1
|
Hartung J, Müller C, Calkhoven CF. The dual role of the TSC complex in cancer. Trends Mol Med 2025; 31:452-465. [PMID: 39488444 DOI: 10.1016/j.molmed.2024.10.009] [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: 08/19/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
The tuberous sclerosis complex (TSC1/TSC2/TBC1D7) primarily functions to inhibit the mechanistic target of rapamycin complex 1 (mTORC1), a crucial regulator of cell growth. Mutations in TSC1 or TSC2 cause tuberous sclerosis complex (TSC), a rare autosomal dominant genetic disorder marked by benign tumors in multiple organs that rarely progress to malignancy. Traditionally, TSC proteins are considered tumor suppressive due to their inhibition of mTORC1 and other mechanisms. However, more recent studies have shown that TSC proteins can also promote tumorigenesis in certain cancer types. In this review, we explore the composition and function of the TSC protein complex, the roles of its individual components in cancer biology, and potential future therapeutic targeting strategies.
Collapse
Affiliation(s)
- Josephine Hartung
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Christine Müller
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Cornelis F Calkhoven
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands.
| |
Collapse
|
2
|
Ryspayeva D, Seyhan AA, MacDonald WJ, Purcell C, Roady TJ, Ghandali M, Verovkina N, El-Deiry WS, Taylor MS, Graff SL. Signaling pathway dysregulation in breast cancer. Oncotarget 2025; 16:168-201. [PMID: 40080721 PMCID: PMC11906143 DOI: 10.18632/oncotarget.28701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
This article provides a comprehensive analysis of the signaling pathways implicated in breast cancer (BC), the most prevalent malignancy among women and a leading cause of cancer-related mortality globally. Special emphasis is placed on the structural dynamics of protein complexes that are integral to the regulation of these signaling cascades. Dysregulation of cellular signaling is a fundamental aspect of BC pathophysiology, with both upstream and downstream signaling cascade activation contributing to cellular process aberrations that not only drive tumor growth, but also contribute to resistance against current treatments. The review explores alterations within these pathways across different BC subtypes and highlights potential therapeutic strategies targeting these pathways. Additionally, the influence of specific mutations on therapeutic decision-making is examined, underscoring their relevance to particular BC subtypes. The article also discusses both approved therapeutic modalities and ongoing clinical trials targeting disrupted signaling pathways. However, further investigation is necessary to fully elucidate the underlying mechanisms and optimize personalized treatment approaches.
Collapse
Affiliation(s)
- Dinara Ryspayeva
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
| | - William J. MacDonald
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Connor Purcell
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Tyler J. Roady
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
| | - Maryam Ghandali
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Nataliia Verovkina
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
- Department of Medicine, Hematology/Oncology Division, Lifespan Health System and Brown University, RI 02903, USA
| | - Martin S. Taylor
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
- Brown Center on the Biology of Aging, Brown University, RI 02903, USA
| | - Stephanie L. Graff
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Department of Medicine, Hematology/Oncology Division, Lifespan Health System and Brown University, RI 02903, USA
| |
Collapse
|
3
|
Liu L, Graff SL, Wang Y. New Emerging Therapies Targeting PI3K/AKT/mTOR/PTEN Pathway in Hormonal Receptor-Positive and HER2-Negative Breast Cancer-Current State and Molecular Pathology Perspective. Cancers (Basel) 2024; 17:16. [PMID: 39796647 PMCID: PMC11718791 DOI: 10.3390/cancers17010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
In hormone receptor-positive and HER2-negative breast cancers, a growing number of revolutionary personalized therapies are in clinical use or trials, such as CDK4/6 inhibitors, immune checkpoint inhibitors, and PIK3CA inhibitors. Those treatment options are largely driven by the presence or absence of genomic alterations in the tumor. Therefore, molecular profiling is often performed during disease progression. The most encountered genomic alterations are in the PI3K/AKT/mTOR/PTEN pathway. This review discusses the genetic alterations associated with the PI3K/AKT/mTOR/PTEN pathway to help clinicians understand drug selection, resistance, or interaction from a molecular pathologist's perspective.
Collapse
Affiliation(s)
- Liu Liu
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Brown University Health, Providence, RI 02903, USA;
- Legorreta Cancer Center, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
| | - Stephanie L. Graff
- Legorreta Cancer Center, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
- Division of Medical Oncology, Rhode Island Hospital and Brown University Health, Providence, RI 02903, USA
| | - Yihong Wang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Brown University Health, Providence, RI 02903, USA;
- Legorreta Cancer Center, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA;
| |
Collapse
|
4
|
Yang L, Xiao Y, Ren S. Identification of common genetic features and pathways involved in pulmonary lymphangioleiomyomatosis and ER-positive breast cancer. Medicine (Baltimore) 2023; 102:e34810. [PMID: 37773865 PMCID: PMC10545372 DOI: 10.1097/md.0000000000034810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/27/2023] [Indexed: 10/01/2023] Open
Abstract
Accumulating evidence suggests that patients with pulmonary lymphangioleiomyomatosis (PLAM) have a markedly higher prevalence of breast cancer (BC) than the general population. However, the underlying pathophysiological mechanisms remain unclear. Therefore, in this study, we employed a bioinformatics approach to understand the association between PLAM and estrogen receptor (ER)-positive BC. The PLAM (GSE12027) and ER-positive BC (GSE42568, GSE29044, and GSE29431) datasets were obtained from the Gene Expression Omnibus database, and GEO2R was used to identify common differentially expressed genes (DEGs) between them. Functional annotation was performed, and a protein-protein interaction (PPI) network was constructed. Hub genes were identified and verified using western blotting and immunohistochemistry. We conducted an immune infiltration analysis; based on the results, selected 102 common DEGs for follow-up analysis. Functional analyses revealed that the DEGs were mostly enriched in cell proliferation, gene expression regulation, and tumor-related pathways. Four hub genes-ESR1, IL6, PLA2G4A, and CAV1-were further analyzed, and CAV1 was revealed to be associated with clinical outcomes and immune infiltration in ER-positive BC. This study proposes a common, possible pathogenesis of PLAM and ER-positive BC. These common pathways and pivotal genes may provide new directions for further mechanistic studies.
Collapse
Affiliation(s)
- Lulu Yang
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ying Xiao
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Siying Ren
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
5
|
Githaka JM, Pirayeshfard L, Goping IS. Cancer invasion and metastasis: Insights from murine pubertal mammary gland morphogenesis. Biochim Biophys Acta Gen Subj 2023; 1867:130375. [PMID: 37150225 DOI: 10.1016/j.bbagen.2023.130375] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/20/2023] [Accepted: 05/02/2023] [Indexed: 05/09/2023]
Abstract
Cancer invasion and metastasis accounts for the majority of cancer related mortality. A better understanding of the players that drive the aberrant invasion and migration of tumors cells will provide critical targets to inhibit metastasis. Postnatal pubertal mammary gland morphogenesis is characterized by highly proliferative, invasive, and migratory normal epithelial cells. Identifying the molecular regulators of pubertal gland development is a promising strategy since tumorigenesis and metastasis is postulated to be a consequence of aberrant reactivation of developmental stages. In this review, we summarize the pubertal morphogenesis regulators that are involved in cancer metastasis and revisit pubertal mammary gland transcriptome profiling to uncover both known and unknown metastasis genes. Our updated list of pubertal morphogenesis regulators shows that most are implicated in invasion and metastasis. This review highlights molecular linkages between development and metastasis and provides a guide for exploring novel metastatic drivers.
Collapse
Affiliation(s)
- John Maringa Githaka
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Leila Pirayeshfard
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Ing Swie Goping
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Department of Oncology, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
6
|
Wang H, Liu B, Chen H, Xu P, Xue H, Yuan J. Dynamic changes of DNA methylation induced by benzo(a)pyrene in cancer. Genes Environ 2023; 45:21. [PMID: 37391844 DOI: 10.1186/s41021-023-00278-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/07/2023] [Indexed: 07/02/2023] Open
Abstract
Benzo(a)pyrene (BaP), the earliest and most significant carcinogen among polycyclic aromatic hydrocarbons (PAHs), has been found in foods, tobacco smoke, and automobiles exhaust, etc. Exposure to BaP induced DNA damage directly, or oxidative stress-related damage, resulting in cell apoptosis and carcinogenesis in human respiratory system, digestive system, reproductive system, etc. Moreover, BaP triggered genome-wide epigenetic alterations by methylation, which might cause disturbances in regulation of gene expression, and thereby induced cancer. It has been proved that BaP reduced genome-wide DNA methylation, and activated proto-oncogene by hypomethylation in the promoter region, but silenced tumor suppressor genes by promoter hypermethylation, resulting in cancer initiation and progression. Here we summarized the changes in DNA methylation in BaP exposure, and revealed the methylation of DNA plays a role in cancer development.
Collapse
Affiliation(s)
- Huizeng Wang
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Bingchun Liu
- Stem Cell Research Center, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Hong Chen
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Peixin Xu
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Huiting Xue
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010010, China.
| | - Jianlong Yuan
- Department of Laboratory Medicine, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China.
| |
Collapse
|
7
|
Bovari-Biri J, Abdelwahab EMM, Garai K, Pongracz JE. Prdx5 in the Regulation of Tuberous Sclerosis Complex Mutation-Induced Signaling Mechanisms. Cells 2023; 12:1713. [PMID: 37443747 PMCID: PMC10340296 DOI: 10.3390/cells12131713] [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: 05/19/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
(1) Background: Tuberous sclerosis complex (TSC) mutations directly affect mTORC activity and, as a result, protein synthesis. In several cancer types, TSC mutation is part of the driver mutation panel. TSC mutations have been associated with mitochondrial dysfunction, tolerance to reactive oxygen species due to increased thioredoxin reductase (TrxR) enzyme activity, tolerance to endoplasmic reticulum (ER) stress, and apoptosis. The FDA-approved drug rapamycin is frequently used in clinical applications to inhibit protein synthesis in cancers. Recently, TrxR inhibitor auranofin has also been involved in clinical trials to investigate the anticancer efficacy of the combination treatment with rapamycin. We aimed to investigate the molecular background of the efficacy of such drug combinations in treating neoplasia modulated by TSC mutations. (2) Methods: TSC2 mutant and TSC2 wild-type (WT) cell lines were exposed to rapamycin and auranofin in either mono- or combination treatment. Mitochondrial membrane potential, TrxR enzyme activity, stress protein array, mRNA and protein levels were investigated via cell proliferation assay, electron microscopy, etc. (3) Results: Auranofin and rapamycin normalized mitochondrial membrane potential and reduced proliferation capacity of TSC2 mutant cells. Database analysis identified peroxiredoxin 5 (Prdx5) as the joint target of auranofin and rapamycin. The auranofin and the combination of the two drugs reduced Prdx5 levels. The combination treatment increased the expression of heat shock protein 70, a cellular ER stress marker. (4) Conclusions: After extensive analyses, Prdx5 was identified as a shared target of the two drugs. The decreased Prdx5 protein level and the inhibition of both TrxR and mTOR by rapamycin and auranofin in the combination treatment made ER stress-induced cell death possible in TSC2 mutant cells.
Collapse
Affiliation(s)
| | | | | | - Judit E. Pongracz
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, 2. Rokus Str, H-7624 Pecs, Hungary
| |
Collapse
|
8
|
Zhu QY, He ZM, Cao WM, Li B. The role of TSC2 in breast cancer: a literature review. Front Oncol 2023; 13:1188371. [PMID: 37251941 PMCID: PMC10213421 DOI: 10.3389/fonc.2023.1188371] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
TSC2 is a tumor suppressor gene as well as a disease-causing gene for autosomal dominant disorder tuberous sclerosis complex (TSC). Research has found that some tumor tissues have lower TSC2 expression levels than normal tissues. Furthermore, low expression of TSC2 is associated with poor prognosis in breast cancer. TSC2 acts as a convergence point of a complex network of signaling pathways and receives signals from the PI3K, AMPK, MAPK, and WNT pathways. It also regulates cellular metabolism and autophagy through inhibition of a mechanistic target of rapamycin complex, which are processes relevant to the progression, treatment, and prognosis of breast cancer. In-depth study of TSC2 functions provides significant guidance for clinical applications in breast cancer, including improving the treatment efficacy, overcoming drug resistance, and predicting prognosis. In this review, protein structure and biological functions of TSC2 were described and recent advances in TSC2 research in different molecular subtypes of breast cancer were summarized.
Collapse
Affiliation(s)
- Qiao-Yan Zhu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Zhe-Min He
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Wen-Ming Cao
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Bei Li
- Department of Geriatric, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
9
|
Lin C, Traets JJH, Vredevoogd DW, Visser NL, Peeper DS. TSC2 regulates tumor susceptibility to TRAIL-mediated T-cell killing by orchestrating mTOR signaling. EMBO J 2023; 42:e111614. [PMID: 36715448 PMCID: PMC9975943 DOI: 10.15252/embj.2022111614] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 12/08/2022] [Accepted: 12/14/2022] [Indexed: 01/31/2023] Open
Abstract
Resistance to cancer immunotherapy continues to impair common clinical benefit. Here, we use whole-genome CRISPR-Cas9 knockout data to uncover an important role for Tuberous Sclerosis Complex 2 (TSC2) in determining tumor susceptibility to cytotoxic T lymphocyte (CTL) killing in human melanoma cells. TSC2-depleted tumor cells had disrupted mTOR regulation following CTL attack, which was associated with enhanced cell death. Wild-type tumor cells adapted to CTL attack by shifting their mTOR signaling balance toward increased mTORC2 activity, circumventing apoptosis, and necroptosis. TSC2 ablation strongly augmented tumor cell sensitivity to CTL attack in vitro and in vivo, suggesting one of its functions is to critically protect tumor cells. Mechanistically, TSC2 inactivation caused elevation of TRAIL receptor expression, cooperating with mTORC1-S6 signaling to induce tumor cell death. Clinically, we found a negative correlation between TSC2 expression and TRAIL signaling in TCGA patient cohorts. Moreover, a lower TSC2 immune response signature was observed in melanomas from patients responding to immune checkpoint blockade. Our study uncovers a pivotal role for TSC2 in the cancer immune response by governing crosstalk between TSC2-mTOR and TRAIL signaling, aiding future therapeutic exploration of this pathway in immuno-oncology.
