1
|
Stokes AE, Clark HM, Edwards JL, Payton RR, Beever JE, Freeman TF, Hessock EA, Schrick FN, Moorey SE. Transcriptome profiles of blastocysts originating from oocytes matured in follicular fluid from preovulatory follicles of greater or lesser maturity. BMC Genomics 2025; 26:339. [PMID: 40186098 PMCID: PMC11969919 DOI: 10.1186/s12864-025-11521-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Oocyte competence for early embryo development relies on intercellular communication between the maturing oocyte and preovulatory follicle. Preovulatory follicle maturity, as indicated by serum estradiol concentration or follicle diameter, has previously been linked to pregnancy, follicular fluid metabolites, cumulus-oocyte metabolism, and oocyte competency for embryo development. Such relationships indicate metabolic and developmental programming of the oocyte based on the preovulatory follicle's physiological status, but downstream impacts on the molecular signature of blastocysts have not been examined. We hypothesized that supplementing maturing oocytes with follicular fluid originating from preovulatory follicles of greater or lesser maturity would impact the transcriptome of resulting blastocysts and indicate metabolic programming of the embryo that originated from the oocyte's maturation environment. The objective was to investigate the effect of follicle maturity on the oocyte by examining the transcriptome of blastocysts originating from oocytes matured in the presence of follicular fluid from preovulatory follicles of greater or lesser maturity. RESULTS In vitro maturing oocytes were supplemented with follicular fluid collected from preovulatory follicles of greater or lesser maturity. Following identical embryo culture procedures, RNA-sequencing was performed on pools of 2 blastocysts (Greater, n = 12; Lesser, n = 15; all with stage code = 7 and quality code = 1). A total of 12,310 genes were identified in blastocysts after filtering to remove lowly abundant genes. There were 113 genes that differed in expression between blastocysts originating from oocytes matured in greater versus lesser maturity follicular fluid (eFDR < 0.01). Although no pathways were significantly enriched with differentially expressed genes, transcriptome profiles suggested improved Wnt/β-catenin signaling, metabolism, and protection from oxidative stress in blastocysts derived from oocytes matured in greater maturity follicular fluid, while potential unregulated cell growth presented in blastocysts resulting from the lesser follicle maturity treatment. CONCLUSIONS Follicular fluid from preovulatory follicles of greater physiological maturity may better prepare maturing oocytes for early embryo development. Furthermore, oocytes matured in follicular fluid from preovulatory follicles of lesser maturity may attempt to overcompensate for nutrient deficit during oocyte maturation, leading to uncontrolled cellular growth and increased oxidative stress.
Collapse
Affiliation(s)
- Allyson E Stokes
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - Hannah M Clark
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - J Lannett Edwards
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - Rebecca R Payton
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - Jon E Beever
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - Trevor F Freeman
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - Emma A Hessock
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - F Neal Schrick
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA
| | - Sarah E Moorey
- Department of Animal Science, University of Tennessee Institute of Agriculture and AgResearch, 2506 River Drive, Knoxville, TN, 37996, USA.
| |
Collapse
|
2
|
Clark A, Villarreal MR, Huang SB, Jayamohan S, Rivas P, Hussain SS, Ybarra M, Osmulski P, Gaczynska ME, Shim EY, Smith T, Gupta YK, Yang X, Delma CR, Natarajan M, Lai Z, Wang LJ, Michalek JE, Higginson DS, Ikeno Y, Ha CS, Chen Y, Ghosh R, Kumar AP. Targeting S6K/NFκB/SQSTM1/Polθ signaling to suppress radiation resistance in prostate cancer. Cancer Lett 2024; 597:217063. [PMID: 38925361 DOI: 10.1016/j.canlet.2024.217063] [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] [Received: 12/15/2023] [Revised: 05/29/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024]
Abstract
In this study we have identified POLθ-S6K-p62 as a novel druggable regulator of radiation response in prostate cancer. Despite significant advances in delivery, radiotherapy continues to negatively affect treatment outcomes and quality of life due to resistance and late toxic effects to the surrounding normal tissues such as bladder and rectum. It is essential to develop new and effective strategies to achieve better control of tumor. We found that ribosomal protein S6K (RPS6KB1) is elevated in human prostate tumors, and contributes to resistance to radiation. As a downstream effector of mTOR signaling, S6K is known to be involved in growth regulation. However, the impact of S6K signaling on radiation response has not been fully explored. Here we show that loss of S6K led to formation of smaller tumors with less metastatic ability in mice. Mechanistically we found that S6K depletion reduced NFκB and SQSTM1 (p62) reporter activity and DNA polymerase θ (POLθ) that is involved in alternate end-joining repair. We further show that the natural compound berberine interacts with S6K in a in a hitherto unreported novel mode and that pharmacological inhibition of S6K with berberine reduces Polθ and downregulates p62 transcriptional activity via NFκB. Loss of S6K or pre-treatment with berberine improved response to radiation in prostate cancer cells and prevented radiation-mediated resurgence of PSA in animals implanted with prostate cancer cells. Notably, silencing POLQ in S6K overexpressing cells enhanced response to radiation suggesting S6K sensitizes prostate cancer cells to radiation via POLQ. Additionally, inhibition of autophagy with CQ potentiated growth inhibition induced by berberine plus radiation. These observations suggest that pharmacological inhibition of S6K with berberine not only downregulates NFκB/p62 signaling to disrupt autophagic flux but also decreases Polθ. Therefore, combination treatment with radiation and berberine inhibits autophagy and alternate end-joining DNA repair, two processes associated with radioresistance leading to increased radiation sensitivity.
Collapse
Affiliation(s)
- Alison Clark
- Departments of Molecular Medicine, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Michelle R Villarreal
- Departments of Molecular Medicine, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Shih-Bo Huang
- Departments of Molecular Medicine, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Sridharan Jayamohan
- Departments of Molecular Medicine, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Paul Rivas
- Departments of Molecular Medicine, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Suleman S Hussain
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Meagan Ybarra
- Departments of Molecular Medicine, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Pawel Osmulski
- Departments of Molecular Medicine, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Maria E Gaczynska
- Departments of Molecular Medicine, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Eun Yong Shim
- Departments of Molecular Medicine, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Tyler Smith
- Departments of Molecular Medicine, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Yogesh K Gupta
- Departments of Greehey Children's Cancer Institute, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA; Department of Biochemistry and Structural Biology, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Xiaoyu Yang
- Departments of Molecular Medicine, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Caroline R Delma
- Departments of Pathology, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Mohan Natarajan
- Departments of Pathology, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Zhao Lai
- Departments of Molecular Medicine, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA; Departments of Greehey Children's Cancer Institute, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA; Departments of Mays Cancer Center, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Li-Ju Wang
- Departments of Greehey Children's Cancer Institute, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Joel E Michalek
- Departments of Mays Cancer Center, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA; Departments of Epidemiology and Biostatistics, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Daniel S Higginson
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yuji Ikeno
- Departments of Pathology, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA; Barshop Institute for Longevity and Aging Studies, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA; Audie L. Murphy VA Hospital (STVHCS), Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Chul Soo Ha
- Departments of Mays Cancer Center, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA; Department of Radiation Oncology, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Yidong Chen
- Departments of Greehey Children's Cancer Institute, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA; Departments of Mays Cancer Center, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA
| | - Rita Ghosh
- Departments of Molecular Medicine, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA; Departments of Urology, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA; Departments of Pharmacology, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA.
