1
|
Ergün S, Aslan S, Demir D, Kayaoğlu S, Saydam M, Keleş Y, Kolcuoğlu D, Taşkurt Hekim N, Güneş S. Beyond Death: Unmasking the Intricacies of Apoptosis Escape. Mol Diagn Ther 2024; 28:403-423. [PMID: 38890247 PMCID: PMC11211167 DOI: 10.1007/s40291-024-00718-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 06/20/2024]
Abstract
Apoptosis, or programmed cell death, maintains tissue homeostasis by eliminating damaged or unnecessary cells. However, cells can evade this process, contributing to conditions such as cancer. Escape mechanisms include anoikis, mitochondrial DNA depletion, cellular FLICE inhibitory protein (c-FLIP), endosomal sorting complexes required for transport (ESCRT), mitotic slippage, anastasis, and blebbishield formation. Anoikis, triggered by cell detachment from the extracellular matrix, is pivotal in cancer research due to its role in cellular survival and metastasis. Mitochondrial DNA depletion, associated with cellular dysfunction and diseases such as breast and prostate cancer, links to apoptosis resistance. The c-FLIP protein family, notably CFLAR, regulates cell death processes as a truncated caspase-8 form. The ESCRT complex aids apoptosis evasion by repairing intracellular damage through increased Ca2+ levels. Antimitotic agents induce mitotic arrest in cancer treatment but can lead to mitotic slippage and tetraploid cell formation. Anastasis allows cells to resist apoptosis induced by various triggers. Blebbishield formation suppresses apoptosis indirectly in cancer stem cells by transforming apoptotic cells into blebbishields. In conclusion, the future of apoptosis research offers exciting possibilities for innovative therapeutic approaches, enhanced diagnostic tools, and a deeper understanding of the complex biological processes that govern cell fate. Collaborative efforts across disciplines, including molecular biology, genetics, immunology, and bioinformatics, will be essential to realize these prospects and improve patient outcomes in diverse disease contexts.
Collapse
Affiliation(s)
- Sercan Ergün
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey.
- Department of Multidisciplinary Molecular Medicine, Institute of Graduate Studies, Ondokuz Mayis University, Samsun, Turkey.
| | - Senanur Aslan
- Department of Multidisciplinary Molecular Medicine, Institute of Graduate Studies, Ondokuz Mayis University, Samsun, Turkey
| | - Dilbeste Demir
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Sümeyye Kayaoğlu
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Mevsim Saydam
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Yeda Keleş
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Damla Kolcuoğlu
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Neslihan Taşkurt Hekim
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
- Department of Multidisciplinary Molecular Medicine, Institute of Graduate Studies, Ondokuz Mayis University, Samsun, Turkey
| | - Sezgin Güneş
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayis University, Samsun, Turkey
- Department of Multidisciplinary Molecular Medicine, Institute of Graduate Studies, Ondokuz Mayis University, Samsun, Turkey
| |
Collapse
|
2
|
Xing Z, Su A, Mi L, Zhang Y, He T, Qiu Y, Wei T, Li Z, Zhu J, Wu W. Withaferin A: A Dietary Supplement with Promising Potential as an Anti-Tumor Therapeutic for Cancer Treatment - Pharmacology and Mechanisms. Drug Des Devel Ther 2023; 17:2909-2929. [PMID: 37753228 PMCID: PMC10519218 DOI: 10.2147/dddt.s422512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/18/2023] [Indexed: 09/28/2023] Open
Abstract
Cancer, as the leading cause of death worldwide, poses a serious threat to human health, making the development of effective tumor treatments a significant challenge. Natural products continue to serve as crucial resources for drug discovery. Among them, Withaferin A (WA), the most active phytocompound extracted from the renowned dietary supplement Withania somnifera (L.) Dunal, exhibits remarkable anti-tumor efficacy. In this manuscript, we aim to comprehensively summarize the pharmacological characteristics of WA as a potential anti-tumor drug candidate, with the objective of contributing to its further development and the discovery of prospective drugs. Through an extensive review of literature from PubMed, Science Direct, and Web of Science, we have gathered substantial evidence showcasing WA's significant anti-tumor effects against a wide range of cancers in both in vitro and in vivo studies. Mechanistically, WA exerts its anti-tumor influence by inducing cell cycle arrest, apoptosis, autophagy, and ferroptosis. Additionally, it inhibits cell proliferation, cancer stem cells, tumor metastasis, and also suppresses epithelial-mesenchymal transition (EMT) and angiogenesis. Several studies have identified direct target proteins of WA, such as vimentin, Hsp90, annexin II and mFAM72A, while BCR-ABL, Mortalin (mtHsp70), Nrf2, and c-MYB are potential targets of WA. Notwithstanding its remarkable anti-tumor efficacy, there are some limitations associated with WA, including potential toxicity and poor oral bioavailability, which need to be addressed when considering it as an anti-tumor candidate agent. Nevertheless, I given its promising anti-tumor attributes, WA remains an encouraging candidate for future drug development. Unveiling the exact target and comprehensive mechanism of WA's action represents a crucial research direction to pursue in the future.
Collapse
Affiliation(s)
- Zhichao Xing
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Anping Su
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Li Mi
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Yujie Zhang
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Ting He
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Yuxuan Qiu
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Tao Wei
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Zhihui Li
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Jingqiang Zhu
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Wenshuang Wu
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|
3
|
Rousta N, Aslan M, Yesilcimen Akbas M, Ozcan F, Sar T, Taherzadeh MJ. Effects of fungal based bioactive compounds on human health: Review paper. Crit Rev Food Sci Nutr 2023; 64:7004-7027. [PMID: 36794421 DOI: 10.1080/10408398.2023.2178379] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Since the first years of history, microbial fermentation products such as bread, wine, yogurt and vinegar have always been noteworthy regarding their nutritional and health effects. Similarly, mushrooms have been a valuable food product in point of both nutrition and medicine due to their rich chemical components. Alternatively, filamentous fungi, which can be easier to produce, play an active role in the synthesis of some bioactive compounds, which are also important for health, as well as being rich in protein content. Therefore, this review presents some important bioactive compounds (bioactive peptides, chitin/chitosan, β-glucan, gamma-aminobutyric acid, L-carnitine, ergosterol and fructooligosaccharides) synthesized by fungal strains and their health benefits. In addition, potential probiotic- and prebiotic fungi were researched to determine their effects on gut microbiota. The current uses of fungal based bioactive compounds for cancer treatment were also discussed. The use of fungal strains in the food industry, especially to develop innovative food production, has been seen as promising microorganisms in obtaining healthy and nutritious food.
Collapse
Affiliation(s)
- Neda Rousta
- Swedish Centre for Resource Recovery, University of Borås, Borås, Sweden
| | - Melissa Aslan
- Swedish Centre for Resource Recovery, University of Borås, Borås, Sweden
- Department of Molecular Biology and Genetics, Gebze Technical University, Gebze-Kocaeli, Turkey
| | - Meltem Yesilcimen Akbas
- Department of Molecular Biology and Genetics, Gebze Technical University, Gebze-Kocaeli, Turkey
| | - Ferruh Ozcan
- Department of Molecular Biology and Genetics, Gebze Technical University, Gebze-Kocaeli, Turkey
| | - Taner Sar
- Swedish Centre for Resource Recovery, University of Borås, Borås, Sweden
| | | |
Collapse
|
4
|
Targeting of the Interleukin-13 Receptor (IL-13R)α2 Expressing Prostate Cancer by a Novel Hybrid Lytic Peptide. Biomolecules 2023; 13:biom13020356. [PMID: 36830725 PMCID: PMC9953383 DOI: 10.3390/biom13020356] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/31/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
The IL-13Rα2 cell surface receptor is highly expressed in tumours such as prostate cancer. In this report, we evaluated the hypothesis that prostate cancer cells with enhanced IL-13Rα2 expression are a suitable target for the hybrid lytic peptide (Pep-1-Phor21) peptide, which is generated by fusing the IL-13Rα2 specific ligand (Pep-1) and a cell membrane disrupting lytic peptide (Phor21). The expression of IL-13Rα2 mRNA and protein in prostate cancer tissues and cell lines was assessed via real-time PCR (RT-PCR) and immunoblotting. The effect of Pep-1-Phor21 on the viability of prostate cancer cells grown in monolayers (2D) and microtissue spheroids (3D) was assessed via CellTox green cytotoxic assay. IL-13Rα2 expression and Pep-1-Phor21-mediated killing were also determined in the cells treated with epigenetic regulators (Trichostatin A (TSA) and 5-aza-2 deoxycytidine (5-Aza-dC)). The hybrid lytic peptide cytotoxic activity correlated with the expression of IL-13Rα2 in prostate cancer cell lines cultured as monolayers (2D) or 3D spheroids. In addition, TSA or 5-Aza-dC treatment of prostate cancer cells, particularly those with low expression of IL-13Rα2, enhanced the cells' sensitivity to the lytic peptide by increasing IL-13Rα2 expression. These results demonstrate that the Pep-1-Phor21 hybrid lytic peptide has potent and selective anticancer properties against IL-13Rα2-expressing prostate cancer cells.
Collapse
|
5
|
Chang JS, Chen CY, Tikhomirov AS, Islam A, Liang RH, Weng CW, Wu WH, Shchekotikhin AE, Chueh PJ. Bis(chloroacetamidino)-Derived Heteroarene-Fused Anthraquinones Bind to and Cause Proteasomal Degradation of tNOX, Leading to c-Flip Downregulation and Apoptosis in Oral Cancer Cells. Cancers (Basel) 2022; 14:cancers14194719. [PMID: 36230644 PMCID: PMC9562014 DOI: 10.3390/cancers14194719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary New-generation anthraquinone derivatives attached with different heterocycles and bearing chloroacetamidines in the side chains have been synthesized to reduce side effects and drug resistance. In this study, we identified the cellular target of the studied compounds through ligand binding assays and in silico simulations. Our results illustrate that the studied compounds bound to and targeted the tumor-associated NADH oxidase (tNOX) in oral cancer cells. tNOX is a growth-related protein and is found to be expressed in cancer cells but not in non-transformed cells, and its knockdown by RNA interference in tumor cells overturns cancer phenotypes, supporting its role in cellular growth. We also identified that tNOX bound to the studied compounds and underwent degradation, which was correlated with apoptosis induction in oral cancer cells. Abstract Anthraquinone-based intercalating compounds, namely doxorubicin and mitoxantrone, have been used clinically based on their capacity to bind DNA and induce DNA damage. However, their applications have been limited by side effects and drug resistance. New-generation anthraquinone derivatives fused with different heterocycles have been chemically synthesized and screened for higher anticancer potency. Among the compounds reported in our previous study, 4,11-bis(2-(2-chloroacetamidine)ethylamino)anthra[2,3-b]thiophene-5,10-dione dihydrochloride (designated 2c) was found to be apoptotic, but the direct cellular target responsible for the cytotoxicity remained unknown. Here, we report the synthesis and anticancer properties of two other derivatives, 4,11-bis(2-(2-chloroacetamidine)ethylamino)naphtho[2,3-f]indole-5,10-dione dihydrochloride (2a) and 4,11-bis(2-(2-chloroacetamidine)ethylamino)-2-methylanthra[2,3-b]furan-5,10-dione dihydrochloride (2b). We sought to identify and validate the protein target(s) of these derivatives in oral cancer cells, using molecular docking simulations and cellular thermal shift assays (CETSA). Our CETSA results illustrate that these derivatives targeted the tumor-associated NADH oxidase (tNOX, ENOX2), and their direct binding downregulated tNOX in p53-functional SAS and p53-mutated HSC-3 cells. Interestingly, the compounds targeted and downregulated tNOX to reduce SIRT1 deacetylase activity and increase Ku70 acetylation, which triggers c-Flip ubiquitination and induces apoptosis in oral cancer cells. Together, our data highlight the potential value of these heteroarene-fused anthraquinones in managing cancer by targeting tNOX and augmenting apoptosis.
Collapse
Affiliation(s)
- Jeng Shiun Chang
- Department of Otolaryngology, Head and Neck Surgery, Jen-Ai Hospital, Taichung 41265, Taiwan
| | - Chien-Yu Chen
- Institute of Biomedical Sciences, National Chung Hsing University, 145 Xingda Rd., Taichung 40227, Taiwan
| | | | - Atikul Islam
- Institute of Biomedical Sciences, National Chung Hsing University, 145 Xingda Rd., Taichung 40227, Taiwan
| | - Ru-Hao Liang
- Institute of Biomedical Sciences, National Chung Hsing University, 145 Xingda Rd., Taichung 40227, Taiwan
| | - Chia-Wei Weng
- Institute of Biomedical Sciences, National Chung Hsing University, 145 Xingda Rd., Taichung 40227, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Wei-Hou Wu
- Institute of Biomedical Sciences, National Chung Hsing University, 145 Xingda Rd., Taichung 40227, Taiwan
| | - Andrey E. Shchekotikhin
- Gause Institute of New Antibiotics, 11 B. Pirogovskaya Street, 119021 Moscow, Russia
- Correspondence: (A.E.S.); (P.J.C.); Tel.: +7-499-246-0228 (A.E.S.); +886-4-22840896 (P.J.C.)
| | - Pin Ju Chueh
- Institute of Biomedical Sciences, National Chung Hsing University, 145 Xingda Rd., Taichung 40227, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, 145 Xingda Rd., Taichung 40227, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan
- Graduate Institute of Basic Medicine, China Medical University, Taichung 40402, Taiwan
- Correspondence: (A.E.S.); (P.J.C.); Tel.: +7-499-246-0228 (A.E.S.); +886-4-22840896 (P.J.C.)
| |
Collapse
|
6
|
Wang XP, Wen B, Zhang XJ, Ma L, Liang XL, Zhang ML. Transcriptome Analysis of Genes Responding to Infection of Leghorn Male Hepatocellular Cells With Fowl Adenovirus Serotype 4. Front Vet Sci 2022; 9:871038. [PMID: 35774982 PMCID: PMC9237548 DOI: 10.3389/fvets.2022.871038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/13/2022] [Indexed: 12/29/2022] Open
Abstract
Fowl adenovirus serotype 4 (FAdV-4) is a highly pathogenic virus with a broad host range that causes huge economic losses for the poultry industry worldwide. RNA sequencing has provided valuable and important mechanistic clues regarding FAdV-4–host interactions. However, the pathogenic mechanism and host's responses after FAdV-4 infection remains limited. In this study, we used transcriptome analysis to identify dynamic changes in differentially expressed genes (DEGs) at five characteristic stages (12, 24, 36, 48, and 60 h) post infection (hpi) with FAdV-4. A total of 8,242 DEGs were identified based on comparison of five infection stages: 0 and 12, 12 and 24, 24 and 36, 36 and 48, and 48 and 60 hpi. In addition, at these five important time points, we found 37 common upregulated or downregulated DEGs, suggesting a common role for these genes in host response to viral infection. The predicted function of these DEGs using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses revealed that these DEGs were associated with viral invasion, host metabolic pathways and host immunosuppression. Interestingly, genes involved in viral invasion, probably EGR1, SOCS3, and THBS1, were related to FAdV-4 infection. Validation of nine randomly selected DEGs using quantitative reverse-transcription PCR produced results that were highly consistent with those of RNA sequencing. This transcriptomic profiling provides valuable information for investigating the molecular mechanisms underlying host–FAdV-4 interactions. These data support the current molecular knowledge regarding FAdV-4 infection and chicken defense mechanisms.
