1
|
Patel SS, Cook RS, Lo JH, Cherry FK, Hoogenboezem EN, Yu F, Francini N, Cassidy NT, McCune JT, Gbur EF, Messier L, Dean TA, Wilson KL, Brantley-Sieders DM, Duvall CL. Induction of Triple-Negative Breast Cancer Cell Death and Chemosensitivity Using mTORC2-Directed RNAi Nanomedicine. CANCER RESEARCH COMMUNICATIONS 2025; 5:458-476. [PMID: 40019775 PMCID: PMC11921867 DOI: 10.1158/2767-9764.crc-24-0261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 11/13/2024] [Accepted: 02/26/2025] [Indexed: 03/01/2025]
Abstract
SIGNIFICANCE We identified an mTORC2/Rictor-directed RNAi nanomedicine that cooperates with chemotherapy to enhance in vivo tumor cell killing in PI3K-active TNBCs.
Collapse
Affiliation(s)
- Shrusti S. Patel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Rebecca S. Cook
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Justin H. Lo
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Fiona K. Cherry
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Ella N. Hoogenboezem
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Nora Francini
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Nina T. Cassidy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Joshua T. McCune
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Eva F. Gbur
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Lisa Messier
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Thomas A. Dean
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Kalin L. Wilson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
2
|
Sun Z, Chung D, Neelon B, Millar-Wilson A, Ethier SP, Xiao F, Zheng Y, Wallace K, Hardiman G. A Bayesian framework for pathway-guided identification of cancer subgroups by integrating multiple types of genomic data. Stat Med 2023; 42:5266-5284. [PMID: 37715500 DOI: 10.1002/sim.9911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/15/2023] [Accepted: 09/05/2023] [Indexed: 09/17/2023]
Abstract
In recent years, comprehensive cancer genomics platforms, such as The Cancer Genome Atlas (TCGA), provide access to an enormous amount of high throughput genomic datasets for each patient, including gene expression, DNA copy number alterations, DNA methylation, and somatic mutation. While the integration of these multi-omics datasets has the potential to provide novel insights that can lead to personalized medicine, most existing approaches only focus on gene-level analysis and lack the ability to facilitate biological findings at the pathway-level. In this article, we propose Bayes-InGRiD (Bayesian Integrative Genomics Robust iDentification of cancer subgroups), a novel pathway-guided Bayesian sparse latent factor model for the simultaneous identification of cancer patient subgroups (clustering) and key molecular features (variable selection) within a unified framework, based on the joint analysis of continuous, binary, and count data. By utilizing pathway (gene set) information, Bayes-InGRiD does not only enhance the accuracy and robustness of cancer patient subgroup and key molecular feature identification, but also promotes biological understanding and interpretation. Finally, to facilitate an efficient posterior sampling, an alternative Gibbs sampler for logistic and negative binomial models is proposed using Pólya-Gamma mixtures of normal to represent latent variables for binary and count data, which yields a conditionally Gaussian representation of the posterior. The R package "INGRID" implementing the proposed approach is currently available in our research group GitHub webpage (https://dongjunchung.github.io/INGRID/).
Collapse
Affiliation(s)
- Zequn Sun
- Department of Preventive Medicine, Northwestern University, Chicago, Illinois
| | - Dongjun Chung
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Brian Neelon
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina
| | | | - Stephen P Ethier
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Feifei Xiao
- Department of Biostatistics, University of Florida, Gainesville, Florida
| | - Yinan Zheng
- Department of Preventive Medicine, Northwestern University, Chicago, Illinois
| | - Kristin Wallace
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Gary Hardiman
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina
- Faculty of Medicine, Health and Life Sciences, School of Biological Sciences and Institute for Global Food Security, Queen's University Belfast, Belfast, UK
| |
Collapse
|
3
|
AKT inhibition sensitizes EVI1 expressing colon cancer cells to irinotecan therapy by regulating the Akt/mTOR axis. Cell Oncol (Dordr) 2022; 45:659-675. [PMID: 35834097 DOI: 10.1007/s13402-022-00690-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2022] [Indexed: 01/03/2023] Open
Abstract
PURPOSE Ecotropic viral integration site 1 (EVI1) is an oncogenic transcription factor that has been attributed to chemotherapy resistance in different cancers. As yet, however, its role in colon cancer drug resistance is not completely understood. Here, we set out to investigate the functional and therapeutic relevance of EVI1 in colon cancer drug resistance. METHODS The EVI1 gene was knocked down in colon cancer cells that were subsequently tested for susceptibility to irinotecan using in vitro assays and in vivo subcutaneous mouse colon cancer models. The effect of EVI1 knockdown on the AKT-mTOR signaling pathway was assessed using cell line models, immunohistochemistry and bioinformatics tools. The anti-proliferative activity of AKT inhibitor GSK690693 and its combination with irinotecan was tested in colon cancer cell line models (2D and 3D). Finally, the therapeutic efficacy of GSK690693 and its combination with irinotecan was evaluated in xenografted EVI1 expressing colon cancer mouse models. RESULTS We found that EVI1 knockdown decreased cancer stem cell-like properties and improved irinotecan responses in both cell line and subcutaneous mouse models. In addition, we found that EVI1 downregulation resulted in inhibition of AKT/mTOR signaling and RICTOR expression. Knocking down RICTOR expression increased the cytotoxic effects of irinotecan in EVI1 downregulated colon cancer cells. Co-treatment with irinotecan and ATP-competitive AKT inhibitor GSK690693 significantly reduced colon cancer cell survival and tumor progression rates. CONCLUSION Inhibition of the AKT signaling cascade by GSK690693 may serve as an alternative to improve the irinotecan response in EVI1-expressing colon cancer cells.
Collapse
|
4
|
Dual mTORC1/2 inhibitor AZD2014 diminishes myeloid-derived suppressor cells accumulation in ovarian cancer and delays tumor growth. Cancer Lett 2021; 523:72-81. [PMID: 34560229 DOI: 10.1016/j.canlet.2021.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/04/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023]
Abstract
Mechanistic target of rapamycin (mTOR) forms two distinct complexes, mTOR complex 1 (mTORC1) and mTORC2. Here we investigated the antitumor effect of dual mTORC1/2 inhibitor AZD2014 on epithelial ovarian cancer (EOC) and its potential effect on immunosuppressive myeloid-derived suppressor cells (MDSCs). Immunohistochemical analysis of mTORC1 and mTORC2 was performed on a human ovarian cancer tissue microarray. High mTORC2 expression level was associated with shorter survival in EOC, whereas mTORC1 was not correlate with patients' prognosis. AZD2014 suppressed mTOR signaling pathway in ovarian cancer cells, inhibited proliferation and induced G1-phase cell cycle arrest and apoptosis. In tumor-bearing mice, AZD2014 treatment limited tumor growth, reduced peritoneal ascites, and prolonged survival. AZD2014 specifically reduced MDSCs migration and accumulation in EOC peritoneal fluid but not in the spleen. Moreover, subsequent AZD2014 treatment after cisplatin chemotherapy delayed EOC recurrence. Collectively, we observed that high mTORC2 expression level in EOC indicated a poor prognosis. Remarkably, in tumor-bearing mice, AZD2014 diminished MDSC accumulation and delayed tumor growth and recurrence.
Collapse
|
5
|
Yoo AH, Collins AGE. How Working Memory and Reinforcement Learning Are Intertwined: A Cognitive, Neural, and Computational Perspective. J Cogn Neurosci 2021; 34:551-568. [PMID: 34942642 DOI: 10.1162/jocn_a_01808] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Reinforcement learning and working memory are two core processes of human cognition and are often considered cognitively, neuroscientifically, and algorithmically distinct. Here, we show that the brain networks that support them actually overlap significantly and that they are less distinct cognitive processes than often assumed. We review literature demonstrating the benefits of considering each process to explain properties of the other and highlight recent work investigating their more complex interactions. We discuss how future research in both computational and cognitive sciences can benefit from one another, suggesting that a key missing piece for artificial agents to learn to behave with more human-like efficiency is taking working memory's role in learning seriously. This review highlights the risks of neglecting the interplay between different processes when studying human behavior (in particular when considering individual differences). We emphasize the importance of investigating these dynamics to build a comprehensive understanding of human cognition.