| | - Addanki P Kumar
- Departments of Molecular Medicine, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA; Departments of Urology, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA; Departments of Pharmacology, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA; Departments of Mays Cancer Center, Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA; Audie L. Murphy VA Hospital (STVHCS), Long School of Medicine, The University of Texas Health San Antonio, TX, 78229, USA.
| |
Collapse
|
3
|
Liu C, Xiao K, Yu C, Lei Y, Lyu K, Tian T, Zhao D, Zhou F, Tang H, Zeng J. A probabilistic knowledge graph for target identification. PLoS Comput Biol 2024; 20:e1011945. [PMID: 38578805 PMCID: PMC11034645 DOI: 10.1371/journal.pcbi.1011945] [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] [Received: 05/20/2023] [Revised: 04/22/2024] [Accepted: 02/24/2024] [Indexed: 04/07/2024] Open
Abstract
Early identification of safe and efficacious disease targets is crucial to alleviating the tremendous cost of drug discovery projects. However, existing experimental methods for identifying new targets are generally labor-intensive and failure-prone. On the other hand, computational approaches, especially machine learning-based frameworks, have shown remarkable application potential in drug discovery. In this work, we propose Progeni, a novel machine learning-based framework for target identification. In addition to fully exploiting the known heterogeneous biological networks from various sources, Progeni integrates literature evidence about the relations between biological entities to construct a probabilistic knowledge graph. Graph neural networks are then employed in Progeni to learn the feature embeddings of biological entities to facilitate the identification of biologically relevant target candidates. A comprehensive evaluation of Progeni demonstrated its superior predictive power over the baseline methods on the target identification task. In addition, our extensive tests showed that Progeni exhibited high robustness to the negative effect of exposure bias, a common phenomenon in recommendation systems, and effectively identified new targets that can be strongly supported by the literature. Moreover, our wet lab experiments successfully validated the biological significance of the top target candidates predicted by Progeni for melanoma and colorectal cancer. All these results suggested that Progeni can identify biologically effective targets and thus provide a powerful and useful tool for advancing the drug discovery process.
Collapse
Affiliation(s)
- Chang Liu
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China
| | - Kaimin Xiao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, Beijing, China
| | - Cuinan Yu
- Machine Learning Department, Silexon AI Technology Co., Ltd., Nanjing, Jiangsu Province, China
| | - Yipin Lei
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China
| | - Kangbo Lyu
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China
| | - Tingzhong Tian
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China
| | - Dan Zhao
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China
| | - Fengfeng Zhou
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, Jilin Province, China
| | - Haidong Tang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Jianyang Zeng
- School of Engineering, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Research Center for Industries of the Future and School of Engineering, Westlake University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
4
|
Kim DE, Roh HS, Kim GH, Bhang DH, Um SH, Singh R, Baek KH. S6K1 deficiency in tumor stroma impairs lung metastasis of melanoma in mice. Biochem Biophys Res Commun 2024; 696:149469. [PMID: 38194806 DOI: 10.1016/j.bbrc.2024.149469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024]
Abstract
Accumulating data suggest that ribosomal protein S6 kinase 1 (S6K1), an effector in the mammalian target of rapamycin (mTOR) pathway, plays pleiotropic roles in tumor progression. However, to date, while the tumorigenic function of S6K1 in tumor cells has been well elucidated, its role in the tumor stroma remains poorly understood. We recently showed that S6K1 mediates vascular endothelial growth factor A (VEGF-A) production in macrophages, thereby supporting tumor angiogenesis and growth. As macrophage-derived VEGF-A is crucial for both tumor cell intravasation and extravasation across the vascular endothelium, our previous findings suggest that stromal S6K1 signaling is required for tumor metastatic spread. Therefore, we aimed to determine the impact of host S6K1 depletion on tumor metastasis using a murine model of pulmonary metastasis (S6k1-/- mice implanted with B16F10 melanoma). The ablation of S6K1 in the host microenvironment significantly reduced the metastasized B16F10 melanoma cells on the lung surface in both spontaneous and intravenous lung metastasis mouse models without affecting the incidence of metastasis to distant lymph nodes. In addition, stromal S6K1 loss decreased the number of tumor cells circulating in the peripheral blood of mice bearing B16F10 xenografts without affecting the vascular leakage induced by VEGF-A in vivo. These observations demonstrate that S6K1 signaling in host cells other than endothelial cells is required to modulate the host microenvironment to facilitate the metastatic spread of tumors via blood circulation, thus revealing its novel role in the tumor stroma during tumor progression.
Collapse
Affiliation(s)
- Da-Eun Kim
- Department of Molecular and Cellular Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Hyun-Soo Roh
- Department of Molecular and Cellular Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Ga-Hee Kim
- Department of Molecular and Cellular Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Dong Ha Bhang
- Department of Molecular and Cellular Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Sung Hee Um
- Department of Molecular and Cellular Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Rohit Singh
- Immuno-oncology Branch, Division of Rare and Refractory Cancer, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Kwan-Hyuck Baek
- Department of Molecular and Cellular Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi, 16419, Republic of Korea.
| |
Collapse
|
5
|
Yang X, Guo J, Li W, Li C, Zhu X, Liu Y, Wu X. PPM1H is down-regulated by ATF6 and dephosphorylates p-RPS6KB1 to inhibit progression of hepatocellular carcinoma. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:164-179. [PMID: 37456776 PMCID: PMC10345229 DOI: 10.1016/j.omtn.2023.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023]
Abstract
We have shown previously that polymorphism of activating transcription factor 6 (ATF6) is associated with susceptibility to hepatocellular carcinoma (HCC). Therefore, genes down-regulated by ATF6 might play a tumor-suppressing role. In the present study, we identified that expression of protein phosphatase magnesium- or manganous-dependent 1H (PPM1H) mRNA and protein can be inhibited by ATF6 in hepatoma cells and mice with liver Atf6 knockdown. Tumor tissues from 134 HCC patients were analyzed by immunohistochemistry, and PPM1H exhibited higher expression levels in adjacent para-cancer tissues than in HCC tissues. Therefore, patients with higher expression of PPM1H had a better prognosis. PPM1H inhibited proliferation, migration, and invasion of hepatoma cells. In addition, PPM1H inhibited induced HCC nodule formation as well as tumor xenograft growth in diethylnitrosamine/CCl4-induced HCC mouse model and nude mouse tumorigenicity assay, respectively. A 3D model of PPM1H was obtained by homology multi-template modeling, and ribosomal protein S6 kinase B1 (RPS6KB1) in the bone morphogenetic protein (BMP)/transforming growth factor β (TGF-β) pathway was screened out as the potential substrate of PPM1H by Rosetta. PPM1H could directly dephosphorylate p-RPS6KB1. To conclude, we discovered RPS6KB1 as a new PPM1H dephosphorylation substrate. PPM1H exhibited a suppressive effect on HCC progression by dephosphorylating p-RPS6KB1.
