1
|
Al-Hawary SIS, Ruzibakieva M, Gupta R, Malviya J, Toama MA, Hjazi A, Alkhayyat MRR, Alsaab HO, Hadi A, Alwaily ER. Detailed role of microRNA-mediated regulation of PI3K/AKT axis in human tumors. Cell Biochem Funct 2024; 42:e3904. [PMID: 38102946 DOI: 10.1002/cbf.3904] [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: 10/24/2023] [Revised: 11/23/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023]
Abstract
The regulation of signal transmission and biological processes, such as cell proliferation, apoptosis, metabolism, migration, and angiogenesis are greatly influenced by the PI3K/AKT signaling pathway. Highly conserved endogenous non-protein-coding RNAs known as microRNAs (miRNAs) have the ability to regulate gene expression by inhibiting mRNA translation or mRNA degradation. MiRNAs serve key role in PI3K/AKT pathway as upstream or downstream target, and aberrant activation of this pathway contributes to the development of cancers. A growing body of research shows that miRNAs can control the PI3K/AKT pathway to control the biological processes within cells. The expression of genes linked to cancers can be controlled by the miRNA/PI3K/AKT axis, which in turn controls the development of cancer. There is also a strong correlation between the expression of miRNAs linked to the PI3K/AKT pathway and numerous clinical traits. Moreover, PI3K/AKT pathway-associated miRNAs are potential biomarkers for cancer diagnosis, therapy, and prognostic evaluation. The role and clinical applications of the PI3K/AKT pathway and miRNA/PI3K/AKT axis in the emergence of cancers are reviewed in this article.
Collapse
Affiliation(s)
| | - Malika Ruzibakieva
- Cell Therapy Department, Institute of Immunology and Human Genomics, Uzbekistan Academy of Science, Tashkent, Uzbekistan
| | - Reena Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Jitendra Malviya
- Department of Life Sciences and Biological Sciences, IES University, Bhopal, Madhya Pradesh, India
| | - Mariam Alaa Toama
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Murtadha Raad Radhi Alkhayyat
- Department of Islamic Studies, College of Art, The Islamic University of Najaf, Najaf, Iraq
- Department of Islamic Studies, College of Art, The Islamic University of Babylon, Babylon, Iraq
- Department of Islamic Studies, College of Art, The Islamic University of Al Diwaniyah, Diwaniyah, Iraq
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif, Saudi Arabia
| | - Ali Hadi
- Department of Medical Laboratories Techniques, Imam Ja'afar Al-Sadiq University, Al-Muthanna, Iraq
| | - Enas R Alwaily
- Microbiology Research Group, Al-Ayen University, Thi-Qar, Iraq
| |
Collapse
|
2
|
Jeon T, Oh UJ, Min J, Kim C. Gene-level dissection of chromosome 3q locus amplification in squamous cell carcinoma of the lung using the nCounter assay. Thorac Cancer 2023; 14:2635-2641. [PMID: 37469197 PMCID: PMC10493484 DOI: 10.1111/1759-7714.15045] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Amplification of the 3q region has been identified as a useful biomarker for the diagnosis and treatment of squamous cell carcinoma (SqCC). This region contains genes such as PIK3CA and YEATS2, which have been linked to the prognosis of SqCC. METHODS The NanoString nCounter assay is a powerful tool for identifying genetic alterations that affect the progression and prognosis of SqCC. The NanoString nCounter assay was used to identify a subgroup of patients with gene level gain in the 3q region. RESULTS Gene level gain in the 3q region was more frequent in SqCC than in adenocarcinoma. We found that genes such as PIK3CA and YEATS2 in the 3q region were associated with the prognosis of SqCC. Therefore, identifying a subgroup of patients with gene level gain in the 3q region using the NanoString nCounter assay can aid in selecting appropriate treatment options and improving prognostic predictions for SqCC patients. CONCLUSION Amplification of the 3q region in SqCC of lung cancer is a useful biomarker for diagnosis and treatment. The NanoString nCounter assay is a powerful tool for identifying specific genetic alterations that affect the progression and prognosis of SqCC. Our study highlights the importance 3q amplification and its associated genes in lung cancer.
Collapse
Affiliation(s)
- Taesung Jeon
- Department of PathologyCollege of Medicine, Korea UniversitySeoulSouth Korea
| | - Uk Jeen Oh
- Department of PathologyCollege of Medicine, Korea UniversitySeoulSouth Korea
| | - Jaeyoung Min
- Department of PathologyCollege of Medicine, Korea UniversitySeoulSouth Korea
| | - Chungyeul Kim
- Department of PathologyCollege of Medicine, Korea UniversitySeoulSouth Korea
| |
Collapse
|
3
|
Lazar V, Raynaud J, Magidi S, Bresson C, Martini JF, Galbraith S, Wunder F, Onn A, Batist G, Girard N, Lassen U, Pramesh CS, Al-Omari A, Ikeda S, Berchem G, Blay JY, Solomon B, Felip E, Tabernero J, Rubin E, Philip T, Porgador A, Berindan-Neagoe I, Schilsky RL, Kurzrock R. Comorbidity between lung cancer and COVID-19 pneumonia: role of immunoregulatory gene transcripts in high ACE2-expressing normal lung. Ther Adv Med Oncol 2022; 14:17588359221133893. [PMID: 36324736 PMCID: PMC9618916 DOI: 10.1177/17588359221133893] [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: 05/04/2022] [Accepted: 10/03/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND SARS-CoV-2 (COVID-19) elicits a T-cell antigen-mediated immune response of variable efficacy. To understand this variability, we explored transcriptomic expression of angiotensin-converting enzyme 2 (ACE2, the SARS-CoV-2 receptor) and of immunoregulatory genes in normal lung tissues from patients with non-small cell lung cancer (NSCLC). METHODS This study used the transcriptomic and the clinical data for NSCLC patients generated during the CHEMORES study [n = 123 primary resected (early-stage) NSCLC] and the WINTHER clinical trial (n = 32 metastatic NSCLC). RESULTS We identified patient subgroups with high and low ACE2 expression (p = 1.55 × 10-19) in normal lung tissue, presumed to be at higher and lower risk, respectively, of developing severe COVID-19 should they become infected. ACE2 transcript expression in normal lung tissues (but not in tumor tissue) of patients with NSCLC was higher in individuals with more advanced disease. High-ACE2 expressors had significantly higher levels of CD8+ cytotoxic T lymphocytes and natural killer cells but with presumably impaired function by high Thymocyte Selection-Associated High Mobility Group Box Protein TOX (TOX) expression. In addition, immune checkpoint-related molecules - PD-L1, CTLA-4, PD-1, and TIGIT - are more highly expressed in normal (but not tumor) lung tissues; these molecules might dampen immune response to either viruses or cancer. Importantly, however, high inducible T-cell co-stimulator (ICOS), which can amplify immune and cytokine reactivity, significantly correlated with high ACE2 expression in univariable analysis of normal lung (but not lung tumor tissue). CONCLUSIONS We report a normal lung immune-tolerant state that may explain a potential comorbidity risk between two diseases - NSCLC and susceptibility to COVID-19 pneumonia. Further, a NSCLC patient subgroup has normal lung tissue expressing high ACE2 and high ICOS transcripts, the latter potentially promoting a hyperimmune response, and possibly leading to severe COVID-19 pulmonary compromise.
Collapse
Affiliation(s)
| | | | - Shai Magidi
- Worldwide Innovative Network (WIN) Association – WIN Consortium, Villejuif, France
| | | | | | | | - Fanny Wunder
- Worldwide Innovative Network (WIN) Association – WIN Consortium, Villejuif, France
| | - Amir Onn
- Sheba Medical Center, Tel-Hashomer, Israel
| | - Gerald Batist
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montréal, Canada
| | | | | | - C. S. Pramesh
- Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | | | | | - Guy Berchem
- Centre Hospitalier Luxembourg and Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Jean-Yves Blay
- Centre Leon Bérard, University Lyon 1, LYRICAN & NETSARC+, Lyon, France
| | | | - Enriqueta Felip
- Vall d’Hebron Hospital Campus and Institute of Oncology, UVic-UCC, Barcelona, Spain
| | | | - Eitan Rubin
- Faculty of Health Sciences Ben-Gurion University of the Negev, Beer-Sheeva, Israel
| | | | - Angel Porgador
- Faculty of Health Sciences Ben-Gurion University of the Negev, Beer-Sheeva, Israel
| | | | | | | |
Collapse
|
4
|
Lazar V, Girard N, Raymond E, Martini JF, Galbraith S, Raynaud J, Bresson C, Solomon B, Magidi S, Nechushtan H, Onn A, Berger R, Chen H, Al-Omari A, Ikeda S, Lassen U, Sekacheva M, Felip E, Tabernero J, Batist G, Spatz A, Pramesh CS, Girard P, Blay JY, Philip T, Berindan-Neagoe I, Porgador A, Rubin E, Kurzrock R, Schilsky RL. Transcriptomics in Tumor and Normal Lung Tissues Identify Patients With Early-Stage Non-Small-Cell Lung Cancer With High Risk of Postsurgery Recurrence Who May Benefit From Adjuvant Therapies. JCO Precis Oncol 2022; 6:e2200072. [PMID: 36108261 PMCID: PMC9489166 DOI: 10.1200/po.22.00072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The prognosis of patients with non-small-cell lung cancer (NSCLC), traditionally determined by anatomic histology and TNM staging, neglects the biological features of the tumor that may be important in determining patient outcome and guiding therapeutic interventions. Identifying patients with NSCLC at increased risk of recurrence after curative-intent surgery remains an important unmet need so that known effective adjuvant treatments can be offered to those at highest risk of recurrence. METHODS Relative gene expression level in the primary tumor and normal bronchial tissues was used to retrospectively assess their association with disease-free survival (DFS) in a cohort of 120 patients with NSCLC who underwent curative-intent surgery. RESULTS Low versus high Digital Display Precision Predictor (DDPP) score (a measure of relative gene expression) was significantly associated with shorter DFS (highest recurrence risk; P = .006) in all patients and in patients with TNM stages 1-2 (P = .00051; n = 83). For patients with stages 1-2 and low DDPP score (n = 29), adjuvant chemotherapy was associated with improved DFS (P = .0041). High co-overexpression of CTLA-4, PD-L1, and ICOS in normal lung (28 of 120 patients) was also significantly associated with decreased DFS (P = .0013), suggesting an immune tolerance to tumor neoantigens in some patients. Patients with DDPP low and immunotolerant normal tissue had the shortest DFS (P = 2.12E-11). CONCLUSION TNM stage, DDPP score, and immune competence status of normal lung are independent prognostic factors in multivariate analysis. Our findings open new avenues for prospective prognostic assessment and treatment assignment on the basis of transcriptomic profiling of tumor and normal lung tissue in patients with NSCLC.
Collapse
Affiliation(s)
- Vladimir Lazar
- Worldwide Innovative Network-WIN Consortium, Villejuif, France
| | - Nicolas Girard
- Institut Curie, Paris, France.,Institut du Thorax Curie-Institut Montsouris, Paris, France
| | | | | | | | - Jacques Raynaud
- Worldwide Innovative Network-WIN Consortium, Villejuif, France
| | | | | | - Shai Magidi
- Worldwide Innovative Network-WIN Consortium, Villejuif, France
| | | | - Amir Onn
- Sheba Medical Center, Tel-Hashomer, Israel
| | | | - Haiquan Chen
- Fudan University Shanghai Cancer Center, Shanghai, China
| | | | | | | | | | - Enriqueta Felip
- Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), UVic-UCC, Barcelona, Spain
| | - Josep Tabernero
- Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), UVic-UCC, Barcelona, Spain
| | - Gerald Batist
- Segal Cancer Center, Jewish General Hospital, McGill University, Montréal, Canada
| | - Alan Spatz
- Segal Cancer Center, Jewish General Hospital, McGill University, Montréal, Canada
| | - C S Pramesh
- Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, India
| | | | - Jean-Yves Blay
- Center Leon-Bérard, Lyon, France.,Unicancer, Paris, France
| | | | | | | | - Eitan Rubin
- Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | | | | |
Collapse
|
5
|
Ding L, Zentner GE, McDonald DJ. Sufficient principal component regression for pattern discovery in transcriptomic data. BIOINFORMATICS ADVANCES 2022; 2:vbac033. [PMID: 35722206 PMCID: PMC9194947 DOI: 10.1093/bioadv/vbac033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/16/2022] [Accepted: 05/04/2022] [Indexed: 01/27/2023]
Abstract
Motivation Methods for the global measurement of transcript abundance such as microarrays and RNA-Seq generate datasets in which the number of measured features far exceeds the number of observations. Extracting biologically meaningful and experimentally tractable insights from such data therefore requires high-dimensional prediction. Existing sparse linear approaches to this challenge have been stunningly successful, but some important issues remain. These methods can fail to select the correct features, predict poorly relative to non-sparse alternatives or ignore any unknown grouping structures for the features. Results We propose a method called SuffPCR that yields improved predictions in high-dimensional tasks including regression and classification, especially in the typical context of omics with correlated features. SuffPCR first estimates sparse principal components and then estimates a linear model on the recovered subspace. Because the estimated subspace is sparse in the features, the resulting predictions will depend on only a small subset of genes. SuffPCR works well on a variety of simulated and experimental transcriptomic data, performing nearly optimally when the model assumptions are satisfied. We also demonstrate near-optimal theoretical guarantees. Availability and implementation Code and raw data are freely available at https://github.com/dajmcdon/suffpcr. Package documentation may be viewed at https://dajmcdon.github.io/suffpcr. Contact daniel@stat.ubc.ca. Supplementary information Supplementary data are available at Bioinformatics Advances online.