Collapse
|
6
|
Zhao L, Zhu L, Oh YT, Qian G, Chen Z, Sun SY. Rictor, an essential component of mTOR complex 2, undergoes caspase-mediated cleavage during apoptosis induced by multiple stimuli. Apoptosis 2021; 26:338-347. [PMID: 33905036 DOI: 10.1007/s10495-021-01676-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2021] [Indexed: 10/21/2022]
Abstract
Caspase-mediated cleavage of proteins ensures the irreversible commitment of cells to undergo apoptosis, and is thus a hallmark of apoptosis. Rapamycin-insensitive companion of mTOR (rictor) functions primarily as a core and essential component of mTOR complex 2 (mTORC2) to critically regulate cellular homeostasis. However, its role in the regulation of apoptosis is largely unknown. In the current study, we found that rictor was cleaved to generate two small fragments at ~ 50 kD and ~ 130 kD in cells undergoing apoptosis upon treatment with different stimuli such as the death ligand, TRAIL, and the small molecule, AZD9291. This cleavage was abolished when caspases were inhibited and could be reproduced when directly incubating rictor protein and caspase-3 in vitro. Furthermore, the cleavage site of caspase-3 on rictor was mapped at D1244 (VGVD). These findings together robustly demonstrate that rictor is a substrate of caspase-3 and undergoes cleavage during apoptosis. These results add new information for understanding the biology of rictor in the regulation of cell survival and growth.
Collapse
Affiliation(s)
- Liqun Zhao
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, 1365-C Clifton Road NE, Atlanta, GA, 30322, USA
| | - Lei Zhu
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, 1365-C Clifton Road NE, Atlanta, GA, 30322, USA
- Research Institute for Pharmaceutical Screening & Evaluation, Wannan Medical College School of Pharmacy and Anhui Province Key Laboratory of Active Biological Macromolecules, Wuhu, Anhui, China
| | - You-Take Oh
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, 1365-C Clifton Road NE, Atlanta, GA, 30322, USA
| | - Guoqing Qian
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, 1365-C Clifton Road NE, Atlanta, GA, 30322, USA
| | - Zhen Chen
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, 1365-C Clifton Road NE, Atlanta, GA, 30322, USA
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, 1365-C Clifton Road NE, Atlanta, GA, 30322, USA.
| |
Collapse
|
7
|
An Immunohistochemical Study of the PTEN/AKT Pathway Involvement in Canine and Feline Mammary Tumors. Animals (Basel) 2021; 11:ani11020365. [PMID: 33535663 PMCID: PMC7912927 DOI: 10.3390/ani11020365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 01/09/2023] Open
Abstract
Simple Summary The PTEN/AKT pathway is involved in several human and animal tumors’ pathogenesis. This study investigates the PTEN/AKT pathway’s biological and prognostic values in canine and feline mammary tumors. PTEN, phospho-AKT (p-AKT) and Rictor expression was determined by immunohistochemistry in canine mammary adenomas and carcinomas and feline mammary carcinomas. In mammary tumors of both species p-Akt was inversely correlated with PTEN expression and positively with Rictor expression; p-Akt and Rictor expression correlated with poorer prognosis. This data could provide a rationale for further studies of this pathway in veterinary oncology due to prognostic and therapeutic implications. Abstract Phosphatase and tensin homolog deleted on chromosome10 (PTEN), phospho-v-Akt murine thymoma viral oncogene homolog (AKT), and the Rapamycin-Insensitive Companion of mTOR (Rictor) expression was investigated by immunohistochemistry in 10 canine mammary adenomas (CMAs), 40 canine mammary carcinomas (CMCs), and 30 feline mammary carcinomas (FMCs). All the CMAs, 25 of 40 CMCs (63%) and 7 of 30 FMCs (23%), were PTEN-positive. In dogs, no CMAs and 15 of 25 CMCs (37%) expressed phospho-AKT (p-AKT), while 24 of 30 FMCs (82%) were p-AKT-positive. One of 10 CMAs (10%), 24 of 40 CMCs (60%) and 20 of 30 FMCs (67%) were Rictor-positive. In the dog, PTEN expression correlated with less aggressive tumors, absence of lymphatic invasion, and longer survival. P-AKT expression correlated with more aggressive subtype, lymphatic invasion, and poorer survival and Rictor expression with lymphatic invasion. In cats, PTEN correlated with less aggressive carcinomas, absence of lymphatic invasion, and better survival. P-AKT and Rictor expression correlated with poorer survival. PTEN expression was inversely correlated with p-AKT and Rictor in both species, while p-AKT positively correlated with Rictor expression. A strong PTEN/AKT pathway involvement in behavior worsening of CMT and FMTs is demonstrated, providing a rationale for further studies of this pathway in veterinary oncology.
Collapse
|
8
|
Hou B, Liu S, Li E, Jiang X. Different Role of Raptor and Rictor in Regulating Rasfonin-Induced Autophagy and Apoptosis in Renal Carcinoma Cells. Chem Biodivers 2020; 17:e2000743. [PMID: 33155352 DOI: 10.1002/cbdv.202000743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/05/2020] [Indexed: 01/18/2023]
Abstract
Both Raptor and Rictor are the key components in the complexes of mammalian target of rapamycin (mTOR), which play a vital role in mediating autophagy. Unlike mTOR, the regulatory role of either Raptor or Rictor in the regulation of autophagic process is relatively less explored. In present study, we found that rasfonin, which isolated from Talaromyces sp. 3656-A1 and was a fungal natural product, activated both caspase-dependent apoptosis and autophagy in ACHN, a renal carcinoma cell line. Knockdown of Raptor decreased both rasfonin-induced autophagic flux and PARP-1 cleavage, and in contrast, Rictor silencing increased apoptosis concomitantly enhancing rasfonin-induced autophagy. Unexpectedly, API-2, which was widely used as an inhibitor of Akt, promoted rasfonin-dependent autophagy in Raptor-depleted but not Rictor-deprived cells. Collectively, these results demonstrated that Raptor and Rictor could play a distinctly regulatory role in rasfonin-enhanced autophagy and apoptosis.
Collapse
Affiliation(s)
- Bolin Hou
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, P. R. China.,University of Chinese Academy of Sciences, Beijing, 100039, P. R. China
| | - Shuchun Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Erwei Li
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Xuejun Jiang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| |
Collapse
|
9
|
Kiptiyah K, Widodo W, Ciptadi G, Aulanni'Am A, Widodo MA, Sumitro SB. 10-gingerol induces oxidative stress through HTR1A in cumulus cells: in-vitro and in-silico studies. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2020; 17:/j/jcim.ahead-of-print/jcim-2019-0042/jcim-2019-0042.xml. [PMID: 32284444 DOI: 10.1515/jcim-2019-0042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 10/24/2019] [Indexed: 01/04/2023]
Abstract
Background We investigated whether 10-gingerol is able to induce oxidative stress in cumulus cells. Methods For the in-vitro research, we used a cumulus cell culture in M199, containing 10-gingerol in various concentrations (0, 12, 16, and 20 µM), and detected oxidative stress through superoxide dismutase (SOD) activity and malondialdehyde (MDA) concentrations, with incubation periods of 24, 48, 72, and 96 h. The obtained results were confirmed by in-silico studies. Results The in-vitro data revealed that SOD activity and MDA concentration increased with increasing incubation periods: SOD activity at 0 µM (1.39 ± 0.24i), 12 µM (16.42 ± 0.35ab), 16 µM (17.28 ± 0.55ab), 20 µM (17.81 ± 0.12a), with a contribution of 71.1%. MDA concentration at 0 µM (17.82 ± 1.39 l), 12 µM (72.99 ± 0.31c), 16 µM (79.77 ± 4.19b), 20 µM (85.07 ± 2.57a), with a contribution of 73.1%. Based on this, the in-silico data uncovered that 10-gingerol induces oxidative stress in cumulus cells by inhibiting HTR1A functions and inactivating GSK3B and AKT-1. Conclusions 10-gingerol induces oxidative stress in cumulus cells through enhancing SOD activity and MDA concentration by inhibiting HTR1A functions and inactivating GSK3B and AKT-1.