Collapse
Affiliation(s)
- Xiaoshuang Yang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, P.R. China
- School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Jianting Guo
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, P.R. China
- School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Wei Li
- Department of Interventional Radiology, Affiliated Hospital of Qingdao University, Shandong 266003, P.R. China
| | - Chunrui Li
- Beijing Cloud Computing Key Technique and Application Key Laboratory, Beijing Computing Center, Beijing 100094, P.R. China
| | - Xilin Zhu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, P.R. China
- School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Ying Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, P.R. China
- School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| | - Xiaopan Wu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, P.R. China
- School of Basic Medicine, Peking Union Medical College, Beijing 100005, P.R. China
| |
Collapse
|
6
|
Prediction of Aflatoxin-B1 (AFB1) Molecular Mechanism Network and Interaction to Oncoproteins Growth Factor in Hepatocellular Carcinoma. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2022. [DOI: 10.22207/jpam.16.3.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Aflatoxin-B1 (AFB1) is a common contaminant for staple foods during the storage process. Chronic exposure to AFB1 is widely known to induce the development of hepatocellular carcinoma (HCC). However, there is a lack of understanding of AFBi role in HCC mechanism. This research aims to identify protein(s) in HCC that might interact with AFB1 and to predict the pathway effected by AFB1. Analyses were performed using bioinformatics tools. SMILES notation of AFB1 was submitted into Swiss Target Prediction. Interaction among predicted proteins were analyzed by using STRING. The 3D structure of target protein was constructed by homology modeling. Reverse docking was performed, and the result was ranked based on binding affinity score. Furthermore, protein interaction network was constructed and analyzed by using Cytoscape. Results showed that three protein groups were predicted as target of AFB1, such as kinases, phosphatases, and G protein-coupled receptor with probability of 46.7%, 20%, and 6.7%, respectively. Seven proteins of kinases were strongly related to HCC, including RAF1, MAPK1, MAPK3, AKT1, EGFR, GSK3B, and mTOR. Reverse docking considered the AKT1-AFB1 as the most potential complex with the lowest affinity score -10.2 kcal.mol-1. It has hydrophobic bonds in Trp80, Val270, Tyr272, Asp292, Thr211, Leu210, Leu264, and Lys268 residues, whereas hydrogen bond in Ser205 residues. Moreover, further analysis demonstrated that interaction of AKT1-AFB1 is related to the metastasis pathway in HCC mechanism.
Collapse
|
7
|
Beyond controlling cell size: functional analyses of S6K in tumorigenesis. Cell Death Dis 2022; 13:646. [PMID: 35879299 PMCID: PMC9314331 DOI: 10.1038/s41419-022-05081-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 01/21/2023]
Abstract
As a substrate and major effector of the mammalian target of rapamycin complex 1 (mTORC1), the biological functions of ribosomal protein S6 kinase (S6K) have been canonically assigned for cell size control by facilitating mRNA transcription, splicing, and protein synthesis. However, accumulating evidence implies that diverse stimuli and upstream regulators modulate S6K kinase activity, leading to the activation of a plethora of downstream substrates for distinct pathobiological functions. Beyond controlling cell size, S6K simultaneously plays crucial roles in directing cell apoptosis, metabolism, and feedback regulation of its upstream signals. Thus, we comprehensively summarize the emerging upstream regulators, downstream substrates, mouse models, clinical relevance, and candidate inhibitors for S6K and shed light on S6K as a potential therapeutic target for cancers.
Collapse
|
8
|
p70 S6 kinase as a therapeutic target in cancers: More than just an mTOR effector. Cancer Lett 2022; 535:215593. [PMID: 35176419 DOI: 10.1016/j.canlet.2022.215593] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/25/2022] [Accepted: 02/06/2022] [Indexed: 11/23/2022]
Abstract
p70 S6 kinase (p70S6K) is best-known for its regulatory roles in protein synthesis and cell growth by phosphorylating its primary substrate, ribosomal protein S6, upon mitogen stimulation. The enhanced expression/activation of p70S6K has been correlated with poor prognosis in some cancer types, suggesting that it may serve as a biomarker for disease monitoring. p70S6K is a critical downstream effector of the oncogenic PI3K/Akt/mTOR pathway and its activation is tightly regulated by an ordered cascade of Ser/Thr phosphorylation events. Nonetheless, it should be noted that other upstream mechanisms regulating p70S6K at both the post-translational and post-transcriptional levels also exist. Activated p70S6K could promote various aspects of cancer progression such as epithelial-mesenchymal transition, cancer stemness and drug resistance. Importantly, novel evidence showing that p70S6K may also regulate different cellular components in the tumor microenvironment will be discussed. Therapeutic targeting of p70S6K alone or in combination with traditional chemotherapies or other microenvironmental-based drugs such as immunotherapy may represent promising approaches against cancers with aberrant p70S6K signaling. Currently, the only clinically available p70S6K inhibitors are rapamycin analogs (rapalogs) which target mTOR. However, there are emerging p70S6K-selective drugs which are going through active preclinical or clinical trial phases. Moreover, various screening strategies have been used for the discovery of novel p70S6K inhibitors, hence bringing new insights for p70S6K-targeted therapy.
Collapse
|
9
|
Majeed ST, Batool A, Majeed R, Bhat NN, Zargar MA, Andrabi KI. mTORC1 induces eukaryotic translation initiation factor 4E interaction with TOS-S6 kinase 1 and its activation. Cell Cycle 2021; 20:839-854. [PMID: 33938392 DOI: 10.1080/15384101.2021.1901038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Eukaryotic translation initiation factor 4E was recently shown to be a substrate of mTORC1, suggesting it may be a mediator of mTORC1 signaling. Here, we present evidence that eIF4E phosphorylated at S209 interacts with TOS motif of S6 Kinase1 (S6K1). We also show that this interaction is sufficient to overcome rapamycin sensitivity and mTORC1 dependence of S6K1. Furthermore, we show that eIF4E-TOS interaction relieves S6K1 from auto-inhibition due to carboxy terminal domain (CTD) and primes it for hydrophobic motif (HM) phosphorylation and activation in mTORC1 independent manner. We conclude that the role of mTORC1 is restricted to engaging eIF4E with S6K1-TOS motif to influence its state of HM phosphorylation and inducing its activation.