Collapse
Affiliation(s)
- Chun‐Pu Lin
- Division of Molecular Oncology and ImmunologyOncode Institute, The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Joleen J H Traets
- Division of Molecular Oncology and ImmunologyOncode Institute, The Netherlands Cancer InstituteAmsterdamThe Netherlands
- Division of Tumor Biology and ImmunologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - David W Vredevoogd
- Division of Molecular Oncology and ImmunologyOncode Institute, The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Nils L Visser
- Division of Molecular Oncology and ImmunologyOncode Institute, The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Daniel S Peeper
- Division of Molecular Oncology and ImmunologyOncode Institute, The Netherlands Cancer InstituteAmsterdamThe Netherlands
| |
Collapse
|
10
|
Li Q, Li Z, Luo T, Shi H. Targeting the PI3K/AKT/mTOR and RAF/MEK/ERK pathways for cancer therapy. MOLECULAR BIOMEDICINE 2022; 3:47. [PMID: 36539659 PMCID: PMC9768098 DOI: 10.1186/s43556-022-00110-2] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/10/2022] [Indexed: 12/24/2022] Open
Abstract
The PI3K/AKT/mTOR and RAF/MEK/ERK pathways are commonly activated by mutations and chromosomal translocation in vital targets. The PI3K/AKT/mTOR signaling pathway is dysregulated in nearly all kinds of neoplasms, with the component in this pathway alternations. RAF/MEK/ERK signaling cascades are used to conduct signaling from the cell surface to the nucleus to mediate gene expression, cell cycle processes and apoptosis. RAS, B-Raf, PI3K, and PTEN are frequent upstream alternative sites. These mutations resulted in activated cell growth and downregulated cell apoptosis. The two pathways interact with each other to participate in tumorigenesis. PTEN alterations suppress RAF/MEK/ERK pathway activity via AKT phosphorylation and RAS inhibition. Several inhibitors targeting major components of these two pathways have been supported by the FDA. Dozens of agents in these two pathways have attracted great attention and have been assessed in clinical trials. The combination of small molecular inhibitors with traditional regimens has also been explored. Furthermore, dual inhibitors provide new insight into antitumor activity. This review will further comprehensively describe the genetic alterations in normal patients and tumor patients and discuss the role of targeted inhibitors in malignant neoplasm therapy. We hope this review will promote a comprehensive understanding of the role of the PI3K/AKT/mTOR and RAF/MEK/ERK signaling pathways in facilitating tumors and will help direct drug selection for tumor therapy.
Collapse
Affiliation(s)
- Qingfang Li
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, Chengdu, China
| | - Zhihui Li
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu, PR China
| | - Ting Luo
- Department of Breast, Cancer Center, West China Hospital, Sichuan University, 610041, Chengdu, P. R. China.
| | - Huashan Shi
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, 610041, Chengdu, P. R. China.
| |
Collapse
|
11
|
Study on the Expression Profile of Autophagy-Related Genes in Colon Adenocarcinoma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7525048. [PMID: 35572821 PMCID: PMC9095386 DOI: 10.1155/2022/7525048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/11/2022] [Accepted: 03/23/2022] [Indexed: 12/24/2022]
Abstract
Colon adenocarcinoma (COAD) is a common digestive tract tumor. Autophagy-related genes (ARGs) may play an obbligato role in the biological processes of COAD. This study was aimed at exploring the role of ARGs in COAD. Clinical data and RNA sequencing data of tumor and healthy samples were obtained from The Cancer Genome Atlas (TCGA), and discrepantly expressed ARGs were screened. Statistical differences of ARGs were performed with Gene Ontology (GO) functional annotation and the Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. Eight ARGs were selected by univariate Cox and multivariate Cox regression. Kaplan–Meier (K-M) and multivariate receiver operating characteristic (multi-ROC) were used to check the fitness of the model. Among 398 COAD samples and 39 normal samples obtained from the TCGA database, 37 differentially expressed ARGs were screened. In the training group, eight prognostics-related ARGs (MTMR14, VAMP3, HSPA8, TSC1, DAPK1, CX3CL1, ATG13, and MAP1LC3C) were identified by Cox regression. A gene signature risk prediction model was constructed base on 8 autophagy-related genes. The survival time of the low-risk group was longer than the high-risk group, and the AUC of the model was 0.794. Univariate and multivariate Cox regression analysis showed that age and riskscore were the independent predictor. In conclusion, the prognosis model we built based one ARGs of COAD patients can estimate the prognosis of patients in clinical treatment.
Collapse
|
12
|
Wang J, Zhang H, Ren W, Guo M, Yu G. EpiMC: Detecting Epistatic Interactions Using Multiple Clusterings. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:243-254. [PMID: 33989157 DOI: 10.1109/tcbb.2021.3080462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Detecting single nucleotide polymorphisms (SNPs) interactions is crucial to identify susceptibility genes associated with complex human diseases in genome-wide association studies. Clustering-based approaches are widely used in reducing search space and exploring potential relationships between SNPs in epistasis analysis. However, these approaches all only use a single measure to filter out nonsignificant SNP combinations, which may be significant ones from another perspective. In this paper, we propose a two-stage approach named EpiMC (Epistatic Interactions detection based on Multiple Clusterings) that employs multiple clusterings to obtain more precise candidate sets and more comprehensively detect high-order interactions based on these sets. In the first stage, EpiMC proposes a matrix factorization based multiple clusterings algorithm to generate multiple diverse clusterings, each of which divide all SNPs into different clusters. This stage aims to reduce the chance of filtering out potential candidates overlooked by a single clustering and groups associated SNPs together from different clustering perspectives. In the next stage, EpiMC considers both the single-locus effects and interaction effects to select high-quality disease associated SNPs, and then uses Jaccard similarity to get candidate sets. Finally, EpiMC uses exhaustive search on the obtained small candidate sets to precisely detect epsitatic interactions. Extensive simulation experiments show that EpiMC has a better performance in detecting high-order interactions than state-of-the-art solutions. On the Wellcome Trust Case Control Consortium (WTCCC) dataset, EpiMC detects several significant epistatic interactions associated with breast cancer (BC) and age-related macular degeneration (AMD), which again corroborate the effectiveness of EpiMC.
Collapse
|
13
|
Bongaarts A, Mijnsbergen C, Anink JJ, Jansen FE, Spliet WGM, den Dunnen WFA, Coras R, Blümcke I, Paulus W, Gruber VE, Scholl T, Hainfellner JA, Feucht M, Kotulska K, Jozwiak S, Grajkowska W, Buccoliero AM, Caporalini C, Giordano F, Genitori L, Söylemezoğlu F, Pimentel J, Jones DTW, Scicluna BP, Schouten-van Meeteren AYN, Mühlebner A, Mills JD, Aronica E. Distinct DNA Methylation Patterns of Subependymal Giant Cell Astrocytomas in Tuberous Sclerosis Complex. Cell Mol Neurobiol 2021; 42:2863-2892. [PMID: 34709498 PMCID: PMC9560915 DOI: 10.1007/s10571-021-01157-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/12/2021] [Indexed: 10/28/2022]
Abstract
Tuberous sclerosis complex (TSC) is a monogenic disorder caused by mutations in either the TSC1 or TSC2 gene, two key regulators of the mechanistic target of the rapamycin complex pathway. Phenotypically, this leads to growth and formation of hamartomas in several organs, including the brain. Subependymal giant cell astrocytomas (SEGAs) are low-grade brain tumors commonly associated with TSC. Recently, gene expression studies provided evidence that the immune system, the MAPK pathway and extracellular matrix organization play an important role in SEGA development. However, the precise mechanisms behind the gene expression changes in SEGA are still largely unknown, providing a potential role for DNA methylation. We investigated the methylation profile of SEGAs using the Illumina Infinium HumanMethylation450 BeadChip (SEGAs n = 42, periventricular control n = 8). The SEGA methylation profile was enriched for the adaptive immune system, T cell activation, leukocyte mediated immunity, extracellular structure organization and the ERK1 & ERK2 cascade. More interestingly, we identified two subgroups in the SEGA methylation data and show that the differentially expressed genes between the two subgroups are related to the MAPK cascade and adaptive immune response. Overall, this study shows that the immune system, the MAPK pathway and extracellular matrix organization are also affected on DNA methylation level, suggesting that therapeutic intervention on DNA level could be useful for these specific pathways in SEGA. Moreover, we identified two subgroups in SEGA that seem to be driven by changes in the adaptive immune response and MAPK pathway and could potentially hold predictive information on target treatment response.
Collapse
Affiliation(s)
- Anika Bongaarts
- Department of Neuro Pathology, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105, Amsterdam, The Netherlands
| | - Caroline Mijnsbergen
- Department of Neuro Pathology, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105, Amsterdam, The Netherlands
| | - Jasper J Anink
- Department of Neuro Pathology, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105, Amsterdam, The Netherlands
| | - Floor E Jansen
- Department of Pediatric Neurology, Brain Center, University Medical Center, Utrecht, The Netherlands
| | - Wim G M Spliet
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wilfred F A den Dunnen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Roland Coras
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Ingmar Blümcke
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Werner Paulus
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Victoria E Gruber
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Theresa Scholl
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Johannes A Hainfellner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Martha Feucht
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Katarzyna Kotulska
- Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw, Poland
| | - Sergiusz Jozwiak
- Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw, Poland.,Department of Child Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Wieslawa Grajkowska
- Department of Pathology, Children's Memorial Health Institute, Warsaw, Poland
| | | | | | - Flavio Giordano
- Department of Neurosurgery, Anna Meyer Children's Hospital, Florence, Italy
| | - Lorenzo Genitori
- Department of Neurosurgery, Anna Meyer Children's Hospital, Florence, Italy
| | - Figen Söylemezoğlu
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - José Pimentel
- Laboratory of Neuropathology, Department of Neurology, Hospital de Santa Maria (CHULN), Lisbon, Portugal
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Brendon P Scicluna
- Center for Experimental & Molecular Medicine and Department of Clinical Epidemiology, Biostatistics & Bioinformatics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Antoinette Y N Schouten-van Meeteren
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Pediatric Oncology, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Angelika Mühlebner
- Department of Neuro Pathology, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105, Amsterdam, The Netherlands.
| | - James D Mills
- Department of Neuro Pathology, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105, Amsterdam, The Netherlands.
| | - Eleonora Aronica
- Department of Neuro Pathology, Amsterdam UMC, Location AMC, University of Amsterdam, Meibergdreef 9, 1105, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| |
Collapse
|
14
|
Wang X, Zhang H, Wang J, Yu G, Cui L, Guo M. EpiHNet: Detecting epistasis by heterogeneous molecule network. Methods 2021; 198:65-75. [PMID: 34555529 DOI: 10.1016/j.ymeth.2021.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/16/2021] [Accepted: 09/16/2021] [Indexed: 12/22/2022] Open
Abstract
Epistasis between single nucleotide polymorphisms (SNPs) plays an important role in elucidating the missing heritability of complex diseases. Diverse approaches have been invented for detecting SNP interactions, but they canonically neglect the important and useful connections between SNPs and other bio-molecules (i.e., miRNAs and lncRNAs). To comprehensively model these disease related molecules, a heterogeneous bio-molecular network based solution EpiHNet is introduced for high-order SNP interactions detection. EpiHNet firstly uses case/control data to construct an SNP statistical network, and meta-path based similarity on the heterogeneous network composed with SNPs, genes, lncRNAs, miRNAs and diseases to define another SNP relational network. The SNP relational network can explore and exploit different associations between molecules and diseases to complement the SNP statistical network and search the significantly associated SNPs. Next, EpiHNet integrates these two networks into a composite network, applies the modularity based clustering with fast search strategy to divide SNP nodes into different clusters. After that, it detects SNP interactions based on SNP combinations derived from each cluster. Synthetic experiments on diverse two-locus and three-locus disease models manifest that EpiHNet outperforms competitive baselines, even without the heterogeneous network. For real WTCCC breast cancer data, EpiHNet also demonstrates expressive results on detecting high-order SNP interactions.
Collapse
Affiliation(s)
- Xin Wang
- School of Software, Shandong University, Jinan, China; Joint SDU-NTU Centre For AI Research (C-FAIR), Shandong University, Jinan, China.
| | - Huiling Zhang
- College of Computer and Information Sciences, Southwest University, Chongqing, China.
| | - Jun Wang
- School of Software, Shandong University, Jinan, China; Joint SDU-NTU Centre For AI Research (C-FAIR), Shandong University, Jinan, China.
| | - Guoxian Yu
- School of Software, Shandong University, Jinan, China; Joint SDU-NTU Centre For AI Research (C-FAIR), Shandong University, Jinan, China.
| | - Lizhen Cui
- School of Software, Shandong University, Jinan, China; Joint SDU-NTU Centre For AI Research (C-FAIR), Shandong University, Jinan, China.
| | - Maozu Guo
- College of Electrical and Information Engineering, Beijing University of Civil Engineering and Architecture, Beijing, China.
| |
Collapse
|
15
|
Herranz C, Mateo F, Baiges A, Ruiz de Garibay G, Junza A, Johnson SR, Miller S, García N, Capellades J, Gómez A, Vidal A, Palomero L, Espín R, Extremera AI, Blommaert E, Revilla‐López E, Saez B, Gómez‐Ollés S, Ancochea J, Valenzuela C, Alonso T, Ussetti P, Laporta R, Xaubet A, Rodríguez‐Portal JA, Montes‐Worboys A, Machahua C, Bordas J, Menendez JA, Cruzado JM, Guiteras R, Bontoux C, La Motta C, Noguera‐Castells A, Mancino M, Lastra E, Rigo‐Bonnin R, Perales JC, Viñals F, Lahiguera A, Zhang X, Cuadras D, van Moorsel CHM, van der Vis JJ, Quanjel MJR, Filippakis H, Hakem R, Gorrini C, Ferrer M, Ugun‐Klusek A, Billett E, Radzikowska E, Casanova Á, Molina‐Molina M, Roman A, Yanes O, Pujana MA. Histamine signaling and metabolism identify potential biomarkers and therapies for lymphangioleiomyomatosis. EMBO Mol Med 2021; 13:e13929. [PMID: 34378323 PMCID: PMC8422079 DOI: 10.15252/emmm.202113929] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 11/12/2022] Open
Abstract
Inhibition of mTOR is the standard of care for lymphangioleiomyomatosis (LAM). However, this therapy has variable tolerability and some patients show progressive decline of lung function despite treatment. LAM diagnosis and monitoring can also be challenging due to the heterogeneity of symptoms and insufficiency of non-invasive tests. Here, we propose monoamine-derived biomarkers that provide preclinical evidence for novel therapeutic approaches. The major histamine-derived metabolite methylimidazoleacetic acid (MIAA) is relatively more abundant in LAM plasma, and MIAA values are independent of VEGF-D. Higher levels of histamine are associated with poorer lung function and greater disease burden. Molecular and cellular analyses, and metabolic profiling confirmed active histamine signaling and metabolism. LAM tumorigenesis is reduced using approved drugs targeting monoamine oxidases A/B (clorgyline and rasagiline) or histamine H1 receptor (loratadine), and loratadine synergizes with rapamycin. Depletion of Maoa or Hrh1 expression, and administration of an L-histidine analog, or a low L-histidine diet, also reduce LAM tumorigenesis. These findings extend our knowledge of LAM biology and suggest possible ways of improving disease management.