Collapse
Affiliation(s)
- Kiptiyah Kiptiyah
- Department of Biology, Maulana Malik Ibrahim Islamic State University of Malang, Malang 65144, Indonesia
| | - Widodo Widodo
- Department of Biology, Brawijaya University of Malang, Malang, Indonesia
| | - Gatot Ciptadi
- Husbandry Faculty, Brawijaya University of Malang, Malang, Indonesia
| | | | - Mohammad A Widodo
- Biomedical Study Programme, Brawijaya University of Malang, Malang, Indonesia
| | - Sutiman B Sumitro
- Department of Biology, Brawijaya University of Malang, Malang, Indonesia
| |
Collapse
|
10
|
Lambert IH, Nielsen D, Stürup S. Impact of the histone deacetylase inhibitor trichostatin A on active uptake, volume-sensitive release of taurine, and cell fate in human ovarian cancer cells. Am J Physiol Cell Physiol 2020; 318:C581-C597. [PMID: 31913698 DOI: 10.1152/ajpcell.00460.2019] [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] [Indexed: 12/29/2022]
Abstract
The histone deacetylase inhibitor trichostatin A (TSA) reduces cell viability in cisplatin-sensitive (A2780WT) and cisplatin-resistant (A2780RES) human ovarian cancer cells due to progression of apoptosis (increased caspase-9 activity), autophagy (increased LC3-II expression), and cell cycle arrest (increased p21 expression). The TSA-mediated effect on p21 and caspase-9 is mainly p53 independent. Cisplatin increases DNA-damage (histone H2AX phosphorylation) in A2780WT cells, whereas cisplatin, due to reduced uptake [inductively coupled-plasma-mass spectrometry (Pt) analysis], has no DNA-damaging effect in A2780RES cells. TSA has no effect on cisplatin accumulation or cisplatin-induced DNA-damage in A2780WT/A2780RES cells. Tracer technique indicates that TSA inhibits the volume-sensitive organic anion channel (VSOAC) in A2780WT/A2780RES cells and that the activity is restored by exogenous H2O2. As TSA reduces NOX4 mRNA accumulation and concomitantly increases catalase mRNA/protein accumulation, we suggest that TSA increases the antioxidative defense in A2780 cells. Inhibition of the kinase mTOR (rapamycin, palomid, siRNA), which is normally associated with cell growth, reduces VSOAC activity synergistically to TSA. However, as TSA increases mTOR activity (phosphorylation of 4EBP1, S6 kinase, S6, ULK1, SGK1), the effect of TSA on VSOAC activity does not reflect the shift in mTOR signaling. Upregulation of the protein expression and activity of the taurine transporter (TauT) is a phenotypic characteristic of A2780RES cells. However, TSA reduces TauT protein expression in A2780RES cells and activity to values seen in A2780WT cells. It is suggested that therapeutic benefits of TSA in A2780 do not imply facilitation of cisplatin uptake but more likely a synergistic activation of apoptosis/autophagy and reduced TauT activity.
Collapse
Affiliation(s)
- Ian Henry Lambert
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Dorthe Nielsen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Stefan Stürup
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Gkountakos A, Pilotto S, Mafficini A, Vicentini C, Simbolo M, Milella M, Tortora G, Scarpa A, Bria E, Corbo V. Unmasking the impact of Rictor in cancer: novel insights of mTORC2 complex. Carcinogenesis 2019; 39:971-980. [PMID: 29955840 DOI: 10.1093/carcin/bgy086] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/16/2018] [Accepted: 06/26/2018] [Indexed: 12/15/2022] Open
Abstract
Genomic alterations affecting components of the mechanistic target of rapamycin (mTOR) pathway are found rather frequently in cancers, suggesting that aberrant pathway activity is implicated in oncogenesis of different tumor types. mTOR functions as the core catalytic kinase of two distinct complexes, mTOR complex 1 (mTORC1) and 2 (mTORC2), which control numerous vital cellular processes. There is growing evidence indicating that Rictor, an essential subunit of the mTORC2 complex, is inappropriately overexpressed across numerous cancer types and this is associated with poor survival. To date, the candidate mechanisms responsible for aberrant Rictor expression described in cancer are two: (i) gene amplification and (ii) epigenetic regulation, mainly by microRNAs. Moreover, different mTOR-independent Rictor-containing complexes with oncogenic role have been documented, revealing alternative routes of Rictor-driven tumorigenesis, but simultaneously, paving the way for identifying novel biomarkers and therapeutic targets. Here, we review the main preclinical and clinical data regarding the role of Rictor in carcinogenesis and metastatic behavior as well as the potentiality of its alteration as a target.
Collapse
Affiliation(s)
- Anastasios Gkountakos
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Sara Pilotto
- Medical Oncology Section, Department of Medicine, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Andrea Mafficini
- ARC-NET Applied Research on Cancer Center, University of Verona, Verona, Italy
| | - Caterina Vicentini
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy.,ARC-NET Applied Research on Cancer Center, University of Verona, Verona, Italy
| | - Michele Simbolo
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Michele Milella
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giampaolo Tortora
- Medical Oncology Section, Department of Medicine, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Aldo Scarpa
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy.,ARC-NET Applied Research on Cancer Center, University of Verona, Verona, Italy
| | - Emilio Bria
- Medical Oncology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Vincenzo Corbo
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy.,ARC-NET Applied Research on Cancer Center, University of Verona, Verona, Italy
| |
Collapse
|
12
|
Ruicci KM, Plantinga P, Pinto N, Khan MI, Stecho W, Dhaliwal SS, Yoo J, Fung K, MacNeil D, Mymryk JS, Barrett JW, Howlett CJ, Nichols AC. Disruption of the RICTOR/mTORC2 complex enhances the response of head and neck squamous cell carcinoma cells to PI3K inhibition. Mol Oncol 2019; 13:2160-2177. [PMID: 31393061 PMCID: PMC6763779 DOI: 10.1002/1878-0261.12558] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 07/28/2019] [Accepted: 08/07/2019] [Indexed: 12/19/2022] Open
Abstract
Phosphoinositide 3-kinase (PI3K) is aberrantly activated in head and neck squamous cell carcinomas (HNSCC) and plays a pivotal role in tumorigenesis by driving Akt signaling, leading to cell survival and proliferation. Phosphorylation of Akt Thr308 by PI3K-PDK1 and Akt Ser473 by mammalian target of rapamycin complex 2 (mTORC2) activates Akt. Targeted inhibition of PI3K is a major area of preclinical and clinical investigation as it reduces Akt Thr308 phosphorylation, suppressing downstream mTORC1 activity. However, inhibition of mTORC1 releases feedback inhibition of mTORC2, resulting in a resurgence of Akt activation mediated by mTORC2. While the role of PI3K-activated Akt signaling is well established in HNSCC, the significance of mTORC2-driven Akt signaling has not been thoroughly examined. Here we explore the expression and function of mTORC2 and its obligate subunit RICTOR in HNSCC primary tumors and cell lines. We find RICTOR to be overexpressed in a subset of HNSCC tumors, including those with PIK3CA or EGFR gene amplifications. Whereas overexpression of RICTOR reduced susceptibility of HNSCC tumor cells to PI3K inhibition, genetic ablation of RICTOR using CRISPR/Cas9 sensitized cells to PI3K inhibition, as well as to EGFR inhibition and cisplatin treatment. Further, mTORC2 disruption led to reduced viability and colony forming abilities of HNSCC cells relative to their parental lines and induced loss of both activating Akt phosphorylation modifications (Thr308 and Ser473). Taken together, our findings establish RICTOR/mTORC2 as a critical oncogenic complex in HNSCC and rationalize the development of an mTORC2-specific inhibitor for use in HNSCC, either combined with agents already under investigation, or as an independent therapy.