Collapse
Affiliation(s)
- Sheikh Tahir Majeed
- Growth Factor Signaling Laboratory, Department of Biotechnology, University of Kashmir, Srinagar, India.,Department of Biotechnology, Central University of Kashmir, Ganderbal, India
| | - Asiya Batool
- Growth Factor Signaling Laboratory, Department of Biotechnology, University of Kashmir, Srinagar, India.,Division of Cancer Pharmacology, Indian Institute of Integrative Medicine, Srinagar, India
| | - Rabiya Majeed
- Growth Factor Signaling Laboratory, Department of Biotechnology, University of Kashmir, Srinagar, India.,Department of Biochemistry, University of Kashmir, Srinagar, India
| | - Nadiem Nazir Bhat
- Growth Factor Signaling Laboratory, Department of Biotechnology, University of Kashmir, Srinagar, India
| | | | - Khurshid Iqbal Andrabi
- Growth Factor Signaling Laboratory, Department of Biotechnology, University of Kashmir, Srinagar, India
| |
Collapse
|
10
|
A New Computational Approach to Evaluating Systemic Gene–Gene Interactions in a Pathway Affected by Drug LY294002. Processes (Basel) 2020. [DOI: 10.3390/pr8101230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In this study, we investigate how drugs systemically affect genes via pathways by integrating information from interactions between chemical compounds and molecular expression datasets, and from pathway information such as gene sets using mathematical models. First, we adopt drug-induced gene expression datasets; then, employ gene set enrichment analysis tools for selecting candidate enrichment pathways; and lastly, implement the inverse algorithm package for identifying gene–gene regulatory networks in a pathway. We tested LY294002-induced datasets of the MCF7 breast cancer cell lines, and found a CELL CYCLE pathway with 101 genes, ERBB signaling pathway consisting of 82 genes, and MTOR pathway consisting of 45 genes. We consider two interactions: quantity strength depending on number of interactions, and quality strength depending on weight of interaction as positive (+) and negative (−) interactions. Our methods revealed ANAPC1-CDK6 (−0.412) and ORC2L- CHEK1(0.951) for the CELL CYCLE pathway; INS-RPS6 (−3.125) and PRKAA2-PRKAA2 (+1.319) for the MTOR pathway; and CBLB-RPS6KB1 (−0.141), RPS6KB1-CBLC (+0.238) for the ERBB signaling pathway to be top quality interactions. Top quantity interactions discovered include 12; the CDC (−,+) gene family for the CELL CYCLE pathway, 20; PIK3 (−), 23; PIK3CG (+) for the MTOR pathway, 11; PAK (−), 10; PIK3 (+) for the ERBB signaling pathway.
Collapse
|
11
|
Athamneh K, Alneyadi A, Alsamri H, Alrashedi A, Palakott A, El-Tarabily KA, Eid AH, Al Dhaheri Y, Iratni R. Origanum majorana Essential Oil Triggers p38 MAPK-Mediated Protective Autophagy, Apoptosis, and Caspase-Dependent Cleavage of P70S6K in Colorectal Cancer Cells. Biomolecules 2020; 10:biom10030412. [PMID: 32155920 PMCID: PMC7175132 DOI: 10.3390/biom10030412] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 02/26/2020] [Accepted: 02/26/2020] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common type of cancer in terms of incidence and mortality worldwide. Here we have investigated the anti-colon cancer potential of Origanum majorana essential oil (OMEO) and its underlying mechanisms of action. We showed that OMEO significantly inhibited the cellular viability and colony growth of human HT-29 colorectal cancer cells. OMEO induced protective autophagy, associated with downregulation of the mTOR/p70S6K pathway, and activated caspase-8 and caspase-9-dependent apoptosis. Blockade of autophagy with 3-methyladenine (3-MA) and chloroquine (CQ), two autophagy inhibitors, potentiated the OMEO-induced apoptotic cell death. Inversely, inhibition of apoptosis with the pan-caspase inhibitor, Z-VAD-FMK, significantly reduced cell death, suggesting that apoptosis represents the main mechanism of OMEO-induced cell death. Mechanistically, we found that OMEO induces protective autophagy and apoptotic cells death via the activation of the p38 MAPK signaling pathway. Pharmacological inhibition of p38 MAPK by the p38 inhibitors SB 202190 and SB 203580 not only significantly decreased apoptotic cell death, but also reduced the autophagy level in OMEO treated HT-29 cells. Strikingly, we found that OMEO also induces p38 MAPK-mediated caspase-dependent cleavage of p70S6K, a protein reported to be overexpressed in colon cancer and associated with drug resistance. Our findings suggest that OMEO inhibits colon cancer through p38 MAPK-mediated protective autophagy and apoptosis associated with caspase-dependent cleavage of p70S6K. To the best of our knowledge, this study is the first to report on the implications of the p38 MAPK signaling pathway in targeting p70S6K to caspase cleavage.
Collapse
Affiliation(s)
- Khawlah Athamneh
- Department of Biology, College of Science, UAE University, United Arab Emirates University, Al-Ain P.O. Box 15551, UAE; (K.A.); (A.A.); (H.A.); (A.A.); (A.P.); (K.A.E.-T.); (Y.A.D.)
| | - Aysha Alneyadi
- Department of Biology, College of Science, UAE University, United Arab Emirates University, Al-Ain P.O. Box 15551, UAE; (K.A.); (A.A.); (H.A.); (A.A.); (A.P.); (K.A.E.-T.); (Y.A.D.)
| | - Halima Alsamri
- Department of Biology, College of Science, UAE University, United Arab Emirates University, Al-Ain P.O. Box 15551, UAE; (K.A.); (A.A.); (H.A.); (A.A.); (A.P.); (K.A.E.-T.); (Y.A.D.)
| | - Asma Alrashedi
- Department of Biology, College of Science, UAE University, United Arab Emirates University, Al-Ain P.O. Box 15551, UAE; (K.A.); (A.A.); (H.A.); (A.A.); (A.P.); (K.A.E.-T.); (Y.A.D.)
| | - Abdulrasheed Palakott
- Department of Biology, College of Science, UAE University, United Arab Emirates University, Al-Ain P.O. Box 15551, UAE; (K.A.); (A.A.); (H.A.); (A.A.); (A.P.); (K.A.E.-T.); (Y.A.D.)
| | - Khaled A. El-Tarabily
- Department of Biology, College of Science, UAE University, United Arab Emirates University, Al-Ain P.O. Box 15551, UAE; (K.A.); (A.A.); (H.A.); (A.A.); (A.P.); (K.A.E.-T.); (Y.A.D.)
- Khalifa Center for Genetic Engineering and Biotechnology, United Arab Emirates University, Al-Ain P.O. Box 15551, UAE
| | - Ali H. Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon;
| | - Yusra Al Dhaheri
- Department of Biology, College of Science, UAE University, United Arab Emirates University, Al-Ain P.O. Box 15551, UAE; (K.A.); (A.A.); (H.A.); (A.A.); (A.P.); (K.A.E.-T.); (Y.A.D.)
| | - Rabah Iratni
- Department of Biology, College of Science, UAE University, United Arab Emirates University, Al-Ain P.O. Box 15551, UAE; (K.A.); (A.A.); (H.A.); (A.A.); (A.P.); (K.A.E.-T.); (Y.A.D.)
