai CH, Shan YS, Cheng HC, Hsu PI, Lee CH, Lee YC, Wang HY, Wang CH, Cheng JQ, Hsiao M, Lu PJ. RUNX3-mediated transcriptional inhibition of Akt suppresses tumorigenesis of human gastric cancer cells. Oncogene 2012; 31:4302-4316. [PMID: 22231444 DOI: 10.1038/onc.2011.596] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 11/01/2011] [Accepted: 11/18/2011] [Indexed: 12/14/2022]
Abstract
Activation of Akt signaling pathway has been suggested involving in chemoresistance, metastasis and tumorigenesis of gastric cancer. However, the mechanism of Akt regulation in gastric cancer is not fully understood. RUNX3, which was first identified as a transcription factor, suppresses gastric tumorigenesis through regulating expression of target genes. Here, we found that restoration of RUNX3 significantly downregulates the protein and mRNA expression of Akt1 in gastric cancer cell lines, AGS and SNU-1. Knockdown of RUNX3 upregulates protein and mRNA expression of Akt1 in normal gastric epithelial cell line, GES-1. The negative correlation of RUNX3 and Akt expression and downstream β-catenin/cyclin D1 effectors was further confirmed in AGS and GES-1 cell lines, as well as clinical specimens of gastric cancer. We identified two RUNX3-binding sites in Akt1 promoter and the binding of RUNX3 on Akt1 promoter significantly inhibits Akt1 expression. The RUNX3-mediated inhibition of Akt1 caused β-catenin protein degradation and then cyclin D1 downregulation. Restoration of cyclin D1 reverses cell growth inhibition and G1 phase arrest induced by RUNX3 in gastric cancer cells. Our results show that loss of RUNX3 expression can enhance the Akt1-mediated signaling pathway and promote the tumorigenesis process in human gastric cancer.
Collapse
Affiliation(s)
- F-C Lin
- Institute of Clinical Medicine, National Cheng Kung University, Medical College, Department of Radiation Oncology, National Cheng Kung University Hospital, Tainan, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|