Collapse
Affiliation(s)
- Kara M. Ruicci
- Department of Otolaryngology – Head and Neck Surgery, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
- Department of Pathology & Laboratory Medicine, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
| | - Paul Plantinga
- Department of Pathology & Laboratory Medicine, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
| | - Nicole Pinto
- Department of Otolaryngology – Head and Neck Surgery, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
| | - Mohammed I. Khan
- Department of Otolaryngology – Head and Neck Surgery, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
| | - William Stecho
- Department of Pathology & Laboratory Medicine, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
| | - Sandeep S. Dhaliwal
- Department of Otolaryngology – Head and Neck Surgery, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
- Department of Oncology, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
| | - John Yoo
- Department of Otolaryngology – Head and Neck Surgery, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
- Department of Oncology, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
| | - Kevin Fung
- Department of Otolaryngology – Head and Neck Surgery, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
- Department of Oncology, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
| | - Danielle MacNeil
- Department of Otolaryngology – Head and Neck Surgery, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
- Department of Oncology, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
| | - Joe S. Mymryk
- Department of Otolaryngology – Head and Neck Surgery, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
- Department of Oncology, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
- Department of Microbiology and Immunology, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
| | - John W. Barrett
- Department of Otolaryngology – Head and Neck Surgery, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
| | - Christopher J. Howlett
- Department of Pathology & Laboratory Medicine, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
| | - Anthony C. Nichols
- Department of Otolaryngology – Head and Neck Surgery, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
- Department of Pathology & Laboratory Medicine, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
- Department of Oncology, Schulich School of Medicine & DentistryWestern UniversityLondonCanada
| |
Collapse
|
13
|
Wang G, Yan Y, Xu N, Hui Y, Yin D. Upregulation of microRNA-424 relieved diabetic nephropathy by targeting Rictor through mTOR Complex2/Protein Kinase B signaling. J Cell Physiol 2019; 234:11646-11653. [PMID: 30637733 DOI: 10.1002/jcp.27822] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/06/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To investigate the role of miR-424 in diabetic nephropathy (DN) and its relationship with Rictor in mammalian target of rapamycin (mTOR) C2/Akt signaling. METHODS The western blot analysis and real-time polymerase chain reaction were used to determine the differential expression of Rictor, mTOR, and miR-424 in DN rats. The upregulation of miR-424 was achieved by caudal vein injection of miR-424 mimics. The renal lesion was evaluated by hematoxylin-eosin staining (H&E) and periodic acid schiff staining. The dual-luciferase reporter assay was conducted to determine the binding target of miR-424. The effect of miR-424 upregulation on apoptosis was detected by the terminal deoxynucleotidyl transferase-mediated 2-Deoxyuridine-5-Triphosphate (dUTP) nick-end labeling assay and western blot analysis. RESULTS A significantly lower expression of miR-424 and a significantly higher expression of Rictor and mTOR were found in renal tissues of DN rats. The upregulation of miR-424 improved renal lesion and DN symptoms of blood glucose level, urine protein level, body weight, creatinine level, blood urea nitrogen, and KW/BW ratio. The upregulation of miR-424 could significantly reduce apoptosis rates of tissue cells by decreasing the expression levels of caspase-3 and Bax as well as increasing the level of Bcl-2. Furthermore, Rictor was the direct target for miR-424, and upregulation of miR-424 inhibited Rictor through Akt signaling in renal tissue of DN rats and high-glucose-treated human glomerular mesangial cells. CONCLUSION miR-424 contributes to alleviating the symptoms in DN rat models by targeting Rictor through mTORC2/Akt signaling.
Collapse
Affiliation(s)
- Guofeng Wang
- Department of endocrinology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First People's Hospital of Lianyungang, The Affiliated Hospital of Kangda College of Nanjing Medical University Lianyungang Clinical College of Nanjing Medical University, Lianyungang, China
| | - Yongxin Yan
- Department of endocrinology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First People's Hospital of Lianyungang, The Affiliated Hospital of Kangda College of Nanjing Medical University Lianyungang Clinical College of Nanjing Medical University, Lianyungang, China
| | - Ning Xu
- Department of endocrinology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First People's Hospital of Lianyungang, The Affiliated Hospital of Kangda College of Nanjing Medical University Lianyungang Clinical College of Nanjing Medical University, Lianyungang, China
| | - Yuan Hui
- Department of endocrinology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First People's Hospital of Lianyungang, The Affiliated Hospital of Kangda College of Nanjing Medical University Lianyungang Clinical College of Nanjing Medical University, Lianyungang, China
| | - Dong Yin
- Department of endocrinology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First People's Hospital of Lianyungang, The Affiliated Hospital of Kangda College of Nanjing Medical University Lianyungang Clinical College of Nanjing Medical University, Lianyungang, China
| |
Collapse
|
14
|
Harachi M, Masui K, Okamura Y, Tsukui R, Mischel PS, Shibata N. mTOR Complexes as a Nutrient Sensor for Driving Cancer Progression. Int J Mol Sci 2018; 19:ijms19103267. [PMID: 30347859 PMCID: PMC6214109 DOI: 10.3390/ijms19103267] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 10/14/2018] [Accepted: 10/14/2018] [Indexed: 02/06/2023] Open
Abstract
Recent advancement in the field of molecular cancer research has clearly revealed that abnormality of oncogenes or tumor suppressor genes causes tumor progression thorough the promotion of intracellular metabolism. Metabolic reprogramming is one of the strategies for cancer cells to ensure their survival by enabling cancer cells to obtain the macromolecular precursors and energy needed for the rapid growth. However, an orchestration of appropriate metabolic reactions for the cancer cell survival requires the precise mechanism to sense and harness the nutrient in the microenvironment. Mammalian/mechanistic target of rapamycin (mTOR) complexes are known downstream effectors of many cancer-causing mutations, which are thought to regulate cancer cell survival and growth. Recent studies demonstrate the intriguing role of mTOR to achieve the feat through metabolic reprogramming in cancer. Importantly, not only mTORC1, a well-known regulator of metabolism both in normal and cancer cell, but mTORC2, an essential partner of mTORC1 downstream of growth factor receptor signaling, controls cooperatively specific metabolism, which nominates them as an essential regulator of cancer metabolism as well as a promising candidate to garner and convey the nutrient information from the surrounding environment. In this article, we depict the recent findings on the role of mTOR complexes in cancer as a master regulator of cancer metabolism and a potential sensor of nutrients, especially focusing on glucose and amino acid sensing in cancer. Novel and detailed molecular mechanisms that amino acids activate mTOR complexes signaling have been identified. We would also like to mention the intricate crosstalk between glucose and amino acid metabolism that ensures the survival of cancer cells, but at the same time it could be exploitable for the novel intervention to target the metabolic vulnerabilities of cancer cells.
Collapse
Affiliation(s)
- Mio Harachi
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Kenta Masui
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Yukinori Okamura
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Ryota Tsukui
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| | - Paul S Mischel
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA.
| | - Noriyuki Shibata
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| |
Collapse
|
15
|
The Effects of Different mTOR Inhibitors in EGFR Inhibitor Resistant Colon Carcinoma Cells. Pathol Oncol Res 2018; 25:1379-1386. [PMID: 29882195 DOI: 10.1007/s12253-018-0434-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/29/2018] [Indexed: 12/25/2022]
Abstract
Several monoclonal antibodies and inhibitors targeting signalling pathways are being used in personalised medicine. Anti-EGFR antibodies seem to be effective, however, therapy resistance often occurs in colon carcinoma cases. mTOR inhibitors (mTORIs) could have a potential role in the breakthrough of therapy resistance. The mTOR activity related protein expression patterns and the in vitro effects of EGFR inhibitors (EGFRIs), mTORIs and their combinations were studied in different colon carcinoma cell lines (with different genetic backgrounds). Alamar Blue test and flow cytometry were used to analyse the in vitro proliferation and apoptotic effects of cetuximab, gefitinib, cisplatin, rapamycin, PP242 and NVP-BEZ235. The expressions of mTOR activity related proteins (p-70S6K, p-S6, Rictor, p-mTOR, Raptor) were studied by Western blot, immunocytochemistry and Duolink staining. The EGFRI resistance of the studied colon carcinoma cell lines related to their known mutations were confirmed, neither gefitinib nor cetuximab inhibited the proliferation or induced apoptosis in vitro. Individual differences in Rictor and Raptor expressions were detected by Western blot and immunocytochemistry beside elevated mTOR activity of these different colon carcinoma cell lines. These expression patterns correlated to the mTORIs sensitivity differences, moreover, mTORIs could enhance the effects of EGFRIs and other in vitro treatments. Our results suggest that mTORI combinations could be helpful in both EGFRI and platinum-based therapy of colon carcinomas. Moreover, we suggest determining both mTOR complex activity and mutations in Akt/mTOR signalling pathways for selecting the appropriate mTORIs and patients in potential future combination treatments.
