1
|
Li D, Xu J, Yang MQ. Gene Regulation Analysis Reveals Perturbations of Autism Spectrum Disorder during Neural System Development. Genes (Basel) 2021; 12:genes12121901. [PMID: 34946850 PMCID: PMC8700980 DOI: 10.3390/genes12121901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 01/21/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder that impedes patients' cognition, social, speech and communication skills. ASD is highly heterogeneous with a variety of etiologies and clinical manifestations. The prevalence rate of ASD increased steadily in recent years. Presently, molecular mechanisms underlying ASD occurrence and development remain to be elucidated. Here, we integrated multi-layer genomics data to investigate the transcriptome and pathway dysregulations in ASD development. The RNA sequencing (RNA-seq) expression profiles of induced pluripotent stem cells (iPSCs), neural progenitor cells (NPCs) and neuron cells from ASD and normal samples were compared in our study. We found that substantially more genes were differentially expressed in the NPCs than the iPSCs. Consistently, gene set variation analysis revealed that the activity of the known ASD pathways in NPCs and neural cells were significantly different from the iPSCs, suggesting that ASD occurred at the early stage of neural system development. We further constructed comprehensive brain- and neural-specific regulatory networks by incorporating transcription factor (TF) and gene interactions with long 5 non-coding RNA(lncRNA) and protein interactions. We then overlaid the transcriptomes of different cell types on the regulatory networks to infer the regulatory cascades. The variations of the regulatory cascades between ASD and normal samples uncovered a set of novel disease-associated genes and gene interactions, particularly highlighting the functional roles of ELF3 and the interaction between STAT1 and lncRNA ELF3-AS 1 in the disease development. These new findings extend our understanding of ASD and offer putative new therapeutic targets for further studies.
Collapse
Affiliation(s)
- Dan Li
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA;
| | - Joshua Xu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA;
- Correspondence: (J.X.); (M.Q.Y.)
| | - Mary Qu Yang
- MidSouth Bioinformatics Center, Joint Bioinformatics Graduate Program of University of Arkansas at Little Rock, University of Arkansas for Medical Sciences, Little Rock, AR 72204, USA
- Correspondence: (J.X.); (M.Q.Y.)
| |
Collapse
|
2
|
Yu CY, Mitrofanova A. Mechanism-Centric Approaches for Biomarker Detection and Precision Therapeutics in Cancer. Front Genet 2021; 12:687813. [PMID: 34408770 PMCID: PMC8365516 DOI: 10.3389/fgene.2021.687813] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
Biomarker discovery is at the heart of personalized treatment planning and cancer precision therapeutics, encompassing disease classification and prognosis, prediction of treatment response, and therapeutic targeting. However, many biomarkers represent passenger rather than driver alterations, limiting their utilization as functional units for therapeutic targeting. We suggest that identification of driver biomarkers through mechanism-centric approaches, which take into account upstream and downstream regulatory mechanisms, is fundamental to the discovery of functionally meaningful markers. Here, we examine computational approaches that identify mechanism-centric biomarkers elucidated from gene co-expression networks, regulatory networks (e.g., transcriptional regulation), protein-protein interaction (PPI) networks, and molecular pathways. We discuss their objectives, advantages over gene-centric approaches, and known limitations. Future directions highlight the importance of input and model interpretability, method and data integration, and the role of recently introduced technological advantages, such as single-cell sequencing, which are central for effective biomarker discovery and time-cautious precision therapeutics.
