1
|
Sheikh KA, Amjad M, Irfan MT, Anjum S, Majeed T, Riaz MU, Jassim AY, Sharif EAM, Ibrahim WN. Exploring TGF-β Signaling in Cancer Progression: Prospects and Therapeutic Strategies. Onco Targets Ther 2025; 18:233-262. [PMID: 39989503 PMCID: PMC11846535 DOI: 10.2147/ott.s493643] [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: 08/29/2024] [Accepted: 11/19/2024] [Indexed: 02/25/2025] Open
Abstract
Cancer persists as a ubiquitous global challenge despite the remarkable advances. It is caused by uncontrolled cell growth and metastasis. The Transforming Growth Factor-beta (TGF-β) signaling pathway is considered a primary regulator of various normal physiological processes in the human body. Recently, factors determining the nature of TGF-β response have received attention, specifically its signaling pathway which can be an attractive therapeutic target for various cancer treatments. The TGF-β receptor is activated by its ligands and undergoes transduction of signals via canonical (SMAD dependent) or non-canonical (SMAD independent) signaling pathways regulating several cellular functions. Furthermore, the cross talk of the TGF-β signaling pathway cross with other signaling pathways has shown the controlled regulation of cellular functions. This review highlights the cross talk between various major signaling pathways and TGF-β. These signaling pathways include Wnt, NF-κB, PI3K/Akt, and Hedgehog (Hh). TGF-β signaling pathway has a dual role at different stages. It can suppress tumor formation at early stages and promote progression at advanced stages. This complex behaviour of TGF-β has made it a promising target for therapeutic interventions. Moreover, many strategies have been designed to control TGF-β signaling pathways at different levels, inhibiting tumor-promoting while enhancing tumor-suppressive effects, each with unique molecular mechanisms and clinical implications. This review also discusses various therapeutic inhibitors including ligand traps, small molecule inhibitors (SMIs), monoclonal antibodies (mAbs), and antisense oligonucleotides which target specific components of TGF-β signaling pathway to inhibit TGF-β signaling and are studied in both preclinical and clinical trials for different types of cancer. The review also highlights the prospect of TGF-β signaling in normal physiology and in the case of dysregulation, TGF-β inhibitors, and different therapeutic effects in cancer therapy along with the perspective of combinational therapies to treat cancer.
Collapse
Affiliation(s)
- Khansa Ali Sheikh
- Department of Biotechnology, Kinnaird College for Women, Lahore, Pakistan
| | - Momna Amjad
- Department of Biotechnology, Kinnaird College for Women, Lahore, Pakistan
| | | | - Sumaira Anjum
- Department of Biotechnology, Kinnaird College for Women, Lahore, Pakistan
| | - Tanveer Majeed
- Department of Biotechnology, Kinnaird College for Women, Lahore, Pakistan
| | - Muhammad Usman Riaz
- School of Computer Science, University College Dublin, Belfield, Dublin 4, Ireland
| | | | - Elham Abdullatif M Sharif
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Wisam Nabeel Ibrahim
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
2
|
An H, Li Y, Li Y, Gong S, Zhu Y, Li X, Zhou S, Wu Y. Advances in Metabolism and Metabolic Toxicology of Quinoxaline 1,4-Di-N-oxides. Chem Res Toxicol 2024; 37:528-539. [PMID: 38507288 DOI: 10.1021/acs.chemrestox.4c00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Quinoxaline 1,4-di-N-Oxides (QdNOs) have been used as synthetic antimicrobial agents in animal husbandry and aquaculture. The metabolism and potential toxicity have been also concerns in recently years. The metabolism investigations showed that there were 8 metabolites of Carbadox (CBX), 34 metabolites of Cyadox (CYA), 33 metabolites of Mequindox (MEQ), 35 metabolites of Olaquindox (OLA), and 56 metabolites of Quinocetone (QCT) in different animals. Among them, Cb3 and Cb8, M6, and O9 are metabolic residual markers of CBX, MEQ and OLA, which are associated with N → O reduction. Toxicity studies revealed that QdNOs exhibited severe tumorigenicity, cytotoxicity, and adrenal toxicity. Metabolic toxicology showed that toxicity of QdNOs metabolites might be related to the N → O group reduction, and some metabolites exhibited higher toxic effects than the precursor, which could provide guidance for further research on the metabolic toxicology of QdNOs and provide a wealth of information for food safety evaluation.
Collapse
Affiliation(s)
- Haoxian An
- College of Life Science, Yantai University, Yantai 264005, People's Republic of China
| | - Yonggang Li
- Hubei Provincial Key Laboratory for Applied Toxicology, Hubei Provincial Center for Disease Control and Prevention, Wuhan 430079, People's Republic of China
| | - Yanshen Li
- College of Life Science, Yantai University, Yantai 264005, People's Republic of China
| | - Shanmin Gong
- College of Life Science, Yantai University, Yantai 264005, People's Republic of China
| | - Ya'ning Zhu
- College of Life Science, Yantai University, Yantai 264005, People's Republic of China
| | - Xinru Li
- College of Life Science, Yantai University, Yantai 264005, People's Republic of China
| | - Shuang Zhou
- NHC Key Laboratory of Food Safety Risk Assessment, Chinese Academy of Medical Science Research Unit (2019RU014), China National Center for Food Safety Risk Assessment, Beijing 100017, People's Republic of China
| | - Yongning Wu
- NHC Key Laboratory of Food Safety Risk Assessment, Chinese Academy of Medical Science Research Unit (2019RU014), China National Center for Food Safety Risk Assessment, Beijing 100017, People's Republic of China
| |
Collapse
|
3
|
Sulaiman AA, Al-Ansari DE, Ali R, Aouida M, Ramotar D. Mft1, identified from a genome-wide screen of the yeast haploid mutants, mediates cell cycle arrest to counteract quinoxaline-induced toxicity. Front Genet 2024; 14:1296383. [PMID: 38283148 PMCID: PMC10811161 DOI: 10.3389/fgene.2023.1296383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/31/2023] [Indexed: 01/30/2024] Open
Abstract
Quinoxaline is a heterocyclic compound with a two-membered ring structure that undergoes redox cycling to produce toxic free radicals. It has antiviral, antibacterial, antifungal, and antitumor activities. However, the biological functions that are involved in mounting a response against the toxic effects of quinoxaline have not been investigated. Herein, we performed a genome-wide screen using the yeast haploid mutant collection and reported the identification of 12 mutants that displayed varying sensitivity towards quinoxaline. No mutant was recovered that showed resistance to quinoxaline. The quinoxaline-sensitive mutants were deleted for genes that encode cell cycle function, as well as genes that belong to other physiological pathways such as the vacuolar detoxification process. Three of the highly sensitive gene-deletion mutants lack the DDC1, DUN1, and MFT1 genes. While Ddc1 and Dun1 are known to perform roles in the cell cycle arrest pathway, the role of Mft1 remains unclear. We show that the mft1Δ mutant is as sensitive to quinoxaline as the ddc1Δ mutant. However, the double mutant ddc1Δ mft1Δ lacking the DDC1 and MFT1 genes, is extremely sensitive to quinoxaline, as compared to the ddc1Δ and mft1Δ single mutants. We further show that the mft1Δ mutant is unable to arrest in the G2/M phase in response to the drug. We conclude that Mft1 performs a unique function independent of Ddc1 in the cell cycle arrest pathway in response to quinoxaline exposure. This is the first demonstration that quinoxaline exerts its toxic effect likely by inducing oxidative DNA damage causing cell cycle arrest. We suggest that clinical applications of quinoxaline and its derivatives should entail targeting cancer cells with defective cell cycle arrest.