Collapse
|
16
|
Simabuco FM, Morale MG, Pavan IC, Morelli AP, Silva FR, Tamura RE. p53 and metabolism: from mechanism to therapeutics. Oncotarget 2018; 9:23780-23823. [PMID: 29805774 PMCID: PMC5955117 DOI: 10.18632/oncotarget.25267] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/06/2018] [Indexed: 11/25/2022] Open
Abstract
The tumor cell changes itself and its microenvironment to adapt to different situations, including action of drugs and other agents targeting tumor control. Therefore, metabolism plays an important role in the activation of survival mechanisms to keep the cell proliferative potential. The Warburg effect directs the cellular metabolism towards an aerobic glycolytic pathway, despite the fact that it generates less adenosine triphosphate than oxidative phosphorylation; because it creates the building blocks necessary for cell proliferation. The transcription factor p53 is the master tumor suppressor; it binds to more than 4,000 sites in the genome and regulates the expression of more than 500 genes. Among these genes are important regulators of metabolism, affecting glucose, lipids and amino acids metabolism, oxidative phosphorylation, reactive oxygen species (ROS) generation and growth factors signaling. Wild-type and mutant p53 may have opposing effects in the expression of these metabolic genes. Therefore, depending on the p53 status of the cell, drugs that target metabolism may have different outcomes and metabolism may modulate drug resistance. Conversely, induction of p53 expression may regulate differently the tumor cell metabolism, inducing senescence, autophagy and apoptosis, which are dependent on the regulation of the PI3K/AKT/mTOR pathway and/or ROS induction. The interplay between p53 and metabolism is essential in the decision of cell fate and for cancer therapeutics.
Collapse
Affiliation(s)
- Fernando M. Simabuco
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Mirian G. Morale
- Center for Translational Investigation in Oncology/LIM24, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
- Department of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Isadora C.B. Pavan
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Ana P. Morelli
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Fernando R. Silva
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rodrigo E. Tamura
- Center for Translational Investigation in Oncology/LIM24, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
- Department of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
17
|
Abstract
The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR)-dependent pathway is one of the most integral pathways linked to cell metabolism, proliferation, differentiation, and survival. This pathway is dysregulated in a variety of diseases, including neoplasia, immune-mediated diseases, and fibroproliferative diseases such as pulmonary fibrosis. The mTOR kinase is frequently referred to as the master regulator of this pathway. Alterations in mTOR signaling are closely associated with dysregulation of autophagy, inflammation, and cell growth and survival, leading to the development of lung fibrosis. Inhibitors of mTOR have been widely studied in cancer therapy, as they may sensitize cancer cells to radiation therapy. Studies also suggest that mTOR inhibitors are promising modulators of fibroproliferative diseases such as idiopathic pulmonary fibrosis (IPF) and radiation-induced pulmonary fibrosis (RIPF). Therefore, mTOR represents an attractive and unique therapeutic target in pulmonary fibrosis. In this review, we discuss the pathological role of mTOR kinase in pulmonary fibrosis and examine how mTOR inhibitors may mitigate fibrotic progression.
Collapse
|
18
|
Chen QY, Jiao DM, Wang J, Hu H, Tang X, Chen J, Mou H, Lu W. miR-206 regulates cisplatin resistance and EMT in human lung adenocarcinoma cells partly by targeting MET. Oncotarget 2017; 7:24510-26. [PMID: 27014910 PMCID: PMC5029718 DOI: 10.18632/oncotarget.8229] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 03/04/2016] [Indexed: 01/15/2023] Open
Abstract
MicroRNAs (miRNAs) play a critical role in drug resistance and epithelial-mesenchymal transition (EMT). The aims of this study were to explore the potential role of miR-206 in governing cisplatin resistance and EMT in lung cancer cells. We found that both lung adenocarcinoma A549 cisplatin-resistant cells (A549/DDP) and H1299 cisplatin-resistant cells (H1299/DDP) acquired mesenchymal features and were along with low expression of miR-206 and high migration and invasion abilities. Ectopic expression of miR-206 mimics inhibited cisplatin resistance, reversed the EMT phenotype, decreased the migration and invasion in these DDP-resistant cells. In contrast, miR-206 inhibitors increased cisplatin resistance, EMT, cell migration and invasion in non-DDP-resistant cells. Furthermore, we found that MET is the direct target of miR-206 in lung cancer cells. Knockdown of MET exhibited an EMT and DDP resistant inhibitory effect on DDP-resistant cells. Conversely, overexpression of MET in non-DDP- resistant cells produced a promoting effect on cell EMT and DDP resistance. In lung adenocarcinoma tissues, we demonstrated that low expression of miR-206 were also correlated with increased cisplatin resistance and MET expression. In addition, we revealed that miR-206 overexpression reduced cisplatin resistance and EMT in DDP-resistant cells, partly due to inactivation of MET/PI3K/AKT/mTOR signaling pathway, and subsequent downregulation of MDR1, ZEB1 and Snail expression. Finally, we found that miR-206 could also sensitize A549/DDP cells to cisplatin in mice model. Taken together, our study implied that activation of miR-206 or inactivation of its target gene pathway could serve as a novel approach to reverse cisplatin resistance in lung adenocarcinomas cells.
Collapse
Affiliation(s)
- Qing-Yong Chen
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| | - De-Min Jiao
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| | - Jian Wang
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| | - Huizhen Hu
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| | - Xiali Tang
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| | - Jun Chen
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| | - Hao Mou
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| | - Wei Lu
- Department of Oncology, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| |
Collapse
|
19
|
Wang L, Qi J, Yu J, Chen H, Zou Z, Lin X, Guo L. Overexpression of Rictor protein in colorectal cancer is correlated with tumor progression and prognosis. Oncol Lett 2017; 14:6198-6202. [PMID: 29113267 DOI: 10.3892/ol.2017.6936] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 07/03/2017] [Indexed: 12/20/2022] Open
Abstract
In order to understand the clinical significance of rapamycin-insensitive companion of mammalian target of rapamycin (Rictor) in colorectal cancer (CRC), 62 CRC tissue samples excised during operations were evaluated by immunohistochemistry. Analysis of the association between the expression level of Rictor protein and clinicopathological parameters demonstrated that the expression level of Rictor in CRC tissues was significantly higher than that in paracarcinoma tissues (P<0.0001). In cellular experiments, this result was further confirmed by comparing differences in Rictor expression between the CRC cell lines HCT116, SW480 and LoVo, and the human normal liver cell line HL-7702. It was also noticed that the expression of Rictor was associated with Dukes stage, lymphatic metastasis and prognosis, as determined by χ2 test, Kaplan-Meier analysis and log-rank test. These results suggest that Rictor may be a novel target for the treatment and prognostic assessment of CRC patients in the future.