Collapse
Affiliation(s)
- Lei Ding
- Department of Statistics, Indiana University, Bloomington, IN 47405, USA
| | - Gabriel E Zentner
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Daniel J McDonald
- Department of Statistics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
6
|
Li H, Li M, Guo H, Lin G, Huang Q, Qiu M. Integrative Analyses of Circulating mRNA and lncRNA Expression Profile in Plasma of Lung Cancer Patients. Front Oncol 2022; 12:843054. [PMID: 35433477 PMCID: PMC9008738 DOI: 10.3389/fonc.2022.843054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Circulating-free RNAs (cfRNAs) have been regarded as potential biomarkers for “liquid biopsy” in cancers. However, the circulating messenger RNA (mRNA) and long noncoding RNA (lncRNA) profiles of lung cancer have not been fully characterized. In this study, we profiled circulating mRNA and lncRNA profiles of 16 lung cancer patients and 4 patients with benign pulmonary nodules. Compared with benign pulmonary nodules, 806 mRNAs and 1,762 lncRNAs were differentially expressed in plasma of lung adenocarcinoma patients. For lung squamous cell carcinomas, 256 mRNAs and 946 lncRNAs were differentially expressed. A total of 231 mRNAs and 298 lncRNAs were differentially expressed in small cell lung cancer. Eleven mRNAs, 51 lncRNAs, and 207 canonical pathways were differentially expressed in lung cancer in total. Forty-five blood samples were collected to verify our findings via performing qPCR. There are plenty of meaningful mRNAs and lncRNAs that were found. MYC, a transcription regulator associated with the stemness of cancer cells, is overexpressed in lung adenocarcinoma. Transforming growth factor beta (TGFB1), which plays pleiotropic roles in cancer progression, was found to be upregulated in lung squamous carcinoma. MALAT1, a well-known oncogenic lncRNA, was also found to be upregulated in lung squamous carcinoma. Thus, this study provided a systematic resource of mRNA and lncRNA expression profiles in lung cancer plasma.
Collapse
Affiliation(s)
- Haoran Li
- Department of Thoracic Surgery, Peking University People’s Hospital, Beijing, China
| | - Mingru Li
- Department of Thoracic Surgery, Aerospace 731 Hospital, Beijing, China
| | - Haifa Guo
- The First Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Guihu Lin
- Department of Thoracic Surgery, Aerospace 731 Hospital, Beijing, China
| | - Qi Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Qi Huang, ; Mantang Qiu,
| | - Mantang Qiu
- Department of Thoracic Surgery, Peking University People’s Hospital, Beijing, China
- *Correspondence: Qi Huang, ; Mantang Qiu,
| |
Collapse
|
7
|
Abstract
This overview of the molecular pathology of lung cancer includes a review of the most salient molecular alterations of the genome, transcriptome, and the epigenome. The insights provided by the growing use of next-generation sequencing (NGS) in lung cancer will be discussed, and interrelated concepts such as intertumor heterogeneity, intratumor heterogeneity, tumor mutational burden, and the advent of liquid biopsy will be explored. Moreover, this work describes how the evolving field of molecular pathology refines the understanding of different histologic phenotypes of non-small-cell lung cancer (NSCLC) and the underlying biology of small-cell lung cancer. This review will provide an appreciation for how ongoing scientific findings and technologic advances in molecular pathology are crucial for development of biomarkers, therapeutic agents, clinical trials, and ultimately improved patient care.
Collapse
Affiliation(s)
- James J Saller
- Departments of Pathology and Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - Theresa A Boyle
- Departments of Pathology and Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| |
Collapse
|
8
|
Friedlander JE, Shen N, Zeng A, Korm S, Feng H. Failure to Guard: Mitochondrial Protein Quality Control in Cancer. Int J Mol Sci 2021; 22:ijms22158306. [PMID: 34361072 PMCID: PMC8348654 DOI: 10.3390/ijms22158306] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/20/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023] Open
Abstract
Mitochondria are energetic and dynamic organelles with a crucial role in bioenergetics, metabolism, and signaling. Mitochondrial proteins, encoded by both nuclear and mitochondrial DNA, must be properly regulated to ensure proteostasis. Mitochondrial protein quality control (MPQC) serves as a critical surveillance system, employing different pathways and regulators as cellular guardians to ensure mitochondrial protein quality and quantity. In this review, we describe key pathways and players in MPQC, such as mitochondrial protein translocation-associated degradation, mitochondrial stress responses, chaperones, and proteases, and how they work together to safeguard mitochondrial health and integrity. Deregulated MPQC leads to proteotoxicity and dysfunctional mitochondria, which contributes to numerous human diseases, including cancer. We discuss how alterations in MPQC components are linked to tumorigenesis, whether they act as drivers, suppressors, or both. Finally, we summarize recent advances that seek to target these alterations for the development of anti-cancer drugs.
Collapse
Affiliation(s)
- Joseph E. Friedlander
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; (J.E.F.); (N.S.); (A.Z.); (S.K.)
| | - Ning Shen
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; (J.E.F.); (N.S.); (A.Z.); (S.K.)
- Department of Medicine, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Aozhuo Zeng
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; (J.E.F.); (N.S.); (A.Z.); (S.K.)
| | - Sovannarith Korm
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; (J.E.F.); (N.S.); (A.Z.); (S.K.)
| | - Hui Feng
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; (J.E.F.); (N.S.); (A.Z.); (S.K.)
- Department of Medicine, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA 02118, USA
- Correspondence: ; Tel.: +1-617-358-4688; Fax: +1-617-358-1599
| |
Collapse
|
9
|
Yuan L, Li JX, Yang Y, Chen Y, Ma TT, Liang S, Bu Y, Yu L, Nan Y. Depletion of MRPL35 inhibits gastric carcinoma cell proliferation by regulating downstream signaling proteins. World J Gastroenterol 2021; 27:1785-1804. [PMID: 33967557 PMCID: PMC8072187 DOI: 10.3748/wjg.v27.i16.1785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/04/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric carcinoma (GC) is a digestive system disease with high morbidity and mortality. However, early clinical detection is difficult, and the therapeutic effect for advanced disease is not satisfactory. Thus, finding new tumor markers and therapeutic targets conducive to the treatment of GC is imperative. MRPL35 is a member of the large subunit family of mitochondrial ribosomal protein. MRPL35 shows the characteristic of oncogene in colorectal cancer and esophageal cancer, which promotes the exploration of the correlation between MRPL35 and GC. We proposed that the expression of MRPL35 might be critical in GC.
AIM To study the effect of MRPL35 knockdown on GC cell proliferation.
METHODS The expression of MRPL35 in GC was evaluated based on data from the public tumor database UALCAN (http://www.ualcan.path.uab.edu). The effect of the expression of MRPL35 on the prognosis was evaluated with KMplot (http://www.kmplot.com). The expression of MRPL35 was assessed on the tissue microarray by immunohistochemistry and the level of MRPL35 mRNA in 25 pairs of clinical GC tissues and matched adjacent tissues was detected by quantitative reverse transcription-polymerase chain reaction. Celigo cell count assay, colony formation assay, and flow cytometry were used to assess the role of MRPL35 in GC cell proliferation and apoptosis in vitro. Additionally, tumor formation experiment in BALB/c nude mice was utilized to determine the effect of MRPL35 on GC cell proliferation. After knockdown of MRPL35, related proteins were identified by isobaric tags for relative and absolute quantification analysis, and the expression of related proteins was detected by Western blot.
RESULTS The expression of MRPL35 was up-regulated in GC (P = 1.77 × 10-4). The Kaplan-Meier plots of the overall survival indicated that high expression of MRPL35 was associated with a poor survival in GC. Compared with adjacent tissues, the expression of MRPL35 in GC tissues was increased, which was related to age (P = 0.03), lymph node metastasis (P = 0.007), and pathological tumor-node-metastasis stage (P = 0.024). Knockdown of MRPL35 inhibited GC cell proliferation and colony formation and induced apoptosis. Animal experiment results showed that knockdown of MRPL35 inhibited tumor formation in BALB/c nude mice. Western blotting analysis showed that after knockdown of MRPL35, the expression of PICK1 and BCL-XL proteins decreased, and that of AGR2 protein increased.
CONCLUSION Collectively, our findings demonstrate that knockdown of MRPL35 inhibits GC cell proliferation through related proteins including PICK1, BCL-XL, and AGR2.
Collapse
Affiliation(s)
- Ling Yuan
- Pharmacy College of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
- Key Laboratory of Hui Ethnic Medicine Modernization of Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Jia-Xin Li
- Pharmacy College of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Yang
- Pharmacy College of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yan Chen
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ting-Ting Ma
- Pharmacy College of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Shuang Liang
- Department of Oncology and Endocrinology, Yinchuan Hospital of Traditional Chinese Medicine Affiliated to Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yang Bu
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Lei Yu
- Department of Infectious Diseases, The Fourth Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Yi Nan
- Key Laboratory of Hui Ethnic Medicine Modernization of Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
10
|
Yao J, Tang YC, Yi B, Yang J, Chai Y, Yin N, Zhang ZX, Wei YJ, Li DC, Zhou J. Signature of gene aberrant alternative splicing events in pancreatic adenocarcinoma prognosis. J Cancer 2021; 12:3164-3179. [PMID: 33976726 PMCID: PMC8100795 DOI: 10.7150/jca.48661] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 03/15/2021] [Indexed: 12/17/2022] Open
Abstract
Alternative splicing (AS), as an effective and universal mechanism of transcriptional regulation, is involved in the development and progression of cancer. Therefore, systematic analysis of alternative splicing in pancreatic adenocarcinoma (PAAD) is warranted. The corresponding clinical information of the RNA-Seq data and PAAD cohort was downloaded from the TCGA data portal. Then, a java application, SpliceSeq, was used to evaluate the RNA splicing pattern and calculate the splicing percentage index (PSI). Differentially expressed AS events (DEAS) were identified based on PSI values between PAAD cancer samples and normal samples of adjacent tissues. Kaplan-Meier and Cox regression analyses were used to assess the association between DEAS and patient clinical characteristics. Unsupervised cluster analysis used to reveal four clusters with different survival patterns. At the same time, GEO and TCGA combined with GTEx to verify the differential expression of AS gene and splicing factor. After rigorous filtering, a total of 45,313 AS events were identified, 1,546 of which were differentially expressed AS events. Nineteen DEAS were found to be associated with OS with a five-year overall survival rate of 0.946. And the subtype clusters results indicate that there are differences in the nature of individual AS that affect clinical outcomes. Results also identified 15 splicing factors associated with the prognosis of PAAD. And the splicing factors ESRP1 and RBM5 played an important role in the PAAD-associated AS events. The PAAD-associated AS events, splicing networks, and clusters identified in this study are valuable for deciphering the underlying mechanisms of AS in PAAD and may facilitate the establishment of therapeutic goals for further validation.
Collapse
Affiliation(s)
- Jun Yao
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Yu-Chen Tang
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Bin Yi
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Jian Yang
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Yun Chai
- Department of Plastic Surgery, Suzhou Municipal Hospital, Suzhou, Jiangsu, 215006, China
| | - Ni Yin
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Zi-Xiang Zhang
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Yi-Jun Wei
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - De-Chun Li
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Jian Zhou
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
- Pancreatic Disease Research Centre, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| |
Collapse
|
11
|
Guerriero I, Monaco G, Coppola V, Orlacchio A. Serum and Glucocorticoid-Inducible Kinase 1 (SGK1) in NSCLC Therapy. Pharmaceuticals (Basel) 2020; 13:ph13110413. [PMID: 33266470 PMCID: PMC7700219 DOI: 10.3390/ph13110413] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) remains the most prevalent and one of the deadliest cancers worldwide. Despite recent success, there is still an urgent need for new therapeutic strategies. It is also becoming increasingly evident that combinatorial approaches are more effective than single modality treatments. This review proposes that the serum and glucocorticoid-inducible kinase 1 (SGK1) may represent an attractive target for therapy of NSCLC. Although ubiquitously expressed, SGK1 deletion in mice causes only mild defects of ion physiology. The frequent overexpression of SGK1 in tumors is likely stress-induced and provides a therapeutic window to spare normal tissues. SGK1 appears to promote oncogenic signaling aimed at preserving the survival and fitness of cancer cells. Most importantly, recent investigations have revealed the ability of SGK1 to skew immune-cell differentiation toward pro-tumorigenic phenotypes. Future studies are needed to fully evaluate the potential of SGK1 as a therapeutic target in combinatorial treatments of NSCLC. However, based on what is currently known, SGK1 inactivation can result in anti-oncogenic effects both on tumor cells and on the immune microenvironment. A first generation of small molecules to inactivate SGK1 has already been already produced.
Collapse
Affiliation(s)
- Ilaria Guerriero
- Biogem Institute for Genetic Research Gaetano Salvatore, Ariano Irpino, 83031 Avellino, Italy; (I.G.); (G.M.)
| | - Gianni Monaco
- Biogem Institute for Genetic Research Gaetano Salvatore, Ariano Irpino, 83031 Avellino, Italy; (I.G.); (G.M.)
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (V.C.); (A.O.); Tel.: +1-614-688-8038 (V.C.); +1-646-552-0641 (A.O.)
| | - Arturo Orlacchio
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (V.C.); (A.O.); Tel.: +1-614-688-8038 (V.C.); +1-646-552-0641 (A.O.)
| |
Collapse
|
12
|
Yin X, Xia J, Sun Y, Zhang Z. CHCHD2 is a potential prognostic factor for NSCLC and is associated with HIF-1a expression. BMC Pulm Med 2020; 20:40. [PMID: 32054470 PMCID: PMC7020603 DOI: 10.1186/s12890-020-1079-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/05/2020] [Indexed: 12/28/2022] Open
Abstract
Background CHCHD2 was identified a novel cell migration-promoting gene, which could promote cell migration and altered cell adhesion when ectopically overexpressed in NIH3T3 fibroblasts, and it was identified as a protein necessary for OxPhos function as well. However, the clinic relevance of CHCHD2 expression in NSCLC remains unclear. Here we assumed that CHCHD2 expression would accompanies the expression of HIF-1α to response hypoxia in the occurrence of NSCLC. Methods In order to verify this hypothesis, correlations among the expression levels of CHCHD2 and HIF-1α were detected and analyzed in 209 pair cases of NSCLC. The expression and location of these molecules were assessed using Immunohistochemistry, immunohistofluorescence, qRT-PCR and western blotting. The differences and correlations of the expression of these two molecules with clinical pathological characteristics in NSCLC were statistically analyzed using Wilcoxon (W) text, Mann-Whitney U, Kruskal-Wallis H and cross-table tests. Kaplan-Meier survival analysis and Cox proportional hazards models were used to estimate the effect of the expression of CHCHD2 and HIF-1α on the patients’ survival. Results Data showed that CHCHD2 and HIF-1α expression were higher in NSCLC than in normal tissues (all P = 0.000). CHCHD2 expression was significantly related with smoking, tumor size, differentiation degree, TNM Stage, lymph metastasis (all P<0.05). The HIF-1α expression was significantly associated with smoking, tumor category, differentiation degree, TNM Stage, Lymph metastasis (all P<0.05). There was a marked correlation of CHCHD2 and HIF-1α expression with histological type, differentiation and lymph metastasis of NSCLC (all P<0.05, rs>0.3). Immunohistofluorescence showed that there were co-localization phenomenon in cytoplasm and nucleus between CHCHD2 and HIF-1α expression. NSCLC patients with higher CHCHD2 and HIF-1α expression had a significantly worse prognosis than those with lower CHCHD2 and HIF-1α expression (all P = 0.0001; log-rank test). The multivariate analysis indicated that CHCHD2 expression was an independent prognostic factor in NSCLC (hazard ratio [HR], 0.492, P = 0.001). Conclusion Our results indicate that over-expression of CHCHD2 would promote the expression of HIF-1α to adapt the hypoxia microenviroment in NSCLC and CHCHD2 could serves as a prognostic biomarker in NSCLC.
