1
|
Ghani A, Singh H, Kumar H, Vaiphei K. MicroRNA expression signature in gastrointestinal stromal tumour & their molecular & histological features. Indian J Med Res 2024; 160:118-127. [PMID: 39382501 PMCID: PMC11463855 DOI: 10.25259/ijmr_2567_22] [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: 12/16/2022] [Indexed: 10/10/2024] Open
Abstract
Background & objectives In gastrointestinal stromal tumour (GIST), not only genetic abnormalities are responsible for adverse clinical events, but epigenetic modifications also play a crucial role. MicroRNA (miRNA) dysregulation plays a significant role in carcinogenesis as miRNAs serve as natural silencer for their targets. Our study aimed to explore the miRNAs expression and its association with molecular and histopathological characteristics of GIST. Methods Fifty GIST samples, including 45 formalin fixed paraffin embedded (FFPE) and fresh tissues were included. Peripheral non-tumour tissues were used as controls. All the cases were confirmed using immunohistochemistry. RNA was extracted using miRNA-specific kit, and the expression was performed using RT-qPCR. The data were evaluated using AriaMx software version 1.5 (Agilent, US). MiRNAs expression was analyzed by using the relative quantification method (ΔΔCT). Results miR-221, miR-222, miR-494 and miR-34a showed significant down-regulation in tumours relative to non-tumour tissues. The expression levels of these miRNAs were significantly down-regulated in c-KIT (proto-oncogene encoding the tyrosine kinase transmembrane receptor)-positive tumours compared to c-KIT-negative. Further analysis revealed that reduced expression was associated with spindle subtypes and gastric localization. However, there was no significant correlation with other histological features. Additionally, miR-221/222, and miR-494 were down-regulated in most of the KIT exon 11 mutant subtypes, while miRNA-34a was associated with platelet derived growth factor receptor alpha (PDGFRA) mutations. Interpretation & conclusions The present study showed that the down-regulation of these miRNAs may help better molecular classification and characterization of GISTs. Our results offer new insight into the association between miRNAs and histological features, enabling a more thorough understanding of GISTs at the molecular level.
Collapse
Affiliation(s)
- Abdul Ghani
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Harvinder Singh
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
- Department of Translational and Regenerative Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Hemanth Kumar
- Department of General Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Kim Vaiphei
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
2
|
Han S, Song M, Wang J, Huang Y, Li Z, Yang A, Sui C, Zhang Z, Qiao J, Yang J. Intelligent identification system of gastric stromal tumors based on blood biopsy indicators. BMC Med Inform Decis Mak 2023; 23:214. [PMID: 37833709 PMCID: PMC10576280 DOI: 10.1186/s12911-023-02324-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND The most prevalent mesenchymal-derived gastrointestinal cancers are gastric stromal tumors (GSTs), which have the highest incidence (60-70%) of all gastrointestinal stromal tumors (GISTs). However, simple and effective diagnostic and screening methods for GST remain a great challenge at home and abroad. This study aimed to build a GST early warning system based on a combination of machine learning algorithms and routine blood, biochemical and tumour marker indicators. METHODS In total, 697 complete samples were collected from four hospitals in Gansu Province, including 42 blood indicators from 318 pretreatment GST patients, 180 samples of gastric polyps and 199 healthy individuals. In this study, three algorithms, gradient boosting machine (GBM), random forest (RF), and logistic regression (LR), were chosen to build GST prediction models for comparison. The performance and stability of the models were evaluated using two different validation techniques: 5-fold cross-validation and external validation. The DeLong test assesses significant differences in AUC values by comparing different ROC curves, the variance and covariance of the AUC value. RESULTS The AUC values of both the GBM and RF models were higher than those of the LR model, and this difference was statistically significant (P < 0.05). The GBM model was considered to be the optimal model, as a larger area was enclosed by the ROC curve, and the axes indicated robust model classification performance according to the accepted model discriminant. Finally, the integration of 8 top-ranked blood indices was proven to be able to distinguish GST from gastric polyps and healthy people with sensitivity, specificity and area under the curve of 0.941, 0.807 and 0.951 for the cross-validation set, respectively. CONCLUSION The GBM demonstrated powerful classification performance and was able to rapidly distinguish GST patients from gastric polyps and healthy individuals. This identification system not only provides an innovative strategy for the diagnosis of GST but also enables the exploration of hidden associations between blood parameters and GST for subsequent studies on the prevention and disease surveillance management of GST. The GST discrimination system is available online for free testing of doctors and high-risk groups at https://jzlyc.gsyy.cn/bear/mobile/index.html .