Collapse
Affiliation(s)
- Lifeng Wang
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Jia Qi
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Jinlong Yu
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Haijin Chen
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Zhaowei Zou
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Xiaohua Lin
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Linlang Guo
- Department of Pathology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| |
Collapse
|
20
|
10-Gingerol as an inducer of apoptosis through HTR1A in cumulus cells: In-vitro and in-silico studies. J Taibah Univ Med Sci 2017; 12:397-406. [PMID: 31435270 PMCID: PMC6695051 DOI: 10.1016/j.jtumed.2017.05.012] [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: 12/19/2016] [Revised: 05/17/2017] [Accepted: 05/21/2017] [Indexed: 12/20/2022] Open
Abstract
Objectives Cumulus cells play a crucial role as essential mediators in the maturation of ova. Ginger contains 10-gingerol, which induces apoptosis in colon cancer cells. Based on this hypothesis, this study aimed to determine whether 10-gingerol is able to induce apoptosis in normal cells, namely, cumulus cells. Methods This study used an in vitro analysis by culturing Cumulus cells in M199 containing 10-gingerol in various concentrations (12, 16, and 20 μM) and later detected early apoptotic activity using an Annexin V-FITC detection kit. Result The in vitro data revealed that the number of apoptosis cells increased along with the period of incubation as follows: 12 μM (63.71% ± 2.192%); 16 μM (74.51% ± 4.596%); and 20 μM (78.795% ± 1.435%). The substance 10-gingerol induces apoptosis in cumulus cells by inhibiting HTR1A functions and inactivating GSK3B and AKT-1. Conclusions These findings indicate that further examination is warranted for 10-gingerol as a contraception agent.
Collapse
Key Words
- 10-Gingerol
- ARG, arginine
- Apoptosis
- Cumulus cells
- FOXO, forkhead box
- GLU, glutamine
- GLY, glycine
- GSK3B, glycogen synthase kinase-3β
- HTR1A
- HTR1A, 5-hydroxytryptamine receptor 1 A
- ILE, ileusine
- ILK, integrin-linked kinase
- In silico
- In vitro
- LYS, lysine
- MDM2, murine double minute clone 2
- MET, methionine
- NO, nitric oxide
- NOS3, nitric oxide synthase 3
- PTEN, phosphatase and tensin homologue delete on chromosome ten
- RICTOR, rapamycin-insensitive companion of mTOR
- TYR, tyrosine
- eNOS, endothelial nitric oxide synthase
- mTOR, mammalian target of rapamycin
- mTORC1, mTOR complex 1
- mTORC2, mTOR complex 2
Collapse
|
21
|
mTORC1 and -2 Coordinate Transcriptional and Translational Reprogramming in Resistance to DNA Damage and Replicative Stress in Breast Cancer Cells. Mol Cell Biol 2017; 37:MCB.00577-16. [PMID: 27956700 DOI: 10.1128/mcb.00577-16] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 12/02/2016] [Indexed: 01/04/2023] Open
Abstract
mTOR coordinates growth signals with metabolic pathways and protein synthesis and is hyperactivated in many human cancers. mTOR exists in two complexes: mTORC1, which stimulates protein, lipid, and ribosome biosynthesis, and mTORC2, which regulates cytoskeleton functions. While mTOR is known to be involved in the DNA damage response, little is actually known regarding the functions of mTORC1 compared to mTORC2 in this regard or the respective impacts on transcriptional versus translational regulation. We show that mTORC1 and mTORC2 are both required to enact DNA damage repair and cell survival, resulting in increased cancer cell survival during DNA damage. Together mTORC1 and -2 enact coordinated transcription and translation of protective cell cycle and DNA replication, recombination, and repair genes. This coordinated transcriptional-translational response to DNA damage was not impaired by rapalog inhibition of mTORC1 or independent inhibition of mTORC1 or mTORC2 but was blocked by inhibition of mTORC1/2. Only mTORC1/2 inhibition reversed cancer cell resistance to DNA damage and replicative stress and increased tumor cell killing and tumor control by DNA damage therapies in animal models. When combined with DNA damage, inhibition of mTORC1/2 blocked transcriptional induction more strongly than translation of DNA replication, survival, and DNA damage response mRNAs.
Collapse
|
22
|
Wang S, Song X, Li X, Zhao X, Chen H, Wang J, Wu J, Gao Z, Qian J, Han B, Bai C, Li Q, Lu D. RICTOR polymorphisms affect efficiency of platinum-based chemotherapy in Chinese non-small-cell lung cancer patients. Pharmacogenomics 2016; 17:1637-1647. [PMID: 27676404 DOI: 10.2217/pgs-2016-0070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
AIM We investigated the association between RICTOR polymorphisms and clinical outcomes of platinum-based chemotherapy for Chinese non-small-cell lung cancer patients. MATERIALS & METHODS Ten tag SNPs were genotyped in 1004 patients to assess their association with clinical benefit, overall survival, progression-free survival, gastrointestinal toxicity, neutropenia, anemia and thrombocytopenia. RESULTS rs6878291 was significantly associated with clinical benefit (odds ratio: 2.037; p = 0.001) and reduced progression-free survival (hazard ratio: 1.461; p = 0.001). Stratified analysis showed that their most significant interaction was in nonsmokers. No association was observed between SNPs and other clinical outcomes. CONCLUSION The study showed evidences for RICTOR polymorphisms' role in platinum-based chemotherapy efficiency, which could provide new insight to lung cancer management.
Collapse
Affiliation(s)
- Shiming Wang
- State Key Laboratory of Genetic Engineering & MOE Key Laboratory of Contemporary Anthropology, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiao Song
- State Key Laboratory of Genetic Engineering & MOE Key Laboratory of Contemporary Anthropology, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China.,Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Xiaoying Li
- State Key Laboratory of Genetic Engineering & MOE Key Laboratory of Contemporary Anthropology, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Crime SceneEvidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai, China
| | - Xueying Zhao
- State Key Laboratory of Genetic Engineering & MOE Key Laboratory of Contemporary Anthropology, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Hongyan Chen
- State Key Laboratory of Genetic Engineering & MOE Key Laboratory of Contemporary Anthropology, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering & MOE Key Laboratory of Contemporary Anthropology, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Junjie Wu
- State Key Laboratory of Genetic Engineering & MOE Key Laboratory of Contemporary Anthropology, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China.,Department of Pneumology, Changhai Hospital of Shanghai, Second Military Medical University, Shanghai, China
| | - Zhiqiang Gao
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ji Qian
- State Key Laboratory of Genetic Engineering & MOE Key Laboratory of Contemporary Anthropology, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Baohui Han
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chunxue Bai
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiang Li
- Department of Pneumology, Changhai Hospital of Shanghai, Second Military Medical University, Shanghai, China
| | - Daru Lu
- State Key Laboratory of Genetic Engineering & MOE Key Laboratory of Contemporary Anthropology, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Doxorubicin impairs cardiomyocyte viability by suppressing transcription factor EB expression and disrupting autophagy. Biochem J 2016; 473:3769-3789. [PMID: 27487838 DOI: 10.1042/bcj20160385] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/03/2016] [Indexed: 12/11/2022]
Abstract
Doxorubicin (DOX) is an effective anti-cancer agent. However, DOX treatment increases patient susceptibility to dilated cardiomyopathy. DOX predisposes cardiomyocytes to insult by suppressing mitochondrial energy metabolism, altering calcium flux, and disrupting proteolysis and proteostasis. Prior studies have assessed the role of macroautophagy in DOX cardiotoxicity; however, limited studies have examined whether DOX mediates cardiac injury through dysfunctions in inter- and/or intra-lysosomal signaling events. Lysosomal signaling and function is governed by transcription factor EB (TFEB). In the present study, we hypothesized that DOX caused myocyte injury by impairing lysosomal function and signaling through negative regulation of TFEB. Indeed, we found that DOX repressed cellular TFEB expression, which was associated with impaired cathepsin proteolytic activity across in vivo, ex vivo, and in vitro models of DOX cardiotoxicity. Furthermore, we observed that loss of TFEB was associated with reduction in macroautophagy protein expression, inhibition of autophagic flux, impairments in lysosomal cathepsin B activity, and activation of cell death. Restoration and/or activation of TFEB in DOX-treated cardiomyocytes prevented DOX-induced suppression of cathepsin B activity, reduced DOX-mediated reactive oxygen species (ROS) overproduction, attenuated activation of caspase-3, and improved cellular viability. Collectively, loss of TFEB inhibits lysosomal autophagy, rendering cardiomyocytes susceptible to DOX-induced proteotoxicity and injury. Our data reveal a novel mechanism wherein DOX primes cardiomyocytes for cell death by depleting cellular TFEB.