Collapse
Affiliation(s)
- Christina Y. Yu
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Antonina Mitrofanova
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
3
|
Zhu Y, Chen QY, Jordan A, Sun H, Roy N, Costa M. RUNX2/miR‑31/SATB2 pathway in nickel‑induced BEAS‑2B cell transformation. Oncol Rep 2021; 46:154. [PMID: 34109987 DOI: 10.3892/or.2021.8105] [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: 01/26/2021] [Accepted: 05/05/2021] [Indexed: 11/05/2022] Open
Abstract
Nickel (Ni) compounds are classified as Group 1 carcinogens by the International Agency for Research on Cancer (IARC) and are known to be carcinogenic to the lungs. In our previous study, special AT‑rich sequence‑binding protein 2 (SATB2) was required for Ni‑induced BEAS‑2B cell transformation. In the present study, a pathway that regulates the expression of SATB2 protein was investigated in Ni‑transformed BEAS‑2B cells using western blotting and RT‑qPCR for expression, and soft agar, migration and invasion assays for cell transformation. Runt‑related transcription factor 2 (RUNX2), a master regulator of osteogenesis and an oncogene, was identified as an upstream regulator for SATB2. Ni induced RUNX2 expression and initiated BEAS‑2B transformation and metastatic potential. Previously, miRNA‑31 was identified as a negative regulator of SATB2 during arsenic‑induced cell transformation, and in the present study it was identified as a downstream target of RUNX2 during carcinogenesis. miR‑31 expression was reduced in Ni‑transformed BEAS‑2B cells, which was required to maintain cancer hallmarks. The expression level of miR‑31 was suppressed by RUNX2 in BEAS‑2B cells, and this increased the expression level of SATB2, initiating cell transformation. Ni caused the repression of miR‑31 by placing repressive marks at its promoter, which in turn increased the expression level of SATB2, leading to cell transformation.
Collapse
Affiliation(s)
- Yusha Zhu
- Department of Environmental Medicine, New York University Grossman School of Medicine, New York, NY 10100, USA
| | - Qiao Yi Chen
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shanxi 710000, P.R. China
| | - Ashley Jordan
- Department of Environmental Medicine, New York University Grossman School of Medicine, New York, NY 10100, USA
| | - Hong Sun
- Department of Environmental Medicine, New York University Grossman School of Medicine, New York, NY 10100, USA
| | - Nirmal Roy
- Department of Environmental Medicine, New York University Grossman School of Medicine, New York, NY 10100, USA
| | - Max Costa
- Department of Environmental Medicine, New York University Grossman School of Medicine, New York, NY 10100, USA
| |
Collapse
|
4
|
Tanabe S, Quader S, Cabral H, Ono R. Interplay of EMT and CSC in Cancer and the Potential Therapeutic Strategies. Front Pharmacol 2020; 11:904. [PMID: 32625096 PMCID: PMC7311659 DOI: 10.3389/fphar.2020.00904] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/03/2020] [Indexed: 02/05/2023] Open
Abstract
The mechanism of epithelial-mesenchymal transition (EMT) consists of the cellular phenotypic transition from epithelial to mesenchymal status. The cells exhibiting EMT exist in cancer stem cell (CSC) population, which is involved in drug resistance. CSCs demonstrating EMT feature remain after cancer treatment, which leads to drug resistance, recurrence, metastasis and malignancy of cancer. In this context, the recent advance of nanotechnology in the medical application has ascended the possibility to target CSCs using nanomedicines. In this review article, we focused on the mechanism of CSCs and EMT, especially into the signaling pathways in EMT, regulation of EMT and CSCs by microRNAs and nanomedicine-based approaches to target CSCs.