Collapse
Affiliation(s)
- Xin Yin
- Department of Radiotherapy, Xianyang Hospital, Yanan University, Xianyang, Shaanxi, 712000, People's Republic of China
| | - Jinghua Xia
- Department of Thoracic Surgery, The Second Affliated Hospital of Air Force Medical University, Xi'an, 710038, China.
| | - Ying Sun
- Department of Thoracic Surgery, The Second Affliated Hospital of Air Force Medical University, Xi'an, 710038, China
| | - Zhipei Zhang
- Department of Thoracic Surgery, The Second Affliated Hospital of Air Force Medical University, Xi'an, 710038, China.
| |
Collapse
|
13
|
Chan LW, Wang F, Meng F, Wang L, Wong SCC, Au JS, Yang S, Cho WC. Multi-scale representation of proteomic data exhibits distinct microRNA regulatory modules in non-smoking female patients with lung adenocarcinoma. Comput Biol Med 2018; 102:51-56. [PMID: 30245277 DOI: 10.1016/j.compbiomed.2018.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/10/2018] [Accepted: 09/10/2018] [Indexed: 11/20/2022]
Abstract
Adenocarcinoma in female non-smokers is an under-explored subgroup of non-small cell lung cancer (NSCLC), in which the molecular mechanism and genetic risk factors remain unclear. We analyzed the protein profiles of plasma samples of 45 patients in this subgroup and 60 non-cancer subjects using surface-enhanced laser desorption/ionization time-of- flight mass spectrometry. Among 85 peaks of mass spectra, the differential expression analysis identified 15 markers based on False Discovery Rate control and the Discrete Wavelet Transforms further selected a cluster of 6 markers that were consistently observed at multiple scales of mass-charge ratios. This marker cluster, corresponding to 7 unique proteins, was able to distinguish the female non-smokers with adenocarcinoma from non-cancer subjects with a value of accuracy of 87.6%. We also predicted the role of competing endogenous RNAs (ceRNAs) in 3 out of these 7 proteins. Other studies reported that these ceRNAs and their targeting microRNAs, miR-206 and miR-613, were significantly associated with NSCLC. This study paves a crucial path for further investigating the genetic markers and molecular mechanism of this special NSCLC subgroup.
Collapse
Affiliation(s)
- Lawrence W Chan
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong.
| | - Fengfeng Wang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong
| | - Fei Meng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong
| | - Lili Wang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong
| | - S C Cesar Wong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong
| | - Joseph S Au
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Sijun Yang
- Institute of Animal Model for Human Diseases, ABSL-3 Laboratory and State Key Lab of Virology, Wuhan University, Wuhan, Hubei, China
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong.
| |
Collapse
|
14
|
Li W, Zhang X, Li Z, Jiang F, Zhao H, Wei B. Identification of genes associated with matrix metalloproteinases in invasive lung adenocarcinoma. Oncol Lett 2018; 16:123-130. [PMID: 29928392 PMCID: PMC6006458 DOI: 10.3892/ol.2018.8683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 02/07/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to identify genes with similar function to that of matrix metalloproteinases (MMPs) in invasive lung adenocarcinoma (AC) and to screen the transcription factors that regulate MMPs. The gene expression dataset GSE2514, including 20 invasive lung AC samples and 19 adjacent normal lung samples, was downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were screened using the limma package in R. Genes with similar function to MMPs were identified by K-means clustering. Their correlations with MMPs were validated using Pearson correlation analysis. The expression of MMPs in lung cancer and normal tissues was evaluated by western blot analysis. Protein-protein interaction (PPI) network and transcriptional regulatory network analyses were performed with Retrieval of Interacting Genes and Database for Annotation, Visualization and Integrated Discovery, respectively. As a result, 269 DEGs were identified between invasive lung AC samples and normal lung samples, including 78 upregulated and 191 downregulated genes. Four MMPs (MMP1, MMP7, MMP9 and MMP12), which were upregulated in lung AC, were clustered into one group with other genes, including NAD(P)H quinone oxidoreductase 1, claudin 3 (CLDN3), S100 calcium-binding protein P, serine protease inhibitor Kazal type 1, collagen type XI α 1 chain, periostin and desmoplakin (DSP), following cluster analysis. Pearson correlation analysis further confirmed correlations between MMP9-CLDN3, MMP9-DSP and MMP12-DSP. PPI network analysis also indicated multiple interactions between MMPs-associated genes. Furthermore, MMPs were commonly regulated by CCAAT/enhancer binding protein α transcription factor. These findings may provide further insight into the mechanisms of MMPs in invasive lung AC.
Collapse
Affiliation(s)
- Weiqing Li
- Department of Thoracic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| | - Xugang Zhang
- Department of Thoracic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| | - Zhitian Li
- Department of Thoracic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| | - Fusheng Jiang
- Department of Thoracic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| | - Hongwei Zhao
- Department of Interventional Treatment, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| | - Bo Wei
- Department of Thoracic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| |
Collapse
|
15
|
Procházková J, Strapáčová S, Svržková L, Andrysík Z, Hýžďalová M, Hrubá E, Pěnčíková K, Líbalová H, Topinka J, Kléma J, Espinosa JM, Vondráček J, Machala M. Adaptive changes in global gene expression profile of lung carcinoma A549 cells acutely exposed to distinct types of AhR ligands. Toxicol Lett 2018; 292:162-174. [PMID: 29704546 DOI: 10.1016/j.toxlet.2018.04.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 03/28/2018] [Accepted: 04/17/2018] [Indexed: 12/19/2022]
Abstract
Exposure to persistent ligands of aryl hydrocarbon receptor (AhR) has been found to cause lung cancer in experimental animals, and lung adenocarcinomas are often associated with enhanced AhR expression and aberrant AhR activation. In order to better understand the action of toxic AhR ligands in lung epithelial cells, we performed global gene expression profiling and analyze TCDD-induced changes in A549 transcriptome, both sensitive and non-sensitive to CH223191 co-treatment. Comparison of our data with results from previously reported microarray and ChIP-seq experiments enabled us to identify candidate genes, which expression status reflects exposure of lung cancer cells to TCDD, and to predict processes, pathways (e.g. ER stress, Wnt/β-cat, IFNɣ, EGFR/Erbb1), putative TFs (e.g. STAT, AP1, E2F1, TCF4), which may be implicated in adaptive response of lung cells to TCDD-induced AhR activation. Importantly, TCDD-like expression fingerprint of selected genes was observed also in A549 cells exposed acutely to both toxic (benzo[a]pyrene, benzo[k]fluoranthene) and endogenous AhR ligands (2-(1H-Indol-3-ylcarbonyl)-4-thiazolecarboxylic acid methyl ester and 6-formylindolo[3,2-b]carbazole). Overall, our results suggest novel cellular candidates, which could help to improve monitoring of AhR-dependent transcriptional activity during acute exposure of lung cells to distinct types of environmental pollutants.
Collapse
Affiliation(s)
- Jiřina Procházková
- Department of Chemistry and Toxicology, Veterinary Research Institute, Brno, Czech Republic
| | - Simona Strapáčová
- Department of Chemistry and Toxicology, Veterinary Research Institute, Brno, Czech Republic
| | - Lucie Svržková
- Department of Chemistry and Toxicology, Veterinary Research Institute, Brno, Czech Republic
| | - Zdeněk Andrysík
- 1 Linda Crnic Institute for Down Syndrome, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Martina Hýžďalová
- Department of Chemistry and Toxicology, Veterinary Research Institute, Brno, Czech Republic
| | - Eva Hrubá
- Department of Chemistry and Toxicology, Veterinary Research Institute, Brno, Czech Republic
| | - Kateřina Pěnčíková
- Department of Chemistry and Toxicology, Veterinary Research Institute, Brno, Czech Republic
| | - Helena Líbalová
- Department of Genetic Toxicology and Nanotoxicology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Topinka
- Department of Genetic Toxicology and Nanotoxicology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiří Kléma
- Department of Computer Science, Czech Technical University in Prague, Czech Republic
| | - Joaquín M Espinosa
- 1 Linda Crnic Institute for Down Syndrome, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Miroslav Machala
- Department of Chemistry and Toxicology, Veterinary Research Institute, Brno, Czech Republic.
| |
Collapse
|
16
|
Gao X, Wu Y, Yu W, Li H. Identification of a seven-miRNA signature as prognostic biomarker for lung squamous cell carcinoma. Oncotarget 2018; 7:81670-81679. [PMID: 27835574 PMCID: PMC5348421 DOI: 10.18632/oncotarget.13164] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 10/19/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Specific biomarkers for outcome prediction of lung squamous cell carcinoma (LUSC) are still lacking. This study assessed the prognostic value of differentially expressed miRNAs of LUSC patients. RESULTS Twelve of the 133 most significantly altered miRNAs were associated with overall survival (OS) across different clinical subclasses of the Cancer Genome Atlas (TCGA) LUSC cohort. A linear prognostic model of seven miRNAs was developed to divide patients into high- and low-risk groups. Patients assigned to the high-risk group exhibited poor OS compared with patients in the low-risk group, which was further validated in the validation cohort and entire LUSC cohort. METHODS MiRNA expression profiles with clinical information of 447 LUSC patients were obtained from TCGA. Most significantly altered miRNAs were identified between tumor and normal samples. Using survival analysis and supervised principal components method, a seven-miRNA signature for prediction of OS of LUSC patients was established. Survival receiver operating characteristic (ROC) analysis was used to assess the performance of survival prediction. The biological relevance of predicted miRNA targets was also analyzed using bioinformatics method. CONCLUSIONS The current study suggests that seven-miRNA signature may have clinical implications in the outcome prediction of LUSC.
Collapse
Affiliation(s)
- Xujie Gao
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center of Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Yupeng Wu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center of Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Wenwen Yu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center of Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Hui Li
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center of Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| |
Collapse
|
17
|
Cabel L, Fuerea A, Lacroix L, Baldini C, Martin P, Hollebecque A, Postel-Vinay S, Varga A, Balheda R, Gazzah A, Michot JM, Marabelle A, Rouleau E, Solary E, De Baere T, Angevin E, Armand JP, Michiels S, Scoazec JY, Ammari S, André F, Soria JC, Massard C, Verlingue L. Efficacy of histology-agnostic and molecularly-driven HER2 inhibitors for refractory cancers. Oncotarget 2018. [PMID: 29515767 PMCID: PMC5839398 DOI: 10.18632/oncotarget.24188] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A targeted therapy is recommended in case of ERBB2 alteration for breast and gastric carcinomas, but miscellaneous other tumor types are ERBB2-altered at low prevalence. Broadening the administration of HER2 inhibitors across tumor types and genomic alterations could benefit to patients with refractory metastatic tumors. Targeted next-generation-sequencing (tNGS) and comparative genomic hybridization array (CGH) have been performed on fresh tumor biopsies of patients included in the MOSCATO-01 and ongoing MOSCATO-02 trials to administrate HER2 inhibitors in case of ERBB2 pathogenic mutation of amplification. Between December 2011 and January 2017 a molecular analysis was performed for 934 patients (759 CGH and 912 tNGS). A novel ERBB2 alteration has been found in 4.7% (n = 44/934), including 1.5% (n = 14/912) ERBB2 mutations, and 4% (n = 30/759) ERBB2 amplifications. A matched HER2 inhibitor was administrated to 70% (31/44) of patients and consisted in trastuzumab plus chemotherapy for 90% of them (28/31). On the 31 evaluable patients, 1 complete response (CR), 10 partial response (PR) and 2 stable disease (SD) >24 weeks were observed accounting for a clinical benefit rate (CBR) of 42% (n = 13/31, 95% CI 25–61%). Besides breast and oesogastric carcinomas, 19 patients affected by 8 different tumor types had a CBR of 25% for ERBB2 mutations (n = 2/8, 95% CI 3%–65%, with 2 PR) and 64% for ERBB2 amplifications (n = 7/11, 95% CI 31%–89%; with 1 CR, 4 PR, 2 SD). ERBB2 genomic alterations were diffuse across metastatic tumor types and signs of efficacy emerged for HER2 targeted treatments, especially in case of ERBB2 amplifications or a p.S310Y ERBB2 mutation.