Collapse
Affiliation(s)
- Carmen Herranz
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Francesca Mateo
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Alexandra Baiges
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Gorka Ruiz de Garibay
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Alexandra Junza
- Department of Electronic EngineeringInstitute of Health Research Pere Virgili (IIPSV)University Rovira i VirgiliTarragonaSpain
- Biomedical Research Network Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)Instituto de Salud Carlos IIIMadridSpain
| | - Simon R Johnson
- National Centre for LymphangioleiomyomatosisNottingham University Hospitals NHS Trust, NottinghamshireDivision of Respiratory MedicineUniversity of NottinghamNottinghamUK
| | - Suzanne Miller
- National Centre for LymphangioleiomyomatosisNottingham University Hospitals NHS Trust, NottinghamshireDivision of Respiratory MedicineUniversity of NottinghamNottinghamUK
| | - Nadia García
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Jordi Capellades
- Department of Electronic EngineeringInstitute of Health Research Pere Virgili (IIPSV)University Rovira i VirgiliTarragonaSpain
- Biomedical Research Network Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)Instituto de Salud Carlos IIIMadridSpain
| | - Antonio Gómez
- Centre for Genomic RegulationBarcelona Institute of Science and TechnologyBarcelonaSpain
- Present address:
Rheumatology Department and Rheumatology Research GroupVall d'Hebron Hospital Research Institute (VHIR)BarcelonaSpain
| | - August Vidal
- Department of PathologyUniversity Hospital of BellvitgeOncobellIDIBELL, L’Hospitalet del LlobregatBarcelonaSpain
- CIBER on Cancer (CIBERONC)Instituto de Salud Carlos IIIMadridSpain
| | - Luis Palomero
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Roderic Espín
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Ana I Extremera
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Eline Blommaert
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Eva Revilla‐López
- Lung Transplant Unit, Pneumology ServiceLymphangioleiomyomatosis ClinicVall d’Hebron University HospitalBarcelonaSpain
| | - Berta Saez
- Lung Transplant Unit, Pneumology ServiceLymphangioleiomyomatosis ClinicVall d’Hebron University HospitalBarcelonaSpain
| | - Susana Gómez‐Ollés
- Lung Transplant Unit, Pneumology ServiceLymphangioleiomyomatosis ClinicVall d’Hebron University HospitalBarcelonaSpain
| | - Julio Ancochea
- Pneumology ServiceLa Princesa Research InstituteUniversity Hospital La PrincesaMadridSpain
| | - Claudia Valenzuela
- Pneumology ServiceLa Princesa Research InstituteUniversity Hospital La PrincesaMadridSpain
| | - Tamara Alonso
- Pneumology ServiceLa Princesa Research InstituteUniversity Hospital La PrincesaMadridSpain
| | - Piedad Ussetti
- Pneumology ServiceUniversity Hospital Clínica Puerta del Hierro, MajadahondaMadridSpain
| | - Rosalía Laporta
- Pneumology ServiceUniversity Hospital Clínica Puerta del Hierro, MajadahondaMadridSpain
| | - Antoni Xaubet
- Pneumology ServiceHospital Clínic de BarcelonaBarcelonaSpain
| | - José A Rodríguez‐Portal
- Medical‐Surgical Unit of Respiratory DiseasesInstitute of Biomedicine of Seville (IBiS)University Hospital Virgen del RocíoSevilleSpain
- Biomedical Research Network Centre in Respiratory Diseases (CIBERES)Instituto de Salud Carlos IIIMadridSpain
| | - Ana Montes‐Worboys
- Biomedical Research Network Centre in Respiratory Diseases (CIBERES)Instituto de Salud Carlos IIIMadridSpain
- Interstitial Lung Disease UnitDepartment of Respiratory MedicineUniversity Hospital of BellvitgeIDIBELLL’Hospitalet del LlobregatBarcelonaSpain
| | - Carlos Machahua
- Biomedical Research Network Centre in Respiratory Diseases (CIBERES)Instituto de Salud Carlos IIIMadridSpain
- Interstitial Lung Disease UnitDepartment of Respiratory MedicineUniversity Hospital of BellvitgeIDIBELLL’Hospitalet del LlobregatBarcelonaSpain
| | - Jaume Bordas
- Biomedical Research Network Centre in Respiratory Diseases (CIBERES)Instituto de Salud Carlos IIIMadridSpain
- Interstitial Lung Disease UnitDepartment of Respiratory MedicineUniversity Hospital of BellvitgeIDIBELLL’Hospitalet del LlobregatBarcelonaSpain
| | - Javier A Menendez
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Josep M Cruzado
- Experimental NephrologyDepartment of Clinical SciencesUniversity of BarcelonaBarcelonaSpain
- Department of NephrologyUniversity Hospital of BellvitgeIDIBELLL’Hospitalet del LlobregatBarcelonaSpain
| | - Roser Guiteras
- Experimental NephrologyDepartment of Clinical SciencesUniversity of BarcelonaBarcelonaSpain
- Department of NephrologyUniversity Hospital of BellvitgeIDIBELLL’Hospitalet del LlobregatBarcelonaSpain
| | - Christophe Bontoux
- Department of PathologyUniversity Hospital Pitié‐SalpêtrièreFaculty of MedicineUniversity of SorbonneParisFrance
| | | | - Aleix Noguera‐Castells
- Biomedical Research Institute “August Pi i Sunyer” (IDIBAPS)Department of MedicineUniversity of BarcelonaBarcelonaSpain
| | - Mario Mancino
- Biomedical Research Institute “August Pi i Sunyer” (IDIBAPS)Department of MedicineUniversity of BarcelonaBarcelonaSpain
| | - Enrique Lastra
- Genetic Counseling UnitDepartment of Medical OncologyUniversity Hospital of BurgosBurgosSpain
| | - Raúl Rigo‐Bonnin
- Clinical LaboratoryUniversity Hospital of BellvitgeIDIBELLL'Hospitalet de LlobregatBarcelonaSpain
| | - Jose C Perales
- Department of Physiological Science IIUniversity of BarcelonaBarcelonaSpain
| | - Francesc Viñals
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
- Department of Physiological Science IIUniversity of BarcelonaBarcelonaSpain
| | - Alvaro Lahiguera
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| | - Xiaohu Zhang
- National Center for Advancing Translational Sciences (NCATS)National Institute of Health (NIH)BethesdaMDUSA
| | - Daniel Cuadras
- Statistics DepartmentFoundation Sant Joan de DéuEspluguesSpain
| | - Coline H M van Moorsel
- Interstitial Lung Disease (ILD) Center of ExcellenceSt. Antonius HospitalNieuwegeinThe Netherlands
| | - Joanne J van der Vis
- Interstitial Lung Disease (ILD) Center of ExcellenceSt. Antonius HospitalNieuwegeinThe Netherlands
| | - Marian J R Quanjel
- Interstitial Lung Disease (ILD) Center of ExcellenceSt. Antonius HospitalNieuwegeinThe Netherlands
| | - Harilaos Filippakis
- Pulmonary and Critical Care MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Razq Hakem
- Princess Margaret Cancer CentreUniversity Health NetworkDepartment of Medical BiophysicsUniversity of TorontoTorontoOntarioCanada
| | - Chiara Gorrini
- Princess Margaret HospitalThe Campbell Family Institute for Breast Cancer ResearchOntario Cancer InstituteUniversity Health NetworkTorontoONCanada
| | - Marc Ferrer
- National Center for Advancing Translational Sciences (NCATS)National Institute of Health (NIH)BethesdaMDUSA
| | - Aslihan Ugun‐Klusek
- Centre for Health, Ageing and Understanding Disease (CHAUD)School of Science and TechnologyNottingham Trent UniversityNottinghamUK
| | - Ellen Billett
- Centre for Health, Ageing and Understanding Disease (CHAUD)School of Science and TechnologyNottingham Trent UniversityNottinghamUK
| | - Elżbieta Radzikowska
- Department of Lung Diseases IIINational Tuberculosis and Lung Disease Research InstituteWarsawPoland
| | - Álvaro Casanova
- Pneumology ServiceUniversity Hospital of HenaresUniversity Francisco de Vitoria, CosladaMadridSpain
| | - María Molina‐Molina
- Biomedical Research Network Centre in Respiratory Diseases (CIBERES)Instituto de Salud Carlos IIIMadridSpain
- Interstitial Lung Disease UnitDepartment of Respiratory MedicineUniversity Hospital of BellvitgeIDIBELLL’Hospitalet del LlobregatBarcelonaSpain
| | - Antonio Roman
- Lung Transplant Unit, Pneumology ServiceLymphangioleiomyomatosis ClinicVall d’Hebron University HospitalBarcelonaSpain
| | - Oscar Yanes
- Department of Electronic EngineeringInstitute of Health Research Pere Virgili (IIPSV)University Rovira i VirgiliTarragonaSpain
- Biomedical Research Network Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM)Instituto de Salud Carlos IIIMadridSpain
| | - Miquel A Pujana
- ProCURECatalan Institute of OncologyOncobellBellvitge Institute for Biomedical Research (IDIBELL)L’Hospitalet del LlobregatBarcelonaSpain
| |
Collapse
|
16
|
Mallela K, Shivananda S, Gopinath KS, Kumar A. Oncogenic role of MiR-130a in oral squamous cell carcinoma. Sci Rep 2021; 11:7787. [PMID: 33833339 PMCID: PMC8032739 DOI: 10.1038/s41598-021-87388-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Aberrant activation of the PI3K/AKT/mTOR pathway is attributed to the pathogenesis of oral squamous cell carcinoma (OSCC). In recent years, increasing evidence suggests the involvement of microRNAs (miRNAs) in oral carcinogenesis by acting as tumor suppressors or oncogenes. TSC1, as a component of the above pathway, regulates several cellular functions such as cell proliferation, apoptosis, migration and invasion. Downregulation of TSC1 is reported in oral as well as several other cancers and is associated with an unfavourable clinical outcome in patients. Here we show that oncogenic miR-130a binds to the 3′UTR of TSC1 and represses its expression. MiR-130a-mediated repression of TSC1 increases cell proliferation, anchorage independent growth and invasion of OSCC cells, which is dependent on the presence of the 3′UTR in TSC1. We observe an inverse correlation between the expression levels of miR-130a and TSC1 in OSCC samples, suggesting that their interaction is physiologically relevant. Delivery of antagomiR-130a to OSCC cells results in a significant decrease in xenograft size. Taken together, the findings of the study indicate that miR-130a-mediated TSC1 downregulation is not only a novel mechanism in OSCC, but also the restoration of TSC1 levels by antagomiR-130a may be a potential therapeutic strategy for the treatment of OSCC.
Collapse
Affiliation(s)
- Karthik Mallela
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | | | | | - Arun Kumar
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
17
|
Mallela K, Kumar A. Role of TSC1 in physiology and diseases. Mol Cell Biochem 2021; 476:2269-2282. [PMID: 33575875 DOI: 10.1007/s11010-021-04088-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/27/2021] [Indexed: 12/15/2022]
Abstract
Since its initial discovery as the gene altered in Tuberous Sclerosis Complex (TSC), an autosomal dominant disorder, the interest in TSC1 (Tuberous Sclerosis Complex 1) has steadily risen. TSC1, an essential component of the pro-survival PI3K/AKT/MTOR signaling pathway, plays an important role in processes like development, cell growth and proliferation, survival, autophagy and cilia development by co-operating with a variety of regulatory molecules. Recent studies have emphasized the tumor suppressive role of TSC1 in several human cancers including liver, lung, bladder, breast, ovarian, and pancreatic cancers. TSC1 perceives inputs from various signaling pathways, including TNF-α/IKK-β, TGF-β-Smad2/3, AKT/Foxo/Bim, Wnt/β-catenin/Notch, and MTOR/Mdm2/p53 axis, thereby regulating cancer cell proliferation, metabolism, migration, invasion, and immune regulation. This review provides a first comprehensive evaluation of TSC1 and illuminates its diverse functions apart from its involvement in TSC genetic disorder. Further, we have summarized the physiological functions of TSC1 in various cellular events and conditions whose dysregulation may lead to several pathological manifestations including cancer.
Collapse
Affiliation(s)
- Karthik Mallela
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - Arun Kumar
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
18
|
Sundar R, Ng A, Zouridis H, Padmanabhan N, Sheng T, Zhang S, Lee MH, Ooi WF, Qamra A, Inam I, Hewitt LC, So JBY, Koh V, Nankivell MG, Langley RE, Allum WH, Cunningham D, Rozen SG, Yong WP, Grabsch HI, Tan P. DNA epigenetic signature predictive of benefit from neoadjuvant chemotherapy in oesophageal adenocarcinoma: results from the MRC OE02 trial. Eur J Cancer 2019; 123:48-57. [PMID: 31655359 DOI: 10.1016/j.ejca.2019.09.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/26/2019] [Accepted: 09/16/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND DNA methylation signatures describing distinct histological subtypes of oesophageal cancer have been reported. We studied DNA methylation in samples from the MRC OE02 phase III trial, which randomised patients with resectable oesophageal cancer to surgery alone (S) or neoadjuvant chemotherapy followed by surgery (CS). AIM The aim of the study was to identify epigenetic signatures predictive of chemotherapy benefit in patients with oesophageal adenocarcinoma (OAC) from the OE02 trial and validate the findings in an independent cohort. METHODS DNA methylation was analysed using the Illumina GoldenGate platform on surgically resected OAC specimens from patients in the OE02 trial. Cox proportional hazard analysis was performed to select probes predictive of survival in the CS arm. Non-negative matrix factorisation was used to perform clustering and delineate DNA methylation signatures. The findings were validated in an independent cohort of patients with gastroesophageal adenocarcinoma treated with neoadjuvant chemotherapy. RESULTS A total of 229 patients with OAC were analysed from the OE02 trial (118 in the CS arm and 111 in the S arm). There was no difference in DNA methylation status between the CS and S arms. A metagene signature was created by dichotomising samples into two clusters. In cluster 1, patients in the CS arm had significant overall survival (OS) benefit (median OS CS: 931 days vs. S: 536 days [HR: 1.54, P = 0.031]). In cluster 2, patients in the CS arm had similar (or worse) OS compared with patients in the S arm (CS: 348 days vs. S: 472 days [HR: 0.70, P = 0.1], and test of interaction was significant (p = 0.005). In the validation cohort (n = 13), there was no difference in DNA methylation status in paired pre- and post-treatment samples. When the epigenetic signature was applied, cluster 1 samples had better OS (median OS, cluster 1: 1174 days vs. cluster 2: 392 days, HR: 3.47, p = 0.059) CONCLUSIONS: This is the first and largest study of DNA methylation in patients with OAC uniformly treated in a randomised phase III trial. We identified an epigenetic signature that may serve as a predictive biomarker for chemotherapy benefit in OAC.