Collapse
Affiliation(s)
- Abdallah Alhaj Sulaiman
- Qatar Foundation, Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Dana E. Al-Ansari
- Qatar Foundation, Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Reem Ali
- Qatar Foundation, Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Mustapha Aouida
- Qatar Foundation, Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Dindial Ramotar
- Qatar Foundation, Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
4
|
Buravchenko GI, Shchekotikhin AE. Quinoxaline 1,4-Dioxides: Advances in Chemistry and Chemotherapeutic Drug Development. Pharmaceuticals (Basel) 2023; 16:1174. [PMID: 37631089 PMCID: PMC10459860 DOI: 10.3390/ph16081174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/08/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
N-Oxides of heterocyclic compounds are the focus of medical chemistry due to their diverse biological properties. The high reactivity and tendency to undergo various rearrangements have piqued the interest of synthetic chemists in heterocycles with N-oxide fragments. Quinoxaline 1,4-dioxides are an example of an important class of heterocyclic N-oxides, whose wide range of biological activity determines the prospects of their practical use in the development of drugs of various pharmaceutical groups. Derivatives from this series have found application in the clinic as antibacterial drugs and are used in agriculture. Quinoxaline 1,4-dioxides present a promising class for the development of new drugs targeting bacterial infections, oncological diseases, malaria, trypanosomiasis, leishmaniasis, and amoebiasis. The review considers the most important methods for the synthesis and key directions in the chemical modification of quinoxaline 1,4-dioxide derivatives, analyzes their biological properties, and evaluates the prospects for the practical application of the most interesting compounds.
Collapse
|
5
|
Quercetin Attenuates Quinocetone-Induced Cell Apoptosis In Vitro by Activating the P38/Nrf2/HO-1 Pathway and Inhibiting the ROS/Mitochondrial Apoptotic Pathway. Antioxidants (Basel) 2022; 11:antiox11081498. [PMID: 36009217 PMCID: PMC9405464 DOI: 10.3390/antiox11081498] [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: 06/22/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 01/25/2023] Open
Abstract
Quinocetone (QCT), a member of the quinoxaline 1,4-di-N-oxides (QdNOs) family, can cause genotoxicity and hepatotoxicity, however, the precise molecular mechanisms of QCT are unclear. This present study investigated the protective effect of quercetin on QCT-induced cytotoxicity and the underlying molecular mechanisms in human L02 and HepG2 cells. The results showed that quercetin treatment (at 7.5–30 μM) significantly improved QCT-induced cytotoxicity and oxidative damage in human L02 and HepG2 cells. Meanwhile, quercetin treatment at 30 μM significantly inhibited QCT-induced loss of mitochondrial membrane potential, an increase in the expression of the CytC protein and the Bax/Bcl-2 ratio, and an increase in caspases-9 and -3 activity, and finally improved cell apoptosis. Quercetin pretreatment promoted the expression of the phosphorylation of p38, Nrf2, and HO-1 proteins. Pharmacological inhibition of p38 significantly inhibited quercetin-mediated activation of the Nrf2/HO-1 pathway. Consistently, pharmacological inhibitions of the Nrf2 or p38 pathways both promoted QCT-induced cytotoxicity and partly abolished the protective effects of quercetin. In conclusion, for the first time, our results reveal that quercetin could improve QCT-induced cytotoxicity and apoptosis by activating the p38/Nrf2/HO-1 pathway and inhibiting the ROS/mitochondrial apoptotic pathway. Our study highlights that quercetin may be a promising candidate for preventing QdNOs-induced cytotoxicity in humans or animals.
Collapse
|
6
|
Li YL, Zhu XM, Chen NF, Chen ST, Yang Y, Liang H, Chen ZF. Anticancer activity of ruthenium(II) plumbagin complexes with polypyridyl as ancillary ligands via inhibiting energy metabolism and GADD45A-mediated cell cycle arrest. Eur J Med Chem 2022; 236:114312. [PMID: 35421660 DOI: 10.1016/j.ejmech.2022.114312] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/10/2022] [Accepted: 03/19/2022] [Indexed: 12/31/2022]
Abstract
To study the antitumor activity and action mechanism of Ru(II) polypyridyl plumbagin (PLN) complexes, four complexes [Ru(PLN)(DMSO)2]Cl (Ru1), [Ru(bpy)2(PLN)](PF6) (bpy is bipyridine) (Ru2), [Ru(phen)2(PLN)](PF6) (phen is 1,10-phenanthroline) (Ru3), and [Ru(DIP)2(PLN)](PF6) (DIP is 4,7-diphenyl-1,10-phenanthroline) (Ru4) were obtained and fully characterized. Lipophilicity, cellular uptake and cytotoxicity of these Ru(II) complexes are in the order of: Ru1<Ru2<Ru3<Ru4. The ancillary polypyridyl ligands affected the bioactivity and action mechanisms of these Ru(II) complexes. Ru3 and Ru4 inhibited energy metabolism by severely impairing mitochondrial respiration and glycolysis processes. Moreover, Ru3 and Ru4 induced DNA damage and the increased expression of GADD45A, which led to cell cycle arrest in G0/G1 phase in MGC-803 cells, while the inactivation of GADD45A attenuated these effects; however, Ru3 or Ru4-induced GADD45A did not affect cell apoptosis. Further studies revealed that Ru3 and Ru4 induced ROS-dependent and caspase-dependent apoptotic cell death by mitochondrial dysfunction, and Ru4 displayed higher potency than Ru3. The in vivo results in MGC-803 xenograft nude mice model also confirmed that Ru4 obviously inhibited tumor growth. Ru4 is a promising candidate to be developed as a chemotherapeutic agent.