Collapse
Affiliation(s)
- Shangjun Han
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Meijuan Song
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Jiarui Wang
- Department of Medical Information and Engineering, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Yalong Huang
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Zuxi Li
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Aijia Yang
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Changsheng Sui
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Zeping Zhang
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Jiling Qiao
- Department of the First Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, People's Republic of China
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China
| | - Jing Yang
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, People's Republic of China.
| |
Collapse
|
3
|
Ravegnini G, Nannini M, Indio V, Serrano C, Gorini F, Astolfi A, Di Vito A, Morroni F, Pantaleo MA, Hrelia P, Angelini S. miRNA Expression May Have Implications for Immunotherapy in PDGFRA Mutant GISTs. Int J Mol Sci 2022; 23:ijms232012248. [PMID: 36293105 PMCID: PMC9603477 DOI: 10.3390/ijms232012248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 12/05/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) harboring mutations in the PDGFRA gene occur in only about 5-7% of patients. The most common PDGFRA mutation is exon 18 D842V, which is correlated with specific clinico-pathological features compared to the other PDGFRA mutated GISTs. Herein, we present a miRNA expression profile comparison of PDGFRA D842V mutant GISTs and PDGFRA with mutations other than D842V (non-D842V). miRNA expression profiling was carried out on 10 patients using a TLDA miRNA array. Then, miRNA expression was followed by bioinformatic analysis aimed at evaluating differential expression, pathway enrichment, and miRNA-mRNA networks. We highlighted 24 differentially expressed miRNAs between D842V and non-D842V GIST patients. Pathway enrichment analysis showed that deregulated miRNAs targeted genes that are mainly involved in the immune response pathways. The miRNA-mRNA networks highlighted a signature of miRNAs/mRNA that could explain the indolent behavior of the D842V mutated GIST. The results highlighted a different miRNA fingerprint in PDGFRA D842V GISTs compared to non-D842Vmutated patients, which could explain the different biological behavior of this GIST subset.
Collapse
Affiliation(s)
- Gloria Ravegnini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Margherita Nannini
- Department of Specialized, Experimental and Diagnostic Medicine, Sant’Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- Division of Oncology, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
| | - Valentina Indio
- Department of Veterinary Medical Sciences, University of Bologna, 40164 Ozzano, Italy
| | - Cesar Serrano
- Sarcoma Translational Research Laboratory, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Hospital Campus, C/ Natzaret 115-117, 08035 Barcelona, Spain
- Department of Medical Oncology, Vall d’Hebron University Hospital, P/Vall d’Hebron 119, 08035 Barcelona, Spain
| | - Francesca Gorini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Annalisa Astolfi
- Department of Specialized, Experimental and Diagnostic Medicine, Sant’Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Aldo Di Vito
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Fabiana Morroni
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Maria Abbondanza Pantaleo
- Department of Specialized, Experimental and Diagnostic Medicine, Sant’Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- Division of Oncology, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Sabrina Angelini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
- Inter-Departmental Center for Health Sciences & Technologies, CIRI-SDV, Alma Mater Studiorum-University of Bologna, 40126 Bologna, Italy
- Correspondence:
| |
Collapse
|
4
|
Ravegnini G, Serrano C, Ricci R, Zhang Q, Terrenato I, Graziosi A, Valori G, Landolfi S, Hrelia P, Angelini S. miRNA landscape in primary tumors and matched metastases in gastrointestinal stromal tumors. Epigenomics 2021; 13:369-377. [PMID: 33432846 DOI: 10.2217/epi-2020-0303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Gastrointestinal stromal tumor management is extremely challenging, particularly the metastatic disease. The underlying mechanism in metastasis spread remains largely unknown. We aimed to characterize miRNAs involved in the metastatic process in gastrointestinal stromal tumor. Material & methods: Eight primary tumors and 18 synchronous metastases were analyzed through miRNA Taqman arrays or assays. Results: miRNAs profiles revealed similar expression in primary site and metastases. Pair-wise correlation coefficient between primary tumor and metastases was significant for each patient (p < 0.0001 for all profiled patients). Conclusion: Our study, the largest including primary tumors and metastases so far performed, highlighted perpetuation of miRNAs features in metastatic lesions and that the primary origin appears to be the main determinant of the metastases miRNA profile.