Collapse
Affiliation(s)
- Xueping P. Wang
- Henan Joint International Research Laboratory of Veterinary Biologics Research and Application, Anyang Institute of Technology, Anyang, China
- *Correspondence: Xueping P. Wang
| | - Bo Wen
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Xiao J. Zhang
- Henan Joint International Research Laboratory of Veterinary Biologics Research and Application, Anyang Institute of Technology, Anyang, China
| | - Lei Ma
- Henan Joint International Research Laboratory of Veterinary Biologics Research and Application, Anyang Institute of Technology, Anyang, China
| | - Xiu L. Liang
- Henan Joint International Research Laboratory of Veterinary Biologics Research and Application, Anyang Institute of Technology, Anyang, China
| | - Ming L. Zhang
- Henan Joint International Research Laboratory of Veterinary Biologics Research and Application, Anyang Institute of Technology, Anyang, China
| |
Collapse
|
7
|
Umar MI, Hassan W, Murtaza G, Buabeid M, Arafa E, Irfan HM, Asmawi MZ, Huang X. The Adipokine Component in the Molecular Regulation of Cancer Cell Survival, Proliferation and Metastasis. Pathol Oncol Res 2021; 27:1609828. [PMID: 34588926 PMCID: PMC8473628 DOI: 10.3389/pore.2021.1609828] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/09/2021] [Indexed: 12/22/2022]
Abstract
A hormonal imbalance may disrupt the rigorously monitored cellular microenvironment by hampering the natural homeostatic mechanisms. The most common example of such hormonal glitch could be seen in obesity where the uprise in adipokine levels is in virtue of the expanding bulk of adipose tissue. Such aberrant endocrine signaling disrupts the regulation of cellular fate, rendering the cells to live in a tumor supportive microenvironment. Previously, it was believed that the adipokines support cancer proliferation and metastasis with no direct involvement in neoplastic transformations and tumorigenesis. However, the recent studies have reported discrete mechanisms that establish the direct involvement of adipokine signaling in tumorigenesis. Moreover, the individual adipokine profile of the patients has never been considered in the prognosis and staging of the disease. Hence, the present manuscript has focused on the reported extensive mechanisms that culminate the basis of poor prognosis and diminished survival rate in obese cancer patients.
Collapse
Affiliation(s)
| | - Waseem Hassan
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan
| | - Manal Buabeid
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Elshaimaa Arafa
- Department of Clinical Sciences, Ajman University, Ajman, United Arab Emirates.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | | | - Mohd Zaini Asmawi
- School of Pharmaceutical Sciences, University of Science Malaysia, Pulau Pinang, Malaysia
| | - Xianju Huang
- College of Pharmacy, South-Central University for Nationalities, Wuhan, China
| |
Collapse
|
8
|
Su Y, Zhou W, Zhang Y, Wang X, Han B. Identification And validation of transcription factor genes involved in prostate cancer metastasis. ALL LIFE 2021. [DOI: 10.1080/26895293.2021.1915394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Yiming Su
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Wenhao Zhou
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yu Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Xiaohai Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Bangmin Han
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
9
|
Paul T, Roy R, Sarkar RD, Sinha S, Biswas N. H 2O 2 mediated FLIP and XIAP down-regulation involves increased ITCH expression and ERK-Akt crosstalk in imatinib resistant Chronic Myeloid Leukemia cell line K562. Free Radic Biol Med 2021; 166:265-276. [PMID: 33631302 DOI: 10.1016/j.freeradbiomed.2021.02.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 01/29/2021] [Accepted: 02/13/2021] [Indexed: 11/19/2022]
Abstract
Regulation of anti-apoptotic protein FLICE-like inhibitory protein (FLIP) and X-linked inhibitor of apoptosis protein (XIAP) remains a crucial step in the cell fate determination and thus targeting these anti-apoptotic proteins could be a viable strategy for the treatment of cancer. However the regulation of FLIP and XIAP is not very well established till date. Here we have shown that ROS decreased XIAP and FLIP by activation of ubiquitin-proteasomal pathway in imatinib resistant K562 cells. Activation of the components of MAPK pathway, ERK and JNK, played a crucial role in XIAP and FLIP degradation because ectopic expression or knock down of ERK and JNK changed the pattern of ROS mediated down-regulation of these two proteins. We have also found that JNK and ERK differentially regulates FLIP and XIAP, respectively. Moreover, our data suggests that activated ERK decreased Akt phosphorylation and thus its binding to and stabilization of XIAP. On the other hand, JNK activation increased E3 ubiquitin ligase ITCH expression and its binding to FLIP which leads to its degradation. Thus, we have, for the first time elucidated that ROS mediated ERK-Akt crosstalk regulates XIAP. We have also shown for the first time that ROS regulates ITCH expression which controls FLIP degradation.
Collapse
Affiliation(s)
- Tamalika Paul
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Rajdeep Roy
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Rupak Dey Sarkar
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Samraj Sinha
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Nabendu Biswas
- Department of Life Sciences, Presidency University, Kolkata, India.
| |
Collapse
|
10
|
Xiang H, Wang S, Kong X, Yu Y, Shen L, Long C, Liu X, Wei GH. c-Fos is upregulated in the genital tubercle of DEHP-induced hypospadiac rats and the prepuce of patients with hypospadias. Syst Biol Reprod Med 2021; 67:193-200. [PMID: 33618583 DOI: 10.1080/19396368.2020.1862356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
This study aimed to investigate the expression of Fos proto-oncogene, AP-1 transcription factor subunit (c-Fos) in the genital tubercle (GT) of rats with di(2-ethylhexyl) phthalate (DEHP)-induced hypospadias and in the prepuce of patients with hypospadias compared with unaffected controls. Pregnant rats were given 750 mg/kg/day DEHP orally from gestational days 12-19. Western blotting showed that c-Fos expression was increased in DEHP-induced hypospadiac male offspring. In addition, 30 prepuce tissue specimens obtained during hypospadias repair surgery were divided into 2 groups: the mild hypospadias group (n = 15) and the severe hypospadias group (n = 15). Fifteen normal prepuce tissue specimens were harvested during elective circumcision as normal controls. Real-time quantitative polymerase chain reaction, western blotting and immunohistochemistry analyses were used to assess c-Fos expression. c-Fos protein levels were higher in the GT of DEHP-induced rats than in that of control rats. c-Fos mRNA and protein levels were also higher in the hypospadias groups than in the control group (p < 0.05, p < 0.001), and c-Fos protein levels were significantly higher in the severe hypospadias group than in the mild hypospadias group (p < 0.01). The expression of c-Fos was increased in both the GT of DEHP-induced hypospadiac rats and the prepuce of hypospadias patients. Thus, c-Fos overexpression might contribute to hypospadias.Abbreviations: DEHP: di(2-ethylhexyl) phthalate; c-Fos: Fos proto-oncogene, AP-1 transcription factor subunit; Mafb: the masculinization-regulatory gene v-maf musculoaponeurotic fibrosarcoma oncogene family, protein B; GT: genital tubercle; ED: embryonic day; AGD: anogenital distance; AGI: anogenital distance index; ED: embryonic day.
Collapse
Affiliation(s)
- Han Xiang
- Department of Urology; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Child Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Shao Wang
- Department of Urology; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Child Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyan Kong
- Department of Urology; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Child Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yihang Yu
- Department of Urology; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Child Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Lianju Shen
- Department of Urology; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Child Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Chunlan Long
- Department of Urology; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Child Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xing Liu
- Department of Urology; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Child Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Guang-Hui Wei
- Department of Urology; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Child Urogenital Development and Tissue Engineering; Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Gammacoronavirus Avian Infectious Bronchitis Virus and Alphacoronavirus Porcine Epidemic Diarrhea Virus Exploit a Cell-Survival Strategy via Upregulation of cFOS to Promote Viral Replication. J Virol 2021; 95:JVI.02107-20. [PMID: 33239458 PMCID: PMC7851560 DOI: 10.1128/jvi.02107-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Coronaviruses have evolved a variety of strategies to optimize cellular microenvironment for efficient replication. In this study, we report the induction of AP-1 transcription factors by coronavirus infection based on genome-wide analyses of differentially expressed genes in cells infected with avian coronavirus infectious bronchitis virus (IBV). Most members of the AP-1 transcription factors were subsequently found to be upregulated during the course of IBV and porcine epidemic diarrhea virus (PEDV) infection of cultured cells as well as in IBV-infected chicken embryos. Further characterization of the induction kinetics and functional roles of cFOS in IBV replication demonstrated that upregulation of cFOS at early to intermediate phases of IBV replication cycles suppresses IBV-induced apoptosis and promotes viral replication. Blockage of nuclear translocation of cFOS by peptide inhibitor NLSP suppressed IBV replication and apoptosis, ruling out the involvement of the cytoplasmic functions of cFOS in the replication of IBV. Furthermore, knockdown of ERK1/2 and inhibition of JNK and p38 kinase activities reduced cFOS upregulation and IBV replication. This study reveals an important function of cFOS in the regulation of coronavirus-induced apoptosis, facilitating viral replication.IMPORTANCE The ongoing pandemic of coronavirus disease 2019 (COVID-19), caused by a newly emerged zoonotic coronavirus (SARS-CoV-2), highlights the importance of coronaviruses as human and animal pathogens and our knowledge gaps in understanding the cellular mechanisms, especially mechanisms shared among human and animal coronaviruses, exploited by coronaviruses for optimal replication and enhanced pathogenicity. This study reveals that upregulation of cFOS, along with other AP-1 transcription factors, as a cell-survival strategy is such a mechanism utilized by coronaviruses during their replication cycles. Through induction and regulation of apoptosis of the infected cells at early to intermediate phases of the replication cycles, subtle but appreciable differences in coronavirus replication efficiency were observed when the expression levels of cFOS were manipulated in the infected cells. As the AP-1 transcription factors are multi-functional, further studies of their regulatory roles in proinflammatory responses may provide new insights into the pathogenesis and virus-host interactions during coronavirus infection.
Collapse
|
12
|
Tu Y, Wu W, Guo Y, Lu F, Li X, Xu D, Zou D, Tu Y, Chai Y, He L. Up-regulation of hsa-miR-221-3p induced by UVB affects proliferation and apoptosis of keratinocytes via Bcl-xL/Bax pathway. PHOTODERMATOLOGY PHOTOIMMUNOLOGY & PHOTOMEDICINE 2021; 37:269-277. [PMID: 33351232 DOI: 10.1111/phpp.12647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 12/19/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Chronic actinic dermatitis (CAD) is a photoallergic skin disease with abnormal hyperplasia. At present, the mechanism of abnormal proliferation is not clear. OBJECTIVE To explore possible mechanism of CAD proliferative lesions. METHODS Immunohistochemistry (IHC) assay and small RNA sequencing were carried out. Quantitative real-time PCR (qRT-PCR) analysis was performed to evaluate expression levels of hsa-miR-221-3p and FOS. The interaction between hsa-miR-221-3p and FOS was identified by dual-luciferase reporter assay. Expression of hsa-miR-221-3p also was detected by qRT-PCR after UVB irradiation. Influences of hsa-miR-221-3p and FOS on cell viability and apoptosis were assessed through a series of functional experiments and rescue experiments. Western blot analysis was used to detect protein expression of fos, Bax, Bcl-xL, and caspase-3. RESULTS Patients with CAD had marked epidermal hyperplasia. The expression of hsa-miR-221-3p was up-regulated in CAD while FOS was significantly down-regulated. Dual-luciferase reporter assay confirmed that hsa-miR-221-3p targeted FOS 3'UTR. Hsa-miR-221-3p induced by UVB ranged from 0 to 30 mJ. Moreover, hsa-miR-221-3p overexpression or FOS knockdown promoted cell proliferation and reduced cell apoptosis. Western blot showed that hsa-miR-221-3p negatively regulated fos, which regulated Bcl-xL/Bax. Cell proliferation caused by hsa-miR-221-3p overexpression or FOS knockdown could be reversed by Bcl-xL inhibitor. CONCLUSION Hsa-miR-221-3p induced by UVB targeted FOS 3'UTR, which played an important role in regulating proliferation and apoptosis of keratinocytes via Bcl-xL/Bax pathway; this may provide a new insight for CAD proliferative lesions.