Collapse
Affiliation(s)
- Raghav Sundar
- Department of Haematology-Oncology, National University Health System, Singapore; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Alvin Ng
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore; Centre for Computational Biology, Duke-NUS Medical School, Singapore; NUS Graduate School for Integrative Sciences and Engineering, Singapore
| | - Hermioni Zouridis
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore; Technology Innovation and Delivery Excellence, AstraZeneca, USA
| | - Nisha Padmanabhan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Taotao Sheng
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Shenli Zhang
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Ming Hui Lee
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Wen Fong Ooi
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Aditi Qamra
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Imran Inam
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Lindsay C Hewitt
- Department of Pathology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Jimmy Bok-Yan So
- Department of Surgery, National University Health System, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Vivien Koh
- Department of Haematology-Oncology, National University Health System, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Ruth E Langley
- MRC Clinical Trials Unit at University College London, London, UK
| | | | - David Cunningham
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Steven G Rozen
- Centre for Computational Biology, Duke-NUS Medical School, Singapore
| | - Wei Peng Yong
- Department of Haematology-Oncology, National University Health System, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Heike I Grabsch
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK; Department of Pathology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, the Netherlands.
| | - Patrick Tan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore; Biomedical Research Council, Agency for Science, Technology and Research, Singapore; SingHealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore.
| |
Collapse
|
19
|
Engin AB, Engin A, Gonul II. The effect of adipocyte-macrophage crosstalk in obesity-related breast cancer. J Mol Endocrinol 2019; 62:R201-R222. [PMID: 30620711 DOI: 10.1530/jme-18-0252] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 01/07/2019] [Indexed: 12/11/2022]
Abstract
Adipose tissue is the primary source of many pro-inflammatory cytokines in obesity. Macrophage numbers and pro-inflammatory gene expression are positively associated with adipocyte size. Free fatty acid and tumor necrosis factor-α involve in a vicious cycle between adipocytes and macrophages aggravating inflammatory changes. Thereby, M1 macrophages form a characteristic 'crown-like structure (CLS)' around necrotic adipocytes in obese adipose tissue. In obese women, CLSs of breast adipose tissue are responsible for both increase in local aromatase activity and aggressive behavior of breast cancer cells. Interlinked molecular mechanisms between adipocyte-macrophage-breast cancer cells in obesity involve seven consecutive processes: Excessive release of adipocyte- and macrophage-derived inflammatory cytokines, TSC1-TSC2 complex-mTOR crosstalk, insulin resistance, endoplasmic reticulum (ER) stress and excessive oxidative stress generation, uncoupled respiration and hypoxia, SIRT1 controversy, the increased levels of aromatase activity and estrogen production. Considering elevated risks of estrogen receptor (E2R)-positive postmenopausal breast cancer growth in obesity, adipocyte-macrophage crosstalk is important in the aforementioned issues. Increased mTORC1 signaling in obesity ensures the strong activation of oncogenic signaling in E2Rα-positive breast cancer cells. Since insulin and insulin-like growth factors have been identified as tumor promoters, hyperinsulinemia is an independent risk factor for poor prognosis in breast cancer despite peripheral insulin resistance. The unpredictable effects of adipocyte-derived leptin-estrogen-macrophage axis, and sirtuin 1 (SIRT1)-adipose-resident macrophage axis in obese postmenopausal patients with breast cancer are unresolved mechanistic gaps in the molecular links between the tumor growth and adipocytokines.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Ipek Isik Gonul
- Department of Pathology, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
20
|
Fiannaca A, La Rosa M, La Paglia L, Urso A. miRTissue: a web application for the analysis of miRNA-target interactions in human tissues. BMC Bioinformatics 2018; 19:434. [PMID: 30497361 PMCID: PMC6266954 DOI: 10.1186/s12859-018-2418-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Background microRNAs act as regulators of gene expression interacting with their gene targets. Current bioinformatics services, such as databases of validated miRNA-target interactions and prediction tools, usually provide interactions without any information about what tissue that interaction is more likely to appear nor information about the type of interactions, causing mRNA degradation or translation inhibition respectively. Results In this work, we introduce miRTissue, a web application that combines validated miRNA-target interactions with statistical correlation among expression profiles of miRNAs, genes and proteins in 15 different human tissues. Validated interactions are taken from the miRTarBase database, while expression profiles are downloaded from The Cancer Genome Atlas repository. As a result, the service provides a tissue-specific characterisation of each couple of miRNA and gene together with its statistical significance (p-value). The inclusion of protein data also allows providing the type of interaction. Moreover, miRTissue offers several views for analysing interactions, focusing for example on the comparison between different cancer types or different tissue conditions. All the results are freely downloadable in the most common formats. Conclusions miRTissue fills a gap concerning current bioinformatics services related to miRNA-target interactions because it provides a tissue-specific context to each validated interaction and the type of interaction itself. miRTissue is easily browsable allowing the user to select miRNAs, genes, cancer types and tissue conditions. The results can be sorted according to p-values to immediately identify those interactions that are more likely to occur in a given tissue. miRTissue is available at http://tblab.pa.icar.cnr.it/mirtissue.html. Electronic supplementary material The online version of this article (10.1186/s12859-018-2418-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Antonino Fiannaca
- CNR-ICAR, National Research Council of Italy, Via Ugo La Malfa, 153, Palermo, 90146, Italy.
| | - Massimo La Rosa
- CNR-ICAR, National Research Council of Italy, Via Ugo La Malfa, 153, Palermo, 90146, Italy
| | - Laura La Paglia
- CNR-ICAR, National Research Council of Italy, Via Ugo La Malfa, 153, Palermo, 90146, Italy
| | - Alfonso Urso
- CNR-ICAR, National Research Council of Italy, Via Ugo La Malfa, 153, Palermo, 90146, Italy
| |
Collapse
|
21
|
Sundaramoorthy S, Devanand P, Ryu MS, Song KY, Noh DY, Lim IK. TIS21 /BTG2 inhibits breast cancer growth and progression by differential regulation of mTORc1 and mTORc2-AKT1-NFAT1-PHLPP2 signaling axis. J Cancer Res Clin Oncol 2018; 144:1445-1462. [PMID: 29808317 DOI: 10.1007/s00432-018-2677-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 05/21/2018] [Indexed: 10/25/2022]
Abstract
PURPOSE It has been reported that PI3K/AKT pathway is altered in various cancers and AKT isoforms specifically regulate cell growth and metastasis of cancer cells; AKT1, but not AKT2, reduces invasion of cancer cells but maintains cancer growth. We propose here a novel mechanism of the tumor suppresser, TIS21/BTG2, that inhibits both growth and invasion of triple negative breast cancer cells via AKT1 activation by differential regulation of mTORc1 and mTORc2 activity. METHODS Transduction of adenovirus carrying TIS21/BTG2 gene and transfection of short interfering RNAs were employed to regulate TIS21/BTG2 gene expression in various cell lines. Treatment of mTOR inhibitors and mTOR kinase assays can evaluate the role of mTORc in the regulation of AKT phosphorylation at S473 residue by TIS21/BTG2 in breast cancer cells. Open data and immunohistochemical analysis were performed to confirm the role of TIS21/BTG2 expression in various human breast cancer tissues. RESULTS We observed that TIS21/BTG2 inhibited mTORc1 activity by reducing Raptor-mTOR interaction along with upregulation of tsc1 expression, which lead to significant reduction of p70S6K activation as opposed to AKT1S473, but not AKT2, phosphorylation via downregulating PHLPP2 (AKT1-specific phosphatase) in breast cancers. TIS21/BTG2-induced pAKTS473 required Rictor-bound mTOR kinase, indicating activation of mTORc2 by TIS21/BTG2 gene. Additionally, the TIS21/BTG2-induced pAKTS473 could reduce expression of NFAT1 (nuclear factor of activated T cells) and its target genes, which regulate cancer microenvironment. CONCLUSIONS TIS21/BTG2 significantly lost in the infiltrating ductal carcinoma, but it can inhibit cancer growth via the TIS21/BTG2-tsc1/2-mTORc1-p70S6K axis and downregulate cancer progression via the TIS21/BTG2-mTORc2-AKT1-NFAT1-PHLPP2 pathway.
Collapse
Affiliation(s)
- Santhoshkumar Sundaramoorthy
- Division of Medical Sciences, BK21 Plus program, Graduate School of Ajou University, Suwon, 16499, Republic of Korea
| | - Preethi Devanand
- Division of Medical Sciences, BK21 Plus program, Graduate School of Ajou University, Suwon, 16499, Republic of Korea
| | - Min Sook Ryu
- Division of Medical Sciences, BK21 Plus program, Graduate School of Ajou University, Suwon, 16499, Republic of Korea
| | - Kye Yong Song
- Department of Pathology, Chung-Ang University College of Medicine, Seoul, 156-756, Republic of Korea
| | - Dong Young Noh
- Department of Surgery, Seoul National University, Seoul, 03080, Republic of Korea
| | - In Kyoung Lim
- Division of Medical Sciences, BK21 Plus program, Graduate School of Ajou University, Suwon, 16499, Republic of Korea.
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea.
| |
Collapse
|
22
|
Zohrap N, Saatci Ö, Ozes B, Coban I, Atay HM, Battaloglu E, Şahin Ö, Bugra K. SIK2 attenuates proliferation and survival of breast cancer cells with simultaneous perturbation of MAPK and PI3K/Akt pathways. Oncotarget 2018; 9:21876-21892. [PMID: 29774109 PMCID: PMC5955149 DOI: 10.18632/oncotarget.25082] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 03/09/2018] [Indexed: 12/15/2022] Open
Abstract
Salt Inducible Kinase2 (SIK2) has been shown to contribute to tumorigenesis in multiple tumor types in a dichotomous manner. However, little is known about its contribution to breast malignancies. Here, we report SIK2 as a potential tumor suppressor in breast cancer whose expression was reduced in tumor tissues and breast cancer cell lines compared to normal counterparts. In vitro loss- and gain-of-function experiments combined with xenograft studies demonstrated that SIK2-mediated attenuation of proliferation and survival of breast cancer cells with parallel inhibition of both Ras/Erk and PI3K/Akt pathways. Our findings elucidated that SIK2 has also an inhibitory role in migration/invasion ability of breast cancer cells through regulation of epithelial mesenchymal transition. Immunostaining of patient tumors revealed that SIK2 protein level is frequently downregulated in invasive mammary carcinomas and negatively correlated with the mitotic activity of the cells in triple negative breast cancers and hormone positive tumors. Strikingly, patient survival analysis indicated that higher levels of SIK2 are significantly associated with better survival, especially in basal breast cancer cases. Overall, our findings suggest SIK2 as a potential tumor suppressor in the control of breast tumorigenesis, at least in part, via inhibiting PI3K/Akt and Ras/ERK signaling cascades simultaneously and a novel prognostic marker, especially in basal subtypes of breast cancer.
Collapse
Affiliation(s)
- Neslihan Zohrap
- Department of Molecular Biology and Genetics, Bogazici University, Istanbul, Turkey
| | - Özge Saatci
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Burcak Ozes
- Department of Molecular Biology and Genetics, Bogazici University, Istanbul, Turkey
| | - Ipek Coban
- Department of Pathology, Istanbul Florence-Nightingale Hospital, Istanbul, Turkey
| | - Hasan Murat Atay
- Department of General Surgery, Gayrettepe Florence-Nightingale Hospital, Istanbul, Turkey
| | - Esra Battaloglu
- Department of Molecular Biology and Genetics, Bogazici University, Istanbul, Turkey
| | - Özgür Şahin
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Kuyas Bugra
- Department of Molecular Biology and Genetics, Bogazici University, Istanbul, Turkey.,Life Sciences Center, Bogazici University, Istanbul, Turkey
| |
Collapse
|
23
|
Escudero-Esparza A, Bartoschek M, Gialeli C, Okroj M, Owen S, Jirström K, Orimo A, Jiang WG, Pietras K, Blom AM. Complement inhibitor CSMD1 acts as tumor suppressor in human breast cancer. Oncotarget 2018; 7:76920-76933. [PMID: 27764775 PMCID: PMC5363559 DOI: 10.18632/oncotarget.12729] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/11/2016] [Indexed: 12/02/2022] Open
Abstract
Human CUB and Sushi multiple domains 1 (CSMD1) is a membrane-bound complement inhibitor suggested to act as a putative tumor suppressor gene, since allelic loss of this region encompassing 8p23 including CSMD1 characterizes various malignancies. Here, we assessed the role of CSMD1 as a tumor suppressor gene in the development of breast cancer in vitro and in vivo. We found that human breast tumor tissues expressed CSMD1 at lower levels compared to that in normal mammary tissues. The decreased expression of CSMD1 was linked to a shorter overall survival of breast cancer patients. We also revealed that expression of CSMD1 in human breast cancer cells BT-20 and MDA-MB-231 significantly inhibited their malignant phenotypes, including migration, adhesion and invasion. Conversely, stable silencing of CSMD1 expression in T47D cells enhanced cancer cell migratory, adherent and clonogenic abilities. Moreover, expression of CSMD1 in the highly invasive MDA-MB-231 cells diminished their signaling potential as well as their stem cell-like properties as assessed by measurement of aldehyde dehydrogenase activity. In a xenograft model, expression of CSMD1 blocked the ability of cancer cells to metastasize to secondary sites in vivo, likely via inhibiting local invasion but not the extravasation into distant tissues. Taken together, these findings demonstrate the role of CSMD1 as a tumor suppressor gene in breast cancer.