Collapse
Affiliation(s)
- Gloria Ravegnini
- Department of Pharmacy & Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Cèsar Serrano
- Sarcoma Translational Research Laboratory, Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain.,Department of Medical Oncology, Vall d'Hebron University Hospital, 08035 Barcelona, Spain
| | - Riccardo Ricci
- UOC di Anatomia Patologica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| | - Qianqian Zhang
- UOC di Anatomia Patologica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, 00168 Rome, Italy
| | - Irene Terrenato
- Biostatistics-Scientific Direction, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Agnese Graziosi
- Department of Pharmacy & Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Giorgia Valori
- Department of Pharmacy & Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Stefania Landolfi
- Department of Pathology, Vall d'Hebron University Hospital, 08035 Barcelona, Spain
| | - Patrizia Hrelia
- Department of Pharmacy & Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Sabrina Angelini
- Department of Pharmacy & Biotechnology, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
5
|
Ravegnini G, Valori G, Zhang Q, Ricci R, Hrelia P, Angelini S. Pharmacogenetics in the treatment of gastrointestinal stromal tumors - an updated review. Expert Opin Drug Metab Toxicol 2020; 16:797-808. [PMID: 32597248 DOI: 10.1080/17425255.2020.1789589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Gastrointestinal stromal tumors (GIST) are the best example of a targeted therapy in solid tumors. The introduction of tyrosine kinase inhibitors (TKIs) deeply improved the prognosis of this tumor. However, a degree of inter-patient variability is still reported in response rates and pharmacogenetics may play an important role in the final clinical outcome. AREAS COVERED In this review, the authors provide an updated overview of the pharmacogenetic literature analyzing the role of polymorphisms in both GIST treatment efficacy and toxicity. EXPERT OPINION Besides the primary role of somatic DNA in dictating the clinical response to TKIs, several polymorphisms influencing their pharmacokinetics and pharmacodynamics have been identified as being potentially involved. In the last 10 years, many potential biomarkers have been proposed to predict clinical response and toxicity after TKI administration. However, the evidence is still too limited to promote a clinical translation. To date, the somatic mutational status represents the main player in clinical response to TKIs in GIST treatment; however, pharmacogenetics could still explain the degree of inter-patient variability observed in GIST patients. A combination of different theoretical approaches, experimental model systems, and statistical methods is clearly needed, in order to translate pharmacogenetics to clinical practice in the near future.
Collapse
Affiliation(s)
- Gloria Ravegnini
- Department of Pharmacy and Biotechnology, University of Bologna , Bologna, Italy
| | - Giorgia Valori
- Department of Pharmacy and Biotechnology, University of Bologna , Bologna, Italy
| | - Qianqian Zhang
- UOC di Anatomia Patologica, Fondazione Policlinico Universitario 'A. Gemelli' IRCCS , Rome, Italy
| | - Riccardo Ricci
- UOC di Anatomia Patologica, Fondazione Policlinico Universitario 'A. Gemelli' IRCCS , Rome, Italy.,Department of Pathology, Universita Cattolica del Sacro Cuore , Rome, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, University of Bologna , Bologna, Italy
| | - Sabrina Angelini
- Department of Pharmacy and Biotechnology, University of Bologna , Bologna, Italy
| |
Collapse
|
6
|
Stefanou IK, Gazouli M, Zografos GC, Toutouzas KG. Role of non-coding RNAs in pathogenesis of gastrointestinal stromal tumors. World J Meta-Anal 2020; 8:233-244. [DOI: 10.13105/wjma.v8.i3.233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/22/2020] [Accepted: 06/28/2020] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are considered the model solid malignancies of targeted therapy after the discovery of imatinib effectiveness against their tyrosine kinase inhibitors. Non-coding RNAs are molecules with no protein coding capacity that play crucial role to several biological steps of normal cell proliferation and differentiation. When the expression of these molecules found to be altered it seems that they affect the process of carcinogenesis in multiple ways, such as proliferation, apoptosis, differentiation, metastasis, and drug resistance. This review aims to provide an overview of the latest research papers and summarize the current evidence about the role of non-coding RNAs in pathogenesis of GISTs, including their potential clinical applications.