Collapse
Affiliation(s)
- Yu-Lan Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, PR China
| | - Xiao-Min Zhu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, PR China
| | - Nan-Feng Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, PR China
| | - Shao-Ting Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, PR China
| | - Yang Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, PR China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, PR China.
| | - Zhen-Feng Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, PR China.
| |
Collapse
|
7
|
Liao X, Chen Y, Lei M, Hou C, Li X, Wang T. Hydrophilic-interaction-based magnetically assisted matrix solid-phase dispersion extraction of carbadox and olaquindox in feeds. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:2080-2089. [PMID: 34599509 DOI: 10.1002/jsfa.11549] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/03/2021] [Accepted: 10/02/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Carbadox and olaquindox have been banned from feeds since 1998 by the EU because of their mutagenic, photoallergic, and carcinogenic effects. Unfortunately, owing to their outstanding effect, they are frequently abused or misused in animal husbandry. There is an urgent need to develop a sensitive and reliable method for monitoring these drugs in animal feeds. RESULTS This work reported a new method of hydrophilic-interaction-based magnetically assisted matrix solid-phase dispersion (MMSPD) extraction coupled with reversed-phase liquid chromatography-mass spectrometry for simultaneous determination of carbadox and olaquindox in animal feeds. 3-Trimethoxysilylpropyl methacrylate (γ-MAPS)-modified attapulgite (ATP) was crosslinked with γ-MAPS-modified iron(II,III) oxide (Fe3 O4 ), 1-vinyl-3-(butyl-4-sulfonate) imidazolium (VBSIm), acrylamide (AM), and N,N'-methylene-bis(acrylamide) (MBA) to synthesize ATP@Fe3 O4 @poly(VBSIm-AM-MBA) particles. The resultant particles were characterized by scanning electron microscopy, energy dispersive spectrometer, transmission electron microscopy, vibrating sample magnetometer, and Fourier transform infrared spectroscopy. Crosslinking of ATP into the magnetic particles has significantly increased the adsorption capacity of the particles. Under optimum conditions, the limits of detection (S/N = 3) were 0.3 μg kg-1 and 0.9 μg kg-1 for carbadox and olaquindox respectively. The intra-day and inter-day recoveries of the spiked targets in feed samples were in the range 83.5-98.3% with relative standard deviations of 1.0-8.3%. CONCLUSION With a simplified procedure and a low amount of sample, the proposed hydrophilic-interaction-based MMSPD method is not only useful for the determination of carbadox and olaquindox in feeds but also holds great promise for the analysis of other polar targets in solid or semisolid matrices. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xibin Liao
- School of Materials and Chemical Engineering, Ningbo University of Technology, Ningbo, China
| | - Yihui Chen
- Ningbo Academy of Inspection and Quarantine, Ningbo, China
| | - Meikang Lei
- Comprehensive Technology and Service Center of Quzhou Customs, Quzhou, China
| | - Chunyan Hou
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Xie Li
- School of Materials and Chemical Engineering, Ningbo University of Technology, Ningbo, China
| | - Tingting Wang
- School of Materials and Chemical Engineering, Ningbo University of Technology, Ningbo, China
| |
Collapse
|
8
|
Im JY, Kim BK, Yoon SH, Cho BC, Baek YM, Kang MJ, Kim N, Gong YD, Won M. DGG-100629 inhibits lung cancer growth by suppressing the NFATc1/DDIAS/STAT3 pathway. Exp Mol Med 2021; 53:643-653. [PMID: 33859351 PMCID: PMC8102629 DOI: 10.1038/s12276-021-00601-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 02/07/2021] [Accepted: 02/15/2021] [Indexed: 02/02/2023] Open
Abstract
DNA damage-induced apoptosis suppressor (DDIAS) promotes the progression of lung cancer and hepatocellular carcinoma through the regulation of multiple pathways. We screened a chemical library for anticancer agent(s) capable of inhibiting DDIAS transcription. DGG-100629 was found to suppress lung cancer cell growth through the inhibition of DDIAS expression. DGG-100629 induced c-Jun NH(2)-terminal kinase (JNK) activation and inhibited NFATc1 nuclear translocation. Treatment with SP600125 (a JNK inhibitor) or knockdown of JNK1 restored DDIAS expression and reversed DGG-100629-induced cell death. In addition, DGG-100629 suppressed the signal transducer and activator of transcription (STAT3) signaling pathway. DDIAS or STAT3 overexpression restored lung cancer cell growth in the presence of DGG-100629. In a xenograft assay, DGG-100629 inhibited tumor growth by reducing the level of phosphorylated STAT3 and the expression of STAT3 target genes. Moreover, DGG-100629 inhibited the growth of lung cancer patient-derived gefitinib-resistant cells expressing NFATc1 and DDIAS. Our findings emphasize the potential of DDIAS blockade as a therapeutic approach and suggest a novel strategy for the treatment of gefitinib-resistant lung cancer.