Collapse
|
24
|
Shull AY, Noonepalle SK, Awan FT, Liu J, Pei L, Bollag RJ, Salman H, Ding Z, Shi H. RPPA-based protein profiling reveals eIF4G overexpression and 4E-BP1 serine 65 phosphorylation as molecular events that correspond with a pro-survival phenotype in chronic lymphocytic leukemia. Oncotarget 2016; 6:14632-45. [PMID: 25999352 PMCID: PMC4546493 DOI: 10.18632/oncotarget.4104] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 04/08/2015] [Indexed: 12/22/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL), the most common adult leukemia, remains incurable despite advancements in treatment regimens over the past decade. Several expression profile studies have been pursued to better understand CLL pathogenesis. However, these large-scale studies only provide information at the transcriptional level. To better comprehend the differential protein changes that take place in CLL, we performed a reverse-phase protein array (RPPA) analysis using 167 different antibodies on B-cell lysates from 18 CLL patients and 6 normal donors. From our analysis, we discovered an enrichment of protein alterations involved with mRNA translation, specifically upregulation of the translation initiator eIF4G and phosphorylation of the cap-dependent translation inhibitor 4E-BP1 at serine 65. Interestingly, 4E-BP1 phosphorylation occurred independently of AKT phosphorylation, suggesting a disconnect between PI3K/AKT pathway activation and 4E-BP1 phosphorylation. Based on these results, we treated primary CLL samples with NVP-BEZ235, a PI3K/mTOR dual inhibitor, and compared its apoptotic-inducing potential against the BTK inhibitor Ibrutinib and the PI3Kδ inhibitor Idelalisib. We demonstrated that treatment with NVP-BEZ235 caused greater apoptosis, greater apoptotic cleavage of eIF4G, and greater dephosphorylation of 4E-BP1 in primary CLL cells. Taken together, these results highlight the potential dependence of eIF4G overexpression and 4E-BP1 phosphorylation in CLL survival.
Collapse
Affiliation(s)
- Austin Y Shull
- Department of Biochemistry & Molecular Biology, Georgia Regents University, Augusta, Georgia, USA.,GRU Cancer Center, Georgia Regents University, Augusta, Georgia, USA
| | - Satish K Noonepalle
- Department of Biochemistry & Molecular Biology, Georgia Regents University, Augusta, Georgia, USA.,GRU Cancer Center, Georgia Regents University, Augusta, Georgia, USA
| | - Farrukh T Awan
- The Ohio State Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Jimei Liu
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, USA
| | - Lirong Pei
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, USA
| | - Roni J Bollag
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, USA.,Department of Pathology, Georgia Regents University, Augusta, Georgia, USA
| | - Huda Salman
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, USA.,Deparment of Medicine, Georgia Regents University, Augusta, Georgia, USA
| | - Zhiyong Ding
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Huidong Shi
- Department of Biochemistry & Molecular Biology, Georgia Regents University, Augusta, Georgia, USA.,GRU Cancer Center, Georgia Regents University, Augusta, Georgia, USA
| |
Collapse
|
25
|
Andorfer P, Heuwieser A, Heinzel A, Lukas A, Mayer B, Perco P. Vascular endothelial growth factor A as predictive marker for mTOR inhibition in relapsing high-grade serous ovarian cancer. BMC SYSTEMS BIOLOGY 2016; 10:33. [PMID: 27090655 PMCID: PMC4836190 DOI: 10.1186/s12918-016-0278-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 04/13/2016] [Indexed: 02/02/2023]
Abstract
Background Development of resistance against first line drug therapy including cisplatin and paclitaxel in high-grade serous ovarian cancer (HGSOC) presents a major challenge. Identifying drug candidates breaking resistance, ideally combined with predictive biomarkers allowing precision use are needed for prolonging progression free survival of ovarian cancer patients. Modeling of molecular processes driving drug resistance in tumor tissue further combined with mechanism of action of drugs provides a strategy for identification of candidate drugs and associated predictive biomarkers. Results Consolidation of transcriptomics profiles and biomedical literature mining results provides 1242 proteins linked with ovarian cancer drug resistance. Integrating this set on a protein interaction network followed by graph segmentation results in a molecular process model representation of drug resistant HGSOC embedding 409 proteins in 24 molecular processes. Utilizing independent transcriptomics profiles with follow-up data on progression free survival allows deriving molecular biomarker-based classifiers for predicting recurrence under first line therapy. Biomarkers of specific relevance are identified in a molecular process encapsulating TGF-beta, mTOR, Jak-STAT and Neurotrophin signaling. Mechanism of action molecular model representations of cisplatin and paclitaxel embed the very same signaling components, and specifically proteins afflicted with the activation status of the mTOR pathway become evident, including VEGFA. Analyzing mechanism of action interference of the mTOR inhibitor sirolimus shows specific impact on the drug resistance signature imposed by cisplatin and paclitaxel, further holding evidence for a synthetic lethal interaction to paclitaxel mechanism of action involving cyclin D1. Conclusions Stratifying drug resistant high grade serous ovarian cancer via VEGFA, and specifically treating with mTOR inhibitors in case of activation of the pathway may allow adding precision for overcoming resistance to first line therapy.
Collapse
Affiliation(s)
- Peter Andorfer
- emergentec biodevelopment GmbH, Gersthofer Strasse 29-31, 1180, Vienna, Austria
| | - Alexander Heuwieser
- emergentec biodevelopment GmbH, Gersthofer Strasse 29-31, 1180, Vienna, Austria
| | - Andreas Heinzel
- emergentec biodevelopment GmbH, Gersthofer Strasse 29-31, 1180, Vienna, Austria
| | - Arno Lukas
- emergentec biodevelopment GmbH, Gersthofer Strasse 29-31, 1180, Vienna, Austria
| | - Bernd Mayer
- emergentec biodevelopment GmbH, Gersthofer Strasse 29-31, 1180, Vienna, Austria
| | - Paul Perco
- emergentec biodevelopment GmbH, Gersthofer Strasse 29-31, 1180, Vienna, Austria.
| |
Collapse
|
26
|
Sørensen BH, Nielsen D, Thorsteinsdottir UA, Hoffmann EK, Lambert IH. Downregulation of LRRC8A protects human ovarian and alveolar carcinoma cells against Cisplatin-induced expression of p53, MDM2, p21Waf1/Cip1, and Caspase-9/-3 activation. Am J Physiol Cell Physiol 2016; 310:C857-73. [PMID: 26984736 PMCID: PMC4935196 DOI: 10.1152/ajpcell.00256.2015] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 03/13/2016] [Indexed: 11/22/2022]
Abstract
The leucine-rich repeat containing 8A (LRRC8A) protein is an essential component of the volume-sensitive organic anion channel (VSOAC), and using pharmacological anion channel inhibitors (NS3728, DIDS) and LRRC8A siRNA we have investigated its role in development of Cisplatin resistance in human ovarian (A2780) and alveolar (A549) carcinoma cells. In Cisplatin-sensitive cells Cisplatin treatment increases p53-protein level as well as downstream signaling, e.g., expression of p21Waf1/Cip1, Bax, Noxa, MDM2, and activation of Caspase-9/-3. In contrast, Cisplatin-resistant cells do not enter apoptosis, i.e., their p53 and downstream signaling are reduced and caspase activity unaltered following Cisplatin exposure. Reduced LRRC8A expression and VSOAC activity are previously shown to correlate with Cisplatin resistance, and here we demonstrate that pharmacological inhibition and transient knockdown of LRRC8A reduce the protein level of p53, MDM2, and p21Waf1/Cip1 as well as Caspase-9/-3 activation in Cisplatin-sensitive cells. Cisplatin resistance is accompanied by reduction in total LRRC8A expression (A2780) or LRRC8A expression in the plasma membrane (A549). Activation of Caspase-3 dependent apoptosis by TNFα-exposure or hyperosmotic cell shrinkage is almost unaffected by pharmacological anion channel inhibition. Our data indicate 1) that expression/activity of LRRC8A is essential for Cisplatin-induced increase in p53 protein level and its downstream signaling, i.e., Caspase-9/-3 activation, expression of p21Waf1/Cip1 and MDM2; and 2) that downregulation of LRRC8A-dependent osmolyte transporters contributes to acquirement of Cisplatin resistance in ovarian and lung carcinoma cells. Activation of LRRC8A-containing channels is upstream to apoptotic volume decrease as hypertonic cell shrinkage induces apoptosis independent of the presence of LRRC8A.