Collapse
Affiliation(s)
- Yunhua Tu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China.,Department of Dermatology, The Second People's Hospital of Guiyang, Guizhou, China
| | - Wenjuan Wu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yanni Guo
- Department of Dermatology, The Second Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Fengyan Lu
- Department of Dermatology, The First People's Hospital of Qujing, Qujing, China
| | - Xing Li
- Department of Dermatology, People's Hospital of Chuxiong Yi Autonomous Prefecture, Chuxiong, China
| | - Dan Xu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Dandan Zou
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ying Tu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yanjie Chai
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li He
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
13
|
Ye Y, Huang Z, Chen M, Mo Y, Mo Z. Luteolin Potentially Treating Prostate Cancer and COVID-19 Analyzed by the Bioinformatics Approach: Clinical Findings and Drug Targets. Front Endocrinol (Lausanne) 2021; 12:802447. [PMID: 35178029 PMCID: PMC8844187 DOI: 10.3389/fendo.2021.802447] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/23/2021] [Indexed: 12/17/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a serious epidemic, characterized by potential mutation and can bring about poor vaccine efficiency. It is evidenced that patients with malignancies, including prostate cancer (PC), may be highly vulnerable to the SARS-CoV-2 infection. Currently, there are no existing drugs that can cure PC and COVID-19. Luteolin can potentially be employed for COVID-19 treatment and serve as a potent anticancer agent. Our present study was conducted to discover the possible drug target and curative mechanism of luteolin to serve as treatment for PC and COVID-19. The differential gene expression of PC cases was determined via RNA sequencing. The application of network pharmacology and molecular docking aimed to exhibit the drug targets and pharmacological mechanisms of luteolin. In this study, we found the top 20 up- and downregulated gene expressions in PC patients. Enrichment data demonstrated anti-inflammatory effects, where improvement of metabolism and enhancement of immunity were the main functions and mechanism of luteolin in treating PC and COVID-19, characterized by associated signaling pathways. Additional core drug targets, including MPO and FOS genes, were computationally identified accordingly. In conclusion, luteolin may be a promising treatment for PC and COVID-19 based on bioinformatics findings, prior to future clinical validation and application.
Collapse
Affiliation(s)
- Yu Ye
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ziyan Huang
- Health Management Department, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Manying Chen
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yongfeng Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zengnan Mo
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- *Correspondence: Zengnan Mo,
| |
Collapse
|
14
|
Hou C, Cai H, Zhu Y, Huang S, Song F, Hou J. Development and Validation of Autophagy-Related Gene Signature and Nomogram for Predicting Survival in Oral Squamous Cell Carcinoma. Front Oncol 2020; 10:558596. [PMID: 33178587 PMCID: PMC7596585 DOI: 10.3389/fonc.2020.558596] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 09/24/2020] [Indexed: 12/19/2022] Open
Abstract
Background Autophagy, a highly conserved self-digesting process, has been deeply involved in the development and progression of oral squamous cell carcinoma (OSCC). However, the prognostic value of autophagy-related genes (ARGs) for OSCC still remains unclear. Our study set out to develop a multigene expression signature based on ARGs for individualized prognosis assessment in OSCC patients. Methods Based on The Cancer Genome Atlas (TCGA) database, we identified prognosis-related ARGs through univariate COX regression analysis. Then we performed the least absolute shrinkage and selection operator (LASSO) regression analysis to identify an optimal autophagy-related multigene signature with the subsequent validation in testing set, GSE41613 and GSE42743 datasets. Results We identified 36 prognosis-related ARGs for OSCC. Subsequently, the multigene signature based on 13 prognostic ARGs was constructed and successfully divided OSCC patients into low and high-risk groups with significantly different overall survival in TCGA training set (p < 0.0001). The autophagy signature remained as an independent prognostic factor for OSCC in univariate and multivariate Cox regression analyses. The area under the curve (AUC) values of the receiver operating characteristic (ROC) curves for 1, 3, and 5-year survival were 0.758, 0.810, 0.798, respectively. Then the gene signature was validated in TCGA testing set, GSE41613 and GSE42743 datasets. Moreover, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and single-sample gene set enrichment analysis (ssGSEA) revealed the underlying biological characteristics and signaling pathways associated with this signature in OSCC. Finally, we constructed a nomogram by combining the gene signature with multiple clinical parameters (age, gender, TNM-stage, tobacco, and alcohol history). The concordance index (C-index) and calibration plots demonstrated favorable predictive performance of our nomogram. Conclusion In summary, we identified and verified a 13-ARGs prognostic signature and nomogram, which provide individualized prognosis evaluation and show insight for potential therapeutic targets for OSCC.
Collapse
Affiliation(s)
- Chen Hou
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Hongshi Cai
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yue Zhu
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Shuojin Huang
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Fan Song
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jinsong Hou
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
15
|
Soliman MM, Elshazly SA, Aldhahrani A. Gamma-irradiation-induced testicular oxidative stress and apoptosis: Mitigation by l-carnitine. J Biochem Mol Toxicol 2020; 34:e22565. [PMID: 32645248 DOI: 10.1002/jbt.22565] [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: 01/25/2020] [Revised: 05/15/2020] [Accepted: 06/17/2020] [Indexed: 01/01/2023]
Abstract
The current study evaluated the potential ameliorative and protective impacts of l-carnitine (L-CAR) against γ-irradiation (RAD)-induced oxidative stress and apoptosis in mice testes. Male Swiss mice were allocated into four groups (n = 7). Group 1 served as a control that received saline intraperitoneally (IP). Group 2 received L-CAR (10 mg/kg bw/day; IP in saline) for 17 days. Group 3 received saline for 17 days and on day 7 exposed to RAD at a dose of 0.1 Gy per day for consecutive 10 days. Group 4 (L-CAR + RAD), received L-CAR same as in group 2 and on day 7 exposed to RAD for consecutive 10 days. Testicular antioxidants (malondialdehyde, MDA; γ-glutamyl-cysteine synthetase, gGCS; and catalase) were altered by γ-irradiation. Preadministration of L-CAR protected γ-irradiated mice from altered changes induced by γ-irradiation. γ-Irradiation affected the mRNA expression of pro-apoptotic, apoptotic, and anti-apoptotic genes (c-jun, c-fos, Bcl-xl, caspase-3, and BAX). All altered genes were ameliorated by prior l-carnitine administration to γ-irradiated mice. Testicular cells showed deformities and edema with congestion in seminiferous tubules and strong immunoreactivity for caspase-9 and a decrease in immunoreactivity of Bcl-2 in histological and immunohistochemical examination. Prior administration of L-CAR to γ-irradiated mice protected this group from reported changes in caspase-9 and Bcl-2 immunostaining. In conclusion, the current study provides evidence for the protective and ameliorative impacts of L-CAR against γ-irradiation-induced testicular oxidative stress and apoptosis at biochemical, molecular, and cellular levels.
Collapse
Affiliation(s)
- Mohamed M Soliman
- Clinical Laboratory Sciences, Turabah University College, Taif University, Turabah, Saudi Arabia.,Biochemistry Department, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Samir A Elshazly
- Biochemistry Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Adil Aldhahrani
- Clinical Laboratory Sciences, Turabah University College, Taif University, Turabah, Saudi Arabia
| |
Collapse
|
16
|
Guo L, Lin M, Cheng Z, Chen Y, Huang Y, Xu K. Identification of key genes and multiple molecular pathways of metastatic process in prostate cancer. PeerJ 2019; 7:e7899. [PMID: 31637138 PMCID: PMC6800981 DOI: 10.7717/peerj.7899] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 09/16/2019] [Indexed: 12/24/2022] Open
Abstract
Background Cancer metastasis is well known as the most adverse outcome and the major cause of mortality in cancer patients, including prostate cancer (PCa). There are no credible predictors, to this day, that can reflect the metastatic ability of localized PCa. In the present study, we firstly identified the differentially expressed genes (DEGs) and molecular pathways involved in the metastaic process of PCa by comparing gene expressions of metastaic PCa with localized PCa directly, with the purpose of identifying potential markers or therapeutic targets. Methods The gene expression profiles (GSE6919 and GSE32269) were downloaded from the Gene Expression Omnibus database, which contained 141 tissue samples, including 87 primary localized PCa samples and 54 metastaic PCa samples. After data processing, DEGs were identified by R language using the Student’s t-test adjusted via the Beniamini–Hochberg method. Subsequently, the gene ontology functional and pathway enrichment analyses of DEGs were performed and the protein–protein interaction network was constructed. Hub genes were identified using the plug-in cytoHubba in Cytoscape software by MCC and degree. Furthermore, validation and prognostic significance analysis of the hub genes were performed by UALCAN and gene expression profiling interactive analysis (GEPIA). Results A total of 90 DEGs were identified between localized and metastaic PCa, which consisted of 47 upregulated and 43 downregulated genes. The enriched functions and pathways of the DEGs include catabolic process, cell cycle, response to steroid hormone, extracellular matrix (ECM)-receptor interaction and vascular smooth muscle contraction. A total of 10 genes were identified as hub genes and biological process analysis of hub genes showed that cell cycle phase, cell division, and mitotic cell cycle process were mainly enriched. The expression of hub genes were confirmed in metastaic PCa when compared with localized PCa tissues by The Cancer Genome Atlas database. Moreover, the disease-free survival analysis of hub genes revealed that these genes may play an important role in invasion, progression or recurrence. Therefore, these hub genes might be the key genes contributed to tumor progression or metastasis in PCa and provide candidate therapeutic targets for PCa. Conclusions The present study identified some DEGs between localized and metastaic PCa tissue samples. These key genes might be potential therapeutic targets and biomarkers for the metastaic process of PCa.
Collapse
Affiliation(s)
- Lihuang Guo
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Mingyue Lin
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Zhenbo Cheng
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Yi Chen
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Yue Huang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Keqian Xu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
17
|
Peng J, Dou YQ, Liang H, He S, Liang XF, Shi LJ. Social Learning of Acquiring Novel Feeding Habit in Mandarin Fish ( Siniperca chuatsi). Int J Mol Sci 2019; 20:ijms20184399. [PMID: 31500232 PMCID: PMC6770103 DOI: 10.3390/ijms20184399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 08/29/2019] [Accepted: 09/02/2019] [Indexed: 12/04/2022] Open
Abstract
Social learning plays important roles in gaining new foraging skills and food preferences. However, the potential role and molecular mechanism of social learning in acquiring new feeding habits is less clear in fish. In the present study, we examined the success rate of feeding habit domestication from live prey fish to dead prey fish, as well as the food intake of dead prey fish in mandarin fish with or without feeders of dead prey fish as demonstrators. Here, we found that mandarin fish can learn from each other how to solve novel foraging tasks, feeding on dead prey fish. In addition, the analysis of gene expressions and signaling pathways of learning through Western blotting and transcriptome sequencing shows that the expression of the c-fos, fra2, zif268, c/ebpd and sytIV genes were significantly increased, and the anorexigenic pomc and leptin a expressions were decreased in fish of the learning group. The phosphorylation levels of protein kinase A (PKA) and Ca2+/calmodulin-dependent protein kinase II (CaMKII) in the learning group were significantly higher than those of the control group, while the phosphorylation level of S6 ribosomal protein (S6) was lower. With the inhibitors of PKA and CaMKII signaling and the chromatin immunoprecipitation (ChIP) assay, we further found that the social learning of new feeding habits in mandarin fish could be attributed to the activation of the CaMKII signaling pathway and then the stimulation of the expression of the c-fos gene, which might be an important transcriptional factor to inhibit the expression of the anorexigenic gene pomc, resulting in the food intake of dead prey fish in mandarin fish. Altogether, our results support the hypothesis that social learning could facilitate the acquisition of novel feeding habits in fish, and it considerably increases the rate of subsequent individual food intake and domestication through the interaction between the learning gene c-fos and the appetite control gene pomc.
Collapse
Affiliation(s)
- Jian Peng
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China
- Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, China
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan 430070, China
| | - Ya-Qi Dou
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China
- Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, China
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan 430070, China
| | - Hui Liang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China
- Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, China
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan 430070, China
| | - Shan He
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China.
- Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, China.