Collapse
Affiliation(s)
- Luc Cabel
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Alina Fuerea
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Ludovic Lacroix
- Laboratory of Translational Research and Biological Resource Center, AMMICA, INSERM US23/CNRS UMS3655 Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France.,Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - Capucine Baldini
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Patricia Martin
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Antoine Hollebecque
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France.,Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Sophie Postel-Vinay
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Andrea Varga
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Rastilav Balheda
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Anas Gazzah
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Jean-Marie Michot
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Aurélien Marabelle
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Etienne Rouleau
- Laboratory of Translational Research and Biological Resource Center, AMMICA, INSERM US23/CNRS UMS3655 Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France.,Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - Eric Solary
- Inserm Unit UMR 1170, Université Paris Saclay, Université Paris-Sud, Gustave Roussy, Villejuif, France
| | - Thierry De Baere
- Department of Interventional Radiology, Gustave Roussy, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Eric Angevin
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Jean-Pierre Armand
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Stefan Michiels
- Service de Biostatistique et d'Épidémiologie, Gustave Roussy, Villejuif, France
| | - Jean Yves Scoazec
- Laboratory of Translational Research and Biological Resource Center, AMMICA, INSERM US23/CNRS UMS3655 Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France.,Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - Samy Ammari
- Department of Radiology, Gustave Roussy, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Fabrice André
- Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France.,Inserm Unit U981, Université Paris Saclay, Université Paris-Sud, Gustave Roussy, Villejuif, France
| | - Jean-Charles Soria
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France.,Inserm Unit U981, Université Paris Saclay, Université Paris-Sud, Gustave Roussy, Villejuif, France
| | - Christophe Massard
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| | - Loic Verlingue
- Drug Development Department (DITEP), Gustave Roussy Department of Medical Oncology, Faculté de Medicine Paris-Sud XI, Villejuif, France
| |
Collapse
|
18
|
Jabs V, Edlund K, König H, Grinberg M, Madjar K, Rahnenführer J, Ekman S, Bergkvist M, Holmberg L, Ickstadt K, Botling J, Hengstler JG, Micke P. Integrative analysis of genome-wide gene copy number changes and gene expression in non-small cell lung cancer. PLoS One 2017; 12:e0187246. [PMID: 29112949 PMCID: PMC5675410 DOI: 10.1371/journal.pone.0187246] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 10/17/2017] [Indexed: 12/27/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) represents a genomically unstable cancer type with extensive copy number aberrations. The relationship of gene copy number alterations and subsequent mRNA levels has only fragmentarily been described. The aim of this study was to conduct a genome-wide analysis of gene copy number gains and corresponding gene expression levels in a clinically well annotated NSCLC patient cohort (n = 190) and their association with survival. While more than half of all analyzed gene copy number-gene expression pairs showed statistically significant correlations (10,296 of 18,756 genes), high correlations, with a correlation coefficient >0.7, were obtained only in a subset of 301 genes (1.6%), including KRAS, EGFR and MDM2. Higher correlation coefficients were associated with higher copy number and expression levels. Strong correlations were frequently based on few tumors with high copy number gains and correspondingly increased mRNA expression. Among the highly correlating genes, GO groups associated with posttranslational protein modifications were particularly frequent, including ubiquitination and neddylation. In a meta-analysis including 1,779 patients we found that survival associated genes were overrepresented among highly correlating genes (61 of the 301 highly correlating genes, FDR adjusted p<0.05). Among them are the chaperone CCT2, the core complex protein NUP107 and the ubiquitination and neddylation associated protein CAND1. In conclusion, in a comprehensive analysis we described a distinct set of highly correlating genes. These genes were found to be overrepresented among survival-associated genes based on gene expression in a large collection of publicly available datasets.
Collapse
Affiliation(s)
- Verena Jabs
- Faculty of Statistics, TU Dortmund University, Dortmund, Germany
| | - Karolina Edlund
- Leibniz Research Centre for Working Environment and Human Factors (IfADo) at Dortmund University, Dortmund, Germany
| | - Helena König
- Faculty of Statistics, TU Dortmund University, Dortmund, Germany
| | | | - Katrin Madjar
- Faculty of Statistics, TU Dortmund University, Dortmund, Germany
| | | | - Simon Ekman
- Department of Oncology, Karolinska University Hospital, Stockholm, Sweden
| | | | - Lars Holmberg
- Regional Cancer Center Uppsala-Örebro, Uppsala, Sweden
- King’s College London, Faculty of Life Sciences and Medicine, Division of Cancer Studies, London, United Kingdom
| | - Katja Ickstadt
- Faculty of Statistics, TU Dortmund University, Dortmund, Germany
| | - Johan Botling
- Dept. of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo) at Dortmund University, Dortmund, Germany
| | - Patrick Micke
- Dept. of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
19
|
Novikova SE, Kurbatov LK, Zavialova MG, Zgoda VG, Archakov AI. [Omics technologies in diagnostics of lung adenocarcinoma]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2017; 63:181-210. [PMID: 28781253 DOI: 10.18097/pbmc20176303181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
To date lung adenocarcinoma (LAC) is the most common type of lung cancer. Numerous studies on LAC biology resulted in identification of crucial mutations in protooncogenes and activating neoplastic transformation pathways. Therapeutic approaches that significantly increase the survival rate of patients with LAC of different etiology have been developed and introduced into clinical practice. However, the main problem in the treatment of LAC is early diagnosis, taking into account both factors and mechanisms responsible in tumor initiation and progression. Identification of a wide biomarker repertoire with high specificity and reliability of detection appears to be a solution to this problem. In this context, proteins with differential expression in normal and pathological condition, suitable for detection in biological fluids are the most promising biomarkers. In this review we have analyzed literature data on studies aimed at search of LAC biomarkers. The major attention has been paid to protein biomarkers as the most promising and convenient subject of clinical diagnosis. The review also summarizes existing knowledge on posttranslational modifications, splice variants, isoforms, as well as model systems and transcriptome changes in LAC.
Collapse
Affiliation(s)
- S E Novikova
- Institute of Biomedical Chemistry, Moscow, Russia
| | - L K Kurbatov
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | - V G Zgoda
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A I Archakov
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
20
|
Differential gene expression profiles according to the Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society histopathological classification in lung adenocarcinoma subtypes. Hum Pathol 2017; 66:188-199. [DOI: 10.1016/j.humpath.2017.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/26/2017] [Accepted: 06/01/2017] [Indexed: 12/29/2022]
|
21
|
Harttrampf AC, Lacroix L, Deloger M, Deschamps F, Puget S, Auger N, Vielh P, Varlet P, Balogh Z, Abbou S, Allorant A, Valteau-Couanet D, Sarnacki S, Gamiche-Rolland L, Meurice G, Minard-Colin V, Grill J, Brugieres L, Dufour C, Gaspar N, Michiels S, Vassal G, Soria JC, Geoerger B. Molecular Screening for Cancer Treatment Optimization (MOSCATO-01) in Pediatric Patients: A Single-Institutional Prospective Molecular Stratification Trial. Clin Cancer Res 2017; 23:6101-6112. [PMID: 28733441 DOI: 10.1158/1078-0432.ccr-17-0381] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 04/21/2017] [Accepted: 07/17/2017] [Indexed: 11/16/2022]
Abstract
Purpose: This single-institutional feasibility study prospectively characterized genomic alterations in recurrent or refractory solid tumors of pediatric patients to select a targeted therapy.Experimental Design: Following treatment failure, patients with signed consent and ages above 6 months, underwent tumor biopsy or surgical resection of primary or metastatic tumor site. These newly acquired samples were analyzed by comparative genomic hybridization array, next-generation sequencing for 75 target genes, whole-exome and RNA sequencing. Biological significance of the alterations and suggestion of most relevant targeted therapies available were discussed in a multidisciplinary tumor board.Results: From December 2012 to January 2016, 75 patients were included, 73 patients underwent 79 interventions, 56 of which were research biopsies with a low complication rate. All patients were pretreated, 37.0% had a brain tumor, and 63.0% had an extra-cranial solid tumor. Median tumor cell content was 70% (range, 0%-100%). Successful molecular analysis in 69 patients detected in 60.9% of patients an actionable alteration in various oncogenic pathways (42.4% with copy-number change, 33.3% with mutation, 2.1% with fusion), and change in diagnosis in three patients. Fourteen patients received 17 targeted therapies; two had received a matched treatment before inclusion.Conclusions: Research biopsies are feasible in advanced pediatric malignancies that exhibit a considerable amount of potentially actionable alterations. Genetic events affecting different cancer hallmarks and limited access to targeted agents within pediatric clinical trials remain the main obstacles that are addressed in our two subsequent precision medicine studies MAPPYACTS and AcSé-ESMART. Clin Cancer Res; 23(20); 6101-12. ©2017 AACR.
Collapse
Affiliation(s)
- Anne C Harttrampf
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Ludovic Lacroix
- Laboratory of Translational Research and Biobank, AMMICA, INSERM US23/CNRS UMS3655, Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France.,Gustave Roussy, Medical Biology and Pathology Department, Villejuif, France
| | - Marc Deloger
- Gustave Roussy, Bioinformatics Platform, AMMICA, INSERM US23/CNRS UMS3655, Villejuif, France
| | | | - Stephanie Puget
- Necker Enfants Malades, Department of Pediatric Neurosurgery, University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Nathalie Auger
- Gustave Roussy, Medical Biology and Pathology Department, Villejuif, France
| | - Philippe Vielh
- Laboratory of Translational Research and Biobank, AMMICA, INSERM US23/CNRS UMS3655, Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France.,Gustave Roussy, Medical Biology and Pathology Department, Villejuif, France
| | - Pascale Varlet
- Sainte-Anne Hospital, Department of Neuropathology, University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Zsofia Balogh
- Laboratory of Translational Research and Biobank, AMMICA, INSERM US23/CNRS UMS3655, Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - Samuel Abbou
- Gustave Roussy, Department of Pediatric and Adolescent Oncology, Villejuif, France
| | - Adrien Allorant
- Gustave Roussy, Biostatistics and Epidemiology, Univers. Paris-Sud, Villejuif, France
| | | | - Sabine Sarnacki
- Necker Enfants Malades, Department of Pediatric Surgery, University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Guillaume Meurice
- Gustave Roussy, Bioinformatics Platform, AMMICA, INSERM US23/CNRS UMS3655, Villejuif, France
| | | | - Jacques Grill
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,Gustave Roussy, Department of Pediatric and Adolescent Oncology, Villejuif, France
| | - Laurence Brugieres
- Gustave Roussy, Department of Pediatric and Adolescent Oncology, Villejuif, France
| | - Christelle Dufour
- Gustave Roussy, Department of Pediatric and Adolescent Oncology, Villejuif, France
| | - Nathalie Gaspar
- Gustave Roussy, Department of Pediatric and Adolescent Oncology, Villejuif, France
| | - Stefan Michiels
- Gustave Roussy, Biostatistics and Epidemiology, Univers. Paris-Sud, Villejuif, France
| | - Gilles Vassal
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,Gustave Roussy, Direction of Clinical Research, Univers. Paris-Sud, Villejuif, France
| | - Jean-Charles Soria
- Gustave Roussy, Department of Innovative Therapeutics and Early Trials, Univers. Paris-Sud, Villejuif, France
| | - Birgit Geoerger
- Vectorology and Anticancer Therapies, UMR 8203, CNRS, Univ. Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif, France. .,Gustave Roussy, Department of Pediatric and Adolescent Oncology, Villejuif, France
| |
Collapse
|
22
|
Pan T, Chen W, Yuan X, Shen J, Qin C, Wang L. miR-944 inhibits metastasis of gastric cancer by preventing the epithelial-mesenchymal transition via MACC1/Met/AKT signaling. FEBS Open Bio 2017; 7:905-914. [PMID: 28680805 PMCID: PMC5494292 DOI: 10.1002/2211-5463.12215] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 02/02/2017] [Accepted: 03/01/2017] [Indexed: 02/05/2023] Open
Abstract
MicroRNAs (miRNAs) are reported to play vital roles in tumor progression. Recently, miR-944 was reported to play either an oncogenic or tumor suppressive role in human cancers. However, the expression of miR-944 and its exact role in gastric cancer (GC) remain unknown. This study aimed to evaluate whether loss of miR-944 could promote the epithelial-mesenchymal transition (EMT) of GC. Reduced expression of miR-944 was identified in 40 pairs of human GC and matched normal tissues by qRT-PCR. Reduced expression of mi-944 was also observed in GC cell lines. Restoration of miR-944 inhibited cell migration and invasion in MGC-803 cells, while its loss facilitated metastasis of SGC-7901 and BGC-823 cells. Notably, miR-944 overexpression prohibited EMT of GC cells in vitro, while miR-944 knockdown had the opposite effect. Bioinformatics software predicted that MACC1 was a direct target of miR-944. We observed negative regulation of miR-944 on MACC1 expression, and direct binding between miR-944 and MACC1 was verified by dual-luciferase assays in HEK293T cells. Restoration of MACC1 resulted in promoted EMT and metastasis in miR-944-overexpressing MGC-803 cells. Loss of MACC1 abrogated the effects of miR-944 knockdown on EMT and metastasis of SGC-7901 cells. We also found that the Met-AKT pathway might be involved in MACC1-mediated EMT. In conclusion, miR-944 acts as an inhibitor of EMT and metastasis of GC by targeting MACC1. This study highlights the potential effects of miR-944 in the prognosis and treatment of GC.
Collapse
Affiliation(s)
- Tao Pan
- Department of Surgical Oncology Sir Run Run Shaw Hospital School of Medicine Zhejiang University Hangzhou China
| | - Wenjun Chen
- Department of Surgical Oncology Sir Run Run Shaw Hospital School of Medicine Zhejiang University Hangzhou China
| | - Xiaoming Yuan
- Department of Surgical Oncology Sir Run Run Shaw Hospital School of Medicine Zhejiang University Hangzhou China
| | - Jun Shen
- Department of Surgical Oncology Sir Run Run Shaw Hospital School of Medicine Zhejiang University Hangzhou China
| | - Chuan Qin
- Department of Surgical Oncology Sir Run Run Shaw Hospital School of Medicine Zhejiang University Hangzhou China
| | - Linbo Wang
- Department of Surgical Oncology Sir Run Run Shaw Hospital School of Medicine Zhejiang University Hangzhou China
| |
Collapse
|
23
|
Wen L, Li Y, Jiang Z, Zhang Y, Yang B, Han F. miR-944 inhibits cell migration and invasion by targeting MACC1 in colorectal cancer. Oncol Rep 2017; 37:3415-3422. [PMID: 28498456 DOI: 10.3892/or.2017.5611] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 04/13/2017] [Indexed: 11/06/2022] Open
Abstract
Dysfunction of microRNAs (miRNAs) is strongly proved to participate in the pathogenesis and tumorigenicity of colorectal cancer (CRC). miR-944 was reported to play either oncogenic or tumor suppressive roles in human cancers. A recent study reported that the levels of miR-944 in recurrent CRC patients were evidently lower than that in non-recurrent cases, suggesting that miR-944 may function as a tumor suppressive miRNA in CRC. Yet, the clinical value and biological function of miR-944 remain rarely known in CRC. In the present study, we present that miR-944 level in CRC tissues is notably reduced compared to matched non-cancerous specimens. Its decreased level is evidently correlated with malignant clinical parameters and poor prognosis of CRC patients. Accordingly, the levels of miR-944 were obviously downregulated in CRC cells. Ectopic expression of miR-944 in CRC cells prominently inhibits the migration and invasion of tumor cells, while miR-944 knockdown increased these effects of CRC cells. Mechanically, miR-944 negatively regulated the metastasis-associated in colon cancer-1 (MACC1) abundance in CRC cells. Herein, MACC1 was found to be a downstream molecule of miR-944 in CRC. An inversely correlation between miR-944 and MACC1 was confirmed in CRC specimens. Furthermore, restoration of MACC1 expression could abrogate the anti-metastatic effects of miR-944 on CRC cells with enhanced cell migration and invasion. MACC1/Met/AKT signaling may be implicated with the function of miR-944 in CRC cells. Altogether, miR-944 potentially act as a prognostic predictor and a drug-target for CRC patients.