Collapse
Affiliation(s)
- Ioannis K Stefanou
- Department of Surgery, Hippocration Hospital Athens, Athens 11527, Greece
| | - Maria Gazouli
- Department of Basic Medical Sciences, Laboratory of Biology, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Georgios C Zografos
- 1st Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Konstantinos G Toutouzas
- 1st Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, Athens 11527, Greece
| |
Collapse
|
7
|
Chen Y, Qin C, Cui X, Geng W, Xian G, Wang Z. miR-4510 acts as a tumor suppressor in gastrointestinal stromal tumor by targeting APOC2. J Cell Physiol 2020; 235:5711-5721. [PMID: 31975384 DOI: 10.1002/jcp.29506] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/09/2020] [Indexed: 12/12/2022]
Abstract
Dysregulation of microRNAs (miRNAs) expression has been demonstrated in gastrointestinal stromal tumor (GIST). In this study, we aimed to determine the differential miRNAs expression in GISTs and explore the functional mechanism of differential miRNAs in GIST cells. We measured differential miRNAs in six pairs of GIST tissues and matched adjacent tissues through a high-throughput sequencing, which was confirmed in 64 pairs of GIST tissues and adjacent tissues by real-time polymerase chain reaction. We found that miR-4510 expression was significantly downregulated in GIST tissues compared to matched control tissues. Luciferase reporter assay identified apolipoprotein C-II (APOC2) as a direct target of miR-4510. Overexpression of miR-4510 inhibited the mRNA and protein expression of APOC2. In addition, overexpression of miR-4510 suppressed GIST cell proliferation, migration, and invasion. Overexpression of miR-4510 also inhibited the phosphorylation of AKT and ERK1/2, reduced the expression of matrix metallopeptidase 2 (MMP2) and MMP9. APOC2 knockdown mimicked the effect of miR-4510 overexpression. Further investigation confirmed that APOC2 was notably upregulated in GIST tissues compared to adjacent control tissues. These results suggested that miR-4510 downregulation could promote GIST progression, including growth, invasion, and metastasis, through increasing APOC2 expression.
Collapse
Affiliation(s)
- Yuan Chen
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Chengkun Qin
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xianping Cui
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Wenmao Geng
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Guozhe Xian
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Zhiyi Wang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
8
|
Prognostic Role of miR-221 and miR-222 Expression in Cancer Patients: A Systematic Review and Meta-Analysis. Cancers (Basel) 2019; 11:cancers11070970. [PMID: 31336701 PMCID: PMC6678869 DOI: 10.3390/cancers11070970] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022] Open
Abstract
Background: A wealth of evidence has shown that microRNAs (miRNAs) can modulate specific genes, increasing our knowledge on the fine-tuning regulation of protein expression. miR-221 and miR-222 have been frequently identified as deregulated across different cancer types; however, their prognostic significance in cancer remains controversial. In view of these considerations, we performed an updated systematic review and meta-analysis of published data investigating the effects of miR-221/222 on overall survival (OS) and other secondary outcomes among cancer patients. A systematic search of PubMed, Web of Knowledge, and Cochrane Library databases was performed. Hazard ratios (HRs) and 95% confidence intervals (95% CIs) were used to assess the strength of association. Results: Fifty studies, analyzing 6086 patients, were included in the systematic review. Twenty-five studies for miR-221 and 17 studies for miR-222 which assessed OS were included in the meta-analysis. High expression of miR-221 and miR-222 significantly predicted poor OS (HR: 1.48, 95% CI: 1.14–1.93, p = 0.003 and HR: 1.90, 95% CI: 1.43–2.54, p < 0.001, respectively). Subgroup analysis revealed that the finding on miR-221 was not as robust as the one on miR-222. Furthermore, high miR-222 expression was also associated with worse progression-free survival and disease-free survival pooled with recurrence-free survival. Conclusions: The meta-analysis demonstrated that high expression of miR-222 is associated with poor prognosis in cancer patients, whereas the significance of miR-221 remains unclear. More work is required to fully elucidate the role of miR-221 and miR-222 in cancer prognosis, particularly in view of the limitations of existing results, including the significant heterogeneity and limited number of studies for some cancers.