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan 430070, China.
| | - Xu-Fang Liang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China
- Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, China
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan 430070, China
| | - Lin-Jie Shi
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan 430070, China
- Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan 430070, China
- Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Wuhan 430070, China
| |
Collapse
|
18
|
Le Clorennec C, Lazrek Y, Dubreuil O, Sampaio C, Larbouret C, Lanotte R, Poul MA, Barret JM, Prost JF, Pèlegrin A, Chardès T. ITCH-dependent proteasomal degradation of c-FLIP induced by the anti-HER3 antibody 9F7-F11 promotes DR5/caspase 8-mediated apoptosis of tumor cells. Cell Commun Signal 2019; 17:106. [PMID: 31443721 PMCID: PMC6708219 DOI: 10.1186/s12964-019-0413-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/05/2019] [Indexed: 12/13/2022] Open
Abstract
Background HER3/ErbB3 receptor deletion or blockade leads to tumor cell apoptosis, whereas its overexpression confers anti-cancer drug resistance through upregulation of protective mechanisms against apoptosis. We produced the anti-HER3 antibody 9F7-F11 that promotes HER3 ubiquitination and degradation via JNK1/2-dependent activation of the E3 ubiquitin ligase ITCH, and that induces apoptosis of cancer cells. Cellular FLICE-like inhibitory protein (c-FLIP) is a key regulator of apoptotic pathways. Here, we wanted to determine the mechanisms underlying the pro-apoptotic effect of 9F7-F11. Methods Anti-HER3 antibody-induced apoptosis was assessed by western blot, and by flow cytometry measurement of Annexin V/7-AAD-labelled tumor cells (BxPC3, MDA-MB-468 and DU145 cell lines). c-FLIP/ITCH interaction and subsequent degradation/ubiquitination were investigated by co-immunoprecipitation of ITCH-silenced vs scramble control cells. The relationship between ITCH-mediated c-FLIP degradation and antibody-induced apoptosis was examined by western blot and flow cytometry of tumor cells, after ITCH RNA interference or by pre-treatment with ITCH chemical inhibitor chlorimipramine (CI). Results Following incubation with 9F7-F11, cancer cell apoptosis occurs through activation of caspase-8, − 9 and − 3 and the subsequent cleavage of poly (ADP-ribose) polymerase (PARP). Moreover we showed that ubiquitination and proteasomal degradation of the anti-apoptotic protein c-FLIP was mediated by USP8-regulated ITCH recruitment. This effect was abrogated by ITCH- and USP8-specific RNA interference (siRNA), or by the ITCH chemical inhibitor CI. Specifically, ITCH silencing or CI blocked 9F7-F11-induced caspase-8-mediated apoptosis of tumor cells, and restored c-FLIP expression. ITCH-silencing or CI concomitantly abrogated HER3-specific antibody-induced apoptosis of Annexin V/7-AAD-labelled BxPC3 cells. 9F7-F11 favored the extrinsic apoptosis pathway by inducing TRAIL-R2/DR5 upregulation and TRAIL expression that promoted the formation of death-inducing signaling complex (DISC), leading to caspase-8-mediated apoptosis. Incubation with 9F7-F11 also induced BID cleavage, BAX upregulation and BIM expression, which initiated the caspase-9/3-mediated mitochondrial death pathway. The anti-HER3 antibody pro-apoptotic effect occurred concomitantly with downregulation of the pro-survival proteins c-IAP2 and XIAP. Conclusions The allosteric non-neuregulin competing modulator 9F7-F11, sensitizes tumor cells to DR5/caspase-8-mediated apoptosis through ITCH-dependent downregulation of c-FLIP. Graphical abstract ![]()
Electronic supplementary material The online version of this article (10.1186/s12964-019-0413-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christophe Le Clorennec
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), F-34298, Montpellier, France.,Present Address: UCSD School of Medicine, Moores Cancer Center, La Jolla, CA, 92093-0815, USA
| | - Yassamine Lazrek
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), F-34298, Montpellier, France.,Present Address: Institut Pasteur de Guyane, F- 97306, Cayenne, France
| | - Olivier Dubreuil
- GamaMabs Pharma SA, Centre Pierre Potier, F-31106, Toulouse, France
| | - Carla Sampaio
- Laboratoire d'Immunologie et d'Immunothérapie des Cancers, EA7269, Université Bourgogne Franche-Comté, F-21000, Dijon, France
| | - Christel Larbouret
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), F-34298, Montpellier, France
| | - Romain Lanotte
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), F-34298, Montpellier, France
| | - Marie-Alix Poul
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), F-34298, Montpellier, France
| | - Jean-Marc Barret
- GamaMabs Pharma SA, Centre Pierre Potier, F-31106, Toulouse, France
| | | | - André Pèlegrin
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), F-34298, Montpellier, France
| | - Thierry Chardès
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), F-34298, Montpellier, France. .,Centre National de la Recherche Scientifique (CNRS), Paris, France.
| |
Collapse
|
19
|
Abstract
Chemerin is a multifunctional adipokine with established roles in inflammation, adipogenesis and glucose homeostasis. Increasing evidence suggest an important function of chemerin in cancer. Chemerin's main cellular receptors, chemokine-like receptor 1 (CMKLR1), G-protein coupled receptor 1 (GPR1) and C-C chemokine receptor-like 2 (CCRL2) are expressed in most normal and tumor tissues. Chemerin's role in cancer is considered controversial, since it is able to exert both anti-tumoral and tumor-promoting effects, which are mediated by different mechanisms like recruiting innate immune defenses or activation of endothelial angiogenesis. For this review article, original research articles on the role of chemerin and its receptors in cancer were considered, which are listed in the PubMed database. Additionally, we included meta-analyses of publicly accessible DNA microarray data to elucidate the association of expression of chemerin and its receptors in tumor tissues with patients' survival.
Collapse
|
20
|
Increased Expression of MicroRNA 551a by c-Fos Reduces Focal Adhesion Kinase Levels and Blocks Tumorigenesis. Mol Cell Biol 2019; 39:MCB.00577-18. [PMID: 30670568 DOI: 10.1128/mcb.00577-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 01/03/2019] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is a recurrent type of cancer among women worldwide. Despite remarkable progress in the prevention, detection, and treatment of breast cancer, it still remains a major chronic problem worldwide and poses significant challenges, like metastasis to distant organs, demanding the need for novel biomarkers and therapeutic targets. Focal adhesion kinase (FAK), a member of the protein tyrosine kinases, has been shown to be expressed in high levels in breast tumors. Of late, FAK has emerged as an impending curative target in breast carcinoma, with few of the small molecular inhibitors reaching the clinical trial stage. In the current study, we established that microRNA 551a (miR-551a) precisely regulates FAK by binding to the complementary sequences in the 3' untranslated region (UTR) of mRNAs of FAK and inhibits its expression in breast carcinoma cell lines. Further, results from human breast carcinoma samples illustrated that miR-551a levels were substantially downregulated in tumor samples, with a concurrent rise in the expression of FAK. Functional experimental studies using miR-551a-overexpressing breast cancer cells and nude mouse xenograft models revealed the tumor suppressor role of miR-551a. We also found that miR-551a expression decreased the invasion and migratory ability of breast carcinoma cells by inhibiting MMP-9 activity. Regulation studies performed utilizing promoter luciferase assays, chromatin immunoprecipitation (ChIP), and electrophoretic mobility shift assay (EMSA) revealed that c-Fos binds to the miR-551a promoter and activates it. Further, we observed a considerable increase in the amount of miR-551a levels upon c-Fos overexpression. All of these results showed that miR-551a can be of clinical relevance in understanding the regulation of FAK in breast tumorigenesis.
Collapse
|
21
|
Wang XP, Qi XF, Yang B, Chen SY, Wang JY. RNA-Seq analysis of duck embryo fibroblast cell gene expression during the early stage of egg drop syndrome virus infection. Poult Sci 2019; 98:404-412. [PMID: 30690613 DOI: 10.3382/ps/pey318] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Egg drop syndrome virus (EDSV), a member of the family Adenoviridae and an economically important pathogen with a broad host range, leads to markedly decreased egg production. However, the molecular mechanism underlying the host-EDSV interaction remains unclear. Here, we performed high-throughput RNA sequencing (RNA-Seq) to study the dynamic changes in host gene expression at 6, 12, and 24 hours post-infection in duck embryo fibroblasts (DEFs) infected with EDSV. Atotal of 441 differentially expressed genes (DEGs) were identified after EDSV infection. Gene Ontology category and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis revealed that these DEGs were associated with multiple biological functions, including signal transduction, host immunity, virus infection, cell apoptosis, cell proliferation, and pathogenicity-related and metabolic process signaling pathways. We screened and identified 12 DEGs for further examination by using qRT-PCR. The qRT-PCR and RNA-Seq results were highly consistent. This study analyzed viral infection and host immunity induced by EDSV infection from a novel perspective, and the results provide valuable information regarding the mechanisms underlying host-EDSV interactions, which will prove useful for the future development of antiviral drugs or vaccines for poultry, thus benefiting the entire poultry industry.
Collapse
Affiliation(s)
- X P Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - X F Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - B Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - S Y Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - J Y Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
22
|
Han L, Yang X, Sun W, Li Z, Ren H, Li B, Zhang R, Zhang D, Shi Z, Liu J, Cao J, Zhang J, Xiong Y. The study of GPX3 methylation in patients with Kashin-Beck Disease and its mechanism in chondrocyte apoptosis. Bone 2018; 117:15-22. [PMID: 30153510 DOI: 10.1016/j.bone.2018.08.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/06/2018] [Accepted: 08/23/2018] [Indexed: 10/28/2022]
Abstract
OBJECTIVE Selenium deficiency is a risk factor for Kashin-Beck Disease (KBD), an endemic osteoarthropathy. Although promoter hypermethylation of glutathione peroxidase 3 (GPX3) (a selenoprotein) has been identified in several cancers, little is known about promoter methylation and expression of GPX3 and their relation to selenium in KBD. The present study was thus conducted to investigate this research question. METHODS Methylation and expressions of GPX3 in whole blood drawn from 288 KBD patients and 362 healthy controls and in chondrocyte cell line were evaluated using methylation-specific PCR and qRT-PCR, respectively. The protein levels of PI3K/Akt/c-fos signaling in the whole blood and chondrocyte cell line were determined with Western blotting. Chondrocytes apoptosis were detected by Hoechst 33342 and Annexin V-FITC/PI staining. RESULTS GPX3 methylation was increased, GPX3 mRNA was decreased, and protein levels in the PI3K/Akt/c-fos signaling pathway were up-regulated in the whole blood collected from KBD patients as compared with healthy controls. Similar results were obtained for chondrocytes injured by oxidative stress. There was a significant, decreasing trend in GPX3 expression across groups of unmethylation, partial methylation, and complete methylation for GPX3, in sequence. Compared with unmethylation group, protein levels in PI3K/Akt/c-fos pathway were enhanced in partial and complete methylation groups. Treatment of chondrocytes with sodium selenite resulted in reduced methylation and increased expression of GPX3 as well as down-regulated level of PI3K/Akt/c-fos proteins. CONCLUSIONS The methylation and expression of GPX3 and expression of PI3K/Akt/c-fos pathway are altered in KBD and these changes are reversible by selenium supplementation.
Collapse
Affiliation(s)
- LiXin Han
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi 710061, P.R. China
| | - XiaoLi Yang
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi 710061, P.R. China
| | - WenYan Sun
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi 710061, P.R. China
| | - ZhaoFang Li
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi 710061, P.R. China
| | - Hao Ren
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi 710061, P.R. China
| | - BaoRong Li
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi 710061, P.R. China
| | - RongQiang Zhang
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi 710061, P.R. China
| | - DanDan Zhang
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi 710061, P.R. China
| | - ZiYun Shi
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi 710061, P.R. China
| | - JiFeng Liu
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi 710061, P.R. China
| | - JunLing Cao
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi 710061, P.R. China
| | - JianJun Zhang
- Department of Epidemiology, Indiana University Richard M. Fairbanks School of Public Health, Indianapolis, 1050 Wishard Boulevard, IN 46202, USA
| | - YongMin Xiong
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Xi'an Jiaotong University Health Science Center, No.76 Yanta West Road, Xi'an, Shaanxi 710061, P.R. China.
| |
Collapse
|
23
|
Hop HT, Arayan LT, Huy TXN, Reyes AWB, Vu SH, Min W, Lee HJ, Rhee MH, Chang HH, Kim S. The Key Role of c-Fos for Immune Regulation and Bacterial Dissemination in Brucella Infected Macrophage. Front Cell Infect Microbiol 2018; 8:287. [PMID: 30186773 PMCID: PMC6110913 DOI: 10.3389/fcimb.2018.00287] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/27/2018] [Indexed: 12/25/2022] Open
Abstract
The cellular oncogene c-Fos (c-Fos) is a component of activator protein 1 (AP1), a master transcriptional regulator of cells. The suppression of c-Fos signaling by siRNA treatment resulted in significant induction of TLR4, which subsequently activates p38 and ERK1/2 mitogen-activated protein kinases (MAPKs) and enhances F-actin polymerization, leading to an increase in B. abortus phagocytosis. During B. abortus infection, c-Fos signaling is induced, which activates the downstream innate-immunity signaling cascade for bacterial clearance. The inhibition of c-Fos signaling led to increased production of interleukin 10 (IL-10), which partially suppressed lysosome-mediated killing, resulting in increased survival of B. abortus inside macrophages. We present evidence of the regulatory role played by the c-Fos pathway in proliferation during B. abortus infection; however, this was independent of the anti-Brucella effect of this pathway. Another finding is the essential contribution of c-Fos/TRAIL to infected-cell necrosis, which is a key event in bacterial dissemination. These data provide the mechanism via which c-Fos participates in host defense mechanisms against Brucella infection and in bacterial dissemination by macrophages.
Collapse
Affiliation(s)
- Huynh T Hop
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Lauren T Arayan
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Tran X N Huy
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Alisha W B Reyes
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Son H Vu
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - WonGi Min
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Hu J Lee
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Man H Rhee
- College of Veterinary Medicine, Kyungpook National University, Daegu, South Korea
| | - Hong H Chang
- Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, South Korea
| | - Suk Kim
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea.,Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, South Korea
| |
Collapse
|
24
|
Yan M, Yang X, Wang H, Shao Q. The critical role of histone lysine demethylase KDM2B in cancer. Am J Transl Res 2018; 10:2222-2233. [PMID: 30210666 PMCID: PMC6129528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/05/2018] [Indexed: 06/08/2023]
Abstract
The discovery of histone demethylases has revealed the dynamic nature of the regulation of histone methylation. KDM2B is an important histone lysine demethylase that removes methyl from H3K36me2 and H3K4me3. It participates in many aspects of normal cellular processes such as cell senescence, cell differentiation and stem cell self-renewal. Recent studies also showed that KDM2B was overexpressed in various types of cancers. This review focuses primarily on the current knowledge of KDM2B and its function in cancer development.