Collapse
Affiliation(s)
| | | | | | - Marcin Okroj
- Department of Medical Biotechnology, Medical University of Gdańsk, Gdańsk, Poland
| | - Sioned Owen
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff University, Cardiff, UK
| | - Karin Jirström
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Akira Orimo
- Department of Pathology and Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff University, Cardiff, UK
| | - Kristian Pietras
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Anna M Blom
- Department of Translational Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
24
|
Woo SU, Sangai T, Akcakanat A, Chen H, Wei C, Meric-Bernstam F. Vertical inhibition of the PI3K/Akt/mTOR pathway is synergistic in breast cancer. Oncogenesis 2017; 6:e385. [PMID: 28991258 PMCID: PMC5668884 DOI: 10.1038/oncsis.2017.86] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/31/2017] [Accepted: 08/24/2017] [Indexed: 02/07/2023] Open
Abstract
Deregulation and activation of the phosphoinositide 3-kinase (PI3K)/Akt/mammalian (or mechanistic) target of rapamycin (mTOR) pathway have a major role in proliferation and cell survival in breast cancer. However, as single agents, mTOR inhibitors have had modest antitumor efficacy. In this study, we evaluated the effects of vertical inhibition of mTOR and Akt in breast cancer cell lines and xenografts. We assessed the effects of mTOR inhibitor rapamycin and Akt inhibitor MK-2206, given as single drugs or in combination, on cell signaling, cell proliferation and apoptosis in a panel of cancer cell lines in vitro. The antitumor efficacy was tested in vivo. We demonstrated that MK-2206 inhibited Akt phosphorylation, cell proliferation and apoptosis in a dose-dependent manner in breast cancer cell lines. Rapamycin inhibited S6 phosphorylation and cell proliferation, and resulted in lower levels of apoptosis induction. Furthermore, the combination treatment inhibited phosphorylation of Akt and S6, synergistically inhibited proliferation and induced apoptosis with a higher efficacy. In vivo combination inhibited tumor growth more than either agent alone. Our data suggest that a combination of Akt and mTOR inhibitors have greater antitumor activity in breast cancer cells, which may be a viable approach to treat patients.
Collapse
Affiliation(s)
- S-U Woo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - T Sangai
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A Akcakanat
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - H Chen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - C Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - F Meric-Bernstam
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
25
|
miR-130a upregulates mTOR pathway by targeting TSC1 and is transactivated by NF-κB in high-grade serous ovarian carcinoma. Cell Death Differ 2017; 24:2089-2100. [PMID: 28800130 DOI: 10.1038/cdd.2017.129] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 06/23/2017] [Accepted: 07/05/2017] [Indexed: 11/08/2022] Open
Abstract
Activation of mammalian target of rapamycin (mTOR) signaling pathway is associated with poor prognosis of epithelial ovarian cancer. The TSC1-TSC2 complex is a critical negative regulator of mTOR signaling. Here, we demonstrated that TSC1 was frequently downregulated in high-grade serous ovarian carcinoma (HGSOC) and low TSC1 expression level is associated with advanced tumor stage. We next identified miR-130a to be a negative regulator of TSC1 by targeting its 3'UTR. miR-130a was overexpressed in HGSOC and could drive proliferation and invasion/metastasis of ovarian cancer cells. miR-130a could also attenuate rapamycin/starvation-induced autophagy. Ectopic TSC1 expression could block the effects of miR-130a on cell proliferation, migration and autophagy. Finally, we found that miR-130a expression could be upregulated by inflammatory factors and was transactivated by NF-κB. Therefore, our findings establish a crosstalk between inflammation and mTOR signaling that is mediated by miR-130a, which might have a pivotal role in the initiation and progression of HGSOC.
Collapse
|
26
|
[Effect of TSC2 gene expression downregulation by lentivirus induced RNA interference on U937 cell line and its mechanism]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2017; 38:612-617. [PMID: 28810331 PMCID: PMC7342282 DOI: 10.3760/cma.j.issn.0253-2727.2017.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
目的 研究下调TSC2基因表达对白血病U937细胞系的生物学作用及其对mTOR通路活性的影响。 方法 选择TSC2高表达的U937细胞系,通过慢病毒介导的RNA干扰技术下调TSC2基因表达;采用CCK-8比色法、细胞集落形成实验和流式细胞术检测其对细胞增殖、分化和凋亡的影响;采用Western blot法和实时荧光定量PCR(RQ-PCR)法检测TSC2表达下调对mTOR通路蛋白表达及活性的影响。 结果 TSC2基因表达降低能够促进U937细胞的增殖和集落形成(P<0.05);能够使U937细胞G1期[(52.53±3.75)%对(75.10±4.33)%,t=6.829,P=0.002]比例明显降低,G2/M期[(22.43±1.00)%对(15.47±1.20)%,t=−5.581,P=0.019]、S期[(25.03±4.34)%对(14.33±0.91)%,t=−5.413,P=0.013]比例升高;对细胞分化和细胞凋亡没有明显影响(P>0.05)。TSC2基因表达下调后,mTOR活性升高,磷酸化的4EBP1和S6K1蛋白活性升高,而AKT蛋白活性没有明显变化;与细胞增殖相关的基因cyclin D1、c-myc表达升高,PTEN基因表达升高,P27KIP基因和凋亡相关基因BCL-XL的表达没有明显的改变。 结论 TSC2基因表达下调可以通过调节mTOR通路活性促进白血病细胞的增殖。
Collapse
|
27
|
Martin KR, Zhou W, Bowman MJ, Shih J, Au KS, Dittenhafer-Reed KE, Sisson KA, Koeman J, Weisenberger DJ, Cottingham SL, DeRoos ST, Devinsky O, Winn ME, Cherniack AD, Shen H, Northrup H, Krueger DA, MacKeigan JP. The genomic landscape of tuberous sclerosis complex. Nat Commun 2017. [PMID: 28643795 PMCID: PMC5481739 DOI: 10.1038/ncomms15816] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a rare genetic disease causing multisystem growth of benign tumours and other hamartomatous lesions, which leads to diverse and debilitating clinical symptoms. Patients are born with TSC1 or TSC2 mutations, and somatic inactivation of wild-type alleles drives MTOR activation; however, second hits to TSC1/TSC2 are not always observed. Here, we present the genomic landscape of TSC hamartomas. We determine that TSC lesions contain a low somatic mutational burden relative to carcinomas, a subset feature large-scale chromosomal aberrations, and highly conserved molecular signatures for each type exist. Analysis of the molecular signatures coupled with computational approaches reveals unique aspects of cellular heterogeneity and cell origin. Using immune data sets, we identify significant neuroinflammation in TSC-associated brain tumours. Taken together, this molecular catalogue of TSC serves as a resource into the origin of these hamartomas and provides a framework that unifies genomic and transcriptomic dimensions for complex tumours.
Collapse
Affiliation(s)
- Katie R Martin
- Center for Cancer and Cell Biology, Van Andel Research Institute, 333 Bostwick Avenue NE, Grand Rapids, Michigan 49503, USA
| | - Wanding Zhou
- Center for Epigenetics, Van Andel Research Institute, 333 Bostwick Avenue NE, Grand Rapids, Michigan 49503, USA
| | - Megan J Bowman
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, 333 Bostwick Avenue NE, Grand Rapids, Michigan 49503, USA
| | - Juliann Shih
- Cancer Program, Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Kit Sing Au
- Department of Pediatrics, University of Texas Health Science Center at Houston-McGovern Medical School, 6431 Fannin, Houston, Texas 77030, USA
| | - Kristin E Dittenhafer-Reed
- Center for Cancer and Cell Biology, Van Andel Research Institute, 333 Bostwick Avenue NE, Grand Rapids, Michigan 49503, USA
| | - Kellie A Sisson
- Center for Cancer and Cell Biology, Van Andel Research Institute, 333 Bostwick Avenue NE, Grand Rapids, Michigan 49503, USA
| | - Julie Koeman
- Cytogenetics and Pathology Core, Van Andel Research Institute, 333 Bostwick Avenue NE, Grand Rapids, Michigan 49503, USA
| | - Daniel J Weisenberger
- Norris Comprehensive Cancer Center, University of Southern California, 1450 Biggy Street, Los Angeles, California 90033, USA
| | - Sandra L Cottingham
- Department of Pathology, Spectrum Health System, 100 Michigan Street NE, Grand Rapids, Michigan 49503, USA
| | - Steven T DeRoos
- Division of Pediatric Neurology, Helen DeVos Children's Hospital, Spectrum Health System, 100 Michigan Street NE, Grand Rapids, Michigan 49503, USA
| | - Orrin Devinsky
- Department of Neurology, New York University School of Medicine, 223 E 34 Street, New York, New York 10016, USA
| | - Mary E Winn
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, 333 Bostwick Avenue NE, Grand Rapids, Michigan 49503, USA
| | - Andrew D Cherniack
- Cancer Program, Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, Massachusetts 02142, USA
| | - Hui Shen
- Center for Epigenetics, Van Andel Research Institute, 333 Bostwick Avenue NE, Grand Rapids, Michigan 49503, USA
| | - Hope Northrup
- Department of Pediatrics, University of Texas Health Science Center at Houston-McGovern Medical School, 6431 Fannin, Houston, Texas 77030, USA
| | - Darcy A Krueger
- Division of Neurology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229, USA
| | - Jeffrey P MacKeigan
- Center for Cancer and Cell Biology, Van Andel Research Institute, 333 Bostwick Avenue NE, Grand Rapids, Michigan 49503, USA.,College of Human Medicine, Michigan State University, 220 Trowbridge Road, East Lansing, Michigan 48824, USA
| |
Collapse
|
28
|
Abstract
Malignancy is the second most common single cause of death observed in organ transplant recipients. The excess cancer risk is related to intensity and duration of immunosuppressive therapy and inversely to recipient age. Immunodeficiency and (chronic/oncogenic) viral infections together constitute a major risk. Nonmelanoma skin cancer, Kaposi sarcoma, and posttransplant lymphoproliferative disease have standardized incidence ratios exceeding 10- or 50-fold. The mammalian target of rapamycin (mTOR) inhibitors, sirolimus and everolimus, are increasingly used after organ transplantation with potential advantages in virus-associated posttransplant malignancies as well as anti-cancer properties. Despite a seemingly clear mechanism of action and solid rationale for their use in cancer therapy, mTORis have met only modest success rates in clinical trials with advanced malignancies except for specific tumors, such as Kaposi sarcoma and mantle cell lymphoma. Because mTORis are primarily cytostatic, not cytotoxic, the observed clinical efficacy is a reflection of disease stabilization rather than tumor regression. Nonmelanoma skin cancers, in particular cutaneous squamous cell carcinoma, have the highest standardized incidence ratios in transplant recipients. Recent meta-analyses and randomized trials on secondary prevention of squamous cell carcinoma observed a reduction in cumulative tumor load, suggesting most benefit to be gained by early conversion to an mTOR inhibitor-based maintenance regime. There is ongoing debate on the mechanisms involved including withdrawal of the carcinogenic effects of calcineurin inhibitors and/or their impact on chronic (oncogenic) viral infections. At present, there is, however, insufficient evidence for the primary use of mTORis as protective agents against most other cancer types.
Collapse
|
29
|
Wang Y, Chen C, Deng Z, Bian E, Huang C, Lei T, Lv X, Liu L, Li J. Repression of TSC1/TSC2 mediated by MeCP2 regulates human embryo lung fibroblast cell differentiation and proliferation. Int J Biol Macromol 2016; 96:578-588. [PMID: 28041914 DOI: 10.1016/j.ijbiomac.2016.12.062] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 12/22/2016] [Accepted: 12/22/2016] [Indexed: 12/12/2022]
Abstract
Pulmonary fibrosis (PF) is a severe inflammatory disease with limited effective treatments. It is known that the transdifferentiation of human embryo lung fibroblast (HELF) cells from pulmonary fibroblasts into myofibroblasts, contributes to the progression of pulmonary fibrogenesis. The tuberous sclerosis proteins TSC1 and TSC2 are two key signaling factors which can suppress cell growth and proliferation. However, the roles of TSC1 and TSC2 in lung fibroblast are unclear. Here, we developed a PF model with bleomycin (BLM) in mice and conducted several simulation experiments in HELF cells. Our study shows that the expression of TSC1 and TSC2 in fibrotic mice lung was reduced and stimulation of HELF cells with TGF-β1 resulted in a down-regulation of TSC1 and TSC2. In addition, overexpression of TSC1 or TSC2 decreased cell proliferation and differentiation. Furthermore, we found that reduced expression of TSC1 and TSC2 caused by TGF-β1 is associated with the promoter methylation status of TSC1 and TSC2. MeCP2, controls an epigenetic pathway that promotes myofibroblast transdifferentiation and fibrosis. We found that expression of TSC1 and TSC2 can be repressed by MeCP2, which regulates HELF cell differentiation and proliferation as myofibroblasts and lead to PF ultimately.
Collapse
Affiliation(s)
- Yuanyuan Wang
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Department of Pharmacology, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, Anhui, China
| | - Chen Chen
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Hefei Binghu Hospital, 3200 Changsha Road, Hefei 230000, Anhui, China
| | - Ziyu Deng
- Department of Scientific and Educational, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, Anhui, China
| | - Erbao Bian
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China
| | - Cheng Huang
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China
| | - Ting Lei
- Department of Pharmacology, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, Anhui, China
| | - Xiongwen Lv
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China
| | - Liping Liu
- Department of Pharmacology, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, Anhui, China
| | - Jun Li
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
30
|
Cho J, Lee J, Kim J, Kim ST, Lee S, Kim SY, Ha SY, Park CK, Lim HY. Loss of Tuberous Sclerosis Complex 2 (TSC2) as a Predictive Biomarker of Response to mTOR Inhibitor Treatment in Patients with Hepatocellular Carcinoma. Transl Oncol 2016; 9:466-471. [PMID: 27751352 PMCID: PMC5067924 DOI: 10.1016/j.tranon.2016.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/22/2016] [Accepted: 08/24/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND: Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death globally. Mechanistic target of rapamycin (mTOR) is frequently up-regulated in HCC and plays an important role in HCC tumorigenesis. Tumors with loss of tuberous sclerosis complex 2 (TSC2), a negative regulator of mTOR signaling, tend to respond well to mTOR inhibitors. We analyzed TSC2 expression status in Korean patients with HCC and evaluated the correlation between TSC2 loss and response to the mTOR inhibitor, everolimus. METHODS: We retrospectively assessed 36 patients with advanced HCC who had received sorafenib at a single center in Korea between 2008 and 2014, and for whom tumor specimens were available for TSC2 immunohistochemical analysis (IHC). Three patient-derived tumor cell lines (PDCs) were analyzed by western blotting to determine TSC2 expression and drug sensitivity to mTOR. RESULTS: Twelve of 36 patients (33.3%) showed low to undetectable levels of TSC2 expression. No significant differences were observed in progression-free survival (PFS) or overall survival with sorafenib treatment based on TSC2 expression status. Two patients were treated with everolimus after sorafenib failure; one patient, with moderate TSC2 expression, experienced stable disease with a PFS of 5.8 months; the other, with high TSC2 expression, experienced rapid progression. PDC models demonstrated that the TSC2-low HCC PDC line was significantly more sensitive to everolimus than the TSC2-high HCC PDC lines. CONCLUSION: Loss of TSC2 may predict improved response to everolimus in HCC patients, but further studies are needed to confirm the predictive role of TSC2 expression for everolimus treatment.