Collapse
|
9
|
Ravegnini G, Serrano C, Simeon V, Sammarini G, Nannini M, Roversi E, Urbini M, Ferrè F, Ricci R, Tarantino G, Pantaleo MA, Hrelia P, Angelini S. The rs17084733 variant in the KIT 3' UTR disrupts a miR-221/222 binding site in gastrointestinal stromal tumour: a sponge-like mechanism conferring disease susceptibility. Epigenetics 2019; 14:545-557. [PMID: 30983504 PMCID: PMC6557610 DOI: 10.1080/15592294.2019.1595997] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Several miRNAs are dysregulated in gastrointestinal stromal tumours (GIST), and miR-221/222 appear to have a prominent role in GIST biology. Therefore, we investigated the role of DNA variants located in miR-221/222 precursor sequences and their target KIT 3'UTR. Ninety-five polymorphisms were analysed in 115 GIST cases and 88 healthy controls. KIT 3'UTR rs17084733 and pri-miR-222 rs75246947 were found significantly associated with GIST susceptibility. Specifically, KIT rs17084733 A allele was more common in GIST, particularly in KIT wild-type (WT) patients (Padj = 0.017). rs17084733 variant is located within one of the three miR-221/222 binding sites in the KIT 3'UTR, resulting in a mismatch in this seed region. Conversely, KIT mRNA levels were lower in patients carrying the variant allele, except for KIT mutant GIST. Luciferase assay data in GIST cells, generated using a construct containing all the three miR-221/222 binding sites, are consistent with KIT mRNA levels in GIST patients. Reporter assay data, generated using a construct containing only the site encompassing rs17084733, confirmed that this is a functional variant disrupting the miR-221/222 binding site. In conclusion, this is the first study investigating the role of SNPs on miR-221/222 precursor sequences and their binding region on KIT 3'UTR in GIST. We identified the KIT variant rs17084733 as a possible novel genetic biomarker for risk of developing KIT-WT GIST. Moreover, our findings suggest the role of one of the three miR-221/222 binding sites on KIT 3'UTR as endogenous sponge, soaking up and subtracting miR-221/222 to the other two sites characterized by a higher affinity.
Collapse
Affiliation(s)
- Gloria Ravegnini
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - César Serrano
- b Medical Oncology Department , Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital , Barcelona , Spain
| | - Vittorio Simeon
- c Medical Statistics Unit , University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Giulia Sammarini
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Margherita Nannini
- d Department of Specialized, Experimental and Diagnostic Medicine, Sant'Orsola-Malpighi Hospital , University of Bologna , Bologna , Italy
| | - Erica Roversi
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Milena Urbini
- e "Giorgio Prodi" Cancer Research Center , University of Bologna , Bologna , Italy
| | - Fabrizio Ferrè
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Riccardo Ricci
- f UOC di Anatomia Patologica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy.,g Department of Pathology , Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giuseppe Tarantino
- e "Giorgio Prodi" Cancer Research Center , University of Bologna , Bologna , Italy
| | - Maria A Pantaleo
- d Department of Specialized, Experimental and Diagnostic Medicine, Sant'Orsola-Malpighi Hospital , University of Bologna , Bologna , Italy.,e "Giorgio Prodi" Cancer Research Center , University of Bologna , Bologna , Italy
| | - Patrizia Hrelia
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Sabrina Angelini
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| |
Collapse
|
10
|
Ravegnini G, Sammarini G, Serrano C, Nannini M, Pantaleo MA, Hrelia P, Angelini S. Clinical relevance of circulating molecules in cancer: focus on gastrointestinal stromal tumors. Ther Adv Med Oncol 2019; 11:1758835919831902. [PMID: 30854029 PMCID: PMC6399766 DOI: 10.1177/1758835919831902] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/30/2018] [Indexed: 12/12/2022] Open
Abstract
In recent years, growing research interest has focused on the so-called liquid biopsy. A simple blood test offers access to a plethora of information, which might be extremely helpful in understanding or characterizing specific diseases. Blood contains different molecules, of which circulating free DNA (cfDNA), circulating tumor DNA (ctDNA), circulating tumor cells (CTCs) and extracellular vesicles (EVs) are the most relevant. Conceivably, these molecules have the potential for tumor diagnosis, monitoring tumor evolution, and evaluating treatment response and pharmacological resistance. This review aims to present a state-of-the-art of recent advances in circulating DNA and circulating RNA in gastrointestinal stromal tumors (GISTs). To date, progress in liquid biopsy has been scarce in GISTs due to several issues correlated with the nature of the pathology. Namely, heterogeneity in primary and secondary mutations in key driver genes has greatly slowed the development and application in GISTs, unlike in other tumor types in which liquid biopsy has already been translated into clinical practice. However, meaningful novel data have shown in recent years a significant clinical potential of ctDNA, CTCs, EVs and circulating RNA in GISTs.