Collapse
Affiliation(s)
- Meina Yan
- Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University Zhenjiang 212013, Jiangsu, China
| | - Xinxin Yang
- Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University Zhenjiang 212013, Jiangsu, China
| | - Hui Wang
- Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University Zhenjiang 212013, Jiangsu, China
| | - Qixiang Shao
- Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University Zhenjiang 212013, Jiangsu, China
| |
Collapse
|
25
|
Humphreys L, Espona-Fiedler M, Longley DB. FLIP as a therapeutic target in cancer. FEBS J 2018; 285:4104-4123. [PMID: 29806737 DOI: 10.1111/febs.14523] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/11/2018] [Accepted: 05/24/2018] [Indexed: 12/13/2022]
Abstract
One of the classic hallmarks of cancer is disruption of cell death signalling. Inhibition of cell death promotes tumour growth and metastasis, causes resistance to chemo- and radiotherapies as well as targeted agents, and is frequently due to overexpression of antiapoptotic proteins rather than loss of pro-apoptotic effectors. FLIP is a major apoptosis-regulatory protein frequently overexpressed in solid and haematological cancers, in which its high expression is often correlated with poor prognosis. FLIP, which is expressed as long (FLIP(L)) and short (FLIP(S)) splice forms, achieves its cell death regulatory functions by binding to FADD, a critical adaptor protein which links FLIP to the apical caspase in the extrinsic apoptotic pathway, caspase-8, in a number of cell death regulating complexes, such as the death-inducing signalling complexes (DISCs) formed by death receptors. FLIP also plays a key role (together with caspase-8) in regulating another form of cell death termed programmed necrosis or 'necroptosis', as well as in other key cellular processes that impact cell survival, including autophagy. In addition, FLIP impacts activation of the intrinsic mitochondrial-mediated apoptotic pathway by regulating caspase-8-mediated activation of the pro-apoptotic Bcl-2 family member Bid. It has been demonstrated that FLIP can not only inhibit death receptor-mediated apoptosis, but also cell death induced by a range of clinically relevant chemotherapeutic and targeted agents as well as ionizing radiation. More recently, key roles for FLIP in promoting the survival of immunosuppressive tumour-promoting immune cells have been discovered. Thus, FLIP is of significant interest as an anticancer therapeutic target. In this article, we review FLIP's biology and potential ways of targeting this important tumour and immune cell death regulator.
Collapse
Affiliation(s)
- Luke Humphreys
- Drug Resistance Group, Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK
| | - Margarita Espona-Fiedler
- Drug Resistance Group, Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK
| | - Daniel B Longley
- Drug Resistance Group, Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK
| |
Collapse
|
26
|
Ryu S, Ahn YJ, Yoon C, Chang JH, Park Y, Kim TH, Howland AR, Armstrong CA, Song PI, Moon AR. The regulation of combined treatment-induced cell death with recombinant TRAIL and bortezomib through TRAIL signaling in TRAIL-resistant cells. BMC Cancer 2018; 18:432. [PMID: 29661248 PMCID: PMC5902847 DOI: 10.1186/s12885-018-4352-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 04/09/2018] [Indexed: 12/14/2022] Open
Abstract
Background Multiple trials have attempted to demonstrate the effective induction of cell death in TRAIL-resistant cancer cells, including using a combined treatment of recombinant TRAIL and various proteasome inhibitors. These studies have yielded limited success, as the mechanism of cell death is currently unidentified. Understanding this mechanism’s driving forces may facilitate the induction of cell death in TRAIL-resistant cancer cells. Methods Three kinds of recombinant soluble TRAIL proteins were treated into TRAIL-resistant cells and TRAIL-susceptible cells, with or without bortezomib, to compare their respective abilities to induce cell death. Recombinant TRAIL was treated with bortezomib to investigate whether this combination treatment could induce tumor regression in a mouse syngeneic tumor model. To understand the mechanism of combined treatment-induced cell death, cells were analyzed by flow cytometry and the effects of various cell death inhibitors on cell death rates were examined. Results ILz:rhTRAIL, a recombinant human TRAIL containing isoleucine zipper hexamerization domain, showed the highest cell death inducing ability both in single treatment and in combination treatment with bortezomib. In both TRAIL-resistant and TRAIL-susceptible cells treated with the combination treatment, an increase in cell death rates was dependent upon both the dose of TRAIL and its intrinsic properties. When a syngeneic mouse tumor model was treated with the combination of ILz:rhTRAIL and bortezomib, significant tumor regression was seen as a result of the effective induction of cancer cell death. The combination treatment-induced cell death was both inhibited by TRAIL blocking antibody and caspase-dependent. However, it was not inhibited by various ER stress inhibitors and autophagy inhibitors. Conclusions The combination treatment with ILz:rhTRAIL and bortezomib was able to induce cell death in both TRAIL-susceptible and TRAIL-resistant cancer cells through the intracellular TRAIL signaling pathway. The efficiency of cell death was dependent on the properties of TRAIL under the environment provided by bortezomib. The combination treatment-induced cell death was not regulated by bortezomib-induced ER stress response or by autophagy. Electronic supplementary material The online version of this article (10.1186/s12885-018-4352-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sunhyo Ryu
- Department of Dermatology, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA
| | - Yun Jeong Ahn
- Department of Biomedical Science and Research Center for Proteinaceous Materials, Chosun University School of Medicine, 309 Pilmoon-Daero, Gwangju, 61452, Republic of Korea
| | - Chakeong Yoon
- Department of Biomedical Science and Research Center for Proteinaceous Materials, Chosun University, Gwangju, South Korea
| | - Jeong Hwan Chang
- Department of Surgery, Chosun University School of Medicine, Gwangju, South Korea.,Present Address: Cheomdan Medical Center, 170 Cheomdanjungang-ro, Gwangsan-gu, Gwangju, 62276, Republic of Korea
| | - Yoonkyung Park
- Department of Biomedical Science and Research Center for Proteinaceous Materials, Chosun University, Gwangju, South Korea
| | - Tae-Hyoung Kim
- Department of Biochemistry, Chosun University School of Medicine, Gwangju, South Korea
| | - Amanda R Howland
- Department of Dermatology, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA
| | - Cheryl A Armstrong
- Department of Dermatology, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA
| | - Peter I Song
- Department of Dermatology, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA. .,Department of Dermatology, University of Colorado Denver Anschutz Medical Campus, 12801 E. 17th Avenue, Aurora, CO, 80045, USA.
| | - Ae Ran Moon
- Department of Biomedical Science and Research Center for Proteinaceous Materials, Chosun University School of Medicine, 309 Pilmoon-Daero, Gwangju, 61452, Republic of Korea. .,Department of Biomedical Science and Research Center for Proteinaceous Materials, Chosun University, Gwangju, South Korea.
| |
Collapse
|
27
|
Huang W, Chen C, Liu X. Hindlimb suspension-induced cell apoptosis in the posterior parietal cortex and lateral geniculate nucleus: corresponding changes in c-Fos protein and the PI3K/Akt signaling pathway. Acta Neurobiol Exp (Wars) 2018. [DOI: 10.21307/ane-2018-020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
28
|
Jeon MY, Min KJ, Woo SM, Seo SU, Kim S, Park JW, Kwon TK. Volasertib Enhances Sensitivity to TRAIL in Renal Carcinoma Caki Cells through Downregulation of c-FLIP Expression. Int J Mol Sci 2017; 18:2568. [PMID: 29186071 PMCID: PMC5751171 DOI: 10.3390/ijms18122568] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 11/28/2017] [Accepted: 11/28/2017] [Indexed: 11/23/2022] Open
Abstract
Polo-like kinase 1 (PLK1) plays major roles in cell cycle control and DNA damage response. Therefore, PLK1 has been investigated as a target for cancer therapy. Volasertib is the second-in class dihydropteridinone derivate that is a specific PLK1 inhibitor. In this study, we examined that combining PLK1 inhibitor with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) would have an additive and synergistic effect on induction of apoptosis in cancer cells. We found that volasertib alone and TRAIL alone had no effect on apoptosis, but the combined treatment of volasertib and TRAIL markedly induced apoptosis in Caki (renal carcinoma), A498 (renal carcinoma) and A549 (lung carcinoma) cells, but not in normal cells (human skin fibroblast cells and mesangial cells). Combined treatment induced accumulation of sub-G1 phase, DNA fragmentation, cleavage of poly (ADP-ribose) polymerase (PARP) and activation of caspase 3 activity in Caki cells. Interestingly, combined treatment induced downregulation of cellular-FLICE-inhibitory protein (c-FLIP) expression and ectopic expression of c-FLIP markedly blocked combined treatment-induced apoptosis. Therefore, this study demonstrates that volasertib may sensitize TRAIL-induced apoptosis in Caki cells via downregulation of c-FLIP.
Collapse
Affiliation(s)
- Mi-Yeon Jeon
- Department of Immunology, School of Mediine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, Korea.
| | - Kyoung-Jin Min
- Department of Immunology, School of Mediine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, Korea.
| | - Seon Min Woo
- Department of Immunology, School of Mediine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, Korea.
| | - Seung Un Seo
- Department of Immunology, School of Mediine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, Korea.
| | - Shin Kim
- Department of Immunology, School of Mediine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, Korea.
| | - Jong-Wook Park
- Department of Immunology, School of Mediine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, Korea.
| | - Taeg Kyu Kwon
- Department of Immunology, School of Mediine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, Korea.
| |
Collapse
|
29
|
Gambogic Acid Induces Cell Apoptosis and Inhibits MAPK Pathway in PTEN -/-/p53 -/- Prostate Cancer Cells In Vitro and Ex Vivo. Chin J Integr Med 2017; 24:109-116. [PMID: 28578487 DOI: 10.1007/s11655-017-2410-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Indexed: 01/28/2023]
Abstract
OBJECTIVE To investigate the effect of gambogic acid (GA) on the growth and cell death of castrate resistant prostate cancer (PC) with phosphate and tension homology (PTEN) and p53 genes deleted in vitro and ex vivo, and elucidate the underlying possible molecular mechanisms. METHODS PTEN-/-/p53-/- PC cells and Los Angeles prostate cancer-4 (LAPC-4) cells were treated with GA for 24 h and 48 h, then cell viability was determined by cell proliferation assay. PTEN-/-/p53-/- PC cells organoids number was calculated under GA treatment for 1 week. In addition, cell titer glo assay was performed to analyze 3 dimensional cell viability of patients derived xenografts (PDX) 170.2 organoids. Flow cytometry was used to detect apoptotic cells treated with GA. And confocal image was performed to detect the apoptotic mitochondrial morphological changes. Apoptotic cell death related protein levels were measured through Western blot (WB) in GA treated cells and organoids. The expression levels of mitogen-activated protein kinases (MAPKs) pathway related ribonucleic acid (RNAs) and proteins were analyzed by reverse transcription polymerase chain reaction (RT-PCR) and WB, respectively. RESULTS The treatment of GA significantly reduced cell viability of PTEN-/-/p53-/- PC cells and LAPC-4 in a time- and concentration-dependent manner. In organoids, GA showed strong inhibition towards organoids' numbers and diameters and continuously led to a complete organoids inhibition with GA 150 nmol/L. Ex vivo results validated that GA 1 μmol/L inhibited 44.6% PDX170.2 organoids growth. As for mechanism, flow cytometry detected continuously increased apoptotic portion under GA treatment from 1.98% to 11.78% (6 h) and 29.94% (8 h, P<0.05). In addition, mitochondrial fragmentation emerged in GA treated cells indicated the mitochondrial apoptotic pathway might be involved. Furthermore, WB detected caspases-3, -9 activation and light chain (LC)-3 conversion with GA treatment. WB revealed decreased activity of MAPK pathway and down-regulation of downstream c-fos oncogene RNA level was detected by RT-PCR before undergoing apoptosis (P<0.05). CONCLUSION GA was a potent anti-tumor compound as for PTEN-/-/p53-/- PC, which contributed to cell apoptosis via inhibition of the MAPK pathway and c-fos.
Collapse
|
30
|
Molecular insights into cancer therapeutic effects of the dietary medicinal phytochemical withaferin A. Proc Nutr Soc 2017; 76:96-105. [PMID: 28162105 DOI: 10.1017/s0029665116002937] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Despite the worldwide research efforts to combat cancer, it remains a leading cause of death. Although various specific kinase inhibitors already have been approved for clinical cancer treatment, occurrence of intrinsic or acquired resistance and intermittent response over longer periods limits long-term success of single kinase-targeted therapies. In this respect, there is a renewed interest in polypharmaceutical natural compounds, which simultaneously target various hyperactivated kinases involved in tumour-inflammation, angiogenesis, cell survival, proliferation, metastasis and angiogenesis. The dietary medicinal phytochemical withaferin A (WA), isolated from Withaferin somnifera (popular Indian name Ashwagandha), holds promise as a novel anti-cancer agent, which targets multiple cell survival kinase pathways, including IκB kinase/NF-κB, PI3 kinase/protein kinase B/mammalian target of rapamycin and mitogen-activated protein kinase/extracellular signal-regulated kinase amongst others. In this review, we propose a novel mechanism of WA-dependent kinase inhibition via electrophilic covalent targeting of cysteine residues in conserved kinase activation domains (kinase cysteinome), which could underlie its pleiotropic therapeutic effects in cancer signalling.
Collapse
|
31
|
Huang Y, Tao Y, Li X, Chang S, Jiang B, Li F, Wang ZM. Bioinformatics analysis of key genes and latent pathway interactions based on the anaplastic thyroid carcinoma gene expression profile. Oncol Lett 2016; 13:167-176. [PMID: 28428828 PMCID: PMC5396846 DOI: 10.3892/ol.2016.5447] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/10/2016] [Indexed: 01/03/2023] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is an aggressive malignant disease in older adults
with a high mortality rate. The present study aimed to examine several key genes and
pathways, which are associated with ATC. The GSE33630 gene expression profile was
downloaded from the Gene Expression Omnibus database, which included 11 ATC and 45
normal thyroid samples. The differentially expressed genes (DEGs) in ATC were
identified using the Limma package in R. The Gene Ontology functions and Kyoto
Encyclopedia of Genes and Genomes pathways of the selected DEGs were enriched using
the Database for Annotation, Visualization and Integrated Discovery. A
protein-protein interaction (PPI) network of the DEGs was constructed to select
significant modules. Furthermore, a latent pathway interactive network was
constructed to select the significant pathways associated with ATC. A total of 665
DEGs in the ATC samples were screened, and four significant modules were selected
from the PPI network. The DEGs in the four modules were enriched in several functions
and pathways. In addition, 29 significant pathways associated with ATC were selected,
and he Toll-like receptor (TLR) signaling pathway, extracellular matrix
(ECM)-receptor interaction and cytokine-cytokine interaction pathway were identified
as important pathways. FBJ murine osteosarcoma viral oncogene homolog (FOS),
chemokine C-X-C motif ligand 10 (CXCL10), collagen type V α1 (COL5A1) and
chemokine (C-C motif) ligand 28 (CCL28) were the key DEGs involved in these
significant pathways. The data obtained in the present study revealed that the TLR
signaling pathway, ECM-receptor interaction and cytokine-cytokine receptor
interaction pathway, and the FOS, CXCL10, COL5A1, COL11A1 and CCL28 genes have
different roles in the progression of ATC, and these may be used as therapeutic
targets for ATC.