- Correspondence: ; Tel.: +971-3-713-6526; Fax: +971-3-7134927
| |
Collapse
|
12
|
Hermawan A, Putri H. Bioinformatics Studies Provide Insight into Possible Target and Mechanisms of Action of Nobiletin against Cancer Stem Cells. Asian Pac J Cancer Prev 2020; 21:611-620. [PMID: 32212785 PMCID: PMC7437309 DOI: 10.31557/apjcp.2020.21.3.611] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 03/06/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Nobiletin treatment on MDA-MB 231 cells reduces the expression of CXC chemokine receptor type 4 (CXCR4), which is highly expressed in cancer stem cell populations in tumor patients. However, the mechanisms of nobiletin in cancer stem cells (CSCs) remain elusive. This study was aimed to explore the potential target and mechanisms of nobiletin in cancer stem cells using bioinformatics approaches. METHODS Gene expression profiles by public COMPARE predicting the sensitivity of tumor cells to nobiletin. Functional annotations on gene lists are carried out with The Database for Annotation, Visualization and Integrated Discovery (DAVID) v6.8, and WEB-based GEne SeT Analysis Toolkit (WebGestalt). The protein-protein interaction (PPI) network was analyzed by STRING-DB and visualized by Cytoscape. RESULTS Microarray analyses reveal many genes involved in protein binding, transcriptional and translational activity. Pathway enrichment analysis revealed breast cancer regulation of estrogen signaling and Wnt/ß-catenin by nobiletin. Moreover, three hub genes, i.e. ESR1, NCOA3, and RPS6KB1 and one significant module were filtered out and selected from the PPI network. CONCLUSION Nobiletin might serve as a lead compound for the development of CSCs-targeted drugs by targeting estrogen and Wnt/ß-catenin signaling. Further studies are needed to explore the full therapeutic potential of nobiletin in cancer stem cells. .
Collapse
Affiliation(s)
| | - Herwandhani Putri
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| |
Collapse
|
13
|
Zhang S, Hu B, Lv X, Chen S, Liu W, Shao Z. The Prognostic Role of Ribosomal Protein S6 Kinase 1 Pathway in Patients With Solid Tumors: A Meta-Analysis. Front Oncol 2019; 9:390. [PMID: 31139572 PMCID: PMC6527894 DOI: 10.3389/fonc.2019.00390] [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: 12/24/2018] [Accepted: 04/26/2019] [Indexed: 12/26/2022] Open
Abstract
Background: Recent studies supported the predictive role of ribosomal protein S6 kinase 1 (S6K1), phosphorylated S6K1 (p-S6K1), and phosphorylated ribosomal protein S6 (p-S6) for the outcome of cancer patients. However, inconsistent results were acquired across different researches. To comprehensively and quantitatively elucidate their prognostic significance in solid malignancies, the current meta-analysis was carried out utilizing the results of clinical studies. Methods: We conducted the literature retrieval by searching PubMed, Web of Science, EMBASE, and Cochrane library to identify eligible publications. Data were collected from included articles to calculate pooled overall survival (OS), disease-free survival (DFS), recurrence-free survival (RFS), and progression-free survival (PFS). Hazard ratios (HRs) with 95% confidence intervals (CIs) served as appropriate parameters to assess prognostic significance. Results: Forty-four original studies were included, of which 7 studies were analyzed for S6K1, 24 for p-S6K1, and 16 for p-S6. The overexpression of p-S6K1 was significantly associated with poorer prognosis of solid tumor patients in OS (HR = 1.706, 95%CI: 1.369–2.125, p < 0.001), DFS (HR = 1.665, 95%CI: 1.002–2.768, p = 0.049). However, prognostic role of p-S6K1 in RFS and PFS was not found. The result also revealed that S6K1 and p-S6 were significantly associated with reduced OS (HR = 1.691, 95%CI: 1.306–2.189, p < 0.001; HR = 2.019, 95%CI: 1.775–2.296, p < 0.001, respectively). Conclusions: The present meta-analysis demonstrated that elevated expression of S6K1, p-S6K1, or p-S6 might indicate worse prognosis of patients with solid tumors, and supported a promising clinical test to predict solid tumor prognosis based on the level of S6K1 pathway.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Binwu Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Songfeng Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weijian Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
The Р60-S6K1 isoform of ribosomal protein S6 kinase 1 is a product of alternative mRNA translation. UKRAINIAN BIOCHEMICAL JOURNAL 2018. [DOI: 10.15407/ubj90.04.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
15
|
Lui KY, Zhao H, Qiu C, Li C, Zhang Z, Peng H, Fu R, Chen HA, Lu MQ. Integrator complex subunit 6 (INTS6) inhibits hepatocellular carcinoma growth by Wnt pathway and serve as a prognostic marker. BMC Cancer 2017; 17:644. [PMID: 28899352 PMCID: PMC5596937 DOI: 10.1186/s12885-017-3628-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 08/28/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Integrator complex subunit 6 (INTS6) was found to play a tumour suppressing role in certain types of solid tumours. In this study, we wanted to determine the expression level of INTS6 in hepatocellular carcinoma (HCC) and evaluate its clinical characteristics and mechanisms in HCC patients (Lui and Lu, European Journal of Cancer, 51:S94, 2015). METHODS First, we used a microarray analysis to explore the mRNA expression levels in HCC and paired normal liver tissues; second, we used qRT-PCR to measure the INTS6 mRNA levels in a cohort of 50 HCC tissues and adjacent normal liver tissues; third, we used Western blot analyses to detect the INTS6 protein levels in 20 paired HCC and normal liver tissues; fourth, we used immunohistochemistry to determine the INTS6 expression levels in 70 archived paraffin-embedded HCC samples. Finally, we investigated the suppressive function of INTS6 in the Wnt pathway. RESULTS Herein, according to the microarray data analysis, the expression levels of INTS6 were dramatically down-regulated in HCC tissues vs. those in normal liver tissues (p<0.05). qRT-PCR and Western blot analyses showed that the INTS6 mRNA and protein expression was significantly down-regulated in tumour tissues compared to the adjacent normal liver tissues (p<0.05). Immunohistochemical assays revealed that decreased INTS6 expression was present in 62.9% (44/70) of HCC patients. Correlation analyses showed that INTS6 expression was significantly correlated with serum alpha-fetoprotein levels (AFP, p =0.004), pathology grade (p =0.005), and tumour recurrence (p =0.04). Kaplan-Meier analysis revealed that patients with low INTS6 expression levels had shorter overall and disease-free survival rates than patients with high INTS6 expression levels (p =0.001 and p =0.001). Multivariate regression analysis indicated that INTS6 was an independent predictor of overall survival and disease-free survival rates. Mechanistically, INTS6 increased WIF-1 expression and then inhibited the Wnt/β-catenin signalling pathway. CONCLUSION The results of our study show that down-regulated INTS6 expression is associated with a poorer prognosis in HCC patients. This newly identified INTS6/WIF-1 axis indicates the molecular mechanism of HCC and may represent a therapeutic target in HCC patients.