Collapse
Affiliation(s)
- Gloria Ravegnini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Giulia Sammarini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - César Serrano
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Margherita Nannini
- Department of Specialized, Experimental and Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Maria A Pantaleo
- Department of Specialized, Experimental and Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Sabrina Angelini
- Department of Pharmacy and Biotechnology, Via Irnerio 48, 40126 Bologna, Italy
| |
Collapse
|
11
|
Teng Y, Li Y, Lin Z, Gao Y, Cao X, Lou X, Lin F, Li Y. Analysis of miRNA expression profiling identifies miR-214-3p as a novel biomarker in sinonasal inverted papilloma. Epigenomics 2018; 10:1541-1553. [PMID: 29952653 DOI: 10.2217/epi-2018-0071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Aim: To analyze the expression profile, diagnostic and clinicopathological significances of miRNAs in sinonasal inverted papilloma (SNIP). Materials & methods: The expression profile of miRNAs was analyzed using a miRNA microarray approach. The potential functions and clinical significances of specific miRNAs were further analyzed by bioinformatics and statistical methods. Results: The microarray assay identified 37 significantly upregulated and 21 downregulated miRNAs in SNIP. Of nine miRNAs randomly selected, the expression levels of seven miRNAs were confirmed by quantitative real-time PCR. The potential target genes of several candidate miRNAs were enriched in some biological processes and cellular signaling pathways related to tumorigenesis. Receiever operating characteristic curve analysis for miR-214-3p indicated an area under the curve of 0.932. Notably, its expression level was significantly decreased in SNIP tissues and associated with SNIP staging and recurrence. Conclusion: MiR-214-3p can possibly serve as a valuable biomarker and a therapeutic target for SNIP.
Collapse
Affiliation(s)
- Yaoshu Teng
- Department of Otorhinolaryngology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 310006 Hangzhou, PR China
- Department of Otorhinolaryngology, The Fourth Clinical Medical College, Zhejiang Chinese Medical University, 310006 Hangzhou, PR China
| | - Yuandong Li
- Department of Otorhinolaryngology, The Fourth Clinical Medical College, Zhejiang Chinese Medical University, 310006 Hangzhou, PR China
| | - Zhihong Lin
- Department of Otorhinolaryngology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 310008 Hangzhou, PR China
| | - Yueqiu Gao
- Department of Otorhinolaryngology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 310006 Hangzhou, PR China
- Department of Otorhinolaryngology, The Fourth Clinical Medical College, Zhejiang Chinese Medical University, 310006 Hangzhou, PR China
| | - Xiaolin Cao
- Department of Otorhinolaryngology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 310006 Hangzhou, PR China
- Department of Otorhinolaryngology, The Fourth Clinical Medical College, Zhejiang Chinese Medical University, 310006 Hangzhou, PR China
| | - Xiangyu Lou
- Department of Otorhinolaryngology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 310006 Hangzhou, PR China
- Department of Otorhinolaryngology, The Fourth Clinical Medical College, Zhejiang Chinese Medical University, 310006 Hangzhou, PR China
| | - Fengchun Lin
- Department of Pathology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 310008 Hangzhou, PR China
| | - Yong Li
- Department of Otorhinolaryngology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 310006 Hangzhou, PR China
- Department of Otorhinolaryngology, The Fourth Clinical Medical College, Zhejiang Chinese Medical University, 310006 Hangzhou, PR China
| |
Collapse
|
12
|
McCarthy JB, El-Ashry D, Turley EA. Hyaluronan, Cancer-Associated Fibroblasts and the Tumor Microenvironment in Malignant Progression. Front Cell Dev Biol 2018; 6:48. [PMID: 29868579 PMCID: PMC5951929 DOI: 10.3389/fcell.2018.00048] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 04/13/2018] [Indexed: 12/16/2022] Open
Abstract
This review summarizes the roles of CAFs in forming a “cancerized” fibrotic stroma favorable to tumor initiation and dissemination, in particular highlighting the functions of the extracellular matrix component hyaluronan (HA) in these processes. The structural complexity of the tumor and its host microenvironment is now well appreciated to be an important contributing factor to malignant progression and resistance-to-therapy. There are multiple components of this complexity, which include an extensive remodeling of the extracellular matrix (ECM) and associated biomechanical changes in tumor stroma. Tumor stroma is often fibrotic and rich in fibrillar type I collagen and hyaluronan (HA). Cancer-associated fibroblasts (CAFs) are a major source of this fibrotic ECM. CAFs organize collagen fibrils and these biomechanical alterations provide highways for invading carcinoma cells either under the guidance of CAFs or following their epithelial to mesenchymal transition (EMT). The increased HA metabolism of a tumor microenvironment instructs carcinoma initiation and dissemination by performing multiple functions. The key effects of HA reviewed here are its role in activating CAFs in pre-malignant and malignant stroma, and facilitating invasion by promoting motility of both CAFs and tumor cells, thus facilitating their invasion. Circulating CAFs (cCAFs) also form heterotypic clusters with circulating tumor cells (CTC), which are considered to be pre-cursors of metastatic colonies. cCAFs are likely required for extravasation of tumors cells and to form a metastatic niche suitable for new tumor colony growth. Therapeutic interventions designed to target both HA and CAFs in order to limit tumor spread and increase response to current therapies are discussed.