Collapse
Affiliation(s)
- Shihori Tanabe
- Division of Risk Assessment, Center for Biological Safety and Research (CBSR), National Institute of Health Science (NIHS), Kawasaki, Japan
- *Correspondence: Shihori Tanabe,
| | - Sabina Quader
- Innovation Centre of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Ryuichi Ono
- Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research (CBSR), National Institute of Health Science (NIHS), Kawasaki, Japan
| |
Collapse
|
5
|
He SY, Xi WJ, Wang X, Xu CH, Cheng L, Liu SY, Meng QQ, Li B, Wang Y, Shi HB, Wang HJ, Wang ZZ. Identification of a Combined RNA Prognostic Signature in Adenocarcinoma of the Lung. Med Sci Monit 2019; 25:3941-3956. [PMID: 31132294 PMCID: PMC6556069 DOI: 10.12659/msm.913727] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Adenocarcinoma of the lung is a type of non-small cell lung cancer (NSCLC). Clinical outcome is associated with tumor grade, stage, and subtype. This study aimed to identify RNA expression profiles, including long noncoding RNA (lncRNA), microRNA (miRNA), and mRNA, associated with clinical outcome in adenocarcinoma of the lung using bioinformatics data. Material/Methods The miRNA and mRNA expression profiles were downloaded from The Cancer Genome Atlas (TCGA) database, and lncRNA expression profiles were downloaded from The Atlas of Noncoding RNAs in Cancer (TANRIC) database. The independent dataset, the Gene Expression Omnibus (GEO) accession dataset, GSE81089, was used. RNA expression profiles were used to identify comprehensive prognostic RNA signatures based on patient survival time. Results From 7,704 lncRNAs, 787 miRNAs, and 28,937 mRNAs of 449 patients, four joint RNA molecular signatures were identified, including RP11-909N17.2, RP11-14N7.2 (lncRNAs), MIR139 (miRNA), KLHDC8B (mRNA). The random forest (RF) classifier was used to test the prediction ability of patient survival risk and showed a good predictive accuracy of 71% and also showed a significant difference in overall survival (log-rank P=0.0002; HR, 3.54; 95% CI, 1.74–7.19). The combined RNA signature also showed good performance in the identification of patient survival in the validation and independent datasets. Conclusions This study identified four RNA sequences as a prognostic molecular signature in adenocarcinoma of the lung, which may also provide an increased understanding of the molecular mechanisms underlying the pathogenesis of this malignancy.
Collapse
Affiliation(s)
- Si-Yu He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Wen-Jing Xi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Xin Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Chao-Han Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Si-Yao Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Qian-Qian Meng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Boyan Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Yahui Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Hong-Bo Shi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Hong-Jiu Wang
- College of Science, Heilongjiang University of Science and Technology, Harbin, Heilongjiang, China (mainland)
| | - Zhen-Zhen Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| |
Collapse
|
6
|
Identification of Transcriptional Signatures of Colon Tumor Stroma by a Meta-Analysis. JOURNAL OF ONCOLOGY 2019; 2019:8752862. [PMID: 31186640 PMCID: PMC6521457 DOI: 10.1155/2019/8752862] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/31/2019] [Indexed: 12/24/2022]
Abstract
Background The tumor stroma plays pivotal roles in influencing tumor growth, invasion, and metastasis. Transcriptional signatures of colon tumor stroma (CTS) are significantly associated with prognosis of colon cancer. Thus, identification of the CTS transcriptional features could be useful for colon cancer diagnosis and therapy. Methods By a meta-analysis of three CTS gene expression profiles datasets, we identified differentially expressed genes (DEGs) between CTS and colon normal stroma. Furthermore, we identified the pathways, upstream regulators, and protein-protein interaction (PPI) network that were significantly associated with the DEGs. Moreover, we analyzed the enrichment levels of immune signatures in CTS. Finally, we identified CTS-associated gene signatures whose expression was significantly associated with prognosis in colon cancer. Results We identified numerous significantly upregulated genes (such as CTHRC1, NFE2L3, SULF1, SOX9, ENC1, and CCND1) and significantly downregulated genes (such as MYOT, ASPA, KIAA2022, ARHGEF37, BCL-2, and PPARGC1A) in CTS versus colon normal stroma. Furthermore, we identified significantly upregulated pathways in CTS that were mainly involved in cellular development, immune regulation, and metabolism, as well as significantly downregulated pathways in CTS that were mostly metabolism-related. Moreover, we identified upstream TFs (such as SUZ12, NFE2L2, RUNX1, STAT3, and SOX2), kinases (such as MAPK14, CSNK2A1, CDK1, CDK2, and CDK4), and master metabolic transcriptional regulators (MMTRs) (such as HNF1A, NFKB1, ZBTB7A, GATA2, and GATA5) regulating the DEGs. We found that CD8+ T cells were more enriched in CTS than in colon normal stroma. Interestingly, we found that many of the DEGs and their regulators were prognostic markers for colon cancer, including CEBPB, PPARGC1, STAT3, MTOR, BCL2, JAK2, and CDK1. Conclusions The identification of CTS-specific transcriptional signatures may provide insights into the tumor microenvironment that mediates the development of colon cancer and has potential clinical implications for colon cancer diagnosis and treatment.