Collapse
Affiliation(s)
- Ka Yin Lui
- Department of Critical Care Medicine, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Hui Zhao
- Department of Hepatic Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Chunhui Qiu
- Department of Hepatic Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Chuo Li
- Obstetric Laboratory, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Zhigang Zhang
- Department of Pathology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Haoran Peng
- Transitional Year, Gwinnentt Medical Center, Lawrenceville, GA, USA
| | - Rongdang Fu
- Department of Hepatic Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Hu-An Chen
- Department of Hepatic Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Min-Qiang Lu
- Department of Hepatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou, 510180, China.
| |
Collapse
|
16
|
Hyperphosphorylation of RPS6KB1, rather than overexpression, predicts worse prognosis in non-small cell lung cancer patients. PLoS One 2017; 12:e0182891. [PMID: 28792981 PMCID: PMC5549961 DOI: 10.1371/journal.pone.0182891] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/26/2017] [Indexed: 02/05/2023] Open
Abstract
RPS6KB1 is the kinase of ribosomal protein S6 which is 70 kDa and is required for protein translation. Although the abnormal activation of RPS6KB1 has been found in types of diseases, its role and clinical significance in non-small cell lung cancer (NSCLC) has not been fully investigated. In this study, we identified that RPS6KB1 was over-phosphorylated (p-RPS6KB1) in NSCLC and it was an independent unfavorable prognostic marker for NSCLC patients. In spite of the frequent expression of total RPS6KB1 and p-RPS6KB1 in NSCLC specimens by immunohistochemical staining (IHC), only p-RPS6KB1 was associated with the clinicopathologic characteristics of NSCLC subjects. Kaplan-Meier survival analysis revealed that the increased expression of p-RPS6KB1 indicated a poorer 5-year overall survival (OS) for NSCLC patients, while the difference between the positive or negative RPS6KB1 group was not significant. Univariate and multivariate Cox regression analysis was then used to confirm the independent prognostic value of p-RPS6KB1. To illustrate the underlying mechanism of RPS6KB1 phosphorylation in NSCLC, LY2584702 was employed to inhibit the RPS6KB1 phosphorylation specifically both in lung adenocarcinoma cell line A549 and squamous cell carcinoma cell line SK-MES-1. As expected, RPS6KB1 dephosphorylation remarkably suppressed cells proliferation in CCK-8 test, and promoted more cells arresting in G0-G1 phase by cell cycle analysis. Moreover, apoptotic A549 cells with RPS6KB1 dephosphorylation increased dramatically, with an elevating trend in SK-MES-1, indicating a potential involvement of RPS6KB1 phosphorylation in inducing apoptosis. In conclusion, our data suggest that RPS6KB1 is over-activated as p-RPS6KB1 in NSCLC, rather than just the total protein overexpressing. The phosphorylation level of RPS6KB1 might be used as a novel prognostic marker for NSCLC patients.
Collapse
|
17
|
Identification and quantification of novel RNA isoforms in horn cancer of Bos indicus by comprehensive RNA-Seq. 3 Biotech 2016; 6:259. [PMID: 28330331 PMCID: PMC5143338 DOI: 10.1007/s13205-016-0577-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/26/2016] [Indexed: 02/07/2023] Open
Abstract
Horn cancer (HC) is a squamous cell carcinoma of horn, commonly observed in Bos indicus of the Asian countries. To elucidate the complexity of alternative splicing present in the HC, high-throughput sequencing and analysis of HC and matching horn normal (HN) tissue were carried out. A total of 535,067 and 849,077 reads were analysed after stringent quality filtering for HN and HC, respectively. Cufflinks pipeline for transcriptome analysis revealed 4786 novel splice isoforms comprising 2432 exclusively in HC, 2055 exclusively in HN and 298 in both the conditions. Based on pathway clustering and in silico verification, 102 novel splice isoforms were selected and further analysed with respect to change in protein sequence using Blastp. Finally, fourteen novel splicing events supported both by Cufflinks and UCSC genome browser were selected and confirmed expression by RT-qPCR. Future studies targeted at in-depth characterization of these potential candidate splice isoforms might be helpful in the development of relevant biomarkers for early diagnosis of HC. The results reported in this study refine the available information on transcriptome repertoire of bovine species and boost the research in the line of development of relevant biomarkers for early diagnosis of HC.
Collapse
|
18
|
Qiu ZX, Sun RF, Mo XM, Li WM. The p70S6K Specific Inhibitor PF-4708671 Impedes Non-Small Cell Lung Cancer Growth. PLoS One 2016; 11:e0147185. [PMID: 26771549 PMCID: PMC4714881 DOI: 10.1371/journal.pone.0147185] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 12/30/2015] [Indexed: 02/05/2023] Open
Abstract
Background As a serine/threonine protein kinase, p70S6K plays an important role in tumor cells. Evidence has revealed overexpression of p70S6K and phosphorylated p70S6K (p-p70S6K) in various tumor tissues, with these proteins identified as independent prognostic markers in non-small cell lung cancer (NSCLC). In this study, we explored the role of the p70S6K specific inhibitor PF-4708671 in NSCLC. Methods Three NSCLC cell lines (A549, SK-MES-1, and NCI-H460) were treated with PF-4708671 at five different concentrations, including 0.1μM, 0.3μM, 1μM, 3μM and 10μM, and protein levels were determined by Western-blot. Then, PF-4708671’s effects were assessed both in vitro (cell proliferation, apoptosis, cell cycle distribution, and invasion) and in vivo. Results The expression levels of p-p70S6K and the downstream effector S6 were significantly reduced by PF-4708671. Diametrically opposite, the downstream protein levels of BAD, Caspase3 and ERK had increased after treatment with PF-4708671. In addition, PF-4708671 drastically inhibited cell proliferation and invasion ability in A549, SK-MES-1 and NCI-H460 cells in vitro, causing cell cycle arrest in G0-G1 phase. Limited effects of PF-4708671 were observed on apoptosis in the three NSCLC cell lines assessed. Importantly, PF-4708671 could inhibit tumorigenesis in nude mice in vivo. Conclusion These findings demonstrated that the p70S6K specific inhibitor PF-4708671 has inhibitory effects on NSCLC tumorigenesis in vitro and in vivo. Therefore, P70S6K should be considered a new potential therapeutic target, and PF-470867 may be used as targeted drug for cancer treatment.