Collapse
Affiliation(s)
- Jinhyun Cho
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jusun Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung Tae Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sujin Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sun Young Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sang Yun Ha
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Cheol-Keun Park
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Ho Yeong Lim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
31
|
Nuñez O, Román A, Johnson SR, Inoue Y, Hirose M, Casanova Á, de Garibay GR, Herranz C, Bueno-Moreno G, Boni J, Mateo F, Petit A, Climent F, Soler T, Vidal A, Sánchez-Mut JV, Esteller M, López JI, García N, Gumà A, Ortega R, Plà MJ, Campos M, Ansótegui E, Molina-Molina M, Valenzuela C, Ussetti P, Laporta R, Ancochea J, Xaubet A, Pollán M, Pujana MA. Study of breast cancer incidence in patients of lymphangioleiomyomatosis. Breast Cancer Res Treat 2016; 156:195-201. [PMID: 26951504 PMCID: PMC4788694 DOI: 10.1007/s10549-016-3737-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/27/2016] [Indexed: 11/30/2022]
Abstract
Molecular evidence has linked the pathophysiology of lymphangioleiomyomatosis (LAM) to that of metastatic breast cancer. Following on this observation, we assessed the association between LAM and subsequent breast cancer. An epidemiological study was carried out using three LAM country cohorts, from Japan, Spain, and the United Kingdom. The number of incident breast cancer cases observed in these cohorts was compared with the number expected on the basis of the country-specific incidence rates for the period 2000–2014. Immunohistochemical studies and exome sequence analysis were performed in two and one tumors, respectively. All cohorts revealed breast cancer standardized incidence ratios (SIRs) ≥ 2.25. The combined analysis of all cases or restricted to pre-menopausal age groups revealed significantly higher incidence of breast cancer: SIR = 2.81, 95 % confidence interval (CI) = 1.32–5.57, P = 0.009; and SIR = 4.88, 95 % CI = 2.29–9.99, P = 0.0007, respectively. Immunohistochemical analyses showed positivity for known markers of lung metastatic potential. This study suggests the existence of increased breast cancer risk among LAM patients. Prospective studies may be warranted to corroborate this result, which may be particularly relevant for pre-menopausal women with LAM.
Collapse
Affiliation(s)
- Olivier Nuñez
- Cancer and Environmental Epidemiology Unit, National Center for Epidemiology, Carlos III Institute of Health, and Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Sinesio Delgado 6, 28029, Madrid, Spain
| | - Antonio Román
- Lung Transplant Unit, Department of Pulmonology, Lymphangioleiomyomatosis Clinic, Vall d'Hebron University Hospital, 08035, Barcelona, Catalonia, Spain
| | - Simon R Johnson
- National Centre for Lymphangioleiomyomatosis, Nottingham University Hospitals NHS Trust, Nottingham, Nottinghamshire, UK Division of Respiratory Medicine and Respiratory Research Unit, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Yoshikazu Inoue
- National Hospital Organization Kinki-Chuo Chest Medical Center, Sakai, 591-8555, Osaka, Japan
| | - Masaki Hirose
- National Hospital Organization Kinki-Chuo Chest Medical Center, Sakai, 591-8555, Osaka, Japan
| | - Álvaro Casanova
- Department of Pneumology, Henares Hospital, 28882, Madrid, Spain
| | - Gorka Ruiz de Garibay
- ProCURE, Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
| | - Carmen Herranz
- ProCURE, Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
| | - Gema Bueno-Moreno
- Department of Biochemistry, Autonomous University of Madrid (UAM), Biomedical Research Institute "Alberto Sols" (Spanish National Research Council (CSIC)-UAM), Hospital La Paz Institute for Health Research (IdiPAZ), 28029, Madrid, Spain
- MD Anderson International Foundation, 28033, Madrid, Spain
| | - Jacopo Boni
- ProCURE, Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
| | - Francesca Mateo
- ProCURE, Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
| | - Anna Petit
- Department of Pathology, University Hospital of Bellvitge, IDIBELL, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
| | - Fina Climent
- Department of Pathology, University Hospital of Bellvitge, IDIBELL, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
| | - Teresa Soler
- Department of Pathology, University Hospital of Bellvitge, IDIBELL, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
| | - August Vidal
- Department of Pathology, University Hospital of Bellvitge, IDIBELL, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
| | - José Vicente Sánchez-Mut
- Cancer Epigenetics and Biology Program, IDIBELL, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
| | - Manel Esteller
- Cancer Epigenetics and Biology Program, IDIBELL, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
- Department of Physiological Sciences II, School of Medicine, University of Barcelona, 08908, Barcelona, Catalonia, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010, Barcelona, Catalonia, Spain
| | - José Ignacio López
- Cruces University Hospital, BioCruces Research Institute, University of the Basque Country, 48903, Barakaldo, Spain
| | - Nadia García
- ProCURE, Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
| | - Anna Gumà
- Department of Radiology, University Hospital of Bellvitge, IDIBELL, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
| | - Raúl Ortega
- Department of Radiology, University Hospital of Bellvitge, IDIBELL, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
| | - María Jesús Plà
- Breast Cancer Functional Unit, Department of Gynecology, University Hospital of Bellvitge, ICO, IDIBELL, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
| | - Miriam Campos
- Breast Cancer Functional Unit, Department of Medical Oncology, ICO, IDIBELL, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
| | - Emilio Ansótegui
- Lung Transplant and Cystic Fibrosis Unit, Hospital Universitario y Politecnico La Fe, 46026, Valencia, Spain
| | - María Molina-Molina
- Department of Pneumology, University Hospital of Bellvitge, IDIBELL, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain
- Consortium for Biomedical Research in Respiratory Diseases (CIBERES), 28029, Madrid, Spain
| | - Claudia Valenzuela
- Department of Pneumology, Instituto de Investigación Sanitaria La Princesa, Hospital La Princesa, 28006, Madrid, Spain
| | - Piedad Ussetti
- Department of Pneumology, University Hospital Clínica Puerta del Hierro, 28222, Madrid, Spain
| | - Rosalía Laporta
- Department of Pneumology, University Hospital Clínica Puerta del Hierro, 28222, Madrid, Spain
| | - Julio Ancochea
- Department of Pneumology, Instituto de Investigación Sanitaria La Princesa, Hospital La Princesa, 28006, Madrid, Spain
| | - Antoni Xaubet
- Consortium for Biomedical Research in Respiratory Diseases (CIBERES), 28029, Madrid, Spain
- Department of Pneumology, Hospital Clinic of Barcelona, August Pi Suñer Biomedical Research Institute (IDIBAPS), 08036, Barcelona, Catalonia, Spain
| | - Marina Pollán
- Cancer and Environmental Epidemiology Unit, National Center for Epidemiology, Carlos III Institute of Health, and Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Sinesio Delgado 6, 28029, Madrid, Spain.
| | - Miguel Angel Pujana
- ProCURE, Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, 08908, Barcelona, Catalonia, Spain.
| |
Collapse
|
32
|
Zhang M, Zhang S, Wen Y, Wang Y, Wei Y, Liu H, Zhang D, Su J, Wang F, Zhang Y. DNA Methylation Patterns Can Estimate Nonequivalent Outcomes of Breast Cancer with the Same Receptor Subtypes. PLoS One 2015; 10:e0142279. [PMID: 26550991 PMCID: PMC4638352 DOI: 10.1371/journal.pone.0142279] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/20/2015] [Indexed: 01/11/2023] Open
Abstract
Breast cancer has various molecular subtypes and displays high heterogeneity. Aberrant DNA methylation is involved in tumor origin, development and progression. Moreover, distinct DNA methylation patterns are associated with specific breast cancer subtypes. We explored DNA methylation patterns in association with gene expression to assess their impact on the prognosis of breast cancer based on Infinium 450K arrays (training set) from The Cancer Genome Atlas (TCGA). The DNA methylation patterns of 12 featured genes that had a high correlation with gene expression were identified through univariate and multivariable Cox proportional hazards models and used to define the methylation risk score (MRS). An improved ability to distinguish the power of the DNA methylation pattern from the 12 featured genes (p = 0.00103) was observed compared with the average methylation levels (p = 0.956) or gene expression (p = 0.909). Furthermore, MRS provided a good prognostic value for breast cancers even when the patients had the same receptor status. We found that ER-, PR- or Her2- samples with high-MRS had the worst 5-year survival rate and overall survival time. An independent test set including 28 patients with death as an outcome was used to test the validity of the MRS of the 12 featured genes; this analysis obtained a prognostic value equivalent to the training set. The predict power was validated through two independent datasets from the GEO database. The DNA methylation pattern is a powerful predictor of breast cancer survival, and can predict outcomes of the same breast cancer molecular subtypes.
Collapse
Affiliation(s)
- Min Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Shaojun Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yanhua Wen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yihan Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yanjun Wei
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Hongbo Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Dongwei Zhang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Jianzhong Su
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Fang Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
33
|
Englund E, Reitsma B, King BC, Escudero-Esparza A, Owen S, Orimo A, Okroj M, Anagnostaki L, Jiang WG, Jirström K, Blom AM. The human complement inhibitor Sushi Domain-Containing Protein 4 (SUSD4) expression in tumor cells and infiltrating T cells is associated with better prognosis of breast cancer patients. BMC Cancer 2015; 15:737. [PMID: 26480818 PMCID: PMC4615997 DOI: 10.1186/s12885-015-1734-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 10/08/2015] [Indexed: 01/22/2023] Open
Abstract
Background The human Sushi Domain-Containing Protein 4 (SUSD4) was recently shown to function as a novel inhibitor of the complement system, but its role in tumor progression is unknown. Methods Using immunohistochemistry and quantitative PCR, we investigated SUSD4 expression in breast cancer tissue samples from two cohorts. The effect of SUSD4 expression on cell migration and invasion was studied in vitro using two human breast cancer cell lines overexpressing SUSD4. Results Tissue stainings revealed that both tumor cells and tumor-infiltrating cells expressed SUSD4. The highest SUSD4 expression was detected in differentiated tumors with decreased rate of metastasis, and SUSD4 expression was associated with improved survival of the patients. Moreover, forced SUSD4 expression in human breast cancer cells attenuated their migratory and invasive traits in culture. SUSD4 expression also inhibited colony formation of human breast cancer cells cultured on carcinoma-associated fibroblasts. Furthermore, large numbers of SUSD4-expressing T cells in the tumor stroma associated with better overall survival of the breast cancer patients. Conclusion Our findings indicate that SUSD4 expression in both breast cancer cells and T cells infiltrating the tumor-associated stroma is useful to predict better prognosis of breast cancer patients.
Collapse
Affiliation(s)
- Emelie Englund
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| | - Bart Reitsma
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| | - Ben C King
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| | - Astrid Escudero-Esparza
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| | - Sioned Owen
- Cardiff's China Medical Research Collaborative (CCMRC), Cardiff University School of Medicine, Cardiff University, Cardiff, UK.
| | - Akira Orimo
- Department of Pathology and Oncology, Juntendo University School of Medicine, Tokyo, Japan.
| | - Marcin Okroj
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden. .,Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology UG‑MUG, Medical University of Gdańsk, 80210, Gdańsk, Poland.
| | - Lola Anagnostaki
- Department of Clinical Pathology, Skåne University Hospital, Malmö, Sweden.
| | - Wen G Jiang
- Cardiff's China Medical Research Collaborative (CCMRC), Cardiff University School of Medicine, Cardiff University, Cardiff, UK.
| | - Karin Jirström
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden.
| | - Anna M Blom
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| |
Collapse
|
34
|
Ruiz de Garibay G, Herranz C, Llorente A, Boni J, Serra-Musach J, Mateo F, Aguilar H, Gómez-Baldó L, Petit A, Vidal A, Climent F, Hernández-Losa J, Cordero Á, González-Suárez E, Sánchez-Mut JV, Esteller M, Llatjós R, Varela M, López JI, García N, Extremera AI, Gumà A, Ortega R, Plà MJ, Fernández A, Pernas S, Falo C, Morilla I, Campos M, Gil M, Román A, Molina-Molina M, Ussetti P, Laporta R, Valenzuela C, Ancochea J, Xaubet A, Casanova Á, Pujana MA. Lymphangioleiomyomatosis Biomarkers Linked to Lung Metastatic Potential and Cell Stemness. PLoS One 2015; 10:e0132546. [PMID: 26167915 PMCID: PMC4500593 DOI: 10.1371/journal.pone.0132546] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 06/17/2015] [Indexed: 12/23/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a rare lung-metastasizing neoplasm caused by the proliferation of smooth muscle-like cells that commonly carry loss-of-function mutations in either the tuberous sclerosis complex 1 or 2 (TSC1 or TSC2) genes. While allosteric inhibition of the mechanistic target of rapamycin (mTOR) has shown substantial clinical benefit, complementary therapies are required to improve response and/or to treat specific patients. However, there is a lack of LAM biomarkers that could potentially be used to monitor the disease and to develop other targeted therapies. We hypothesized that the mediators of cancer metastasis to lung, particularly in breast cancer, also play a relevant role in LAM. Analyses across independent breast cancer datasets revealed associations between low TSC1/2 expression, altered mTOR complex 1 (mTORC1) pathway signaling, and metastasis to lung. Subsequently, immunohistochemical analyses of 23 LAM lesions revealed positivity in all cases for the lung metastasis mediators fascin 1 (FSCN1) and inhibitor of DNA binding 1 (ID1). Moreover, assessment of breast cancer stem or luminal progenitor cell biomarkers showed positivity in most LAM tissue for the aldehyde dehydrogenase 1 (ALDH1), integrin-ß3 (ITGB3/CD61), and/or the sex-determining region Y-box 9 (SOX9) proteins. The immunohistochemical analyses also provided evidence of heterogeneity between and within LAM cases. The analysis of Tsc2-deficient cells revealed relative over-expression of FSCN1 and ID1; however, Tsc2-deficient cells did not show higher sensitivity to ID1-based cancer inhibitors. Collectively, the results of this study reveal novel LAM biomarkers linked to breast cancer metastasis to lung and to cell stemness, which in turn might guide the assessment of additional or complementary therapeutic opportunities for LAM.