Collapse
Affiliation(s)
- Joo-Young Im
- grid.249967.70000 0004 0636 3099Personalized Genomic Medicine Research Center, KRIBB, Daejeon, 34141 Korea
| | - Bo-Kyung Kim
- grid.249967.70000 0004 0636 3099Personalized Genomic Medicine Research Center, KRIBB, Daejeon, 34141 Korea
| | - Sung-Hoon Yoon
- grid.418982.e0000 0004 5345 5340National Center for Efficacy Evaluation for Respiratory Disease Product, Korea Institute of Toxicology, Jeongeup, Jeollabuk-do 56212 Korea ,grid.412786.e0000 0004 1791 8264Department of Human and Environmental Toxicology, University of Science and Technology (UST), Daejeon, 34113 Korea
| | - Byoung Chul Cho
- grid.15444.300000 0004 0470 5454Division of Medical Oncology, Yonsei University College of Medicine, Seoul, 03722 Korea
| | - Yu Mi Baek
- Therna Therapeutics, Yangcheon-ro, Gangseo-gu, Seoul 05029 Korea
| | - Mi-Jung Kang
- grid.249967.70000 0004 0636 3099Personalized Genomic Medicine Research Center, KRIBB, Daejeon, 34141 Korea
| | - Nayeon Kim
- grid.255168.d0000 0001 0671 5021Innovative Drug Library Research Center, Department of Chemistry, College of Science, Dongguk University, Seoul, 04620 Korea
| | - Young-Dae Gong
- grid.255168.d0000 0001 0671 5021Innovative Drug Library Research Center, Department of Chemistry, College of Science, Dongguk University, Seoul, 04620 Korea
| | - Misun Won
- grid.249967.70000 0004 0636 3099Personalized Genomic Medicine Research Center, KRIBB, Daejeon, 34141 Korea ,grid.412786.e0000 0004 1791 8264Deparment of Functional Genomics, University of Science and Technology (UST), Daejeon, 34113 Korea
| |
Collapse
|
9
|
Zhang YL, Wen XD, Guo X, Huang SQ, Wang TT, Zhou PT, Li W, Zhou LF, Hu YH. Progesterone suppresses the progression of colonic carcinoma by increasing the activity of the GADD45α/JNK/c‑Jun signalling pathway. Oncol Rep 2021; 45:95. [PMID: 33846816 PMCID: PMC8054317 DOI: 10.3892/or.2021.8046] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/25/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third most diagnosed cancer worldwide. Progesterone is associated with a decreased risk of CRC and leads to a favourable prognosis. However, the specific mechanism by which progesterone suppresses malignant progression remains to be elucidated. In the present study, the level of progesterone was first analysed in 77 patients with CRC, and immunohistochemistry was performed to detect the expression of progesterone receptor (PGR) in the paired specimens. The correlations between progesterone, PGR and CRC prognosis were assessed. A Cell Counting Kit-8 assay was then used to detect proliferation of the CRC cells. Flow cytometry was performed to estimate apoptosis and to evaluate the cycle of the CRC cells. A xenograft tumour model was established in nude mice to assess the role of progesterone in tumour growth. Finally, a PCR microarray was used to screen differentially expressed genes to further interpret the mechanism by which progesterone inhibits the malignant progression of CRC. It was found that low expression of progesterone and PGR were significantly associated with poor prognosis of CRC. In addition, progesterone suppressed CRC cell proliferation by arresting the cell cycle and inducing apoptosis in vitro. Moreover, the inhibitory role of progesterone in tumour growth was verified in vivo. Further investigation showed that the level of growth arrest and DNA damage-inducible protein α (GADD45α) was up-regulated by progesterone, and this was followed by the activation of the JNK pathway. Progesterone increased the activity of the JNK pathway via GADD45α to inhibit proliferation by arresting the cell cycle and inducing apoptosis, thereby suppressing the malignant progression of CRC. Therefore, it can be concluded that progesterone and PGR might act as inhibiting factors for poor prognosis of CRC.
Collapse
Affiliation(s)
- Yao-Lei Zhang
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, P.R. China
| | - Xu-Dong Wen
- Department of Gastroenterology, Chengdu First People's Hospital, Chengdu, Sichuan 610016, P.R. China
| | - Xin Guo
- Central Laboratory, General Hospital of Western Theater Command, Chengdu, Sichuan 610016, P.R. China
| | - Shang-Qing Huang
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, P.R. China
| | - Ting-Ting Wang
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, P.R. China
| | - Pei-Ting Zhou
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, P.R. China
| | - Wei Li
- Central Laboratory, General Hospital of Western Theater Command, Chengdu, Sichuan 610016, P.R. China
| | - Long-Fu Zhou
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, P.R. China
| | - Yong-He Hu
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan 610031, P.R. China
| |
Collapse
|
10
|
Olaquindox-Induced Liver Damage Involved the Crosstalk of Oxidative Stress and p53 In Vivo and In Vitro. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8835207. [PMID: 33381272 PMCID: PMC7762677 DOI: 10.1155/2020/8835207] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/27/2020] [Accepted: 12/03/2020] [Indexed: 01/21/2023]
Abstract
Olaquindox (OLA), a member of the quinoxaline-N,N-dioxide family, has been widely used as a growth-promoting feed additive and treatment for bacterial infections. The toxicity has been a major concern, and the precise molecular mechanism remains poorly understood. The present study was aimed at investigating the roles of oxidative stress and p53 in OLA-caused liver damage. In a mouse model, OLA administration could markedly cause liver injury as well as the induction of oxidative stress and activation of p53. Antioxidant N-acetylcysteine (NAC) inhibited OLA-induced oxidative stress and p53 activation in vivo. Furthermore, knockout of the p53 gene could significantly inhibit OLA-induced liver damage by inhibiting oxidative stress and the mitochondria apoptotic pathway, compared to the p53 wild-type liver tissue. The cell model in vitro further demonstrated that p53 knockout or knockdown in the HCT116 cell and L02 cell significantly inhibited cell apoptosis and increased cell viability, presented by suppressing ROS production, oxidative stress, and the Nrf2/HO-1 pathway. Moreover, loss of p53 decreased OLA-induced mitochondrial dysfunction and caspase activations, with the evidence of inhibited activation of phosphorylation- (p-) p38 and p-JNK and upregulated cell autophagy via activation of the LC3 and Beclin1 pathway in HCT116 and L02 cells. Taken together, our findings provided a support that p53 primarily played a proapoptotic role in OLA-induced liver damage against oxidative stress and mitochondrial dysfunction, which were largely dependent on suppression of the JNK/p38 pathway and upregulation of the autophagy pathway via activation of LC3 and Beclin1.