Collapse
Affiliation(s)
- Zhi-Xin Qiu
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rong-Fei Sun
- Department of Respiratory Medicine, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Xian-Ming Mo
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei-Min Li
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
- * E-mail:
| |
Collapse
|
19
|
Cai C, Chen QB, Han ZD, Zhang YQ, He HC, Chen JH, Chen YR, Yang SB, Wu YD, Zeng YR, Qin GQ, Liang YX, Dai QS, Jiang FN, Wu SL, Zeng GH, Zhong WD, Wu CL. miR-195 Inhibits Tumor Progression by Targeting RPS6KB1 in Human Prostate Cancer. Clin Cancer Res 2015; 21:4922-34. [PMID: 26080838 DOI: 10.1158/1078-0432.ccr-15-0217] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 06/10/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate the involvement of hsa-miRNA-195-5p (miR-195) in progression and prognosis of human prostate cancer. EXPERIMENTAL DESIGN qRT-PCR was performed to detect miR-195 expression in both prostate cancer cell lines and clinical tissue samples. Its clinical significance was statistically analyzed. The roles of miR-195 and its candidate target gene, ribosomal protein S6 kinase, 70 kDa, polypeptide 1 (RPS6KB1) in prostate cancer progression were confirmed on the basis of both in vitro and in vivo systems. RESULTS miR-195 downregulation in prostate cancer tissues was significantly associated with high Gleason score (P = 0.001), positive metastasis failure (P < 0.001), and biochemical recurrence (BCR, P < 0.001). Survival analysis identified miR-195 as an independent prognostic factor for BCR-free survival of prostate cancer patients (P = 0.022). Then, we confirmed the tumor suppressive role of miR-195 through prostate cancer cell invasion, migration, and apoptosis assays in vitro, along with tumor xenograft growth, angiogenesis, and invasion in vivo according to both gain-of-function and loss-of-function experiments. In addition, RPS6KB1 was identified as a novel direct target of miR-195 through proteomic expression profiling combined with bioinformatic target prediction and luciferase reporter assay. Moreover, the reexpression and knockdown of RPS6KB1 could respectively rescue and imitate the effects induced by miR-195. Importantly, RPS6KB1 expression was closely correlated with aggressive progression and poor prognosis in prostate cancer patients as opposed to miR-195. Furthermore, we identified MMP-9, VEGF, BAD, and E-cadherin as the downstream effectors of miR-195-RPS6KB1 axis. CONCLUSION The newly identified miR-195-RPS6KB1 axis partially illustrates the molecular mechanism of prostate cancer progression and represents a novel potential therapeutic target for prostate cancer treatment.
Collapse
Affiliation(s)
- Chao Cai
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, and Guangdong Key Laboratory of Urology, Guangzhou, China
| | - Qing-Biao Chen
- Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou, China
| | - Zhao-Dong Han
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yan-Qiong Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui-Chan He
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, and Guangdong Key Laboratory of Urology, Guangzhou, China. Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jia-Hong Chen
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yan-Ru Chen
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Sheng-Bang Yang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yong-Ding Wu
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yan-Ru Zeng
- Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou, China. Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Guo-Qiang Qin
- Central Hospital of Panyu District, Guangzhou, China
| | - Yu-Xiang Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qi-Shan Dai
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Fu-Neng Jiang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shu-lin Wu
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Guo-Hua Zeng
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, and Guangdong Key Laboratory of Urology, Guangzhou, China
| | - Wei-De Zhong
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, and Guangdong Key Laboratory of Urology, Guangzhou, China. Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou, China. 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 and Harvard Medical School, Boston, Massachusetts. Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
20
|
Bostner J, Karlsson E, Eding CB, Perez-Tenorio G, Franzén H, Konstantinell A, Fornander T, Nordenskjöld B, Stål O. S6 kinase signaling: tamoxifen response and prognostic indication in two breast cancer cohorts. Endocr Relat Cancer 2015; 22:331-43. [PMID: 25972244 DOI: 10.1530/erc-14-0513] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Detection of signals in the mammalian target of rapamycin (mTOR) and the estrogen receptor (ER) pathways may be a future clinical tool for the prediction of adjuvant treatment response in primary breast cancer. Using immunohistological staining, we investigated the value of the mTOR targets p70-S6 kinase (S6K) 1 and 2 as biomarkers for tamoxifen benefit in two independent clinical trials comparing adjuvant tamoxifen with no tamoxifen or 5 years versus 2 years of tamoxifen treatment. In addition, the prognostic value of the S6Ks was evaluated. We found that S6K1 correlated with proliferation, HER2 status, and cytoplasmic AKT activity, whereas high protein expression levels of S6K2 and phosphorylated (p) S6K were more common in ER-positive, and low-proliferative tumors with pAKT-s473 localized to the nucelus. Nuclear accumulation of S6K1 was indicative of a reduced tamoxifen effect (hazard ratio (HR): 1.07, 95% CI: 0.53-2.81, P=0.84), compared with a significant benefit from tamoxifen treatment in patients without tumor S6K1 nuclear accumulation (HR: 0.42, 95% CI: 0.29-0.62, P<0.00001). Also S6K1 and S6K2 activation, indicated by pS6K-t389 expression, was associated with low benefit from tamoxifen (HR: 0.97, 95% CI: 0.50-1.87, P=0.92). In addition, high protein expression of S6K1, independent of localization, predicted worse prognosis in a multivariate analysis, P=0.00041 (cytoplasm), P=0.016 (nucleus). In conclusion, the mTOR-activated kinases S6K1 and S6K2 interfere with proliferation and response to tamoxifen. Monitoring their activity and intracellular localization may provide biomarkers for breast cancer treatment, allowing the identification of a group of patients less likely to benefit from tamoxifen and thus in need of an alternative or additional targeted treatment.
Collapse
Affiliation(s)
- Josefine Bostner
- Department of Clinical and Experimental MedicineDepartment of OncologyDepartment of Clinical and Experimental MedicineDivision of Dermatology, Linköping University, SE-58185 Linköping, SwedenDepartment of OncologyKarolinska University Hospital, Karolinska Institute, SE-17176 Stockholm, Sweden
| | - Elin Karlsson
- Department of Clinical and Experimental MedicineDepartment of OncologyDepartment of Clinical and Experimental MedicineDivision of Dermatology, Linköping University, SE-58185 Linköping, SwedenDepartment of OncologyKarolinska University Hospital, Karolinska Institute, SE-17176 Stockholm, Sweden
| | - Cecilia Bivik Eding
- Department of Clinical and Experimental MedicineDepartment of OncologyDepartment of Clinical and Experimental MedicineDivision of Dermatology, Linköping University, SE-58185 Linköping, SwedenDepartment of OncologyKarolinska University Hospital, Karolinska Institute, SE-17176 Stockholm, Sweden
| | - Gizeh Perez-Tenorio
- Department of Clinical and Experimental MedicineDepartment of OncologyDepartment of Clinical and Experimental MedicineDivision of Dermatology, Linköping University, SE-58185 Linköping, SwedenDepartment of OncologyKarolinska University Hospital, Karolinska Institute, SE-17176 Stockholm, Sweden