Collapse
Affiliation(s)
- James B McCarthy
- Department of Laboratory Medicine and Pathology, Masonic Comprehensive Cancer Center, Minneapolis, MN, United States
| | - Dorraya El-Ashry
- Department of Laboratory Medicine and Pathology, Masonic Comprehensive Cancer Center, Minneapolis, MN, United States
| | - Eva A Turley
- London Regional Cancer Program, Department of Oncology, Biochemistry and Surgery, Schulich School of Medicine and Dentistry, Lawson Health Research Institute, Western University, London, ON, Canada
| |
Collapse
|
13
|
Wang Y, Li J, Kuang D, Wang X, Zhu Y, Xu S, Chen Y, Cheng H, Zhao Q, Duan Y, Wang G. miR-148b-3p functions as a tumor suppressor in GISTs by directly targeting KIT. Cell Commun Signal 2018; 16:16. [PMID: 29661252 PMCID: PMC5902930 DOI: 10.1186/s12964-018-0228-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/06/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Gain-of-function mutations and overexpression of KIT are characteristic features of gastrointestinal stromal tumor (GIST). Dysregulation in miRNA expression may lead to KIT overexpression and tumorigenesis. METHODS miRNA microarray analysis and real-time PCR were used to determine the miRNA expression profiles in a cohort of 69 clinical samples including 50 CD117IHC+/KITmutation GISTs and 19 CD117IHC-/wild-type GISTs. GO enrichment and KEGG pathway analyses were performed to reveal the predicted targets of the dysregulated miRNAs. Of the dysregulated miRNAs whose expression was inversely correlated with that of KIT miRNAs were predicted by bioinformatics analysis and confirmed by luciferase reporter assay. Cell counting kit-8 (CCK-8) and flow cytometry were used to measure the cell proliferation, cycle arrest and apoptosis. Wound healing and transwell assays were used to evaluate migration and invasion. A xenograft BALB/c nude mouse model was applied to investigate the tumorigenesis in vivo. Western blot and qRT-PCR were used to investigate the protein and mRNA levels of KIT and its downstream effectors including ERK, AKT and STAT3. RESULTS Of the six miRNAs whose expression was inversely correlated with that of KIT, we found that miR-148b-3p was significantly downregulated in the CD117IHC+/KITmutation GIST cohort. This miRNA was subsequently found to inhibit proliferation, migration and invasion of GIST882 cells. Mechanistically, miR-148b-3p was shown to regulate KIT expression through directly binding to the 3'-UTR of the KIT mRNA. Restoration of miR-148b-3p expression in GIST882 cells led to reduced expression of KIT and the downstream effectors proteins ERK, AKT and STAT3. However, overexpression of KIT reversed the inhibitory effect of miR-148b-3p on cell proliferation, migration and invasion. Furthermore, we found that reduced miR-148b-3p expression correlated with poor overall survival (OS) and disease-free survival (DFS) in GIST patients. CONCLUSION miR-148b-3p functions as an important regulator of KIT expression and a potential prognostic biomarker for GISTs.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Dadao, Wuhan, 430030, People's Republic of China
| | - Jun Li
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Dadao, Wuhan, 430030, People's Republic of China
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Dong Kuang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Dadao, Wuhan, 430030, People's Republic of China
| | - Xiaoyan Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Dadao, Wuhan, 430030, People's Republic of China
| | - Yuanli Zhu
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Dadao, Wuhan, 430030, People's Republic of China
| | - Sanpeng Xu
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Dadao, Wuhan, 430030, People's Republic of China
| | - Yaobing Chen
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Dadao, Wuhan, 430030, People's Republic of China
| | - Henghui Cheng
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Dadao, Wuhan, 430030, People's Republic of China
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Yaqi Duan
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Dadao, Wuhan, 430030, People's Republic of China.