Collapse
|
11
|
Zeng X, Wang H, He D, Jia W, Ma R. LIMD1 Increases the Sensitivity of Lung Adenocarcinoma Cells to Cisplatin via the GADD45α/p38 MAPK Signaling Pathway. Front Oncol 2020; 10:969. [PMID: 32754438 PMCID: PMC7365921 DOI: 10.3389/fonc.2020.00969] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/18/2020] [Indexed: 11/30/2022] Open
Abstract
Objective: To investigate the effect of LIM domain-containing protein 1 (LIMD1) on the sensitivity of lung adenocarcinoma cells to cisplatin and explore the mechanism. Methods: A549 and H1299 cells were transfected with lentivirus to establish LIMD1-overexpressing cell lines and their respective controls. The protein expression of DNA damage-inducible 45 alpha (GADD45α) and p38 mitogen-activated protein kinase (MAPK) was detected by Western blot. The survival of A549-vec, A549-LIMD1, H1299-vec, and H1299-LIMD1 cells after cisplatin treatment was observed by CCK-8, and the viability was calculated accordingly. Then, SB203580 was used to inhibit the activity of the p38 MAPK signaling pathway, after which the survival of A549-vec, A549-LIMD1, H1299-vec, and H1299-LIMD1 cells in response to cisplatin was observed again by CCK-8, and the viability was calculated accordingly. Results: When LIMD1 was overexpressed in A549 and H1299 cells, the levels of GADD45α and p-p38 MAPK were increased, but total p38 MAPK expression showed no significant change. After adding 30 μM cisplatin, the optical density (OD) values of A549-LIMD1 and H1299-LIMD1 cells were significantly lower than those of their respective controls at 24, 48, and 72 h. The viability of A549-LIMD1 and H1299-LIMD1 cells was significantly lower than that of their respective controls at all the times tested (p < 0.05). The Western blot results showed that the expression of apoptotic proteins cleaved caspase 3 and cleaved PARP in cisplatin-treated A549-LIDM1 and H1299-LIMD1 cells was significantly higher than that in their respective control cells. Flow cytometry showed that the apoptosis rates of A549-LIMD1 and H1299-LIMD1 cells were significantly higher than those of their respective controls (p < 0.05). SB203580 significantly inhibited the activation of the p38 MAPK signaling pathway in lung adenocarcinoma cells; however, neither the OD values nor the viability of A549-LIMD1 cells and H1299-LIMD1 cells showed no significant difference from those of their controls at 24, 48, and 72 h after cisplatin and SB203580 treatment (p > 0.05 for both). Western blot analysis showed that after SB203580 was added, the expression of cleaved caspase 3 and cleaved PARP in A549-LIMD1 and H1299-LIMD1 cells presented no significant difference compared with that in their respective controls. Conclusion: LIMD1 increases the sensitivity of lung adenocarcinoma cells to cisplatin by activating the GADD45α/p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Xiaofei Zeng
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Hong Wang
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Dongsheng He
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Weikun Jia
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Ruidong Ma
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
12
|
Kang YM, Lan A, Huang YH, Hsu KM, Chao Y, Lan KL. Identification of key genes and pathways associated with topotecan treatment using multiple bioinformatics tools. J Chin Med Assoc 2020; 83:446-453. [PMID: 32243271 DOI: 10.1097/jcma.0000000000000313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The goal of this study is to determine critical genes and pathways associated with topotecan using publicly accessible bioinformatics tools. METHODS Topotecan signatures were downloaded from the Library of Integrated Network-Based Cellular Signatures (LINCS) database (http://www.ilincs.org/ilincs/). Differentially expressed genes (DEGs) were defined as genes that appeared at least three times with p values <0.05 and a fold change of ≥50% (|log2FC| ≥ 0.58). Hub genes were identified by evaluating the following parameters using a protein-protein interaction network: node degrees, betweenness, and eigenfactor scores. Hub genes and the top-40 DEGs by |log2FC| were used to generate a Venn diagram, and key genes were identified. Functional and pathway enrichment analysis was performed using the Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. Information on ovarian cancer patients derived from The Cancer Genome Atlas (TCGA) database was analyzed, and the effect of topotecan on the protein expression was examined by Western blotting. RESULTS Eleven topotecan signatures were downloaded, and 65 upregulated and 87 downregulated DEGs were identified. Twenty-one hub genes were identified. We identified eight key genes as upregulated genes, including NFKBIA, IKBKB, GADD45A, CDKN1A, and HIST2H2BE, while EZH2, CDC20, and CDK7 were identified as downregulated genes, which play critical roles in the cell cycle and carcinogenesis in KEGG analysis. In the TCGA analysis, the CDKN1A+/EZH2- group had the longest median survival, while the CDKN1A-/EZH2+ group had the shortest median survival. Topotecan-treated murine ovarian (MOSEC), colorectal (CT26), and lung (LLC) cancer cell lines displayed upregulated CDKN1A encoding p21 and downregulated Ezh2. CONCLUSION Using publicly accessible bioinformatics tools, we evaluated key genes and pathways related to topotecan and examined the key genes using the TCGA database and in vitro studies.
Collapse
Affiliation(s)
- Yu-Mei Kang
- Division of Radiation Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Alexander Lan
- School of Life Science, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Yen-Hua Huang
- Institute of Biomedical Bioinformatics, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Kai-Mei Hsu
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yee Chao
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Keng-Li Lan
- Division of Radiation Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| |
Collapse
|
13
|
Shi Y, Wang X, Wang N, Li FF, You YL, Wang SQ. The effect of polysaccharides from Cibotium barometz on enhancing temozolomide-induced glutathione exhausted in human glioblastoma U87 cells, as revealed by 1H NMR metabolomics analysis. Int J Biol Macromol 2020; 156:471-484. [PMID: 32243933 DOI: 10.1016/j.ijbiomac.2020.03.243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/18/2020] [Accepted: 03/30/2020] [Indexed: 02/06/2023]
Abstract
Glioblastoma (GBM) is the most malignant central nervous system tumor, with poor prognosis. Temozolomide (TMZ) has been used as a first-line drug for the treatment of GBM for over a decade, but its treatment benefits are limited by acquired resistance. Polysaccharides from Cibotium barometz (CBPs) are polysaccharides purified from the root of Cibotium barometz (L.) J. Sm., possessing sensitizing activity. The purpose of this study was to investigate the anti-cancer effect of CBP from different processing methods on U87 cells using a 1H NMR-based metabolic approach, complemented with qRT-PCR and flow cytometry, to identify potential markers and discover the targets to explore the underlying mechanism. Cibotium barometz is usually processed under sand heating in clinical applications. Polysaccharides from both the processed (PCBP) and raw (RCBP) C. barometz were prepared, and the effect on enhancing the sensitivity to TMZ was investigated in vitro. CBP can significantly increase the toxicity of TMZ to the U87 cell line, promote apoptosis, enhance cell cycle changes, and arrest cells in S phase, and RCBP demonstrated better activity. Multivariate statistical analyses, such as principal component analysis (PCA) and orthogonal projection to latent structure with discriminant analysis (OPLS-DA), were used to identify metabolic biomarkers, and 12 metabolites in the cell extract samples were clearly identified as altered after RCBP exposure. NMR-based cell metabolomics provided a holistic method for the identification of CBP's apoptosis-enhancing mechanisms and the exploration of its potential applications in preclinical and clinical studies.