Collapse
Affiliation(s)
- Yun Huang
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Yiming Tao
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Xinying Li
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Shi Chang
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Bo Jiang
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Feng Li
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Zhi-Ming Wang
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| |
Collapse
|
32
|
Tabuchi Y, Uchiyama H, Zhao QL, Yunoki T, Andocs G, Nojima N, Takeda K, Ishikawa K, Hori M, Kondo T. Effects of nitrogen on the apoptosis of and changes in gene expression in human lymphoma U937 cells exposed to argon-based cold atmospheric pressure plasma. Int J Mol Med 2016; 37:1706-14. [PMID: 27121589 DOI: 10.3892/ijmm.2016.2574] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 04/07/2016] [Indexed: 11/06/2022] Open
Abstract
Cold atmospheric pressure plasma (CAP) is known as a source of biologically active agents, such as reactive oxygen species (ROS) and reactive nitrogen species (RNS). In the present study, we examined the effects of nitrogen (N2) on the apoptosis of and changes in gene expression in human lymphoma U937 cells exposed to argon (Ar)-CAP. Enormous amounts of hydroxyl (·OH) radicals in aqueous solution were produced using Ar‑CAP generated using a 20 kHz low frequency at 18 kV with a flow rate of 2 l/min. The increase in the levels of ·OH radicals was significantly attenuated by the addition of N2 to Ar gas. On the other hand, the level of total nitrate/nitrite in the supernatant was significantly elevated in the Ar + N2-CAP‑exposed U937 cells. When the cells were exposed to Ar‑CAP, a significant increase in apoptosis was observed, whereas apoptosis was markedly decreased in the cells exposed to Ar + N2-CAP. Microarray and pathway analyses revealed that a newly identified gene network containing a number of heat shock proteins (HSPs), anti-apoptotic genes, was mainly associated with the biological function of the prevention of apoptosis. Quantitative PCR revealed that the expression levels of HSPs were significantly elevated in the cells exposed to Ar + N2-CAP than those exposed to Ar‑CAP. These results indicate that N2 gas in Ar‑CAP modifies the ratio of ROS to RNS, and suppresses the apoptosis induced by Ar‑CAP. The modulation of gaseous conditions in CAP may thus prove to be useful for future clinical applications, such as for switching from a sterilizing mode to cytocidal effect for cancer cells.
Collapse
Affiliation(s)
- Yoshiaki Tabuchi
- Division of Molecular Genetics Research, Life Science Research Center, University of Toyama, Toyama 930-0194, Japan
| | | | - Qing-Li Zhao
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Tatsuya Yunoki
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Gabor Andocs
- Tateyama Machine Co., Ltd., Toyama 930-1305, Japan
| | | | - Keigo Takeda
- Plasma Nanotechnology Research Center, Nagoya University, Nagoya 464-8601, Japan
| | - Kenji Ishikawa
- Plasma Nanotechnology Research Center, Nagoya University, Nagoya 464-8601, Japan
| | - Masaru Hori
- Plasma Nanotechnology Research Center, Nagoya University, Nagoya 464-8601, Japan
| | - Takashi Kondo
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
33
|
2'-Hydroxy-4-methylsulfonylchalcone enhances TRAIL-induced apoptosis in prostate cancer cells. Anticancer Drugs 2016; 26:74-84. [PMID: 25192452 DOI: 10.1097/cad.0000000000000163] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Prostate cancer is the most common malignant cancer in men and the second leading cause of cancer deaths. Previously, we have shown that 2'-hydroxy-4-methylsulfonylchalcone (RG003) induced apoptosis in prostate cancer cell lines PC-3 and DU145. Although tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anticancer agent, some cancer cells are resistant to TRAIL treatment. PC-3 and LNCaP prostatic cancer cell lines have been reported to be resistant to TRAIL-induced apoptosis. Here, we show for the first time that RG003 overcomes TRAIL resistance in prostate cancer cells. RG003 can enhance TRAIL-induced apoptosis through DR5 upregulation and downregulation of Bcl-2, PI3K/Akt, NF-κB, and cyclooxygenase-2 (COX-2) survival pathways. When used in combined treatment, RG003 and TRAIL amplified TRAIL-induced activation of apoptosis effectors and particularly activation of caspase-8 and the executioner caspase-3, leading to increased poly-ADP-ribose polymerase cleavage and DNA fragmentation in prostate cancer cells. Furthermore, we showed that RG003 reduced COX-2 expression in cells. Previously, we showed that COX-2 was involved in resistance to an apoptosis mechanism; then, its inhibition by RG003 could render cells more sensitive to TRAIL treatment. We showed that nuclear factor-κB activation was inhibited after RG003 treatment. This inhibition was correlated with reduction in COX-2 expression and induction of apoptosis. Overall, we conclude, for the first time, that RG003 can enhance TRAIL-induced apoptosis in human prostate cancer cells. The significance of our in-vitro study with RG003 and TRAIL combined is very encouraging, suggesting the relevance of testing this combined treatment in xenograft animal models.
Collapse
|
34
|
Shankar E, Song K, Corum SL, Bane KL, Wang H, Kao HY, Danielpour D. A Signaling Network Controlling Androgenic Repression of c-Fos Protein in Prostate Adenocarcinoma Cells. J Biol Chem 2016; 291:5512-5526. [PMID: 26786102 DOI: 10.1074/jbc.m115.694877] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Indexed: 01/04/2023] Open
Abstract
The transcription factor c-Fos controls many important cellular processes, including cell growth and apoptosis. c-Fos expression is rapidly elevated in the prostate upon castration-mediated androgen withdrawal through an undefined mechanism. Here we show that androgens (5α-dihydrotestosterone and R1881) suppress c-Fos protein and mRNA expression induced by 12-O-tetradecanoylphorbol-13-acetate (TPA) or EGF in human prostate cancer (PCa) cell lines. Such suppression transpires through a transcriptional mechanism, predominantly at the proximal serum response element of the c-fos promoter. We show that androgen signaling suppresses TPA-induced c-Fos expression through repressing a PKC/MEK/ERK/ELK-1 signaling pathway. Moreover, our results support the hypothesis that p38(MAPK), PI3K, and PKCδ are involved in the androgenic regulation of c-Fos through controlling MEK/ERK. Stable silencing of c-Fos and PKCδ with shRNAs suggests that R1881 promotes cell death induced by low-dose TPA through a mechanism that is dependent on both PKCδ and loss of c-Fos expression. Reciprocally, loss of either PKCδ or c-Fos activates p38(MAPK) while suppressing the activation of ERK1/2. We also provide the first demonstration that R1881 permits cell death induced by low-dose TPA in the LNCaP androgen-dependent PCa cell line and that TPA-induced cell death is independent of exogenous androgen in the castration-resistant variants of LNCaP, C4-2 and C4-2B. Acquisition of androgen-independent killing by TPA correlates with activation of p38(MAPK), suppression of ERK1/2, and loss of c-Fos. These results provide new insights into androgenic control of c-Fos and use of PKC inhibitors in PCa therapy.
Collapse
Affiliation(s)
| | - Kyung Song
- Division of General Medical Sciences-Oncology
| | | | - Kara L Bane
- Division of General Medical Sciences-Oncology
| | | | - Hung-Ying Kao
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106 and; From the Case Comprehensive Cancer Center
| | - David Danielpour
- Division of General Medical Sciences-Oncology,; Department of Pharmacology, and; the Department of Urology, University Hospitals of Cleveland, Cleveland, Ohio 44106; From the Case Comprehensive Cancer Center,.
| |
Collapse
|
35
|
Platais C, Hakami F, Darda L, Lambert DW, Morgan R, Hunter KD. The role of HOX genes in head and neck squamous cell carcinoma. J Oral Pathol Med 2015; 45:239-47. [PMID: 26661059 DOI: 10.1111/jop.12388] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2015] [Indexed: 12/13/2022]
Abstract
Recent decades have witnessed the publication of numerous studies reporting alterations in the genome and transcriptome of head and neck squamous cell carcinoma (HNSCC). Currently, the utilisation of these alterations as biomarkers and targets for therapy is limited and new, useful molecular characteristics are being sought. Many of the published HNSCC gene expression profiles demonstrate alterations in the expression of HOX genes. These are a family of Homeobox-containing genes which are involved in developmental patterning and morphogenesis in the embryo, and which are often aberrantly expressed in cancer. The 39 HOX genes found in the human genome are arranged in four paralogous groups at different chromosomal loci. These control a wide range of cellular processes, including proliferation and migration, which are relevant in the context of cancer development. In this review article, we will outline the biology of HOX genes in relation to cancer and summarise the accumulating evidence for their role in the development of HNSCC and the possibility that they could be a therapeutic target in this malignancy. We will also identify areas where our current understanding is weak to focus future work and appraise the ongoing strategies for pharmacological intervention.
Collapse
Affiliation(s)
- Christopher Platais
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Fahad Hakami
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK.,Department of Pathology and Laboratory Medicine, King Abdulaziz Medical City-WR, Jeddah, Saudi Arabia
| | - Lav Darda
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Daniel W Lambert
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Richard Morgan
- Institute of Cancer Therapeutics, University of Bradford, Bradford, UK
| | - Keith D Hunter
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK.,Department of Oral Pathology and Biology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
36
|
Ge R, Wang Z, Wu S, Zhuo Y, Otsetov AG, Cai C, Zhong W, Wu CL, Olumi AF. Metformin represses cancer cells via alternate pathways in N-cadherin expressing vs. N-cadherin deficient cells. Oncotarget 2015; 6:28973-87. [PMID: 26359363 PMCID: PMC4745705 DOI: 10.18632/oncotarget.5023] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 08/13/2015] [Indexed: 12/12/2022] Open
Abstract
Metformin has emerged as a potential anticancer agent. Here, we demonstrate that metformin plays an anti-tumor role via repressing N-cadherin, independent of AMPK, in wild-type N-cadherin cancer cells. Ectopic-expression of N-cadherin develops metformin-resistant cancer cells, while suppression of N-cadherin sensitizes cancer to metformin. Manipulation of AMPK expression does not alter sensitivity of cancer to metformin. We show that NF-kappaB is a downstream molecule of N-cadherin and metformin regulates NF-kappaB signaling via suppressing N-cadherin. Moreover, we also suggest that TWIST1 is an upstream molecule of N-cadherin/NF-kappaB signaling and manipulation of TWIST1 expression changes the sensitivity of cancer cells to metformin. In contrast to the cells that express N-cadherin, in N-cadherin deficient cells, metformin plays an anti-tumor role via activation of AMPK. Ectopic expression of N-cadherin makes cancer more resistant to metformin. Therefore, we suggest that metformin's anti-cancer therapeutic effect is mediated through different molecular mechanism in wild-type vs. deficient N-cadherin cancer cells. At last, we selected 49 out of 984 patients' samples with prostatic cancer after radical prostatectomy (selection criteria: Gleason score ≥ 7 and all patients taking metformin) and showed levels of N-cadherin, p65 and AMPK could predict post-surgical recurrence in prostate cancer after treatment of metformin.
Collapse
Affiliation(s)
- Rongbin Ge
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zongwei Wang
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shulin Wu
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yangjia Zhuo
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Aleksandar G. Otsetov
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Chao Cai
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Weide Zhong
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chin-Lee Wu
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Aria F. Olumi
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
37
|
Hsu PY, Hsu HK, Hsiao TH, Ye Z, Wang E, Profit AL, Jatoi I, Chen Y, Kirma NB, Jin VX, Sharp ZD, Huang THM. Spatiotemporal control of estrogen-responsive transcription in ERα-positive breast cancer cells. Oncogene 2015; 35:2379-89. [PMID: 26300005 PMCID: PMC4865474 DOI: 10.1038/onc.2015.298] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 06/20/2015] [Accepted: 07/05/2015] [Indexed: 12/29/2022]
Abstract
Recruitment of transcription machinery to target promoters for aberrant gene expression has been well studied, but underlying control directed by distant-acting enhancers remains unclear in cancer development. Our previous study demonstrated that distant estrogen response elements (DEREs) located on chromosome 20q13 are frequently amplified and translocated to other chromosomes in ERα-positive breast cancer cells. In this study, we used three-dimensional interphase fluorescence in situ hybridization to decipher spatiotemporal gathering of multiple DEREs in the nucleus. Upon estrogen stimulation, scattered 20q13 DEREs were mobilized to form regulatory depots for synchronized gene expression of target loci. A chromosome conformation capture assay coupled with chromatin immunoprecipitation further uncovered that ERα-bound regulatory depots are tethered to heterochromatin protein 1 (HP1) for coordinated chromatin movement and histone modifications of target loci, resulting in transcription repression. Neutralizing HP1 function dysregulated the formation of DERE-involved regulatory depots and transcription inactivation of candidate tumor-suppressor genes. Deletion of amplified DEREs using the CRISPR/Cas9 genomic-editing system profoundly altered transcriptional profiles of proliferation-associated signaling networks, resulting in reduction of cancer cell growth. These findings reveal a formerly uncharacterized feature wherein multiple copies of the amplicon congregate as transcriptional units in the nucleus for synchronous regulation of function-related loci in tumorigenesis. Disruption of their assembly can be a new strategy for treating breast cancers and other malignancies.