Collapse
|
7
|
Sikdar S, Datta S. A novel statistical approach for identification of the master regulator transcription factor. BMC Bioinformatics 2017; 18:79. [PMID: 28148240 PMCID: PMC5288875 DOI: 10.1186/s12859-017-1499-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/27/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Transcription factors are known to play key roles in carcinogenesis and therefore, are gaining popularity as potential therapeutic targets in drug development. A 'master regulator' transcription factor often appears to control most of the regulatory activities of the other transcription factors and the associated genes. This 'master regulator' transcription factor is at the top of the hierarchy of the transcriptomic regulation. Therefore, it is important to identify and target the master regulator transcription factor for proper understanding of the associated disease process and identifying the best therapeutic option. METHODS We present a novel two-step computational approach for identification of master regulator transcription factor in a genome. At the first step of our method we test whether there exists any master regulator transcription factor in the system. We evaluate the concordance of two ranked lists of transcription factors using a statistical measure. In case the concordance measure is statistically significant, we conclude that there is a master regulator. At the second step, our method identifies the master regulator transcription factor, if there exists one. RESULTS In the simulation scenario, our method performs reasonably well in validating the existence of a master regulator when the number of subjects in each treatment group is reasonably large. In application to two real datasets, our method ensures the existence of master regulators and identifies biologically meaningful master regulators. An R code for implementing our method in a sample test data can be found in http://www.somnathdatta.org/software . CONCLUSION We have developed a screening method of identifying the 'master regulator' transcription factor just using only the gene expression data. Understanding the regulatory structure and finding the master regulator help narrowing the search space for identifying biomarkers for complex diseases such as cancer. In addition to identifying the master regulator our method provides an overview of the regulatory structure of the transcription factors which control the global gene expression profiles and consequently the cell functioning.
Collapse
Affiliation(s)
- Sinjini Sikdar
- Department of Biostatistics, University of Florida, Gainesville, FL, 32611, USA
| | - Susmita Datta
- Department of Biostatistics, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
8
|
Yepes S, Torres MM, López-Kleine L. Regulatory network reconstruction reveals genes with prognostic value for chronic lymphocytic leukemia. BMC Genomics 2015; 16:1002. [PMID: 26606983 PMCID: PMC4659237 DOI: 10.1186/s12864-015-2189-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 11/03/2015] [Indexed: 11/10/2022] Open
Abstract
Background The clinical course of chronic lymphocytic leukemia (CLL) is highly variable; some patients follow an indolent course, but others progress to a more advanced stage. The mutational status of rearranged immunoglobulin heavy chain variable (IGVH) genes in CLL is a feature that is widely recognized for dividing patients into groups that are related to their prognoses. However, the regulatory programs associated with the IGVH statuses are poorly understood, and markers that can precisely predict survival outcomes have yet to be identified. Methods In this study, (i) we reconstructed gene regulatory networks in CLL by applying an information-theoretic approach to the expression profiles of 5 cohorts. (ii) We applied master regulator analysis (MRA) to these networks to identify transcription factors (TFs) that regulate an IGVH mutational status signature. The IGVH mutational status signature was developed by searching for differentially expressed genes between the IGVH mutational statuses in numerous CLL cohorts. (iii) To evaluate the biological implication of the inferred regulators, prognostic values were determined using time to treatment (TTT) and overall survival (OS) in two different cohorts. Results A robust IGVH expression signature was obtained, and various TFs emerged as regulators of the signature in most of the reconstructed networks. The TF targets expression profiles exhibited significant differences with respect to survival, which allowed the definition of a reduced profile with a high value for OS. TCF7 and its targets stood out for their roles in progression. Conclusion TFs and their targets, which were obtained merely from inferred regulatory associations, have prognostic implications and reflect a regulatory context for prognosis. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2189-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sally Yepes
- Facultad de Ciencias, Departamento de Ciencias Biológicas, Universidad de los Andes, Bogotá D.C., Colombia.