Collapse
Affiliation(s)
- Liqiang Wen
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yingru Li
- Department of Gastroenterology, Hernia and Abdominal Wall Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Zhipeng Jiang
- Department of Gastroenterology, Hernia and Abdominal Wall Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Yuchao Zhang
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Bin Yang
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Fanghai Han
- Department of Gastrointestinal Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
24
|
miR-944 acts as a prognostic marker and promotes the tumor progression in endometrial cancer. Biomed Pharmacother 2017; 88:902-910. [DOI: 10.1016/j.biopha.2017.01.117] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/09/2017] [Accepted: 01/19/2017] [Indexed: 12/11/2022] Open
|
25
|
Massard C, Michiels S, Ferté C, Le Deley MC, Lacroix L, Hollebecque A, Verlingue L, Ileana E, Rosellini S, Ammari S, Ngo-Camus M, Bahleda R, Gazzah A, Varga A, Postel-Vinay S, Loriot Y, Even C, Breuskin I, Auger N, Job B, De Baere T, Deschamps F, Vielh P, Scoazec JY, Lazar V, Richon C, Ribrag V, Deutsch E, Angevin E, Vassal G, Eggermont A, André F, Soria JC. High-Throughput Genomics and Clinical Outcome in Hard-to-Treat Advanced Cancers: Results of the MOSCATO 01 Trial. Cancer Discov 2017; 7:586-595. [DOI: 10.1158/2159-8290.cd-16-1396] [Citation(s) in RCA: 415] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/06/2017] [Accepted: 03/07/2017] [Indexed: 11/16/2022]
|
26
|
Hou M, Cheng Z, Shen H, He S, Li Y, Pan Y, Feng C, Chen X, Zhang Y, Lin M, Wang L, Ke Z. High expression of CTHRC1 promotes EMT of epithelial ovarian cancer (EOC) and is associated with poor prognosis. Oncotarget 2016; 6:35813-29. [PMID: 26452130 PMCID: PMC4742143 DOI: 10.18632/oncotarget.5358] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 09/21/2015] [Indexed: 12/18/2022] Open
Abstract
Collagen triple helix repeat-containing 1 (CTHRC1) is aberrantly overexpressed in multiple malignant tumors. However, the expression characteristics and function of CTHRC1 in epithelial ovarian cancer (EOC) remain unclear. We found that CTHRC1 expression was up-regulated in the paraffin-embedded EOC tissues compared to borderline or benign tumor tissues. CTHRC1 expression was positively correlated with tumor size (p = 0.008), menopause (p = 0.037), clinical stage (p = 0.002) and lymph node metastasis (p < 0.001) and was also an important prognostic factor for the overall survival of EOC patients, as revealed by Kaplan-Meier analysis. CTHRC1 increased the invasive capabilities of EOC cells in vitro by activating the Wnt/β-catenin signaling pathway. We showed that ectopic transfection of CTHRC1 in EOC cells up-regulated the expression of EMT markers such as N-cadherin and vimentin, and EMT-associated transcriptional factor Snail. Knockdown of CTHRC1 expression in EOC cells resulted in down-regulation of N-cadherin, vimentin, Snail and translocation of β-catenin. Collectively, CTHRC1 may promote EOC metastasis through the induction of EMT process and serve as a potential biomarker for prognosis as well as a target for therapy.
Collapse
Affiliation(s)
- Minzhi Hou
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China.,Department of Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China
| | - Zhiqiang Cheng
- Department of Pathology, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Shenzhen, Guangdong, P.R. China
| | - Hongwei Shen
- Department of Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China
| | - Shanyang He
- Department of Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China
| | - Yang Li
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China
| | - Yunping Pan
- Department of Stomatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China
| | - Chongjin Feng
- Department of Stomatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China
| | - Xinlin Chen
- Department of Preventive Medicine and Biostatistics, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Yang Zhang
- Biomedical Engineering, University of Texas at El Paso, El Paso, Texas, USA
| | - Millicent Lin
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging (CIMI), California NanoSystems Institute (CNSI), University of California, Los Angeles, California, USA
| | - Liantang Wang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China
| |
Collapse
|
27
|
Girard L, Rodriguez-Canales J, Behrens C, Thompson DM, Botros IW, Tang H, Xie Y, Rekhtman N, Travis WD, Wistuba II, Minna JD, Gazdar AF. An Expression Signature as an Aid to the Histologic Classification of Non-Small Cell Lung Cancer. Clin Cancer Res 2016; 22:4880-4889. [PMID: 27354471 PMCID: PMC5492382 DOI: 10.1158/1078-0432.ccr-15-2900] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 06/12/2016] [Indexed: 12/15/2022]
Abstract
PURPOSE Most non-small cell lung cancers (NSCLC) are now diagnosed from small specimens, and classification using standard pathology methods can be difficult. This is of clinical relevance as many therapy regimens and clinical trials are histology dependent. The purpose of this study was to develop an mRNA expression signature as an adjunct test for routine histopathologic classification of NSCLCs. EXPERIMENTAL DESIGN A microarray dataset of resected adenocarcinomas (ADC) and squamous cell carcinomas (SCC) was used as the learning set for an ADC-SCC signature. The Cancer Genome Atlas (TCGA) lung RNAseq dataset was used for validation. Another microarray dataset of ADCs and matched nonmalignant lung was used as the learning set for a tumor versus nonmalignant signature. The classifiers were selected as the most differentially expressed genes and sample classification was determined by a nearest distance approach. RESULTS We developed a 62-gene expression signature that contained many genes used in immunostains for NSCLC typing. It includes 42 genes that distinguish ADC from SCC and 20 genes differentiating nonmalignant lung from lung cancer. Testing of the TCGA and other public datasets resulted in high prediction accuracies (93%-95%). In addition, a prediction score was derived that correlates both with histologic grading and prognosis. We developed a practical version of the Classifier using the HTG EdgeSeq nuclease protection-based technology in combination with next-generation sequencing that can be applied to formalin-fixed paraffin-embedded (FFPE) tissues and small biopsies. CONCLUSIONS Our RNA classifier provides an objective, quantitative method to aid in the pathologic diagnosis of lung cancer. Clin Cancer Res; 22(19); 4880-9. ©2016 AACR.
Collapse
Affiliation(s)
- Luc Girard
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas. Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jaime Rodriguez-Canales
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carmen Behrens
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - Hao Tang
- Department of Clinical Science, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yang Xie
- Department of Clinical Science, University of Texas Southwestern Medical Center, Dallas, Texas. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Natasha Rekhtman
- Department of Thoracic Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William D Travis
- Department of Thoracic Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John D Minna
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas. Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas. Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Adi F Gazdar
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas. Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
28
|
TFAP2C promotes lung tumorigenesis and aggressiveness through miR-183- and miR-33a-mediated cell cycle regulation. Oncogene 2016; 36:1585-1596. [PMID: 27593936 DOI: 10.1038/onc.2016.328] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/05/2016] [Accepted: 07/26/2016] [Indexed: 01/04/2023]
Abstract
Non-small cell lung cancer (NSCLC) remains one of the leading causes of death worldwide, and thus new molecular targets need to be identified to improve treatment efficacy. Although epidermal growth factor receptor (EGFR)/KRAS mutation-driven lung tumorigenesis is well understood, the mechanism of EGFR/KRAS-independent signal activation remains elusive. Enhanced TFAP2C (transcription factor activating enhancer-binding protein 2C) expression is associated with poor prognosis in some types of cancer patients, but little is known of its relation with the pathogenesis of lung cancer. In the present study, we found that TFAP2C overexpression was associated with cell cycle activation and NSCLC cell tumorigenesis. Interestingly, TFAP2C blocked AKAP12-mediated cyclin D1 inhibition by inducing the overexpression of oncogenic microRNA (miRNA)-183 and simultaneously activated cyclin-dependent kinase 6-mediated cell cycle progression by downregulating tumor-suppressive miRNA-33a. In a mouse xenograft model, TFAP2C promoted lung tumorigenesis and disease aggressiveness via the miR-183 and miR-33a pathways. The study provides a mechanism of mitogenic and oncogenic signaling via two functionally opposed miRNAs and suggests that TFAP2C-induced cell cycle hyperactivation contributes to lung tumorigenesis.
Collapse
|
29
|
Liu J, Ding X, Yuan L, Zhang X. Identification of pterygium-related long non-coding RNAs and expression profiling by microarray analysis. Int J Mol Med 2016; 38:529-36. [DOI: 10.3892/ijmm.2016.2641] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/02/2016] [Indexed: 11/06/2022] Open
|
30
|
Lazar V, Rubin E, Depil S, Pawitan Y, Martini JF, Gomez-Navarro J, Yver A, Kan Z, Dry JR, Kehren J, Validire P, Rodon J, Vielh P, Ducreux M, Galbraith S, Lehnert M, Onn A, Berger R, Pierotti MA, Porgador A, Pramesh CS, Ye DW, Carvalho AL, Batist G, Le Chevalier T, Morice P, Besse B, Vassal G, Mortlock A, Hansson J, Berindan-Neagoe I, Dann R, Haspel J, Irimie A, Laderman S, Nechushtan H, Al Omari AS, Haywood T, Bresson C, Soo KC, Osman I, Mata H, Lee JJ, Jhaveri K, Meurice G, Palmer G, Lacroix L, Koscielny S, Eterovic KA, Blay JY, Buller R, Eggermont A, Schilsky RL, Mendelsohn J, Soria JC, Rothenberg M, Scoazec JY, Hong WK, Kurzrock R. A simplified interventional mapping system (SIMS) for the selection of combinations of targeted treatments in non-small cell lung cancer. Oncotarget 2016; 6:14139-52. [PMID: 25944621 PMCID: PMC4546456 DOI: 10.18632/oncotarget.3741] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/02/2015] [Indexed: 02/06/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of death worldwide. Targeted monotherapies produce high regression rates, albeit for limited patient subgroups, who inevitably succumb. We present a novel strategy for identifying customized combinations of triplets of targeted agents, utilizing a simplified interventional mapping system (SIMS) that merges knowledge about existent drugs and their impact on the hallmarks of cancer. Based on interrogation of matched lung tumor and normal tissue using targeted genomic sequencing, copy number variation, transcriptomics, and miRNA expression, the activation status of 24 interventional nodes was elucidated. An algorithm was developed to create a scoring system that enables ranking of the activated interventional nodes for each patient. Based on the trends of co-activation at interventional points, combinations of drug triplets were defined in order to overcome resistance. This methodology will inform a prospective trial to be conducted by the WIN consortium, aiming to significantly impact survival in metastatic NSCLC and other malignancies.
Collapse
Affiliation(s)
- Vladimir Lazar
- Gustave-Roussy Cancer Center, Villejuif, France.,WIN Consortium, Villejuif, France
| | - Eitan Rubin
- Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | - Jean-François Martini
- Pfizer Oncology Research, San Diego, CA, USA.,Pfizer Oncology, Pfizer Inc, New York, NY, USA
| | | | - Antoine Yver
- AstraZeneca Pharmaceuticals LP, Global Medicines Development, Gaithersburg, MD, USA.,Oncology iMED, Waltham, MA, USA.,Oncology iMED, Macclesfield, Cheshire, UK
| | - Zhengyin Kan
- Pfizer Oncology Research, San Diego, CA, USA.,Pfizer Oncology, Pfizer Inc, New York, NY, USA
| | - Jonathan R Dry
- AstraZeneca Pharmaceuticals LP, Global Medicines Development, Gaithersburg, MD, USA.,Oncology iMED, Waltham, MA, USA.,Oncology iMED, Macclesfield, Cheshire, UK
| | | | | | - Jordi Rodon
- Vall d'Hebron Institute of Oncology Universitat Autonoma de Barcelona, Barcelona, Spain
| | | | - Michel Ducreux
- Gustave-Roussy Cancer Center, Villejuif, France.,University Paris-Sud, Kremlin-Bicetre, France
| | - Susan Galbraith
- AstraZeneca Pharmaceuticals LP, Global Medicines Development, Gaithersburg, MD, USA.,Oncology iMED, Waltham, MA, USA.,Oncology iMED, Macclesfield, Cheshire, UK
| | - Manfred Lehnert
- Takeda Pharmaceuticals International Co., Cambridge, MA, USA
| | - Amir Onn
- Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | | | | | | | | | - Ding-wei Ye
- Fudan University Shanghai Cancer Center, Shanghai, China
| | | | - Gerald Batist
- Segal Cancer Centre at the Jewish General Hospital, McGill University, Montreal, QC, Canada
| | | | | | | | | | - Andrew Mortlock
- AstraZeneca Pharmaceuticals LP, Global Medicines Development, Gaithersburg, MD, USA.,Oncology iMED, Waltham, MA, USA.,Oncology iMED, Macclesfield, Cheshire, UK
| | | | - Ioana Berindan-Neagoe
- University of Medicine and Pharmacy Iuliu Hatieganu, Cluj-Napoca, Romania.,Ion Chiricuta Oncology Institut, Cluj-Napoca, Romania
| | - Robert Dann
- General Electric Healthcare, Westborough, MA, USA
| | | | - Alexandru Irimie
- University of Medicine and Pharmacy Iuliu Hatieganu, Cluj-Napoca, Romania.,Ion Chiricuta Oncology Institut, Cluj-Napoca, Romania
| | | | | | | | - Trent Haywood
- Blue Cross Blue Shield Association, Chicago, IL, USA
| | | | | | - Iman Osman
- New York University Langone Medical Center, NY, USA
| | - Hilario Mata
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jack J Lee
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Gary Palmer
- Foundation Medicine Inc., Cambridge, MA, USA
| | | | | | | | | | - Richard Buller
- Pfizer Oncology Research, San Diego, CA, USA.,Pfizer Oncology, Pfizer Inc, New York, NY, USA
| | - Alexander Eggermont
- Gustave-Roussy Cancer Center, Villejuif, France.,University Paris-Sud, Kremlin-Bicetre, France
| | | | - John Mendelsohn
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jean-Charles Soria
- Gustave-Roussy Cancer Center, Villejuif, France.,University Paris-Sud, Kremlin-Bicetre, France
| | - Mace Rothenberg
- Pfizer Oncology Research, San Diego, CA, USA.,Pfizer Oncology, Pfizer Inc, New York, NY, USA
| | - Jean-Yves Scoazec
- Gustave-Roussy Cancer Center, Villejuif, France.,University Paris-Sud, Kremlin-Bicetre, France
| | - Waun Ki Hong
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
31
|
Marien E, Meister M, Muley T, del Pulgar TG, Derua R, Spraggins JM, Van de Plas R, Vanderhoydonc F, Machiels J, Binda MM, Dehairs J, Willette-Brown J, Hu Y, Dienemann H, Thomas M, Schnabel PA, Caprioli RM, Lacal JC, Waelkens E, Swinnen JV. Phospholipid profiling identifies acyl chain elongation as a ubiquitous trait and potential target for the treatment of lung squamous cell carcinoma. Oncotarget 2016; 7:12582-97. [PMID: 26862848 PMCID: PMC4914306 DOI: 10.18632/oncotarget.7179] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 01/04/2016] [Indexed: 01/19/2023] Open
Abstract
Lung cancer is the leading cause of cancer death. Beyond first line treatment, few therapeutic options are available, particularly for squamous cell carcinoma (SCC). Here, we have explored the phospholipidomes of 30 human SCCs and found that they almost invariably (in 96.7% of cases) contain phospholipids with longer acyl chains compared to matched normal tissues. This trait was confirmed using in situ 2D-imaging MS on tissue sections and by phospholipidomics of tumor and normal lung tissue of the L-IkkαKA/KA mouse model of lung SCC. In both human and mouse, the increase in acyl chain length in cancer tissue was accompanied by significant changes in the expression of acyl chain elongases (ELOVLs). Functional screening of differentially expressed ELOVLs by selective gene knockdown in SCC cell lines followed by phospholipidomics revealed ELOVL6 as the main elongation enzyme responsible for acyl chain elongation in cancer cells. Interestingly, inhibition of ELOVL6 drastically reduced colony formation of multiple SCC cell lines in vitro and significantly attenuated their growth as xenografts in vivo in mouse models. These findings identify acyl chain elongation as one of the most common traits of lung SCC discovered so far and pinpoint ELOVL6 as a novel potential target for cancer intervention.