Collapse
Affiliation(s)
- P-Y Hsu
- Department of Molecular Medicine, Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - H-K Hsu
- Department of Molecular Medicine, Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - T-H Hsiao
- Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Department of Medical Research, Taichung Veterans General Hospital, Taichung City, Taiwan
| | - Z Ye
- Department of Molecular Medicine, Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - E Wang
- Department of Cellular and Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - A L Profit
- Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Department of Pathology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - I Jatoi
- Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Department of Surgery, The University of Texas Health Science Center at San Antonio, San Antonio TX, USA
| | - Y Chen
- Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Department of Epidemiology and Biostatistics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - N B Kirma
- Department of Molecular Medicine, Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - V X Jin
- Department of Molecular Medicine, Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Department of Epidemiology and Biostatistics, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Z D Sharp
- Department of Molecular Medicine, Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - T H-M Huang
- Department of Molecular Medicine, Institute of Biotechnology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy and Research Center, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
38
|
Nishikawa Y, Okuzaki D, Fukushima K, Mukai S, Ohno S, Ozaki Y, Yabuta N, Nojima H. Withaferin A Induces Cell Death Selectively in Androgen-Independent Prostate Cancer Cells but Not in Normal Fibroblast Cells. PLoS One 2015; 10:e0134137. [PMID: 26230090 PMCID: PMC4521694 DOI: 10.1371/journal.pone.0134137] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 07/06/2015] [Indexed: 12/28/2022] Open
Abstract
Withaferin A (WA), a major bioactive component of the Indian herb Withania somnifera, induces cell death (apoptosis/necrosis) in multiple types of tumor cells, but the molecular mechanism underlying this cytotoxicity remains elusive. We report here that 2 μM WA induced cell death selectively in androgen-insensitive PC-3 and DU-145 prostate adenocarcinoma cells, whereas its toxicity was less severe in androgen-sensitive LNCaP prostate adenocarcinoma cells and normal human fibroblasts (TIG-1 and KD). WA also killed PC-3 cells in spheroid-forming medium. DNA microarray analysis revealed that WA significantly increased mRNA levels of c-Fos and 11 heat-shock proteins (HSPs) in PC-3 and DU-145, but not in LNCaP and TIG-1. Western analysis revealed increased expression of c-Fos and reduced expression of the anti-apoptotic protein c-FLIP(L). Expression of HSPs such as HSPA6 and Hsp70 was conspicuously elevated; however, because siRNA-mediated depletion of HSF-1, an HSP-inducing transcription factor, reduced PC-3 cell viability, it is likely that these heat-shock genes were involved in protecting against cell death. Moreover, WA induced generation of reactive oxygen species (ROS) in PC-3 and DU-145, but not in normal fibroblasts. Immunocytochemistry and immuno-electron microscopy revealed that WA disrupted the vimentin cytoskeleton, possibly inducing the ROS generation, c-Fos expression and c-FLIP(L) suppression. These observations suggest that multiple events followed by disruption of the vimentin cytoskeleton play pivotal roles in WA-mediated cell death.
Collapse
Affiliation(s)
- Yukihiro Nishikawa
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, 3–1 Yamadaoka, Suita City, Osaka, 565–0871, Japan
| | - Daisuke Okuzaki
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, 3–1 Yamadaoka, Suita City, Osaka, 565–0871, Japan
- DNA-chip Development Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, 3–1 Yamadaoka, Suita City, Osaka, 565–0871, Japan
| | - Kohshiro Fukushima
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, 3–1 Yamadaoka, Suita City, Osaka, 565–0871, Japan
| | - Satomi Mukai
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, 3–1 Yamadaoka, Suita City, Osaka, 565–0871, Japan
| | - Shouichi Ohno
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, 3–1 Yamadaoka, Suita City, Osaka, 565–0871, Japan
| | - Yuki Ozaki
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, 3–1 Yamadaoka, Suita City, Osaka, 565–0871, Japan
| | - Norikazu Yabuta
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, 3–1 Yamadaoka, Suita City, Osaka, 565–0871, Japan
| | - Hiroshi Nojima
- Department of Molecular Genetics, Research Institute for Microbial Diseases, Osaka University, 3–1 Yamadaoka, Suita City, Osaka, 565–0871, Japan
- DNA-chip Development Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, 3–1 Yamadaoka, Suita City, Osaka, 565–0871, Japan
- * E-mail:
| |
Collapse
|
39
|
Venza M, Visalli M, Oteri R, Agliano F, Morabito S, Teti D, Venza I. The overriding of TRAIL resistance by the histone deacetylase inhibitor MS-275 involves c-myc up-regulation in cutaneous, uveal, and mucosal melanoma. Int Immunopharmacol 2015; 28:313-21. [PMID: 26122536 DOI: 10.1016/j.intimp.2015.06.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/18/2015] [Accepted: 06/22/2015] [Indexed: 12/15/2022]
Abstract
Malignant melanoma is a highly aggressive tumor which may occur in the skin, eye, and mucous membranes. The prognosis of melanoma remains poor in spite of therapeutic advances, emphasizing the importance of innovative treatment modalities. Currently, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is showing promising clinical responses, however its use is hampered by intrinsic or acquired melanoma resistance to apoptosis. Recently, we showed that the combination of TRAIL with the class I-specific histone deacetylase inhibitor (HDACi) MS-275 was a privileged way to override TRAIL resistance through down-regulation of cellular Fas-associated death domain (FADD)-like interleukin-1beta-converting enzyme-inhibitory protein (c-FLIP). Here, we elucidated the underlying mechanism and provided evidence that a crucial step in the c-FLIP downregulation triggered by MS-275 implies the up-regulation of c-myc, a transcriptional repressor of c-FLIP. Notably, MS-275 caused H3 histone acetylation at the promoter of c-myc and increased its binding to the c-FLIP promoter, that in turn led to reduced c-FLIP gene transcription. Knockdown of c-myc prevented the MS-275-mediated downregulation of c-FLIP and hindered TRAIL-plus MS-275-induced apoptosis. Findings reported here provide additional knowledge tools for a more aware and effective molecular therapy of melanoma.
Collapse
Affiliation(s)
- Mario Venza
- Department of Experimental Specialized Medical and Surgical and Odontostomatology Sciences, University of Messina, Messina, Italy
| | - Maria Visalli
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Rosaria Oteri
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Federica Agliano
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Silvia Morabito
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Diana Teti
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.
| | - Isabella Venza
- Department of Experimental Specialized Medical and Surgical and Odontostomatology Sciences, University of Messina, Messina, Italy
| |
Collapse
|
40
|
Iron supplementation effectively suppresses gastrocnemius muscle lesions to improve exercise capacity in chronic heart failure rats with anemia. Nutrition 2015; 31:1038-44. [PMID: 26059380 DOI: 10.1016/j.nut.2015.02.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 02/13/2015] [Accepted: 02/27/2015] [Indexed: 12/18/2022]
Abstract
OBJECTIVE For patients with chronic heart failure (CHF), exertional fatigue is one of the most common and debilitating symptoms. However, the poor relationship between heart dysfunction and exercise capacity has been ascribed to peripheral abnormalities. Several previous studies confirmed that iron supplementation could significantly improve the exercise capacity of patients with CHF, although they did not analyze effects in the musculoskeletal system. The aim of this study was to investigate the effect of iron treatment on gastrocnemius muscles of CHF rats with anemia. METHODS Male Sprague-Dawley rats were subjected to coronary ligation to induce heart failure. At the same time, blood (1-1.5 mL) was withdrawn from the retro-orbital plexus once every week to induce anemia. After 6 wk of this process, iron dextran was administered to the CHF rats with anemia (CHFa rats) at the dose of 8, 16, 32, or 64 mg/kg every 2 d for 2 wk. RESULTS Iron dextran (8 mg/kg every 2 d) effectively improved hemodynamic parameters (P < 0.05) compared with CHFa rats. Similarly, this dose of iron dextran significantly reduced the ratio of heart weight to body weight (P < 0.01), whereas it significantly increased the distance run (m) to exhaustion (P < 0.01). Iron dextran effectively inhibited sarcoplasmic vacuolation and muscle atrophy of gastrocnemius muscles in CHFa rats, as evaluated by pathologic examinations. Other iron treatments, however, were found to be ineffective on the same parameters, so particular focus was placed on the iron dextran (8 mg/kg every 2 d) group in subsequent analyses. Consistently, phospho-p38 in gastrocnemius muscles of CHFa rats was markedly suppressed by iron dextran. Additionally, iron dextran significantly decreased c-fos and c-jun and up-regulated cellular FLICE-inhibitory protein expression levels.
Collapse
|
41
|
Zhang T, Fan X, Song L, Ren L, Ma E, Zhang S, Ren L, Zheng Y, Zhang J. c-Fos is involved in inhibition of human bladder carcinoma T24 cells by brazilin. IUBMB Life 2015; 67:175-81. [PMID: 25865820 DOI: 10.1002/iub.1357] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 01/20/2015] [Indexed: 11/11/2022]
Abstract
Crude brazilin extract from Sappan wood has demonstrated strong anti tumor activity in the mouse model of human bladder carcinoma and clinical trial for intravesical therapy. Purified brazilin was confirmed the most active molecule in inhibition of bladder carcinoma T24 cells. Brazilin decreased proliferation and viability of T24 cells in a dose- and time-dependent manner, with a calculated LC50 of 32 µg/mL. More than 1,000 of genes were found upregulated and down regulated by brazilin treatment in digital gene expression profiling. Gene ontology analysis indicated that stress response, apoptosis, and cell cycle regulatory pathways were highly enriched. Among the regulated genes, c-Fos was the most and specifically upregulated. Overexpression of c-Fos in T24 cells resulted in tumor cell specific changes in cell morphology and viability. Over expression of stress-responsive gene, HSP70, and other highly upregulated genes did not have any effect on cell growth. Brazilin may inhibit T24 cell growth and trigger cell death through a c-Fos-mediated and tumor cell specific signaling pathway. Further studies of its down stream mediators may help to identify better tumor cell type specific drug targets.
Collapse
Affiliation(s)
- Tingting Zhang
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, China
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kang D, Choi HJ, Kang S, Kim SY, Hwang YS, Je S, Han Z, Kim JH, Song JJ. Ratio of phosphorylated HSP27 to nonphosphorylated HSP27 biphasically acts as a determinant of cellular fate in gemcitabine-resistant pancreatic cancer cells. Cell Signal 2015; 27:807-17. [PMID: 25615626 DOI: 10.1016/j.cellsig.2015.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/15/2014] [Accepted: 01/13/2015] [Indexed: 01/24/2023]
Abstract
Gemcitabine has been used most commonly as an anticancer drug to treat advanced pancreatic cancer patients. However, intrinsic or acquired resistance of pancreatic cancer to gemcitabine was also developed, which leads to very low five-year survival rates. Here, we investigated whether cellular levels of HSP27 phosphorylation act as a determinant of cellular fate with gemcitabine. In addition we have demonstrated whether HSP27 downregulation effectively could overcome the acquisition of gemcitabine resistance by using transcriptomic analysis. We observed that gemcitabine induced p38/HSP27 phosphorylation and caused acquired resistance. After acquisition of gemcitabine resistance, cancer cells showed higher activity of NF-κB. NF-κB activity, as well as colony formation in gemcitabine-resistant pancreatic cancer cells, was significantly decreased by HSP27 downregulation and subsequent TRAIL treatment, showing that HSP27 was a common network mediator of gemcitabine/TRAIL-induced cell death. After transcriptomic analysis, gene fluctuation after HSP27 downregulation was very similar to that of pancreatic cancer cells susceptible to gemcitabine, and then in opposite position to that of acquired gemcitabine resistance, which makes it possible to downregulate HSP27 to overcome the acquired gemcitabine resistance to function as an overall survival network inhibitor. Most importantly, we demonstrated that the ratio of phosphorylated HSP27 to nonphosphorylated HSP27 rather than the cellular level of HSP27 itself acts biphasically as a determinant of cellular fate in gemcitabine-resistant pancreatic cancer cells.
Collapse
Affiliation(s)
- Dongxu Kang
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, Jilin Province, PR China
| | - Hye Jin Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sujin Kang
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Young Kim
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Sic Hwang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Suyeon Je
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Zhezhu Han
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, Jilin Province, PR China
| | - Joo-Hang Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Jae J Song
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
43
|
Pellegrini KL, Han T, Bijol V, Saikumar J, Craciun FL, Chen WW, Fuscoe JC, Vaidya VS. MicroRNA-155 deficient mice experience heightened kidney toxicity when dosed with cisplatin. Toxicol Sci 2014; 141:484-92. [PMID: 25015656 DOI: 10.1093/toxsci/kfu143] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The development of nephrotoxicity limits the maximum achievable dosage and treatment intervals for cisplatin chemotherapy. Therefore, identifying mechanisms that regulate this toxicity could offer novel methods to optimize cisplatin delivery. MicroRNAs are capable of regulating many different genes, and can influence diverse cellular processes, including cell death and apoptosis. We previously observed miR-155 to be highly increased following ischemic or toxic injury to the kidneys and, therefore, sought to determine whether mice deficient in miR-155 would respond differently to kidney injury. We treated C57BL/6 and miR-155(-/-) mice with 20 mg/kg of cisplatin and found a significantly higher level of kidney injury in the miR-155(-/-) mice. Genome-wide expression profiling and bioinformatic analysis indicated the activation of a number of canonical signaling pathways relating to apoptosis and oxidative stress over the course of the injury, and identified potential upstream regulators of these effects. One predicted upstream regulator was c-Fos, which has two confirmed miR-155 binding sites in its 3' UTR and, therefore, can be directly regulated by miR-155. We established that the miR-155(-/-) mice had significantly higher levels of c-Fos mRNA and protein than the C57BL/6 mice at 72 h after cisplatin exposure. These data indicate a role for miR-155 in the cisplatin response and suggest that targeting of c-Fos could be investigated to reduce cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Kathryn L Pellegrini
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tao Han
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas
| | - Vanesa Bijol
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Janani Saikumar
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Florin L Craciun
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - William W Chen
- Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts
| | - James C Fuscoe
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas
| | - Vishal S Vaidya
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts Laboratory of Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts
| |
Collapse
|
44
|
Tilli TM, Bellahcène A, Castronovo V, Gimba ERP. Changes in the transcriptional profile in response to overexpression of the osteopontin-c splice isoform in ovarian (OvCar-3) and prostate (PC-3) cancer cell lines. BMC Cancer 2014; 14:433. [PMID: 24928374 PMCID: PMC4075779 DOI: 10.1186/1471-2407-14-433] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 05/23/2014] [Indexed: 12/16/2022] Open
Abstract
Background Especially in human tumor cells, the osteopontin (OPN) primary transcript is subject to alternative splicing, generating three isoforms termed OPNa, OPNb and OPNc. We previously demonstrated that the OPNc splice variant activates several aspects of the progression of ovarian and prostate cancers. The goal of the present study was to develop cell line models to determine the impact of OPNc overexpression on main cancer signaling pathways and thus obtain insights into the mechanisms of OPNc pro-tumorigenic roles. Methods Human ovarian and prostate cancer cell lines, OvCar-3 and PC-3 cells, respectively, were stably transfected to overexpress OPNc. Transcriptomic profiling was performed on these cells and compared to controls, to identify OPNc overexpression-dependent changes in gene expression levels and pathways by qRT-PCR analyses. Results Among 84 genes tested by using a multiplex real-time PCR Cancer Pathway Array approach, 34 and 16, respectively, were differentially expressed between OvCar-3 and PC-3 OPNc-overexpressing cells in relation to control clones. Differentially expressed genes are included in all main hallmarks of cancer, and several interacting proteins have been identified using an interactome network analysis. Based on marked up-regulation of Vegfa transcript in response to OPNc overexpression, we partially validated the array data by demonstrating that conditioned medium (CM) secreted from OvCar-3 and PC-3 OPNc-overexpressing cells significantly induced endothelial cell adhesion, proliferation and migration, compared to CM secreted from control cells. Conclusions Overall, the present study elucidated transcriptional changes of OvCar-3 and PC-3 cancer cell lines in response to OPNc overexpression, which provides an assessment for predicting the molecular mechanisms by which this splice variant promotes tumor progression features.