| | - Maria Mercedes Torres
- Facultad de Ciencias, Departamento de Ciencias Biológicas, Universidad de los Andes, Bogotá D.C., Colombia.
| | - Liliana López-Kleine
- Departamento de Estadística, Universidad Nacional de Colombia, Bogotá D.C., Colombia.
| |
Collapse
|
9
|
Cantini L, Isella C, Petti C, Picco G, Chiola S, Ficarra E, Caselle M, Medico E. MicroRNA-mRNA interactions underlying colorectal cancer molecular subtypes. Nat Commun 2015; 6:8878. [PMID: 27305450 PMCID: PMC4660217 DOI: 10.1038/ncomms9878] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 10/12/2015] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) transcriptional subtypes have been recently identified by gene expression profiling. Here we describe an analytical pipeline, microRNA master regulator analysis (MMRA), developed to search for microRNAs potentially driving CRC subtypes. Starting from a microRNA–mRNA tumour expression data set, MMRA identifies candidate regulator microRNAs by assessing their subtype-specific expression, target enrichment in subtype mRNA signatures and network analysis-based contribution to subtype gene expression. When applied to a CRC data set of 450 samples, assigned to subtypes by 3 different transcriptional classifiers, MMRA identifies 24 candidate microRNAs, in most cases downregulated in the stem/serrated/mesenchymal (SSM) poor prognosis subtype. Functional validation in CRC cell lines confirms downregulation of the SSM subtype by miR-194, miR-200b, miR-203 and miR-429, which share target genes and pathways mediating this effect. These results show that, by combining statistical tests, target prediction and network analysis, MMRA effectively identifies microRNAs functionally associated to cancer subtypes. Colorectal cancer subtypes can be distinguished by their different biological and molecular properties. Here the authors present microRNA Master Regulator Analysis, a tool to identify microRNAs driving subtype-specific gene expression and cancer variation.
Collapse
Affiliation(s)
- Laura Cantini
- Department of Oncology, Università degli Studi di Torino, S.P. 142, km 3, 95-10060 Candiolo, Italy.,Department of Control and Computer Engineering, Politecnico di Torino, Cso Duca degli Abruzzi 24, 10129 Torino, Italy.,Istituto Nazionale Biostrutture e Biosistemi-Consorzio Interuniversitario, Viale delle Medaglie d'Oro, 305-00136 Roma, Italy
| | - Claudio Isella
- Department of Oncology, Università degli Studi di Torino, S.P. 142, km 3, 95-10060 Candiolo, Italy.,Candiolo Cancer Institute, FPO IRCCS, S.P. 142, km 3, 95-10060 Candiolo, Italy
| | - Consalvo Petti
- Candiolo Cancer Institute, FPO IRCCS, S.P. 142, km 3, 95-10060 Candiolo, Italy
| | - Gabriele Picco
- Department of Oncology, Università degli Studi di Torino, S.P. 142, km 3, 95-10060 Candiolo, Italy.,Candiolo Cancer Institute, FPO IRCCS, S.P. 142, km 3, 95-10060 Candiolo, Italy
| | - Simone Chiola
- Department of Oncology, Università degli Studi di Torino, S.P. 142, km 3, 95-10060 Candiolo, Italy.,Candiolo Cancer Institute, FPO IRCCS, S.P. 142, km 3, 95-10060 Candiolo, Italy
| | - Elisa Ficarra
- Department of Control and Computer Engineering, Politecnico di Torino, Cso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Michele Caselle
- Department of Physics and INFN, Università degli Studi di Torino, via P.Giuria 1, I-10125 Turin, Italy
| | - Enzo Medico
- Department of Oncology, Università degli Studi di Torino, S.P. 142, km 3, 95-10060 Candiolo, Italy.,Candiolo Cancer Institute, FPO IRCCS, S.P. 142, km 3, 95-10060 Candiolo, Italy
| |
Collapse
|