Collapse
Affiliation(s)
- Yue Shi
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Xiao Wang
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Ning Wang
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Fei-Fei Li
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Yu-Lin You
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Shu-Qi Wang
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
| |
Collapse
|
14
|
Bin Y, Ding Y, Xiao W, Liao A. RASSF1A: A promising target for the diagnosis and treatment of cancer. Clin Chim Acta 2020; 504:98-108. [PMID: 31981586 DOI: 10.1016/j.cca.2020.01.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/16/2020] [Accepted: 01/16/2020] [Indexed: 02/07/2023]
Abstract
The Ras association domain family 1 isoform A (RASSF1A), a tumor suppressor, regulates several tumor-related signaling pathways and interferes with diverse cellular processes. RASSF1A is frequently demonstrated to be inactivated by hypermethylation in numerous types of solid cancers. It is also associated with lymph node metastasis, vascular invasion, and chemo-resistance. Therefore, reactivation of RASSF1A may be a viable strategy to block tumor progress and reverse drug resistance. In this review, we have summarized the clinical value of RASSF1A for screening, staging, and therapeutic management of human malignancies. We also highlighted the potential mechanism of RASSF1A in chemo-resistance, which may help identify novel drugs in the future.
Collapse
Affiliation(s)
- Yuling Bin
- Digestive System Department, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Yong Ding
- Department of Vascular Surgery, Zhongshan Hospital, Institue of Vascular Surgery, Fudan University, Shanghai 200032, China
| | - Weisheng Xiao
- Digestive System Department, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China
| | - Aijun Liao
- Digestive System Department, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
15
|
Silk fibroin peptide suppresses proliferation and induces apoptosis and cell cycle arrest in human lung cancer cells. Acta Pharmacol Sin 2019; 40:522-529. [PMID: 29921888 DOI: 10.1038/s41401-018-0048-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 05/20/2018] [Indexed: 12/21/2022]
Abstract
Silkworm cocoon was recorded to cure carbuncle in the Compendium of Materia Medica. Previous studies have demonstrated that the supplemental silk protein sericin exhibits anticancer activity. In the present study, we investigated the effects of silk fibroin peptide (SFP) extracted from silkworm cocoons against human lung cancer cells in vitro and in vivo and its possible anticancer mechanisms. SFP that we prepared had high content of glycine (~ 30%) and showed a molecular weight of ~ 10 kDa. Intragastric administration of SFP (30 g/kg/d) for 14 days did not affect the weights, vital signs, routine blood indices, and blood biochemical parameters in mice. MTT assay showed that SFP dose-dependently inhibited the growth of human lung cancer A549 and H460 cells in vitro with IC50 values of 9.921 and 9.083 mg/mL, respectively. SFP also dose-dependently suppressed the clonogenic activity of the two cell lines. In lung cancer H460 xenograft mice, intraperitoneal injection of SFP (200 or 500 mg/kg/d) for 40 days significantly suppressed the tumor growth, but did not induce significant changes in the body weight. We further examined the effects of SFP on cell cycle and apoptosis in H460 cells using flow cytometry, which revealed that SFP-induced cell cycle arrest at the S phase, and then promoted cell apoptosis. We demonstrated that SFP (20-50 mg/mL) dose-dependently downregulates Bcl-2 protein expression and upregulates Bax protein in H460 cells during cell apoptosis. The results suggest that SFP should be studied further as a novel therapeutic agent for the treatment of lung cancer.
Collapse
|
16
|
BW18, a C-21 steroidal glycoside, exerts an excellent anti-leukemia activity through inducing S phase cell cycle arrest and apoptosis via MAPK pathway in K562 cells. Biomed Pharmacother 2019; 112:108603. [DOI: 10.1016/j.biopha.2019.108603] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 01/10/2023] Open
|
17
|
Dai C, Xiao X, Li J, Ciccotosto GD, Cappai R, Tang S, Schneider-Futschik EK, Hoyer D, Velkov T, Shen J. Molecular Mechanisms of Neurotoxicity Induced by Polymyxins and Chemoprevention. ACS Chem Neurosci 2019; 10:120-131. [PMID: 30362702 DOI: 10.1021/acschemneuro.8b00300] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neurotoxicity is one major unwanted side-effects associated with polymyxin (i.e., colistin and polymyxin B) therapy. Clinically, colistin neurotoxicity is characterized by neurological symptoms including dizziness, visual disturbances, vertigo, confusion, hallucinations, seizures, ataxia, and facial and peripheral paresthesias. Pathologically, colistin-induced neurotoxicity is characterized by cell injury and death in neuronal cell. This Review covers our current understanding of polymyxin-induced neurotoxicity, its underlying mechanisms, and the discovery of novel neuroprotective agents to limit this neurotoxicity. In recent years, an increasing body of literature supports the notion that polymyxin-induced nerve damage is largely related to oxidative stress and mitochondrial dysfunction. P53, PI3K/Akt, and MAPK pathways are also involved in colistin-induced neuronal cell death. The activation of the redox homeostasis pathways such as Nrf2/HO-1 and autophagy have also been shown to play protective roles against polymyxin-induced neurotoxicity. These pathways have been demonstrated to be upregulated by neuroprotective agents including curcumin, rapamycin and minocycline. Further research is needed toward the development of novel polymyxin formulations in combination with neuroprotective agents to ameliorate this unwanted adverse effect during polymyxins therapy in patients.