Collapse
Affiliation(s)
- Eyra Marien
- KU Leuven – University of Leuven, LKI - Leuven Cancer Institute, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven, Belgium
| | - Michael Meister
- Thoraxklinik at University Hospital Heidelberg, Translational Research Unit, Heidelberg, Germany
- TLRC-H – Translational Lung Research Center Heidelberg, Member of The German Center for Lung Research, Heidelberg, Germany
| | - Thomas Muley
- Thoraxklinik at University Hospital Heidelberg, Translational Research Unit, Heidelberg, Germany
- TLRC-H – Translational Lung Research Center Heidelberg, Member of The German Center for Lung Research, Heidelberg, Germany
| | | | - Rita Derua
- KU Leuven – University of Leuven, Department of Cellular and Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, Leuven, Belgium
| | - Jeffrey M. Spraggins
- Vanderbilt University Medical Center, Department of Biochemistry and Mass Spectrometry Research Center, Nashville, TN, USA
| | - Raf Van de Plas
- Vanderbilt University Medical Center, Department of Biochemistry and Mass Spectrometry Research Center, Nashville, TN, USA
- Delft University of Technology, Delft Center for Systems and Control, Delft, The Netherlands
| | - Frank Vanderhoydonc
- KU Leuven – University of Leuven, LKI - Leuven Cancer Institute, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven, Belgium
| | - Jelle Machiels
- KU Leuven – University of Leuven, LKI - Leuven Cancer Institute, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven, Belgium
| | - Maria Mercedes Binda
- KU Leuven – University of Leuven, LKI - Leuven Cancer Institute, Department of Oncology, Abdominal Surgical Oncology, Leuven, Belgium
| | - Jonas Dehairs
- KU Leuven – University of Leuven, LKI - Leuven Cancer Institute, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven, Belgium
| | - Jami Willette-Brown
- National Cancer Institute, Centre for Cancer Research, Cancer and Inflammation Program, Frederick, MD, USA
| | - Yinling Hu
- National Cancer Institute, Centre for Cancer Research, Cancer and Inflammation Program, Frederick, MD, USA
| | - Hendrik Dienemann
- TLRC-H – Translational Lung Research Center Heidelberg, Member of The German Center for Lung Research, Heidelberg, Germany
- Thoraxklinik at University Hospital Heidelberg, Department of Surgery, Heidelberg, Germany
| | - Michael Thomas
- TLRC-H – Translational Lung Research Center Heidelberg, Member of The German Center for Lung Research, Heidelberg, Germany
- Thoraxklinik at University Hospital Heidelberg, Department of Thoracic Oncology, Heidelberg, Germany
| | - Philipp A. Schnabel
- TLRC-H – Translational Lung Research Center Heidelberg, Member of The German Center for Lung Research, Heidelberg, Germany
- University of The Saarland, Institut für Allgemeine und Spezielle Pathologie, Homburg/Saar, Germany
| | - Richard M. Caprioli
- Vanderbilt University Medical Center, Department of Biochemistry and Mass Spectrometry Research Center, Nashville, TN, USA
| | - Juan Carlos Lacal
- Fundación Jiménez Díaz, Division of Translational Oncology, Madrid, Spain
| | - Etienne Waelkens
- KU Leuven – University of Leuven, Department of Cellular and Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, Leuven, Belgium
| | - Johannes V. Swinnen
- KU Leuven – University of Leuven, LKI - Leuven Cancer Institute, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven, Belgium
| |
Collapse
|
32
|
Schwaederle M, Parker BA, Schwab RB, Daniels GA, Piccioni DE, Kesari S, Helsten TL, Bazhenova LA, Romero J, Fanta PT, Lippman SM, Kurzrock R. Precision Oncology: The UC San Diego Moores Cancer Center PREDICT Experience. Mol Cancer Ther 2016; 15:743-52. [PMID: 26873727 DOI: 10.1158/1535-7163.mct-15-0795] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 02/06/2016] [Indexed: 11/16/2022]
Abstract
By profiling their patients' tumors, oncologists now have the option to use molecular results to match patients with drug(s) based on specific biomarkers. In this observational study, 347 patients with solid advanced cancers and next-generation sequencing (NGS) results were evaluated. Outcomes for patients who received a "matched" versus "unmatched" therapy following their NGS results were compared. Eighty-seven patients (25%) were treated with a "matched" therapy, 93 (26.8%) with an "unmatched" therapy. More patients in the matched group achieved stable disease (SD) ≥ 6 months/partial response (PR)/complete response (CR), 34.5% vs. 16.1%, (P ≤ 0.020 multivariable or propensity score methods). Matched patients had a longer median progression-free survival (PFS; 4.0 vs. 3.0 months, P = 0.039 in the Cox regression model). In analysis using PFS1 (PFS on the prior line of therapy) as a comparator to PFS after NGS, as expected, the unmatched group demonstrated a PFS2 significantly shorter than PFS1 (P = 0.009); however, this shortening was not observed in the matched patients (P = 0.595). Furthermore, 45.3% of the matched patients (24/53) had a PFS2/PFS1 ratio ≥1.3 compared with 19.3% of patients (11/57) in the unmatched group (P = 0.004 univariable and P ≥ 0.057 in multivariable/propensity score analysis). Patients with a "matching-score" (the number of matched drugs divided by the number of aberrations; unmatched patients had a score of zero) > 0.2 had a median overall survival (OS) of 15.7 months compared with 10.6 months when their matching-score was ≤ 0.2, (P = 0.040 in the Cox regression model). Matched versus unmatched patients had higher rates of SD ≥ 6 months/PR/CR and longer PFS, and improvement in OS correlated with a higher matching score in multivariable analysis. Mol Cancer Ther; 15(4); 743-52. ©2016 AACR.
Collapse
Affiliation(s)
- Maria Schwaederle
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UCSD Moores Cancer Center, La Jolla, California.
| | - Barbara A Parker
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UCSD Moores Cancer Center, La Jolla, California
| | - Richard B Schwab
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UCSD Moores Cancer Center, La Jolla, California
| | - Gregory A Daniels
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UCSD Moores Cancer Center, La Jolla, California
| | - David E Piccioni
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UCSD Moores Cancer Center, La Jolla, California
| | - Santosh Kesari
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UCSD Moores Cancer Center, La Jolla, California
| | - Teresa L Helsten
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UCSD Moores Cancer Center, La Jolla, California
| | - Lyudmila A Bazhenova
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UCSD Moores Cancer Center, La Jolla, California
| | - Julio Romero
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UCSD Moores Cancer Center, La Jolla, California
| | - Paul T Fanta
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UCSD Moores Cancer Center, La Jolla, California
| | - Scott M Lippman
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UCSD Moores Cancer Center, La Jolla, California
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UCSD Moores Cancer Center, La Jolla, California
| |
Collapse
|
33
|
Pelosi G, Fabbri A, Papotti M, Rossi G, Cavazza A, Righi L, Tamborini E, Perrone F, Settanni G, Busico A, Testi MA, Maisonneuve P, De Braud F, Garassino M, Valeri B, Sonzogni A, Pastorino U. Dissecting Pulmonary Large-Cell Carcinoma by Targeted Next Generation Sequencing of Several Cancer Genes Pushes Genotypic-Phenotypic Correlations to Emerge. J Thorac Oncol 2015; 10:1560-9. [DOI: 10.1097/jto.0000000000000658] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
34
|
Begum S, Hayashi M, Ogawa T, Jabboure FJ, Brait M, Izumchenko E, Tabak S, Ahrendt SA, Westra WH, Koch W, Sidransky D, Hoque MO. An integrated genome-wide approach to discover deregulated microRNAs in non-small cell lung cancer: Clinical significance of miR-23b-3p deregulation. Sci Rep 2015; 5:13236. [PMID: 26314549 PMCID: PMC4551983 DOI: 10.1038/srep13236] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 05/18/2015] [Indexed: 02/06/2023] Open
Abstract
In spite of significant technical advances, genesis and progression of non-small cell lung cancer (NSCLC) remain poorly understood. We undertook an integrated genetic approach to discover novel microRNAs that were deregulated in NSCLCs. A total 119 primary NSCLCs with matched normal were analyzed for genome-wide copy number changes. We also tested a subset of matched samples by microRNA expression array, and integrated them to identify microRNAs positioned in allelic imbalance area. Our findings support that most of the identified deregulated microRNAs (miR-21, miR-23b, miR-31, miR-126, miR-150, and miR-205) were positioned in allelic imbalance areas. Among microRNAs tested in independent 114 NSCLCs, overexpression of miR-23b was revealed to be a significantly poor prognostic factor of recurrence free survival (HR = 2.40, P = 0.005, 95%CI: 1.32–4.29) and overall survival (HR = 2.35, P = 0.005, 95%CI: 1.30–4.19) in multivariable analysis. In addition, overexpression of miR-23b in H1838 cell line significantly increased cell proliferation, while inhibition of miR-23b in H1437 and H1944 cell lines significantly decreased cell doubling time. In summary, integration of genomic analysis and microRNA expression profiling could identify novel cancer-related microRNAs, and miR-23b could be a potential prognostic marker for early stage NSCLCs. Further biological studies of miR-23b are warranted for the potential development of targeted therapy.
Collapse
Affiliation(s)
- Shahnaz Begum
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, 21231 USA
| | - Masamichi Hayashi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, Maryland, 21231 USA
| | - Takenori Ogawa
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, Maryland, 21231 USA
| | - Fayez J Jabboure
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, Maryland, 21231 USA
| | - Mariana Brait
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, Maryland, 21231 USA
| | - Evgeny Izumchenko
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, Maryland, 21231 USA
| | - Sarit Tabak
- Rosetta Genomics Ltd. 10 Plaut St., Rehovot, Israel, 76706
| | - Steven A Ahrendt
- Department of Surgery, Division of Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213 USA
| | - William H Westra
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, 21231 USA
| | - Wayne Koch
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, Maryland, 21231 USA
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, Maryland, 21231 USA
| | - Mohammad O Hoque
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, Maryland, 21231 USA.,Department of Urology, Johns Hopkins University, Baltimore, Maryland, 21231 USA.,Department of Oncology, Johns Hopkins University, Baltimore, Maryland, 21231 USA
| |
Collapse
|
35
|
Li Y, Xiao X, Ji X, Liu B, Amos CI. RNA-seq analysis of lung adenocarcinomas reveals different gene expression profiles between smoking and nonsmoking patients. Tumour Biol 2015; 36:8993-9003. [PMID: 26081616 DOI: 10.1007/s13277-015-3576-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/15/2015] [Indexed: 02/07/2023] Open
Abstract
Lung adenocarcinoma is caused by the combination of genetic and environmental effects, and smoking plays an important role in the disease development. Exploring the gene expression profile and identifying genes that are shared or vary between smokers and nonsmokers with lung adenocarcinoma will provide insights into the etiology of this complex cancer. We obtained RNA-seq data from paired normal and tumor tissues from 34 nonsmoking and 34 smoking patients with lung adenocarcinoma (GEO: GSE40419). R Bioconductor, edgeR, was adopted to conduct differential gene expression analysis between paired normal and tumor tissues. A generalized linear model was applied to identify genes that were differentially expressed in nonsmoker and smoker patients as well as genes that varied between these two groups. We identified 2273 genes that showed differential expression with FDR < 0.05 and |logFC| >1 in nonsmoker tumor versus normal tissues; 3030 genes in the smoking group; and 1967 genes were common to both groups. Sixty-eight and 70% of the identified genes were downregulated in nonsmoking and smoking groups, respectively. The 20 genes such as SPP1, SPINK1, and FAM83A with largest fold changes in smokers also showed similar large and highly significant fold changes in nonsmokers and vice versa, showing commonalities in expression changes for adenocarcinomas in both smokers and nonsmokers for these genes. We also identified 175 genes that were significantly differently expressed between tumor samples from nonsmoker and smoker patients. Gene expression profile varied substantially between smoker and nonsmoker patients with lung adenocarcinoma. Smoking patients overall showed far more complicated disease mechanism and have more dysregulation in their gene expression profiles. Our study reveals pathogenetic differences in smoking and nonsmoking patients with lung adenocarcinoma from transcriptome analysis. We provided a list of candidate genes for further study for disease detection and treatment in both smoking and nonsmoking patients with lung adenocarcinoma.