Collapse
Affiliation(s)
- Gorka Ruiz de Garibay
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Carmen Herranz
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Alicia Llorente
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Jacopo Boni
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Jordi Serra-Musach
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Francesca Mateo
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Helena Aguilar
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Laia Gómez-Baldó
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Anna Petit
- Department of Pathology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - August Vidal
- Department of Pathology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Fina Climent
- Department of Pathology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | | | - Álex Cordero
- Cancer Epigenetics and Biology Program, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Eva González-Suárez
- Cancer Epigenetics and Biology Program, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - José Vicente Sánchez-Mut
- Cancer Epigenetics and Biology Program, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Manel Esteller
- Cancer Epigenetics and Biology Program, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
- Department of Physiological Sciences II, School of Medicine, University of Barcelona, Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
| | - Roger Llatjós
- Department of Pathology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Mar Varela
- Department of Pathology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - José Ignacio López
- Cruces University Hospital, BioCruces Research Institute, University of the Basque Country, Barakaldo, Spain
| | - Nadia García
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Ana I. Extremera
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Anna Gumà
- Department of Radiology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Raúl Ortega
- Department of Radiology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - María Jesús Plà
- Department of Gynecology, University Hospital of Bellvitge, Breast Cancer Unit, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Adela Fernández
- Department of Medical Oncology, Breast Cancer Unit, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Sònia Pernas
- Department of Medical Oncology, Breast Cancer Unit, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Catalina Falo
- Department of Medical Oncology, Breast Cancer Unit, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Idoia Morilla
- Department of Medical Oncology, Breast Cancer Unit, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Miriam Campos
- Department of Medical Oncology, Breast Cancer Unit, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Miguel Gil
- Department of Medical Oncology, Breast Cancer Unit, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
| | - Antonio Román
- Department of Pulmonology, Lung Transplant Unit, Lymphangioleiomyomatosis (LAM) Clinic, Vall d'Hebron University Hospital, Barcelona, Catalonia, Spain
| | - María Molina-Molina
- Department of Pneumology, University Hospital of Bellvitge, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
- Biomedical Research Centre Network for Respiratory Diseases (CIBERES), Madrid, Spain
| | - Piedad Ussetti
- Department of Pneumology, University Hospital Clínica Puerta del Hierro, Madrid, Spain
| | - Rosalía Laporta
- Department of Pneumology, University Hospital Clínica Puerta del Hierro, Madrid, Spain
| | - Claudia Valenzuela
- Department of Pneumology, Instituto de Investigación Sanitaria La Princesa, Hospital La Princesa, Madrid, Spain
| | - Julio Ancochea
- Department of Pneumology, Instituto de Investigación Sanitaria La Princesa, Hospital La Princesa, Madrid, Spain
| | - Antoni Xaubet
- Biomedical Research Centre Network for Respiratory Diseases (CIBERES), Madrid, Spain
- Department of Pneumology, Hospital Clinic of Barcelona, Agusti Pi Suñer Biomedical Research Institute (IDIBAPS), Barcelona, Catalonia, Spain
| | | | - Miguel Angel Pujana
- Program Against Cancer Therapeutic Resistance (ProCURE), Breast Cancer and Systems Biology, Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Catalonia, Spain
- * E-mail:
| |
Collapse
|
35
|
Huynh H, Hao HX, Chan SL, Chen D, Ong R, Soo KC, Pochanard P, Yang D, Ruddy D, Liu M, Derti A, Balak MN, Palmer MR, Wang Y, Lee BH, Sellami D, Zhu AX, Schlegel R, Huang A. Loss of Tuberous Sclerosis Complex 2 (TSC2) Is Frequent in Hepatocellular Carcinoma and Predicts Response to mTORC1 Inhibitor Everolimus. Mol Cancer Ther 2015; 14:1224-35. [PMID: 25724664 DOI: 10.1158/1535-7163.mct-14-0768] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 02/17/2015] [Indexed: 11/16/2022]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer deaths worldwide and hyperactivation of mTOR signaling plays a pivotal role in HCC tumorigenesis. Tuberous sclerosis complex (TSC), a heterodimer of TSC1 and TSC2, functions as a negative regulator of mTOR signaling. In the current study, we discovered that TSC2 loss-of-function is common in HCC. TSC2 loss was found in 4 of 8 HCC cell lines and 8 of 28 (28.6%) patient-derived HCC xenografts. TSC2 mutations and deletions are likely to be the underlying cause of TSC2 loss in HCC cell lines, xenografts, and primary tumors for most cases. We further demonstrated that TSC2-null HCC cell lines and xenografts had elevated mTOR signaling and, more importantly, were significantly more sensitive to RAD001/everolimus, an mTORC1 inhibitor. These preclinical findings led to the analysis of TSC2 status in HCC samples collected in the EVOLVE-1 clinical trial of everolimus using an optimized immunohistochemistry assay and identified 15 of 139 (10.8%) samples with low to undetectable levels of TSC2. Although the sample size is too small for formal statistical analysis, TSC2-null/low tumor patients who received everolimus tended to have longer overall survival than those who received placebo. Finally, we performed an epidemiology survey of more than 239 Asian HCC tumors and found the frequency of TSC2 loss to be approximately 20% in Asian HBV(+) HCC. Taken together, our data strongly argue that TSC2 loss is a predictive biomarker for the response to everolimus in HCC patients.
Collapse
Affiliation(s)
- Hung Huynh
- Laboratory of Molecular Endocrinology, Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore.
| | - Huai-Xiang Hao
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Stephen L Chan
- State Key Laboratory in Oncology in South China, Department of Clinical Oncology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, PR China
| | - David Chen
- Oncology Global Development, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | - Richard Ong
- Laboratory of Molecular Endocrinology, Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore
| | - Khee Chee Soo
- Laboratory of Molecular Endocrinology, Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore
| | - Panisa Pochanard
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - David Yang
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - David Ruddy
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Manway Liu
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Adnan Derti
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Marissa N Balak
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Michael R Palmer
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Yan Wang
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Benjamin H Lee
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Dalila Sellami
- Oncology Global Development, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | - Andrew X Zhu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Robert Schlegel
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Alan Huang
- Oncology Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts.
| |
Collapse
|
36
|
Huang KT, Huang YH, Li P, He B, Chen ZK, Yu X, Chen JO, Zhang QY, Shi HQ, Shan YF. Correlation between tuberous sclerosis complex 2 and glycogen synthase kinase 3 beta levels, and outcomes of patients with hepatocellular carcinoma treated by hepatectomy. Hepatol Res 2014; 44:1142-50. [PMID: 24119083 DOI: 10.1111/hepr.12256] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 08/16/2013] [Accepted: 09/30/2013] [Indexed: 12/13/2022]
Abstract
AIM Tuberous sclerosis complex 2 (TSC2), a tumor suppressor, may play an essential role in the regulation of cell growth and cell survival under energy stress conditions. In addition, TSC2 may act in concert with Wnt and energy signals by additional phosphorylation of glycogen synthase kinase 3β (GSK3β) to regulate cell growth. The expression levels and function of TSC2 and GSK3β in hepatocellular carcinoma (HCC) remain unclear. METHODS The protein levels of TSC2 and GSK3β were measured by immunohistochemistry in normal liver (n = 20), HCC (n = 80) and pericancerous tissues (n = 80). The correlations between TSC2, and GSK3β levels, clinicopathological features and patient survival were also analyzed. RESULTS The protein levels of TSC2 and GSK3β in HCC tissues were significantly lower than that in normal liver tissues and pericancerous tissues (P < 0.05). Decreased TSC2 and GSK3β expression was found to be significantly correlated with advanced clinicopathological characteristics and poor prognosis. The results also showed that TSC2 protein levels were associated with GSK3β expression in HCC specimens. CONCLUSION This is the first demonstration that the decreases in TSC2 and GSK3β levels may be associated with vascular invasion, histological grade and tumor-node-metastasis classification.
Collapse
Affiliation(s)
- Ka-Te Huang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Lindqvist BM, Wingren S, Motlagh PB, Nilsson TK. Whole genome DNA methylation signature of HER2-positive breast cancer. Epigenetics 2014; 9:1149-62. [PMID: 25089541 DOI: 10.4161/epi.29632] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In order to obtain a comprehensive DNA methylation signature of HER2-positive breast cancer (HER2+ breast cancer), we performed a genome-wide methylation analysis on 17 HER2+ breast cancer and compared with ten normal breast tissue samples using the Illumina Infinium HumanMethylation450 BeadChip (450K). In HER2+ breast cancer, we found altered DNA methylation in genes involved in multicellular development, differentiation and transcription. Within these genes, we observed an overrepresentation of homeobox family genes, including several genes that have not been previously reported in relation to cancer (DBX1, NKX2-6, SIX6). Other affected genes included several belonging to the PI3K and Wnt signaling pathways. Notably, HER2, AKT3, HK1, and PFKP, genes for which altered methylation has not been previously reported, were also identified in this analysis. In total, we report 69 candidate biomarker genes with maximum differential methylation in HER2+ breast cancer. External validation of gene expression in a selected group of these genes (n = 13) revealed lowered mean gene expression in HER2+ breast cancer. We analyzed DNA methylation in six top candidate genes (AKR1B1, INA, FOXC2, NEUROD1, CDKL2, IRF4) using EpiTect Methyl II Custom PCR Array and confirmed the 450K array findings. Future clinical studies focusing on these genes, as well as on homeobox-containing genes and HER2, AKT3, HK1, and PFKP, are warranted which could provide further insights into the biology of HER2+ breast cancer.
Collapse
Affiliation(s)
- Breezy M Lindqvist
- School of Health and Medical Sciences; Örebro University; Örebro, Sweden
| | - Sten Wingren
- School of Health and Medical Sciences; Örebro University; Örebro, Sweden
| | - Parviz B Motlagh
- Department of Medical Biosciences/Clinical Chemistry; Umeå University; Umeå, Sweden
| | - Torbjörn K Nilsson
- Department of Medical Biosciences/Clinical Chemistry; Umeå University; Umeå, Sweden
| |
Collapse
|
38
|
Abstract
Neuroblastoma (NB) is the most common extracranial malignant solid tumors of childhood, and the majority of these high-risk tumors is resistant to nearly all the treatments and has a significantly worse outcome. The mammalian target of rapamycin (mTOR) plays a critical role in oncogenesis and cancer progression of many tumors. This review will describe the function of mTOR, its genetic regulation in pediatric neuroblastoma, and its value as a target for inhibition by anticancer agents for patients with NB.
Collapse
Affiliation(s)
- Hong Mei
- 1Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | | | | | | |
Collapse
|
39
|
Dressler AC, Hudelist G, Fink-Retter A, Gschwantler-Kaulich D, Pfeiler G, Rosner M, Hengstschläger M, Singer CF. Tuberin and p27 expression in breast cancer patients with or without BRCA germline mutations. J Cancer Res Clin Oncol 2013; 139:1349-55. [PMID: 23689538 DOI: 10.1007/s00432-013-1443-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 04/16/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND Tuberin, the protein product of tuberous sclerosis gene 2 (TSC2), is the functional component of the TSC1/TSC2 complex and regulates cell cycle through activation of the cyclin-dependent kinase inhibitor p27. The transcriptional regulation of p27 is, however, also linked to a functional BRCA protein, since in BRCA1 mutant breast cancer cells, which lack the ability to repair DNA damages by homologous recombination, p27 is down-regulated. We have therefore investigated the expression of both tuberin and p27 in normal breast tissue, and in malignant epithelium from women with and without a BRCA mutation. MATERIALS AND METHODS immunohistochemistry was used to compare p27 and tuberin protein expression in 26 BRCA1 and 2 mutation carriers, in 53 matched breast cancer patients without a family history, and in 74 benign breast tissues in a case-control study. RESULTS Tuberin and p27 protein expression were significantly more common in benign when compared to malignant breast tissue (p = 0.01 and p = 0.03), but no difference was observed when sporadic and BRCA-mutated breast cancer specimen were compared. Tuberin and p27 were positively correlated with each other (p = 0.0017, r = 0.2527). Furthermore, p27 expression was positively correlated with ER and PR, and negatively correlated with tumor size. The expression of tuberin and p27 in breast cancer was not correlated with clinical outcome. CONCLUSION Our results suggest that tuberin and p27 are aberrantly expressed in malignant tissue, but their expression does not appear to be dependent on the BRCA mutation state of a breast cancer patient.
Collapse
Affiliation(s)
- Anne Catharina Dressler
- Clinical Division of Gynaecology and Gynaecological Oncology, Department of Obstretrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Lesma E, Ancona S, Orpianesi E, Grande V, Di Giulio AM, Gorio A. Chromatin remodeling by rosuvastatin normalizes TSC2-/meth cell phenotype through the expression of tuberin. J Pharmacol Exp Ther 2013; 345:180-8. [PMID: 23426956 DOI: 10.1124/jpet.113.203141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a multi-systemic syndrome caused by mutations in TSC1 or TSC2 gene. In TSC2-null cells, Rheb, a member of the Ras family of GTPases, is constitutively activated. Statins inhibit 3-hydroxy-3-methylglutaryl coenzyme A reductase and block the synthesis of isoprenoid lipids with inhibition of Rheb farnesylation and RhoA geranylgeranylation. The effects of rosuvastatin on the function of human TSC2(-/-) and TSC2(-/meth) α-actin smooth muscle (ASM) cells have been investigated. The TSC2(-/-) and TSC2(-/meth) ASM cells, previously isolated in our laboratory from the renal angiomyolipoma of two TSC patients, do not express tuberin and bear loss of heterozigosity caused by a double hit on TSC2 and methylation of TSC2 promoter, respectively. Exposure to rosuvastatin affected TSC2(-/meth) ASM cell growth and promoted tuberin expression by acting as a demethylating agent. This occurred without changes in interleukin release. Rosuvastatin also reduced RhoA activation in TSC2(-/meth) ASM cells, and it required coadministration with the specific mTOR (mammalian target of rapamycin) inhibitor rapamycin to be effective in TSC2(-/-) ASM cells. Rapamycin enhanced rosuvastatin effect in inhibiting cell proliferation in TSC2(-/-) and TSC2(-/meth) ASM cells. Rosuvastatin alone did not alter phosphorylation of S6 and extracellular signal-regulated kinase (ERK), and at the higher concentration, rosuvastatin and rapamycin slightly decreased ERK phosphorylation. These results suggest that rosuvastatin may potentially represent a treatment adjunct to the therapy with mTOR inhibitors now in clinical development for TSC. In particular, rosuvastatin appears useful when the disease is originated by epigenetic defects.