Collapse
Affiliation(s)
- Chongshan Dai
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, P. R. China
| | - Xilong Xiao
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, P. R. China
| | - Jichang Li
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150000, P. R. China
| | - Giuseppe D. Ciccotosto
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Roberto Cappai
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Shusheng Tang
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, P. R. China
| | - Elena K. Schneider-Futschik
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Daniel Hoyer
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, Victoria 3052, Australia
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Tony Velkov
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150000, P. R. China
| | - Jianzhong Shen
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, P. R. China
| |
Collapse
|
18
|
Xiao S, Wong NK, Li J, Lin Y, Zhang Y, Ma H, Mo R, Zhang Y, Yu Z. Analysis of in situ Transcriptomes Reveals Divergent Adaptive Response to Hyper- and Hypo-Salinity in the Hong Kong Oyster, Crassostrea hongkongensis. Front Physiol 2018; 9:1491. [PMID: 30416453 PMCID: PMC6212563 DOI: 10.3389/fphys.2018.01491] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/02/2018] [Indexed: 02/05/2023] Open
Abstract
Crassostrea hongkongensis, a commercially valuable aquaculture species dwelling in estuaries along the coast of the South China Sea, is remarkable for its eurysalinity traits that enable its successful colonization of diverse osmotic niches ranging from near freshwater to seawater. In order to elucidate how this oyster copes with coastal waters with immense salinity differences, we performed in situ transcriptomic analysis (RNA-seq) to characterize the global expression patterns of oysters distributed across naturally formed salinity gradients in Zhenhai Bay along the northern coast of the South China Sea. Principal component analysis reveals distinct expression profiles of oysters living in the extreme conditions of hypo-salinity and hyper-salinity. Compared with the situation of optimal salinity for oyster growth, hypo-salinity mainly regulated expression of genes involved in FoxO and oxytocin signaling, tight junction and several immune pathways, while hyper-salinity altered gene expression implicated in amino acid metabolism, AMPK and PI3K-AKt signaling pathways, demonstrating the complexity and plasticity of transcriptomic expression underpinning oyster eurysalinity. Furthermore, the expression patterns of several genes correlated with salinity gradients reveals the fine-tuned coordination of molecular networks necessary for adaptive homeostasis in C. hongkongensis. In conclusion, a striking capacity and distinct patterns of transcriptomic expression contribute to eurysalinity adaptation in C. hongkongensis, which provides new mechanistic insights into the adaptive plasticity and resilience of marine mollusks.
Collapse
Affiliation(s)
- Shu Xiao
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Nai-Kei Wong
- State Key Discipline of Infection Diseases, Shenzhen Third People's Hospital, Shenzhen, China
| | - Jun Li
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Yue Lin
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Yuehuan Zhang
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Haitao Ma
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Riguan Mo
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Yang Zhang
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Ziniu Yu
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
19
|
Liu H, Ren C, Han D, Huang H, Zou R, Zhang H, Xu Y, Gong X, Zhang X, Li Y. UPLC-MS/MS Method for Simultaneous Determination of Three Major Metabolites of Mequindox in Holothurian. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2018; 2018:2768047. [PMID: 29805832 PMCID: PMC5902003 DOI: 10.1155/2018/2768047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/10/2018] [Indexed: 06/08/2023]
Abstract
This study developed an ultraperformance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method for the detection of three major metabolites of mequindox, including 3-methyl-quinoxaline-2-carboxylic acid, 1-desoxymequindox, and 1,4-bisdesoxymequindox (MQCA, 1-DMEQ, and BDMEQ), in holothurian. Target analytes were simplified with ultrasound-assisted acidolysis extracted without complicated enzymolysis steps. After that, each sample was centrifuged and purified by an Oasis MAX cartridge. Then, the processed samples were separated and monitored by UPLC-MS/MS. This developed method has been validated according to FDA criteria. At fortified levels of 2, 10, and 20 μg/kg, recoveries ranged from 82.5% to 93.5% with the intraday RSD less than 7.27% and interday RSD less than 11.8%. The limit of detection (LOD) of all the three metabolites ranged from 0.21 to 0.48 μg/kg, while the limit of quantification (LOQ) ranged from 0.79 to 1.59 μg/kg. On application to commercial samples, 14 of 20 samples were detected positive for the three target analytes, with positive rate at 70 percentage. The result indicated that this method was specific, sensitive, and suitable for the quantification and conformation of the three major metabolites of MEQ in holothurian.
Collapse
Affiliation(s)
- Huihui Liu
- Shandong Marine Resource and Environment Research Institute, Laboratory of Restoration for Marine Ecology, Yantai 264006, China
| | - Chuanbo Ren
- Shandong Marine Resource and Environment Research Institute, Laboratory of Restoration for Marine Ecology, Yantai 264006, China
| | - Dianfeng Han
- Shandong Marine Resource and Environment Research Institute, Laboratory of Restoration for Marine Ecology, Yantai 264006, China
| | - Hui Huang
- Shandong Marine Resource and Environment Research Institute, Laboratory of Restoration for Marine Ecology, Yantai 264006, China
| | - Rongjie Zou
- Shandong Marine Resource and Environment Research Institute, Laboratory of Restoration for Marine Ecology, Yantai 264006, China
| | - Huawei Zhang
- Shandong Marine Resource and Environment Research Institute, Laboratory of Restoration for Marine Ecology, Yantai 264006, China
| | - Yingjiang Xu
- Shandong Marine Resource and Environment Research Institute, Laboratory of Restoration for Marine Ecology, Yantai 264006, China
| | - Xianghong Gong
- Shandong Marine Resource and Environment Research Institute, Laboratory of Restoration for Marine Ecology, Yantai 264006, China
| | - Xiuzhen Zhang
- Shandong Marine Resource and Environment Research Institute, Laboratory of Restoration for Marine Ecology, Yantai 264006, China
| | - Yanshen Li
- College of Life Science, Yantai University, Yantai 264005, China
| |
Collapse
|
20
|
Liu J, Jiang G, Mao P, Zhang J, Zhang L, Liu L, Wang J, Owusu L, Ren B, Tang Y, Li W. Down-regulation of GADD45A enhances chemosensitivity in melanoma. Sci Rep 2018; 8:4111. [PMID: 29515153 PMCID: PMC5841426 DOI: 10.1038/s41598-018-22484-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 02/15/2018] [Indexed: 11/08/2022] Open
Abstract
Melanoma is a malignant skin cancer with considerable drug resistance. Increased expression of DNA repair genes have been reported in melanoma, and this contributes to chemotherapy resistance. GADD45A is involved in DNA repair, cell cycle arrest and apoptosis in response to physiologic or environmental stresses. In this study, we investigated the role of GADD45A in chemotherapy response. Firstly, the mRNA expression of profiled DNA repair genes in cisplatin-treated melanoma cells was detected by RT2 profilerTM PCR array. We found the expression of GADD45A upregulated in a dose- and time- dependent manner. In addition, suppression of GADD45A sensitized melanoma cells to cisplatin and enhanced cisplatin-induced DNA damage. Flow cytometry revealed that downregulating GADD45A released cells from cisplatin-induced G2/M arrest and increased apoptosis. By using a MEK inhibitor, GADD45A was shown to be regulated by MAPK-ERK pathway following cisplatin treatment. Thus, the induction of GADD45A might play important roles in chemotherapy response in human melanoma cancer and could serve as a novel molecular target for melanoma therapy.