| | - Hanna Franzén
- Department of Clinical and Experimental MedicineDepartment of OncologyDepartment of Clinical and Experimental MedicineDivision of Dermatology, Linköping University, SE-58185 Linköping, SwedenDepartment of OncologyKarolinska University Hospital, Karolinska Institute, SE-17176 Stockholm, Sweden
| | - Aelita Konstantinell
- Department of Clinical and Experimental MedicineDepartment of OncologyDepartment of Clinical and Experimental MedicineDivision of Dermatology, Linköping University, SE-58185 Linköping, SwedenDepartment of OncologyKarolinska University Hospital, Karolinska Institute, SE-17176 Stockholm, Sweden
| | - Tommy Fornander
- Department of Clinical and Experimental MedicineDepartment of OncologyDepartment of Clinical and Experimental MedicineDivision of Dermatology, Linköping University, SE-58185 Linköping, SwedenDepartment of OncologyKarolinska University Hospital, Karolinska Institute, SE-17176 Stockholm, Sweden
| | - Bo Nordenskjöld
- Department of Clinical and Experimental MedicineDepartment of OncologyDepartment of Clinical and Experimental MedicineDivision of Dermatology, Linköping University, SE-58185 Linköping, SwedenDepartment of OncologyKarolinska University Hospital, Karolinska Institute, SE-17176 Stockholm, Sweden
| | - Olle Stål
- Department of Clinical and Experimental MedicineDepartment of OncologyDepartment of Clinical and Experimental MedicineDivision of Dermatology, Linköping University, SE-58185 Linköping, SwedenDepartment of OncologyKarolinska University Hospital, Karolinska Institute, SE-17176 Stockholm, Sweden
| |
Collapse
|
21
|
Janaki Ramaiah M, Lavanya A, Honarpisheh M, Zarea M, Bhadra U, Bhadra MP. MiR-15/16 complex targets p70S6 kinase 1 and controls cell proliferation in MDA-MB-231 breast cancer cells. Gene 2014; 552:255-64. [PMID: 25261849 DOI: 10.1016/j.gene.2014.09.052] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 09/18/2014] [Accepted: 09/22/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND MicroRNAs are small non-coding RNAs that regulate post-transcriptional mRNA expression by binding to 3' untranslated region (3'-UTR) of the complementary mRNA sequence resulting in translational repression and gene silencing. They act as negative regulators of gene expression and play a pivotal role in regulating apoptosis and cell proliferation. Studies have shown that miRNAs interact with p53 by regulating the activity and function of p53 through direct repression or its regulators. Mammalian target of rapamycin (mTOR) is an evolutionary conserved check point protein kinase that plays a major effect in the control of cell division via protein synthesis regulation. mTOR regulates protein synthesis through phosphorylation and inactivation of 4E-BP1 and through phosphorylation and activation of S6 kinase 1 (S6K1). These two downstream effectors of mTOR control cell growth and metabolism. In mammals, mTOR protein kinase is the central node in the nutrient and growth factor signaling and p53 plays a critical role in sensing genotoxic stress. Activation of p53 inhibits mTOR activity, which in turn regulates its downstream targets providing a cross talk among both the signaling machinery. MicroRNA-15 and 16 belong to a common precursor family and are highly conserved. Deletion or downregulation of these two microRNAs has been shown to accelerate cell division by modulating the expression of the genes involved in controlling cell cycle progression. These microRNAs may function as tumor suppressors and act on the downstream targets of p53 signaling pathway. To have a better insight of the role of miR-15/16 in regulating the cross talk of p53 and mTOR, we performed an in depth study in MDA-MB-231 breast cancer cells by performing a gain-of-function analysis with lentiviral plasmids expressing microRNA-15 and 16. METHODS The effect of individual microRNAs on RPS6KB1 was examined by using 3'-UTR clones via luciferase based assays. The cell cycle effects were observed by flow-cytometric analysis. Reverse transcription PCR was used to explore the expression of mTOR and RPS6KB1 in cells transfected with miR-15/16. RESULTS Overexpression of miR-15/16 led to inhibition of cell proliferation causing G1 cell cycle arrest as well as caspase-3 dependent apoptosis. Forced expression of miR-15/16 might lead to decrease in mRNA level of RPS6KB1, mTOR. The effect was a complete reversal after treatment with anti-miRs against miR-15/16 proving the specificity of the expression. In addition, the dual luciferase reporter assays indicated a clear decrease in luciferase gene expression in cells transfected with lentiviral based miR-15 and 16 plasmids indicating that miR-15/16 directly targets RPS6KB1 through its 3'-UTR binding. Further, these microRNAs also inhibit epithelial to mesenchymal transition (EMT) by targeting key proteins such as Twist1 and EZH2 clearly demonstrating its crucial role in controlling cell proliferation. CONCLUSION This study suggests that exogenous microRNA-15/16 can target RPS6KB1, control cell proliferation and cause apoptosis in caspase-dependent manner even in the absence of functional p53.
Collapse
Affiliation(s)
- M Janaki Ramaiah
- Centre for Chemical Biology, Indian Institute of Chemical Technology (IICT), Tarnaka, Hyderabad, India; School of Chemical and Biotechnology, SASTRA University, Tirumalaisamudram, Thanjavur, India.
| | - A Lavanya
- Centre for Chemical Biology, Indian Institute of Chemical Technology (IICT), Tarnaka, Hyderabad, India.
| | - Mohsen Honarpisheh
- Centre for Chemical Biology, Indian Institute of Chemical Technology (IICT), Tarnaka, Hyderabad, India.
| | - Mojtaba Zarea
- Centre for Chemical Biology, Indian Institute of Chemical Technology (IICT), Tarnaka, Hyderabad, India.
| | - Utpal Bhadra
- Centre For Cellular and Molecular Biology (CCMB), Uppal Road, Hyderabad, India.
| | - Manika Pal Bhadra
- Centre for Chemical Biology, Indian Institute of Chemical Technology (IICT), Tarnaka, Hyderabad, India.
| |
Collapse
|
22
|
Retinoic acid receptor-related receptor alpha (RORalpha) is a prognostic marker for hepatocellular carcinoma. Tumour Biol 2014; 35:7603-10. [DOI: 10.1007/s13277-014-2007-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Accepted: 04/22/2014] [Indexed: 10/25/2022] Open
|
23
|
Huang YH, Chen ZK, Huang KT, Li P, He B, Guo X, Zhong JQ, Zhang QY, Shi HQ, Song QT, Yu ZP, Shan YF. Decreased expression of LKB1 correlates with poor prognosis in hepatocellular carcinoma patients undergoing hepatectomy. Asian Pac J Cancer Prev 2014; 14:1985-8. [PMID: 23679304 DOI: 10.7314/apjcp.2013.14.3.1985] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM To study any correlation of LKB1 expression with prognosis in hepatocellular carcinoma (HCC) cases. METHODS A total of 70 HCC patients and 20 primary intrahepatic stone patients in the first affiliated hospital of Wenzhou Medical College were enrolled in this study. LKB1 expression was detected by immunohistochemistry. Patients were followed-up and prognostic factors were evaluated. RESULT LKB1 expression was decreased in the HCC samples. Loss of LKB1 expression in HCC was significantly related to histologic grade (P=0.010), vascular invasion (P=0.025) and TMN stage (P=0.011). Patients showing negative LKB1 expression had a significantly shorter disease-free and overall survival than those with positive expression (P = 0.001, P=0.000, respectively). Multivariate Cox regression analysis indicated that LKB1 expression level was an independent factor of survival (P = 0.033). CONCLUSION HCC patients with decreased expression LKB1 have a poor prognosis. The loss of LKB1 expression is correlated with a lower survival rate.
Collapse
Affiliation(s)
- Yue-Han Huang
- Department of Hepatobiliary, The First Affiliated Hospital of Wenzhou Medical College, Wenzhou, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|