Collapse
Affiliation(s)
- Elena Lesma
- Laboratories of Pharmacology, Department of Health Sciences, Università degli Studi di Milano, via di Rudini', 8, 20142 Milano, Italy.
| | | | | | | | | | | |
Collapse
|
41
|
Hoffman AE, Demanelis K, Fu A, Zheng T, Zhu Y. Association of AMP-activated protein kinase with risk and progression of non-Hodgkin lymphoma. Cancer Epidemiol Biomarkers Prev 2013; 22:736-44. [PMID: 23396962 DOI: 10.1158/1055-9965.epi-12-1014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Metabolic dysregulation has been identified as an "emerging hallmark" of cancer. The heterotrimeric AMP-activated protein kinase (AMPK) complex is a central regulator of the metabolic system and an important component of the mTOR pathway and the p53 axis, making it uniquely positioned to influence carcinogenesis through its canonical functions in the metabolic arena, as well as through more traditional mechanisms such as regulation of apoptosis and angiogenesis. METHODS We conducted a population-based genetic association study to examine the impact of mutations in AMPK subunit genes on risk of non-Hodgkin lymphoma (NHL). We also analyzed public microarray data to determine the expression of AMPK in NHL cells and to assess the influence of AMPK expression on overall survival in patients with NHL. RESULTS We identified an AMPK subunit haplotype, which was significantly associated with NHL [OR, 5.44, 95% confidence interval (CI), 2.15-13.75] in women with no family history of cancer. Haplotypes in two subunits, PRKAA2 and PRKAG3, were nominally associated with the follicular and diffuse large B-cell lymphoma histologic subtypes, respectively, although these associations did not retain statistical significance after correction for multiple comparisons. Further, both of these subunits were differentially expressed (P < 0.05) in one or more lymphoma cell type, and higher expression of two versions of the AMPK-β subunit was significantly associated with increased 5-year survival among patients with NHL (P = 0.001 and P = 0.021). CONCLUSION These results provide evidence for AMPK involvement in the pathogenesis and progression of NHL. IMPACT These findings may lead to a novel area of research into NHL treatment and chemoprevention.
Collapse
Affiliation(s)
- Aaron E Hoffman
- Department of Epidemiology, Tulane School of Public Health and Tropical Medicine and Tulane Cancer Center, New Orleans, LA, USA
| | | | | | | | | |
Collapse
|
42
|
Nasr Z, Robert F, Porco JA, Muller WJ, Pelletier J. eIF4F suppression in breast cancer affects maintenance and progression. Oncogene 2012; 32:861-71. [PMID: 22484424 DOI: 10.1038/onc.2012.105] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Levels of eukaryotic initiation factor 4E (eIF4E) are frequently elevated in human cancers and in some instances have been associated with poor prognosis and outcome. Here we utilize transgenic and allograft breast cancer models to demonstrate that increased mammalian target of rapamycin (mTOR) signalling can be a significant contributor to breast cancer progression in vivo. Suppressing mTOR activity, as well as levels and activity of the downstream translation regulators, eIF4E and eIF4A, delayed breast cancer progression, onset of associated pulmonary metastasis in vivo and breast cancer cell invasion and migration in vitro. Translation of vascular endothelial growth factor (VEGF), matrix metallopeptidase 9 (MMP9) and cyclin D1 mRNAs, which encode products associated with the metastatic phenotype, is inhibited upon eIF4E suppression. Our results indicate that the mTOR/eIF4F axis is an important contributor to tumor maintenance and progression programs in breast cancer. Targeting this pathway may be of therapeutic benefit.
Collapse
Affiliation(s)
- Z Nasr
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
43
|
Feng FM, Yang J, Zhang H, Zhao L, Yu J, Li SL, Chen KS. Effect of a TSC2 antisense oligonucleotide on cell proliferation and apoptosis in human esophageal cancer cell line EC9706. Shijie Huaren Xiaohua Zazhi 2011; 19:3302-3307. [DOI: 10.11569/wcjd.v19.i32.3302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To observe the impact of a tuberous sclerosis complex 2 (TSC2)-specific antisense oligonucleotide (ASODN) on cell proliferation and apoptosis in human esophageal cancer cell line EC9706.
METHODS: A TSC2 ASODN at different concentrations (5, 10 and 15 μmol/L) was transfected into EC9706 cells using cationic liposomes. A sense oligonucleotide (15 μmol/L) and nonsense oligonucleotide (15 μmol/L) were used as negative controls, and non-transfected EC9706 cells were used as a normal control. At 24, 48 and 72 h after transfection, MTT and TUNEL assays were used to examine the proliferation and apoptosis of EC9706 cells, respectively, and immunocytochemistry and RT-PCR were used to determine the expression of TSC2.
RESULTS: Compared to the three control groups, transfection of the TSC2 ASODN increased proliferation, decreased apoptosis (13.11 ± 0.13, 9.31 ± 0.29, 4.38 ± 0.43 vs 16.46 ± 0.43, 16.63 ± 0.34, 16.23 ± 0.45, all P < 0.05), and reduced the expression of TSC2 mRNA and protein (FmRNA = 260.23, 572.22, 1004.35, Fprotein = 50.60, 330.69, 1221.28, all P < 0.05) in EC9706 cells in a time- and concentration-dependent manner. Transfection with a dose of 15 μmol/L for 72 h exhibited the most strong effect (P < 0.05).
CONCLUSION: The TSC2 ASODN could enhance proliferation and inhibit apoptosis in human esophageal cancer cell line EC9706.
Collapse
|
44
|
Bartlett JMS. Biomarkers and patient selection for PI3K/Akt/mTOR targeted therapies: current status and future directions. Clin Breast Cancer 2011; 10 Suppl 3:S86-95. [PMID: 21115427 DOI: 10.3816/cbc.2010.s.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt/ mammalian target of rapamycin (mTOR) pathway regulates a broad spectrum of physiologic and pathologic processes. In breast cancer mutation, amplification, deletion, methylation, and posttranslational modifications lead to significant dysregulation of this pathway leading to more aggressive and potentially drug-resistant disease. Multiple novel agents, targeting different nodes within the pathway are currently under development by both commercial and academic partners. The key to the successful validation of these markers is selection of the appropriate patient groups using biomarkers. This article reviews current progress in this area, highlighting the key molecular alterations described in genes within the PI3K/Akt/mTOR pathway that may have an effect on response to current and future therapeutic interventions. Herein, gaps in current knowledge are highlighted and suggestions for future research directions given that may facilitate biomarker development in partnership with current drug development.
Collapse
Affiliation(s)
- John M S Bartlett
- Endocrine Cancer Group and Edinburgh Breakthrough Breast Cancer Laboratory, Edinburgh University,Western General Hospital, Crewe Road South, Edinburgh, UK.
| |
Collapse
|
45
|
Somatotropin-mediated gene expression profiling of differentially displayed ESTs during lactation in Indian buffalo (Bubalus bubalis). J DAIRY RES 2011; 78:326-34. [PMID: 21774858 DOI: 10.1017/s0022029911000380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The study of bovine mammary gland functional genomics requires appropriate cDNA library collections to access gene expression patterns from different developmental and physiological stages. The present study was undertaken with the objective to identify candidate genes involved in the process of increased milk synthesis following 0, 48 and 96 h of recombinant bovine somatotropin (rbST) treatment to Surti buffalo (Bubalus bubalis) through differential display reverse transcriptase PCR (DDRT-PCR). Of a total 50 sequenced DD bands, 64% of ESTs were differentially expressed (appeared only in post-treatment samples, i.e. 48 h and 96 h) and 36% were up-regulated after rbST treatment. Of the ESTs 32%were found to be located on Bos taurus chromosome 24 (equivalent to buffalo chromosome 22), whereas 16% of ESTs could not be mapped, indicating that they are specific to buffalo. Quantitative real time PCR assay of 15 ESTs revealed transcript level surge in 13 ESTs, and decline in one EST, while one showed up-regulation in expression level at 48 h while down-regulation at 96 h. This study indicates more than 30 novel transcripts, with unknown function, involved in increased milk synthesis and also the involvement of many more genes in the physiology of milk production than once thought.
Collapse
|
46
|
Lacher MD, Pincheira RJ, Castro AF. Consequences of interrupted Rheb-to-AMPK feedback signaling in tuberous sclerosis complex and cancer. Small GTPases 2011; 2:211-216. [PMID: 22145093 DOI: 10.4161/sgtp.2.4.16703] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 06/02/2011] [Accepted: 06/06/2011] [Indexed: 02/06/2023] Open
Abstract
Rheb is a small GTPase primarily known for activating mammalian target of rapamycin complex 1 (mTORC1) and promoting cell growth in response to insulin and nutrients (amino acids, glucose). Shortage of glucose activates adenosine 5'-monophosphate-activated protein kinase (AMPK), which induces catabolic processes that produce ATP and suppresses energy-consuming anabolic reactions. As part of the latter response, AMPK activates the TSC1-TSC2 tumor suppressor complex, which in turn inhibits Rheb, thereby reducing mTORC1 activity and consequently suppressing protein synthesis. We recently identified an mTORC1-independent Rheb-to-AMPK feedback signaling loop in Tsc2-null in vitro models of Tuberous Sclerosis Complex (TSC). In addition to activating AMPK, Rheb reduced the nuclear levels of the cyclin-dependent kinase inhibitor p27(KIP1) (p27). Importantly, Rheb-mediated repression of p27 correlated with activation of Cdk2 and cell proliferation, and with tumor formation by TSC cells. Considering that AMPK was previously reported to regulate stability and subcellular localization of p27, we hypothesize that Rheb regulates p27 in TSC cells by activating AMPK. This article discusses how Rheb-to-AMPK, and p27 signaling may impact on disease progression and treatment of TSC, including sporadic lymphangioleiomyomatosis (S-LAM) and malignancies.
Collapse
Affiliation(s)
- Markus D Lacher
- Helen Diller Family Comprehensive Cancer Center; University of California; San Francisco, CA USA
| | | | | |
Collapse
|
47
|
Targeting phosphatidylinositol 3 kinase (PI3K)-Akt beyond rapalogs. Target Oncol 2011; 6:103-17. [DOI: 10.1007/s11523-011-0176-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Accepted: 03/09/2011] [Indexed: 01/20/2023]
|
48
|
O'Regan R, Hawk NN. mTOR inhibition in breast cancer: unraveling the complex mechanisms of mTOR signal transduction and its clinical implications in therapy. Expert Opin Ther Targets 2011; 15:859-72. [PMID: 21476875 DOI: 10.1517/14728222.2011.575362] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION The mammalian target of rapamycin (mTOR)/PI3K/Akt pathway is altered in breast cancer cells, as demonstrated by mutations in both the upstream and downstream regulators of mTOR, including phosphatase and tensin homolog deleted in chromosome 10 (PTEN) loss or Akt/PI3K activation, and potentially in the mTOR protein itself. This contributes to increased cell proliferation, as well as growth-factor independence and endocrine resistance. Thus, mTOR inhibition holds considerable promise as a rational therapeutic strategy in breast cancer. AREAS COVERED This review describes how dysregulation of the mTOR pathway in breast cancer may contribute to breast cancer pathogenesis, as well as discussing preclinical and clinical data that support mTOR inhibitor therapy. EXPERT OPINION Direct blockade of the mTOR pathway is a new and intriguing area in breast cancer therapy, with the potential to modulate growth-factor and estrogen-dependent and -independent pathways, that contribute to the pathogenesis and progression of breast tumors. mTOR inhibitors demonstrate significant biologic activity with manageable toxicities, in combination with hormonal therapy and chemotherapy, in both the neoadjuvant and metastatic breast cancer settings.
Collapse
Affiliation(s)
- Ruth O'Regan
- Emory University School of Medicine, Winship Cancer Institute, Department of Hematology and Medical Oncology, 1365 Clifton Road, Atlanta, GA 30322, USA.
| | | |
Collapse
|
49
|
Wander SA, Hennessy BT, Slingerland JM. Next-generation mTOR inhibitors in clinical oncology: how pathway complexity informs therapeutic strategy. J Clin Invest 2011; 121:1231-41. [PMID: 21490404 DOI: 10.1172/jci44145] [Citation(s) in RCA: 331] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) is a PI3K-related kinase that regulates cell growth, proliferation, and survival via mTOR complex 1 (mTORC1) and mTORC2. The mTOR pathway is often aberrantly activated in cancers. While hypoxia, nutrient deprivation, and DNA damage restrain mTORC1 activity, multiple genetic events constitutively activate mTOR in cancers. Here we provide a brief overview of the signaling pathways up- and downstream of mTORC1 and -2, and discuss the insights into therapeutic anticancer targets - both those that have been tried in the clinic with limited success and those currently under clinical development - that knowledge of these pathways gives us.
Collapse
Affiliation(s)
- Seth A Wander
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | | | | |
Collapse
|
50
|
The long and winding road to rational treatment of cancer associated with LKB1/AMPK/TSC/mTORC1 signaling. Oncogene 2011; 30:2289-303. [PMID: 21258412 DOI: 10.1038/onc.2010.630] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The liver kinase B1 (LKB1)/adenosine mono-phosphate-activated protein kinase (AMPK)/tuberous sclerosis complex (TSC)/mammalian target of rapamycin (mTOR) complex (mTORC1) cassette constitutes a canonical signaling pathway that integrates information on the metabolic and nutrient status and translates this into regulation of cell growth. Alterations in this pathway are associated with a wide variety of cancers and hereditary hamartoma syndromes, diseases in which hyperactivation of mTORC1 has been described. Specific mTORC1 inhibitors have been developed for clinical use, and these drugs have been anticipated to provide efficient treatment for these diseases. In the present review, we provide an overview of the metabolic LKB1/AMPK/TSC/mTORC1 pathway, describe how its aberrant signaling associates with cancer development, and indicate the difficulties encountered when biochemical data are extrapolated to provide avenues for rational treatment of disease when targeting this signaling pathway. A careful examination of preclinical and clinical studies performed with rapamycin or derivatives thereof shows that although results are encouraging, we are only half way in the long and winding road to design rationale treatment targeted at the LKB1/AMPK/TSC/mTORC1 pathway. Inherited cancer syndromes associated with this pathway such as the Peutz-Jeghers syndrome and TSC, provide perfect models to study the relationship between genetics and disease phenotype, and to delineate the complexities that underlie translation of biochemical and genetical information to clinical management, and thus provide important clues for devising novel rational medicine for cancerous diseases in general.
Collapse
|