Collapse
Affiliation(s)
- Yafang Li
- Department of Biomedical Data Science, Dartmouth College, 74 College Street, Vail 716A, Hanover, NH, 03755, USA
| | - Xiangjun Xiao
- Department of Biomedical Data Science, Dartmouth College, 74 College Street, Vail 716A, Hanover, NH, 03755, USA
| | - Xuemei Ji
- Department of Biomedical Data Science, Dartmouth College, 74 College Street, Vail 716A, Hanover, NH, 03755, USA
| | - Bin Liu
- Department of Genetics, Center for Genetics and Genomics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. Unit 1010, Houston, 77030, TX, USA
| | - Christopher I Amos
- Department of Biomedical Data Science, Dartmouth College, 74 College Street, Vail 716A, Hanover, NH, 03755, USA.
| |
Collapse
|
36
|
Ali A, Bhatti MZ, Shah AS, Duong HQ, Alkreathy HM, Mohammad SF, Khan RA, Ahmad A. Tumor-suppressive p53 Signaling Empowers Metastatic Inhibitor KLF17-dependent Transcription to Overcome Tumorigenesis in Non-small Cell Lung Cancer. J Biol Chem 2015; 290:21336-51. [PMID: 25911104 DOI: 10.1074/jbc.m114.635730] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Indexed: 11/06/2022] Open
Abstract
Metastasis, which is controlled by concerted action of multiple genes, is a complex process and is an important cause of cancer death. Krüppel-like factor 17 (KLF17) is a negative regulator of metastasis and epithelial-mesenchymal transition (EMT) during cancer progression. However, the underlying molecular mechanism and biological relevance of KLF17 in cancer cells are poorly understood. Here, we show that tumor suppressor protein p53 plays an integral role to induce KLF17 expression in non-small cell lung cancer (NSCLC). p53 is recruited to the KLF17 promoter and results in the formation of p53-DNA complex. p53 enhances binding of p300 and favors histone acetylation on the KLF17 promoter. Mechanistically, p53 physically interacts with KLF17 and thereby enhances the anti-metastatic function of KLF17. p53 empowers KLF17-mediated EMT genes transcription via enhancing physical association of KLF17 with target gene promoters. Nutlin-3 recruits KLF17 to EMT target gene promoters and results in the formation of KLF17-DNA complex via a p53-dependent pathway. p53 depletion abrogates DNA binding affinity of KLF17 to EMT target gene promoters. KLF17 is critical for p53 cellular activities in NSCLC. Importantly, KLF17 enhances p53 transcription to generate a novel positive feedback loop. KLF17 depletion accelerates lung cancer cell growth in response to chemotherapy. Mechanistically, we found that KLF17 increases the expression of tumor suppressor genes p53, p21, and pRB. Functionally, KLF17 required p53 to suppress cancer cell invasion and migration in NSCLC. In conclusion, our study highlights a novel insight into the anti-EMT effect of KLF17 via a p53-dependent pathway in NSCLC, and KLF17 may be a new therapeutic target in NSCLC with p53 status.
Collapse
Affiliation(s)
- Amjad Ali
- From the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China, the Department of Biotechnology, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Muhammad Zeeshan Bhatti
- From the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China, the Department of Biotechnology, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Abdus Saboor Shah
- From the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Hong-Quan Duong
- the Institute of Research and Development, Duy Tan University, K7/25 Quang Trung, Danang 59000, Vietnam
| | - Huda Mohammad Alkreathy
- the Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudia Arabia
| | - Shah Faisal Mohammad
- the Laboratory of Enzymology and Molecular Evolution, State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China, the Department of Biotechnology, Faculty of Biological Sciences, University of Science and Technology Bannu, Khyber Pakhtunkhwa, Pakistan
| | - Rahmat Ali Khan
- the Department of Biotechnology, Faculty of Biological Sciences, University of Science and Technology Bannu, Khyber Pakhtunkhwa, Pakistan
| | - Ayaz Ahmad
- the Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China, and the Department of Biotechnology, Abdul Wali Khan University, Mardan 23200, Pakistan
| |
Collapse
|
37
|
Rodon J, Soria JC, Berger R, Batist G, Tsimberidou A, Bresson C, Lee JJ, Rubin E, Onn A, Schilsky RL, Miller WH, Eggermont AM, Mendelsohn J, Lazar V, Kurzrock R. Challenges in initiating and conducting personalized cancer therapy trials: perspectives from WINTHER, a Worldwide Innovative Network (WIN) Consortium trial. Ann Oncol 2015; 26:1791-8. [PMID: 25908602 DOI: 10.1093/annonc/mdv191] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 04/13/2015] [Indexed: 12/11/2022] Open
Abstract
Advances in 'omics' technology and targeted therapeutic molecules are together driving the incorporation of molecular-based diagnostics into the care of patients with cancer. There is an urgent need to assess the efficacy of therapy determined by molecular matching of patients with particular targeted therapies. WINTHER is a clinical trial that uses cutting edge genomic and transcriptomic assays to guide treatment decisions. Through the lens of this ambitious multinational trial (five countries, six sites) coordinated by the Worldwide Innovative Networking Consortium for personalized cancer therapy, we discovered key challenges in initiation and conduct of a prospective, omically driven study. To date, the time from study concept to activation has varied between 19 months at Gustave Roussy Cancer Campus in France to 30 months at the Segal Cancer Center, McGill University (Canada). It took 3+ years to be able to activate US sites due to national regulatory hurdles. Access to medications proposed by the molecular analysis remains a major challenge, since their availability through active clinical trials is highly variable over time within sites and across the network. Rules regarding the off-label use of drugs, or drugs not yet approved at all in some countries, pose a further challenge, and many biopharmaceutical companies lack a simple internal mechanism to supply the drugs even if they wish to do so. These various obstacles should be addressed to test and then implement precision medicine in cancer.
Collapse
Affiliation(s)
- J Rodon
- Vall D'Hebron Institute of Oncology and Universitat Autonoma de Barcelona, Barcelona, Spain
| | - J C Soria
- Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - R Berger
- Oncology Institute, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - G Batist
- Segal Cancer Center, Jewish General Hospital Mcgill University, Montreal Quebec Consortium de Recherche en Oncologie Clinique, Quebec, Canada
| | - A Tsimberidou
- The University of Texas MD Anderson Cancer Center, Houston, USA
| | | | - J J Lee
- The University of Texas MD Anderson Cancer Center, Houston, USA
| | - E Rubin
- The National Institute of Biotechnology in the Negev, Ben Gurion University, Beer-Sheva, Israel
| | - A Onn
- Oncology Institute, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - R L Schilsky
- American Society of Clinical Oncology (ASCO), Alexandria
| | - W H Miller
- Segal Cancer Center, Jewish General Hospital Mcgill University, Montreal Quebec Consortium de Recherche en Oncologie Clinique, Quebec, Canada
| | - A M Eggermont
- Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - J Mendelsohn
- The University of Texas MD Anderson Cancer Center, Houston, USA
| | - V Lazar
- Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - R Kurzrock
- Center for Personalized Cancer Therapy, UC San Diego-Moores Cancer Center, La Jolla, USA
| |
Collapse
|
38
|
Marien E, Meister M, Muley T, Fieuws S, Bordel S, Derua R, Spraggins J, Van de Plas R, Dehairs J, Wouters J, Bagadi M, Dienemann H, Thomas M, Schnabel PA, Caprioli RM, Waelkens E, Swinnen JV. Non-small cell lung cancer is characterized by dramatic changes in phospholipid profiles. Int J Cancer 2015; 137:1539-48. [PMID: 25784292 PMCID: PMC4503522 DOI: 10.1002/ijc.29517] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 02/16/2015] [Accepted: 03/05/2015] [Indexed: 12/18/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer death globally. To develop better diagnostics and more effective treatments, research in the past decades has focused on identification of molecular changes in the genome, transcriptome, proteome, and more recently also the metabolome. Phospholipids, which nevertheless play a central role in cell functioning, remain poorly explored. Here, using a mass spectrometry (MS)-based phospholipidomics approach, we profiled 179 phospholipid species in malignant and matched non-malignant lung tissue of 162 NSCLC patients (73 in a discovery cohort and 89 in a validation cohort). We identified 91 phospholipid species that were differentially expressed in cancer versus non-malignant tissues. Most prominent changes included a decrease in sphingomyelins (SMs) and an increase in specific phosphatidylinositols (PIs). Also a decrease in multiple phosphatidylserines (PSs) was observed, along with an increase in several phosphatidylethanolamine (PE) and phosphatidylcholine (PC) species, particularly those with 40 or 42 carbon atoms in both fatty acyl chains together. 2D-imaging MS of the most differentially expressed phospholipids confirmed their differential abundance in cancer cells. We identified lipid markers that can discriminate tumor versus normal tissue and different NSCLC subtypes with an AUC (area under the ROC curve) of 0.999 and 0.885, respectively. In conclusion, using both shotgun and 2D-imaging lipidomics analysis, we uncovered a hitherto unrecognized alteration in phospholipid profiles in NSCLC. These changes may have important biological implications and may have significant potential for biomarker development.
Collapse
Affiliation(s)
- Eyra Marien
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, KU Leuven-University of Leuven, Leuven, Belgium
| | - Michael Meister
- Thoraxklinik at University Hospital Heidelberg, Translational Research Unit, Heidelberg, Germany.,TLRC-H - Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research, Heidelberg, Germany
| | - Thomas Muley
- Thoraxklinik at University Hospital Heidelberg, Translational Research Unit, Heidelberg, Germany.,TLRC-H - Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research, Heidelberg, Germany
| | - Steffen Fieuws
- Department of Public Health and Primary Care, I-Biostat KU Leuven-University of Leuven and Universiteit Hasselt, Leuven, Belgium
| | - Sergio Bordel
- Department of Chemical and Biological Engineering, Systems Biology Group, Chalmers University of Technology, Gothenburg, Sweden
| | - Rita Derua
- Department of Cellular and Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven - University of Leuven, Leuven, Belgium
| | - Jeffrey Spraggins
- Department of Biochemistry and Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, TN
| | - Raf Van de Plas
- Department of Biochemistry and Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, TN.,Delft University of Technology, Delft Center for Systems and Control, CD Delft, The Netherlands
| | - Jonas Dehairs
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, KU Leuven-University of Leuven, Leuven, Belgium
| | - Jens Wouters
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, KU Leuven-University of Leuven, Leuven, Belgium
| | - Muralidhararao Bagadi
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, KU Leuven-University of Leuven, Leuven, Belgium
| | - Hendrik Dienemann
- TLRC-H - Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research, Heidelberg, Germany.,Department of Surgery, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Thomas
- TLRC-H - Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research, Heidelberg, Germany.,Department of Thoracic Oncology, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Philipp A Schnabel
- TLRC-H - Translational Lung Research Center Heidelberg, Member of the German Center for Lung Research, Heidelberg, Germany.,University Hospital Heidelberg, Institute of Pathology, Heidelberg, Germany
| | - Richard M Caprioli
- Department of Biochemistry and Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, TN
| | - Etienne Waelkens
- Department of Cellular and Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven - University of Leuven, Leuven, Belgium
| | - Johannes V Swinnen
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, KU Leuven-University of Leuven, Leuven, Belgium
| |
Collapse
|
39
|
Wood SL, Pernemalm M, Crosbie PA, Whetton AD. Molecular histology of lung cancer: from targets to treatments. Cancer Treat Rev 2015; 41:361-75. [PMID: 25825324 DOI: 10.1016/j.ctrv.2015.02.008] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 02/02/2015] [Accepted: 02/13/2015] [Indexed: 01/06/2023]
Abstract
Lung cancer is the leading cause of cancer-related death worldwide with a 5-year survival rate of less than 15%, despite significant advances in both diagnostic and therapeutic approaches. Combined genomic and transcriptomic sequencing studies have identified numerous genetic driver mutations that are responsible for the development of lung cancer. In addition, molecular profiling studies identify gene products and their mutations which predict tumour responses to targeted therapies such as protein tyrosine kinase inhibitors and also can offer explanation for drug resistance mechanisms. The profiling of circulating micro-RNAs has also provided an ability to discriminate patients in terms of prognosis/diagnosis and high-throughput DNA sequencing strategies are beginning to elucidate cell signalling pathway mutations associated with oncogenesis, including potential stem cell associated pathways, offering the promise that future therapies may target this sub-population, preventing disease relapse post treatment and improving patient survival. This review provides an assessment of molecular profiling within lung cancer concerning molecular mechanisms, treatment options and disease-progression. Current areas of development within lung cancer profiling are discussed (i.e. profiling of circulating tumour cells) and future challenges for lung cancer treatment addressed such as detection of micro-metastases and cancer stem cells.