Collapse
Affiliation(s)
- Belinda Halling Sørensen
- Department of Biology, Section of Cell Biology and Physiology, The August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| | - Dorthe Nielsen
- Department of Biology, Section of Cell Biology and Physiology, The August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| | - Unnur Arna Thorsteinsdottir
- Department of Biology, Section of Cell Biology and Physiology, The August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| | - Else Kay Hoffmann
- Department of Biology, Section of Cell Biology and Physiology, The August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| | - Ian Henry Lambert
- Department of Biology, Section of Cell Biology and Physiology, The August Krogh Building, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Carr TD, Feehan RP, Hall MN, Rüegg MA, Shantz LM. Conditional disruption of rictor demonstrates a direct requirement for mTORC2 in skin tumor development and continued growth of established tumors. Carcinogenesis 2015; 36:487-97. [PMID: 25740823 DOI: 10.1093/carcin/bgv012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 02/15/2015] [Indexed: 12/31/2022] Open
Abstract
Activation of signaling dependent on the mammalian target of rapamycin (mTOR) has been demonstrated in a variety of human malignancies, and our previous work suggests that mTOR complex (mTORC) 1 and mTORC2 may play unique roles in skin tumorigenesis. The purpose of these studies was to investigate the function of mTORC2-dependent pathways in skin tumor development and the maintenance of established tumors. Using mice that allow spatial and temporal control of mTORC2 in epidermis by conditional knockout of its essential component Rictor, we studied the effect of mTORC2 loss on both epidermal proliferation and chemical carcinogenesis. The results demonstrate that mTORC2 is dispensable for both normal epidermal proliferation and the hyperproliferative response to treatment with tetradecanoyl phorbol acetate (TPA). In contrast, deletion of epidermal Rictor prior to initiation in DMBA/TPA chemical carcinogenesis was sufficient to dramatically delay tumor development and resulted in reduced tumor number and size compared with control groups. Silencing of Rictor expression in tumor-bearing animals triggered regression of established tumors and increased caspase-3 cleavage without changes in proliferation. In vitro experiments demonstrate an increased sensitivity to caspase-dependent apoptosis in the absence of rictor, which is dependent on mTORC2 signaling. These studies demonstrate that mTORC2 activation is essential for keratinocyte survival, and suggest that inhibition of mTORC2 has value in chemoprevention by eliminating carcinogen-damaged cells during the early stages of tumorigenesis, and in therapy of existing tumors by restricting critical pro-survival pathways.
Collapse
Affiliation(s)
- Theresa D Carr
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA and
| | - Robert P Feehan
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA and
| | - Michael N Hall
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | - Markus A Rüegg
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | - Lisa M Shantz
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA and
| |
Collapse
|
28
|
Selvarajah J, Elia A, Carroll VA, Moumen A. DNA damage-induced S and G2/M cell cycle arrest requires mTORC2-dependent regulation of Chk1. Oncotarget 2015; 6:427-40. [PMID: 25460505 PMCID: PMC4381605 DOI: 10.18632/oncotarget.2813] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 11/14/2014] [Indexed: 12/25/2022] Open
Abstract
mTOR signalling is commonly dysregulated in cancer. Concordantly, mTOR inhibitors have demonstrated efficacy in a subset of tumors and are in clinical trials as combination therapies. Although mTOR is associated with promoting cell survival after DNA damage, the exact mechanisms are not well understood. Moreover, since mTOR exists as two complexes, mTORC1 and mTORC2, the role of mTORC2 in cancer and in the DNA damage response is less well explored. Here, we report that mTOR protein levels and kinase activity are transiently increased by DNA damage in an ATM and ATR-dependent manner. We show that inactivation of mTOR with siRNA or pharmacological inhibition of mTORC1/2 kinase prevents etoposide-induced S and G2/M cell cycle arrest. Further results show that Chk1, a key regulator of the cell cycle arrest, is important for this since ablation of mTOR prevents DNA damage-induced Chk1 phosphorylation and decreases Chk1 protein production. Furthermore, mTORC2 was essential and mTORC1 dispensable, for this role. Importantly, we show that mTORC1/2 inhibition sensitizes breast cancer cells to chemotherapy. Taken together, these results suggest that breast cancer cells may rely on mTORC2-Chk1 pathway for survival and provide evidence that mTOR kinase inhibitors may overcome resistance to DNA-damage based therapies in breast cancer.
Collapse
Affiliation(s)
- Jogitha Selvarajah
- Cardiovascular and Cell Sciences Research Institute, St George's University of London, Cranmer Terrace, UK
| | - Androulla Elia
- Cardiovascular and Cell Sciences Research Institute, St George's University of London, Cranmer Terrace, UK
| | - Veronica A. Carroll
- Cardiovascular and Cell Sciences Research Institute, St George's University of London, Cranmer Terrace, UK
| | - Abdeladim Moumen
- Division of Medical Biotechnology, MAscIR Institution, Rabat, Morocco
| |
Collapse
|
29
|
Wen SY, Li CH, Zhang YL, Bian YH, Ma L, Ge QL, Teng YC, Zhang ZG. Rictor is an independent prognostic factor for endometrial carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:2068-2078. [PMID: 24966915 PMCID: PMC4069916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 04/10/2014] [Indexed: 06/03/2023]
Abstract
Early-stage endometrial carcinoma (EC) patients have a high cure rate; however, those with high-risk factors may have poor prognosis. Thus, there is an urgent need for searching for new prognostic molecules to more accurately predict survival of patients. We detected the Rictor mRNA expression level in 30 fresh EC tissue and 17 normal endometrial tissue samples with real-time quantitative RT-PCR and Rictor protein expression level in 134 (test cohort) and 115 (validation cohort) paraffin tissue samples by immunohistochemistry, analyzed the correlation between variables and overall survival (OS) using Cox proportional hazards regression, compared the prognostic accuracy of Rictor with other clinicopathological risk factors by logistic regression. The results showed that Rictor mRNA expression of EC is higher than that of normal endometrium; Rictor protein expression level was closely correlated with FIGO stage, grade and vascular invasion in both cohorts; a univariate analysis showed that the pathological type, stage, grade, vascular invasion, lymphatic metastasis and Rictor were predictors of OS in both cohorts; furthermore, multivariate Cox proportional hazards regression analysis indicated that vascular invasion and Rictor were independent prognostic factors for EC in both cohorts; an ROX curve comparison showed that the area under the curve (AUC) for Rictor combined with other clinicopathological prognostic factors was higher than any individual factor or other clinicopathological prognostic factors' combination. Based on the above data, we concluded that Rictor is an independent prognostic factor for EC. It combined with other clinicopathological risk factors was a stronger prognostic model than individual risk factor or their combination.
Collapse
Affiliation(s)
- Shan-Yun Wen
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai, China
- Department of Obstetrics and Gynecology, Shanghai Songjiang District Central HospitalShanghai, China
| | - Chang-Hua Li
- Department of Obstetrics & Gynecology, Huai’an First People’s Hospital, Nanjing Medical UniversityHuai’an, Jiangsu, PR China
| | - Yan-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Yu-Hai Bian
- Department of General Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong UniversityShanghai, China
| | - Li Ma
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai, China
| | - Qiu-Lin Ge
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai, China
| | - Yin-Cheng Teng
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| |
Collapse
|