Collapse
Affiliation(s)
- Jia Liu
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Guoqiang Jiang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Ping Mao
- Department of General Surgery, The people's Hospital of Liaoning Province, Shenyang, 110016, Liaoning, China
| | - Jing Zhang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Lin Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Likun Liu
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Jia Wang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Lawrence Owusu
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Baoyin Ren
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Yawei Tang
- Department of Immunology, Dalian Medical University, Dalian, 116044, China
| | - Weiling Li
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, Liaoning, China.
| |
Collapse
|
21
|
Zhang H, Qu W, Tao Y, Chen D, Xie S, Huang L, Liu Z, Pan Y, Yuan Z. A Convenient and Sensitive LC-MS/MS Method for Simultaneous Determination of Carbadox- and Olaquindox-Related Residues in Swine Muscle and Liver Tissues. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2018; 2018:2834049. [PMID: 30079259 PMCID: PMC6022315 DOI: 10.1155/2018/2834049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/24/2018] [Indexed: 05/09/2023]
Abstract
This paper presents a convenient and sensitive LC-MS/MS method for the simultaneous determination of carbadox and olaquindox residues, including desoxyolaquindox (DOLQ), desoxycarbadox (DCBX), quinoxaline-2-carboxylic acid (QCA), 3-methyl-quinoxaline-2-carboxylic acid (MQCA), and the glycine conjugates of QCA and MQCA (namely, QCA-glycine and MQCA-glycine, resp.) in swine muscle and liver tissues. Tissue samples were extracted with 2% metaphosphoric acid in 20% methanol and cleaned up by solid-phase extraction (SPE) on a mixed-mode anion-exchange column (Oasis MAX). Analysis was performed on a C18 column by detection with mass spectrometry in the multiple reaction monitoring (MRM) mode. The limits of detection (LODs) of the six analytes were determined to be 0.01 μg·kg-1 to 0.25 μg·kg-1, and the limits of quantification (LOQs) were 0.02 μg·kg-1 to 0.5 μg·kg-1. The total recoveries of the six analytes in all tissues were higher than 79.1% with the RSD% less than 9.2%. The developed method can determine the real residue level of QCA and MQCA, whether they are present in free form or as glycine conjugates in tissues, together with the carcinogenic desoxy metabolites DCBX and DOLQ with high recovery. Therefore, this method was suitable for routine analysis of residue control programmes and the residue depletion study of CBX and OLQ on swine.
Collapse
Affiliation(s)
- Heying Zhang
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Wei Qu
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yanfei Tao
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Dongmei Chen
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Shuyu Xie
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Lingli Huang
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zhenli Liu
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yuanhu Pan
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zonghui Yuan
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- National Reference Laboratory of Veterinary Drug Residues and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| |
Collapse
|
22
|
Wu D, Huang CJ, Jiao XF, Ding ZM, Zhang JY, Chen F, Wang YS, Li X, Huo LJ. Olaquindox disrupts tight junction integrity and cytoskeleton architecture in mouse Sertoli cells. Oncotarget 2017; 8:88630-88644. [PMID: 29179463 PMCID: PMC5687633 DOI: 10.18632/oncotarget.20289] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 07/26/2017] [Indexed: 02/06/2023] Open
Abstract
Sertoli cells, by creating an immune-privileged and nutrition supporting environment, maintain mammalian spermatogenesis and thereby holds the heart of male fertility. Olaquindox, an effective feed additive in livestock industry, could potentially expose human into the risk of biological hazards due to its genotoxicity and cytotoxicity, highlighting the significance of determining its bio-safety regarding human reproduction. Herein, we deciphered the detrimental effects of olaquindox on male fertility by mechanistically unraveling how olaquindox intervenes blood-testis barrier in mouse. Olaquindox (400 μg/ml) exposure significantly compromised tight junction permeability function, decreased or dislocated the junction proteins (e.g., ZO-1, occludin and N-cadherin) and attenuated mTORC2 signaling pathway in primary Sertoli cells. Furthermore, olaquindox disrupted F-actin architecture through interfering with the expression of actin branching protein complex (CDC42-N-WASP-Arp3) and actin bunding protein palladin. Olaquindox also triggered severely DNA damage and apoptosis while inhibiting autophagic flux in Sertoli cell presumably due to the exacerbated generation of reactive oxygen species (ROS). Pre-treatment with antioxidant N-acetylcysteine effectively ameliorated olaquindox-induced exhaustion of ZO-1 and N-Cadherin proteins, DNA damage and apoptosis. More significantly, olaquindox disrupted the epigenetic status in Sertoli cells with hypermethylation and concomitantly hypoacetylation of H3K9 and H3K27. Overall, our study determines olaquindox targets Sertoli cells to affect BTB function through tight junction proteins and F-actin orgnization, which might disrupt the process of spermatogenesis.
Collapse
Affiliation(s)
- Di Wu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Wuhan 430070, Hubei, China
| | - Chun-Jie Huang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Wuhan 430070, Hubei, China
| | - Xiao-Fei Jiao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Wuhan 430070, Hubei, China
| | - Zhi-Ming Ding
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Wuhan 430070, Hubei, China
| | - Jia-Yu Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Wuhan 430070, Hubei, China
| | - Fan Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Wuhan 430070, Hubei, China
| | - Yong-Sheng Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Wuhan 430070, Hubei, China
| | - Xiang Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Wuhan 430070, Hubei, China
| | - Li-Jun Huo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China.,Department of Hubei Province Engineering Research Center in Buffalo Breeding and Products, Wuhan 430070, Hubei, China
| |
Collapse
|