Collapse
Affiliation(s)
| | | | | | - Etel R P Gimba
- Coordenação de Pesquisa, Programa de Carcinogênese Molecular, Instituto Nacional de Câncer (INCa)/Programa de Pós Graduação Stricto Sensu em Oncologia do INCa, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
45
|
Apoptosis and molecular targeting therapy in cancer. BIOMED RESEARCH INTERNATIONAL 2014; 2014:150845. [PMID: 25013758 PMCID: PMC4075070 DOI: 10.1155/2014/150845] [Citation(s) in RCA: 753] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/11/2014] [Indexed: 12/22/2022]
Abstract
Apoptosis is the programmed cell death which maintains the healthy survival/death balance in metazoan cells. Defect in apoptosis can cause cancer or autoimmunity, while enhanced apoptosis may cause degenerative diseases. The apoptotic signals contribute into safeguarding the genomic integrity while defective apoptosis may promote carcinogenesis. The apoptotic signals are complicated and they are regulated at several levels. The signals of carcinogenesis modulate the central control points of the apoptotic pathways, including inhibitor of apoptosis (IAP) proteins and FLICE-inhibitory protein (c-FLIP). The tumor cells may use some of several molecular mechanisms to suppress apoptosis and acquire resistance to apoptotic agents, for example, by the expression of antiapoptotic proteins such as Bcl-2 or by the downregulation or mutation of proapoptotic proteins such as BAX. In this review, we provide the main regulatory molecules that govern the main basic mechanisms, extrinsic and intrinsic, of apoptosis in normal cells. We discuss how carcinogenesis could be developed via defective apoptotic pathways or their convergence. We listed some molecules which could be targeted to stimulate apoptosis in different cancers. Together, we briefly discuss the development of some promising cancer treatment strategies which target apoptotic inhibitors including Bcl-2 family proteins, IAPs, and c-FLIP for apoptosis induction.
Collapse
|
46
|
Giampietri C, Petrungaro S, Padula F, D'Alessio A, Marini ES, Facchiano A, Filippini A, Ziparo E. Autophagy modulators sensitize prostate epithelial cancer cell lines to TNF-alpha-dependent apoptosis. Apoptosis 2014; 17:1210-22. [PMID: 22923157 DOI: 10.1007/s10495-012-0752-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
TNF-alpha levels in prostate cancer correlate with the extent of disease and are significantly elevated in the metastatic stage. TNF receptor superfamily controls two distinct signalling cascades, leading to opposite effects, i.e. apoptosis and survival; in prostate cancer TNF-alpha-mediated signalling induces cell survival and resistance to therapy. The apoptosis of prostate epithelial cancer cells LNCaP and PC3 was investigated upon treatment with the autophagy inhibitor 3-methyladenine and the autophagy inducer rapamycin, in combination with TNF-alpha. Cells were exposed to these molecules for 18, 24 and 48 h. Autophagy was assessed via LC3 Western blot analysis; propidium iodide and TUNEL stainings followed by flow cytometry or caspase-8 and caspase-3 activation assays were performed to evaluate apoptosis. TNF-alpha-induced apoptosis was potentiated by 3-methyladenine in the androgen-responsive LNCaP cells, whereas no effect was observed in the androgen-insensitive PC3 cells. Interestingly such pro-apoptosis effect in LNCaP cells was associated with reduced c-Flip levels through proteasomal degradation via increased reactive oxygen species production and p38 activation; such c-Flip reduction was reversed in the presence of either the proteasome inhibitor MG132 or the reactive oxygen species scavenger N-acetyl-cysteine. Conversely in PC3 but not in LNCaP cells, rapamycin stimulated TNF-alpha-dependent apoptosis; such effect was associated with reduced c-Flip promoter activity and FoxO3a activation. We conclude that TNF-alpha-induced apoptosis may be potentiated, in prostate cancer epithelial cells, through autophagy modulators. Increased sensitivity to TNF-alpha-dependent apoptosis correlates with reduced c-Flip levels which are consequent to a post-transcriptional and a transcriptional mechanism in LNCaP and PC3 cells respectively.
Collapse
Affiliation(s)
- Claudia Giampietri
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics-Section of Histology and Medical Embryology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Via A. Scarpa, 14, 00161 Rome, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Oliveira-Ferrer L, Rößler K, Haustein V, Schröder C, Wicklein D, Maltseva D, Khaustova N, Samatov T, Tonevitsky A, Mahner S, Jänicke F, Schumacher U, Milde-Langosch K. c-FOS suppresses ovarian cancer progression by changing adhesion. Br J Cancer 2013; 110:753-63. [PMID: 24322891 PMCID: PMC3915133 DOI: 10.1038/bjc.2013.774] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 11/07/2013] [Accepted: 11/13/2013] [Indexed: 01/11/2023] Open
Abstract
Background: C-Fos was initially described as oncogene, but was associated with favourable prognosis in ovarian cancer (OvCa) patients. The molecular and functional aspects underlying this effect are still unknown. Methods: Using stable transfectants of SKOV3 and OVCAR8 cells, proliferation, migration, invasion and apoptotic potential of c-FOS-overexpressing clones and controls were compared. Adherence to components of the extracellular matrix was analysed in static assays, and adhesion to E-selectin, endothelial and mesothelial cells in dynamic flow assays. The effect of c-FOS in vivo was studied after intraperitoneal injection of SKOV3 clones into SCID mice, and changes in gene expression were determined by microarray analysis. Results: Tumour growth after injection into SCID mice was strongly delayed by c-FOS overexpression, with reduction of lung metastases and circulating tumour cells. In vitro, c-FOS had only weak influence on proliferation and migration, but was strongly pro-apoptotic. Adhesion to components of the extracellular matrix (collagen I, IV) and to E-selectin, endothelial and mesothelial cells was significantly reduced in c-FOS-overexpressing OvCa cells. This corresponds to deregulation of adhesion proteins and glycosylation enzymes in microarray analysis. Conclusion: In addition to its known pro-apoptotic effect, c-FOS might influence OvCa progression by changing the adhesion of OvCa cells to peritoneal surfaces.
Collapse
Affiliation(s)
- L Oliveira-Ferrer
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - K Rößler
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - V Haustein
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - C Schröder
- Department of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - D Wicklein
- Department of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - D Maltseva
- SRC Bioclinicum, Ugreshskaya Street 2/85, Moscow 115088, Russia
| | - N Khaustova
- SRC Bioclinicum, Ugreshskaya Street 2/85, Moscow 115088, Russia
| | - T Samatov
- SRC Bioclinicum, Ugreshskaya Street 2/85, Moscow 115088, Russia
| | - A Tonevitsky
- Institute of General Pathology and Pathophysiology, Baltijskaya Street 8, Moscow 125315, Russia
| | - S Mahner
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - F Jänicke
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - U Schumacher
- Department of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| | - K Milde-Langosch
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, D-20246, Hamburg, Germany
| |
Collapse
|
48
|
Yang H, Chen X, Wang X, Li Y, Chen S, Qian X, Wang R, Chen L, Han W, Ruan A, Du Q, Olumi AF, Zhang X. Inhibition of PP2A activity confers a TRAIL-sensitive phenotype during malignant transformation. Mol Cancer Res 2013; 12:217-27. [PMID: 24296757 DOI: 10.1158/1541-7786.mcr-13-0441] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED TRAIL is a promising anticancer agent because it induces apoptosis in the majority of human cancer cells but spares the normal cells. To determine the mechanistic nature of how normal cells acquire a TRAIL-sensitive phenotype during the process of malignant transformation, an experimental cell system was developed by sequential introduction of human telomerase reverse transcriptase and SV40 T antigens (large and small) into normal human prostatic epithelial cells (PrEC). This model system demonstrated that inhibition of protein phosphatase 2A (PP2A), either by SV40 small T antigen, okadaic acid, Calyculin A, or PP2A catalytic subunit siRNA, sensitized normal human PrEC and immortalized cells to TRAIL-induced apoptosis. Moreover, sensitization occurred during the premalignant period of tumorigenesis and PP2A exerted its antiapoptotic activity by negatively regulating c-Fos/AP-1. In addition, low-dose okadaic acid treatment sensitized TRAIL-resistant cancer cells to TRAIL, suggesting that PP2A inhibitors could be used as an enhancer of apoptosis induced by TRAIL or TRAIL-like agents. These data indicate that downregulation of PP2A activity is a critical step for normal cells to acquire a TRAIL-sensitive phenotype during tumorigenesis and that the level of PP2A activity may foretell cellular sensitivity to TRAIL-induced apoptosis. IMPLICATIONS Inhibition of PP2A is a key determinant in acquiring TRAIL sensitivity during tumorigenesis, with c-Fos/AP-1 as an essential mediator.
Collapse
Affiliation(s)
- Hongmei Yang
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Previous studies demonstrated that proteasome inhibition sensitizes TRAIL resistant prostate cancer cells to TRAIL-mediated apoptosis via stabilization of the active p18 subunit of caspase-8. The present study investigated the impact of proteasome inhibition on caspase-8 stability, ubiquitination, trafficking, and activation in cancer cells. Using caspase-8 deficient neuroblastoma (NB7) cells for reconstituting non-cleavable mutant forms of caspase-8, we demonstrated that the non-cleavable forms of caspase-8 are capable of inducing apoptosis comparably to wild-type caspase-8, in response to proteasome inhibitor and GST-TRAIL. Moreover in the LNCaP human prostate cancer cells, caspase-8 polyubiquitination occurs after TRAIL stimulation and caspase-8 processing. Subcellular fractionation analysis revealed caspase-8 activity in both cytosol and plasma membrane fractions in both NB7 reconstituted caspase-8 cell lines, as well the LNCaP prostate cancer cells. The present results suggest that caspase-8 stabilization through proteasome inhibition leads to reactivation of the extrinsic pathway of apoptosis and identify E3 ligase mediating caspase-8 polyubiquitination, as a novel molecular target. Inhibition of this E3 ligase in combination with TRAIL towards restoring apoptosis signaling activation may have potential therapeutic significance in resistant tumors.
Collapse
|
50
|
Abstract
Cellular FLICE (FADD-like IL-1beta-converting enzyme)-inhibitory protein (c-FLIP) is a major resistance factor and critical anti-apoptotic regulator that inhibits tumor necrosis factor-alpha (TNF-alpha), Fas-L, and TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis as well as chemotherapy-triggered apoptosis in malignant cells. c-FLIP is expressed as long (c-FLIP(L)), short (c-FLIP(S)), and c-FLIP(R) splice variants in human cells. c-FLIP binds to FADD and/or caspase-8 or -10 in a ligand-dependent and-independent fashion, which in turn prevents death-inducing signaling complex (DISC) formation and subsequent activation of the caspase cascade. Moreover, c-FLIP(L) and c-FLIP(S) are known to have multifunctional roles in various signaling pathways, as well as activating and/or upregulating several cytoprotective signaling molecules. Upregulation of c-FLIP has been found in various tumor types, and its downregulation has been shown to restore apoptosis triggered by cytokines and various chemotherapeutic agents. Hence, c-FLIP is an important target for cancer therapy. For example, small interfering RNAs (siRNAs) that specifically knockdown the expression of c-FLIP(L) in diverse human cancer cell lines augmented TRAIL-induced DISC recruitment and increased the efficacy of chemotherapeutic agents, thereby enhancing effector caspase stimulation and apoptosis. Moreover, small molecules causing degradation of c-FLIP as well as decreasing mRNA and protein levels of c-FLIP(L) and c-FLIP(S) splice variants have been found, and efforts are underway to develop other c-FLIP-targeted cancer therapies. This review focuses on (1) the functional role of c-FLIP splice variants in preventing apoptosis and inducing cytokine and drug resistance; (2) the molecular mechanisms that regulate c-FLIP expression; and (3) strategies to inhibit c-FLIP expression and function.
Collapse
|