Collapse
Affiliation(s)
- Steven L Wood
- Faculty Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Wolfson Molecular Imaging Centre, Manchester M20 3LJ, UK.
| | - Maria Pernemalm
- Faculty Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Wolfson Molecular Imaging Centre, Manchester M20 3LJ, UK; Karolinska Institutet, Department of Oncology and Pathology, SciLifeLab, Tomtebodavägen 23A, 17165 Solna, Sweden
| | - Philip A Crosbie
- Faculty Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Wolfson Molecular Imaging Centre, Manchester M20 3LJ, UK
| | - Anthony D Whetton
- Faculty Institute of Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Wolfson Molecular Imaging Centre, Manchester M20 3LJ, UK
| |
Collapse
|
40
|
Suo C, Hrydziuszko O, Lee D, Pramana S, Saputra D, Joshi H, Calza S, Pawitan Y. Integration of somatic mutation, expression and functional data reveals potential driver genes predictive of breast cancer survival. Bioinformatics 2015; 31:2607-13. [PMID: 25810432 DOI: 10.1093/bioinformatics/btv164] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 03/16/2015] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Genome and transcriptome analyses can be used to explore cancers comprehensively, and it is increasingly common to have multiple omics data measured from each individual. Furthermore, there are rich functional data such as predicted impact of mutations on protein coding and gene/protein networks. However, integration of the complex information across the different omics and functional data is still challenging. Clinical validation, particularly based on patient outcomes such as survival, is important for assessing the relevance of the integrated information and for comparing different procedures. RESULTS An analysis pipeline is built for integrating genomic and transcriptomic alterations from whole-exome and RNA sequence data and functional data from protein function prediction and gene interaction networks. The method accumulates evidence for the functional implications of mutated potential driver genes found within and across patients. A driver-gene score (DGscore) is developed to capture the cumulative effect of such genes. To contribute to the score, a gene has to be frequently mutated, with high or moderate mutational impact at protein level, exhibiting an extreme expression and functionally linked to many differentially expressed neighbors in the functional gene network. The pipeline is applied to 60 matched tumor and normal samples of the same patient from The Cancer Genome Atlas breast-cancer project. In clinical validation, patients with high DGscores have worse survival than those with low scores (P = 0.001). Furthermore, the DGscore outperforms the established expression-based signatures MammaPrint and PAM50 in predicting patient survival. In conclusion, integration of mutation, expression and functional data allows identification of clinically relevant potential driver genes in cancer. AVAILABILITY AND IMPLEMENTATION The documented pipeline including annotated sample scripts can be found in http://fafner.meb.ki.se/biostatwiki/driver-genes/. CONTACT yudi.pawitan@ki.se SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Chen Suo
- School of Life Sciences, Peking University, Beijing, China, Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Olga Hrydziuszko
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Donghwan Lee
- Department of Statistics, Ewha Womans University, Seoul, South Korea
| | - Setia Pramana
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden, Department of Computational Statistics, Institute of Statistics, Jakarta, Indonesia and
| | - Dhany Saputra
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Himanshu Joshi
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Stefano Calza
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Yudi Pawitan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
41
|
Schwaederlé M, Lazar V, Validire P, Hansson J, Lacroix L, Soria JC, Pawitan Y, Kurzrock R. VEGF-A Expression Correlates with TP53 Mutations in Non-Small Cell Lung Cancer: Implications for Antiangiogenesis Therapy. Cancer Res 2015; 75:1187-90. [PMID: 25672981 DOI: 10.1158/0008-5472.can-14-2305] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 12/26/2014] [Indexed: 12/11/2022]
Abstract
Bevacizumab is one of the most widely used antiangiogenic drugs in oncology, but the overall beneficial effects of this VEGF-A targeting agent are relatively modest, in part due to the lack of a biomarker to select patients most likely to respond favorably. Several molecular aberrations in cancer influence angiogenesis, including mutations in the tumor suppressor gene TP53, which occur frequently in many human malignancies. In this study, we present a multiple regression analysis of transcriptomic data in 123 patients with non-small cell lung cancer (NSCLC) showing that TP53 mutations are associated with higher VEGF-A expression (P = 0.006). This association was interesting given a recent retrospective study showing longer progression-free survival in patients with diverse tumors who receive bevacizumab, if tumors harbor mutant TP53 instead of wild-type TP53. Thus, our current findings linking TP53 mutation with VEGF-A upregulation offered a mechanistic explanation for why patients exhibit improved outcomes after bevacizumab treatment when their tumors harbor mutant TP53 versus wild-type TP53. Overall, this work warrants further evaluation of TP53 as a ready biomarker to predict bevacizumab response in NSCLC and possibly other tumor types.
Collapse
Affiliation(s)
- Maria Schwaederlé
- Center for Personalized Cancer Therapy, UCSD Moores Cancer Center, La Jolla, California.
| | | | | | | | | | | | | | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, UCSD Moores Cancer Center, La Jolla, California
| |
Collapse
|
42
|
Large cell carcinoma of the lung: a tumor in search of an author. A clinically oriented critical reappraisal. Lung Cancer 2015; 87:226-31. [PMID: 25620799 DOI: 10.1016/j.lungcan.2015.01.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 01/01/2015] [Accepted: 01/09/2015] [Indexed: 12/19/2022]
Abstract
Large cell carcinoma (LCC) is a merely descriptive term indicating a subtype of lung cancer with no specific features of small-cell lung cancer (SCLC), adenocarcinoma (ADC) or squamous cell carcinoma (SQC). This diagnosis is allowed on surgical specimens only, whereas its counterpart in biopsy/cytology samples is non-small-cell lung carcinoma (NSCLC), not otherwise specified (NOS). Although these two terms do not fulfill the same concept, they can be interchangeable synonyms at the clinical level, reflecting, in different ways, the inability to define a specific subtype. Immunohistochemistry (IHC), next generation sequencing (NGS) analysis and, historically, electron microscopy have been unveiling diverse cell differentiation lineages in LCC, resulting in LCC-favor ADC, LCC-favor SQC and LCC-favor large-cell neuroendocrine carcinoma (LCNEC), the latter hopefully to be included into the neuroendocrine tumor (NET) group in the future. Paradoxically, however, the interpretation issues of LCC/NSCLC-NOS are not diminishing, but even increasing albeight an accurate diagnosis is oncologically required and crucial. Also, rare LCC/NSCLC-NOS cases exhibiting null/unclear phenotype, are difficult to classify, and this terminology could be maintained for the sake of classification (basically these tumors are serendipitous ADC, as also confirmed by the lack of p40). In this review article, seven relevant issues to LCC have been addressed by using a question-answer methodology, with final key points discussing major interpretation issues. In conclusion, most LCC/NSCLC-NOS may be eventually re-classified and addressed by exploiting IHC and/or molecular testing to satisfy the criteria of precision medicine (the right drug, to the right patient, at the right time).
Collapse
|
43
|
Shi X, Liang W, Yang W, Xia R, Song Y. Decorin is responsible for progression of non-small-cell lung cancer by promoting cell proliferation and metastasis. Tumour Biol 2014; 36:3345-54. [PMID: 25524578 DOI: 10.1007/s13277-014-2968-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/09/2014] [Indexed: 12/30/2022] Open
Abstract
Decorin, a member of the small leucine-rich proteoglycans family, exists and plays multifunctional roles in stromal and epithelial cells. Emerging evidences showed that decorin is dysregulated expression in a wide variety of human tumors and affects a broad biology process of cancer cells, including growth, metastasis, and angiogenesis. Recent studies demonstrated that decorin could affect A549 proliferation though decreasing TGF-β1, cycling D1 expression and increasing P53 and P21 expression. However, limited data are available on the effect of decorin on metastasis of non-small-cell lung cancer (NSCLC) cell lines and how decorin impacts metastasis is still unknown. In this study, we identified decorin mRNA expression through Oncomine database and verified the expression of decorin mRNA and protein in 50 patients who underwent primary surgical resection of a NSCLC in the Department of Thoracic Surgery, Jinling Hospital, Nanjing University School of Medicine, China, between September 2013 and March 2014 by quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) and Western blot. Also, the correlationship between decorin and the NSCLC patients' clinical characteristics or survival ( www.kmplot.com ) was analyzed. Via ectopic expression analyses and Western blot, the roles of decorin in proliferation, metastasis, and the underline mechanism for decorin expression were further explored. We found that decorin was downregulated in NSCLC tissues compared with the adjacent normal lung tissues or normal tissues. Additionally, the expression of decorin was correlated with tumor size, lymph node metastasis, tumor stage, and prognosis. We also showed that overexpression of decorin could inhibit NSCLC cell lines proliferation and metastasis. Through Western blot analysis, we identified that E-cadherin and vascular endothelial growth factor (VEGF) are two key factors responsible for the growth arrest and metastasis inhibition induced by decorin in NSCLC. Our results indicated that decorin plays crucial roles in NSCLC against carcinogenesis and progression. Decorin might be a predictive factor and an attractive therapeutic target for NSCLC patients.
Collapse
Affiliation(s)
- Xuefei Shi
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China,
| | | | | | | | | |
Collapse
|
44
|
Ilie M, Nunes M, Blot L, Hofman V, Long-Mira E, Butori C, Selva E, Merino-Trigo A, Vénissac N, Mouroux J, Vrignaud P, Hofman P. Setting up a wide panel of patient-derived tumor xenografts of non-small cell lung cancer by improving the preanalytical steps. Cancer Med 2014; 4:201-11. [PMID: 25470237 PMCID: PMC4329004 DOI: 10.1002/cam4.357] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 08/26/2014] [Accepted: 08/31/2014] [Indexed: 12/30/2022] Open
Abstract
With the ongoing need to improve therapy for non–small cell lung cancer (NSCLC) there has been increasing interest in developing reliable preclinical models to test novel therapeutics. Patient-derived tumor xenografts (PDX) are considered to be interesting candidates. However, the establishment of such model systems requires highly specialized research facilities and introduces logistic challenges. We aimed to establish an extensive well-characterized panel of NSCLC xenograft models in the context of a long-distance research network after careful control of the preanalytical steps. One hundred fresh surgically resected NSCLC specimens were shipped in survival medium at room temperature from a hospital-integrated biobank to animal facilities. Within 24 h post-surgery, tumor fragments were subcutaneously xenografted into immunodeficient mice. PDX characterization was performed by histopathological, immunohistochemical, aCGH and next-generation sequencing approaches. For this model system, the tumor take rate was 35%, with higher rates for squamous carcinoma (60%) than for adenocarcinoma (13%). Patients for whom PDX tumors were obtained had a significantly shorter disease-free survival (DFS) compared to patients for whom no PDX tumors (P = 0.039) were obtained. We established a large panel of PDX NSCLC models with a high frequency of mutations (29%) in EGFR, KRAS, NRAS, MEK1, BRAF, PTEN, and PI3KCA genes and with gene amplification (20%) of c-MET and FGFR1. This new patient-derived NSCLC xenograft collection, established regardless of the considerable time required and the distance between the clinic and the animal facilities, recapitulated the histopathology and molecular diversity of NSCLC and provides stable and reliable preclinical models for human lung cancer research.
Collapse
Affiliation(s)
- Marius Ilie
- Laboratory of Clinical and Experimental Pathology, Louis Pasteur Hospital, Nice, France; IRCAN Team 3, Inserm U1081/UMR CNRS 7284, Faculty of Medicine of Nice, University of Nice Sophia Antipolis, Nice, France; Faculty of Medicine, University of Nice Sophia-Antipolis, Nice, France; Hospital Related Biobank BB-0033-00025, Louis Pasteur Hospital, Nice, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Yao F, Liu H, Li Z, Zhong C, Fang W. Down-regulation of LATS2 in non-small cell lung cancer promoted the growth and motility of cancer cells. Tumour Biol 2014; 36:2049-57. [DOI: 10.1007/s13277-014-2812-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 11/04/2014] [Indexed: 12/25/2022] Open
|
46
|
Pelosi G, Papotti M, Rindi G, Scarpa A. Unraveling tumor grading and genomic landscape in lung neuroendocrine tumors. Endocr Pathol 2014; 25:151-64. [PMID: 24771462 DOI: 10.1007/s12022-014-9320-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Currently, grading in lung neuroendocrine tumors (NETs) is inherently defined by the histological classification based on cell features, mitosis count, and necrosis, for which typical carcinoids (TC) are low-grade malignant tumors with long life expectation, atypical carcinoids (AC) intermediate-grade malignant tumors with more aggressive clinical behavior, and large cell NE carcinomas (LCNEC) and small cell lung carcinomas (SCLC) high-grade malignant tumors with dismal prognosis. While Ki-67 antigen labeling index, highlighting the proportion of proliferating tumor cells, has largely been used in digestive NETs for assessing prognosis and assisting therapy decisions, the same marker does not play an established role in the diagnosis, grading, and prognosis of lung NETs. Next generation sequencing techniques (NGS), thanks to their astonishing ability to process in a shorter timeframe up to billions of DNA strands, are radically revolutionizing our approach to diagnosis and therapy of tumors, including lung cancer. When applied to single genes, panels of genes, exome, or the whole genome by using either frozen or paraffin tissues, NGS techniques increase our understanding of cancer, thus realizing the bases of precision medicine. Data are emerging that TC and AC are mainly altered in chromatin remodeling genes, whereas LCNEC and SCLC are also mutated in cell cycle checkpoint and cell differentiation regulators. A common denominator to all lung NETs is a deregulation of cell proliferation, which represents a biological rationale for morphologic (mitoses and necrosis) and molecular (Ki-67 antigen) parameters to successfully serve as predictors of tumor behavior (i.e., identification of pathological entities with clinical correlation). It is envisaged that a novel grading system in lung NETs based on the combined assessment of mitoses, necrosis, and Ki-67 LI may offer a better stratification of prognostic classes, realizing a bridge between molecular alterations, morphological features, and clinical behavior.
Collapse
Affiliation(s)
- Giuseppe Pelosi
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy,
| | | | | | | |
Collapse
|
47
|
SUZ12 is involved in progression of non-small cell lung cancer by promoting cell proliferation and metastasis. Tumour Biol 2014; 35:6073-82. [PMID: 24633887 DOI: 10.1007/s13277-014-1804-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 02/26/2014] [Indexed: 01/21/2023] Open
Abstract
The suppressor of zeste-12 protein (SUZ12), a core component of Polycomb repressive complex 2 (PRC2), is implicated in transcriptional silencing by generating di- and tri-methylation of lysine 27 on histone H3 (H3K27Me3). Although SUZ12 is known to be of great importance in several human cancer tumorigenesis, limited data are available on the expression profile and functional role of SUZ12 in non-small cell lung cancer (NSCLC). Here, we determined the expression level of SUZ12 in 40 paired clinical NSCLC tissues and adjacent normal tissues by quantitative reverse-transcription polymerase chain reaction (qRT-PCR). The results showed that SUZ12 was anomalously expressed in NSCLC tissues compared to adjacent noncancerous tissues (P<0.05) and was highly correlated to tumor size, lymph node metastasis, and clinical stages (P<0.05). Additionally, siRNA-mediated knockdown of SUZ12 could inhibit tumor cell growth, migration, and invasion, indicating that SUZ12 might function as an oncogene in NSCLC initiation and progression. Furthermore, we found that SUZ12 silencing significantly reduced the expression levels of transcription factor transcription factor E2F1 (E2F1) as well as potential metastasis promoters Rho-associated, coiled-coil-containing protein kinase 1 (ROCK1) and roundabout homolog 1 (ROBO1) through Western blot analysis. Altogether, we provide evidences suggesting that SUZ12 is an oncogene in NSCLC and can regulate NSCLC cells proliferation and metastasis partly via reducing E2F1, ROCK1, and ROBO1. Thus, SUZ12 may represent a new potential diagnostic marker for NSCLC and may be a novel therapeutic target for NSCLC intervention.
Collapse
|