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Guoping Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Dadao, Wuhan, 430030, People's Republic of China.
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
14
|
Kupcinskas J. Small Molecules in Rare Tumors: Emerging Role of MicroRNAs in GIST. Int J Mol Sci 2018; 19:E397. [PMID: 29385688 PMCID: PMC5855619 DOI: 10.3390/ijms19020397] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 01/24/2018] [Accepted: 01/24/2018] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of gastrointestinal tract. GISTs have very different clinical phenotypes and underlying molecular characteristics that are not yet completely understood. microRNAs (miRNAs) have been shown to participate in carcinogenesis pathways through post-transcriptional regulation of gene expression in different tumors. Over the last years emerging evidence has highlighted the role of miRNAs in GISTs. This review provides an overview of original research papers that analyze miRNA deregulation patterns, functional role, diagnostic, therapeutic and prognostic implications in GIST as well as provides directions for further research in the field.
Collapse
Affiliation(s)
- Juozas Kupcinskas
- Institute for Digestive Research, Academy of Medicine, Lithuanian University of Health Sciences, Eiveniu str. 2, LT-50009 Kaunas, Lithuania.
- Department of Gastroenterology, Academy of Medicine, Lithuanian University of Health Sciences, Eiveniu str. 2, LT-50009 Kaunas, Lithuania.
| |
Collapse
|
15
|
Li Z, Yang B, Weng X, Tse G, Chan MTV, Wu WKK. Emerging roles of MicroRNAs in osteonecrosis of the femoral head. Cell Prolif 2017; 51. [PMID: 29131454 DOI: 10.1111/cpr.12405] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 08/28/2017] [Indexed: 12/14/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is one of the most common orthopaedic diseases. The exact pathogenic mechanism of ONFH is still unknown. MicroRNAs (miRNAs) are a class of non-coding RNAs that negatively modulate gene expression at post-transcriptional level. An increasing number of studies have shown that miRNAs play crucial roles in different physiological processes, including development, cell proliferation, differentiation and metabolism. Recently, multiple studies demonstrated that miRNAs are involved in the pathogenesis of ONFH. In this review, we summarize dysregulated miRNAs and their functions in ONFH. Furthermore, we discuss their potential clinical applications for diagnosis and treatment of ONFH.
Collapse
Affiliation(s)
- Zheng Li
- Department of Orthopedics Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Yang
- Department of Orthopedics Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xisheng Weng
- Department of Orthopedics Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gary Tse
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | - Matthew T V Chan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong
| | - William Ka Kei Wu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong.,State Key Laboratory of Digestive Disease and LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
16
|
Ravegnini G, Sammarini G, Nannini M, Pantaleo MA, Biasco G, Hrelia P, Angelini S. Gastrointestinal stromal tumors (GIST): Facing cell death between autophagy and apoptosis. Autophagy 2017; 13:452-463. [PMID: 28055310 DOI: 10.1080/15548627.2016.1256522] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Autophagy and apoptosis are 2 fundamental biological mechanisms that may cooperate or be antagonistic, although both are involved in deciding the fate of cells in physiological or pathological conditions. These 2 mechanisms coexist simultaneously in cells and share common upstream signals and stimuli. Autophagy and apoptosis play pivotal roles in cancer development. Autophagy plays a key function in maintaining tumor cell survival by providing energy during unfavorable metabolic conditions through its recycling mechanism, and supporting the high energy requirement for metabolism and growth. This review focuses on gastrointestinal stromal tumors and cell death through autophagy and apoptosis, taking into account the involvement of both of these processes in tumor development and growth and as mechanisms of drug resistance. We also focus on the crosstalk between autophagy and apoptosis as an emerging field with major implications for the development of novel therapeutic options.
Collapse
Affiliation(s)
- Gloria Ravegnini
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna Italy
| | - Giulia Sammarini
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna Italy
| | - Margherita Nannini
- b Department of Specialized , Experimental and Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna , Bologna , Italy
| | - Maria A Pantaleo
- b Department of Specialized , Experimental and Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna , Bologna , Italy.,c "Giorgio Prodi" Cancer Research Center, University of Bologna , Bologna , Italy
| | - Guido Biasco
- b Department of Specialized , Experimental and Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna , Bologna , Italy.,c "Giorgio Prodi" Cancer Research Center, University of Bologna , Bologna , Italy
| | - Patrizia Hrelia
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna Italy
| | - Sabrina Angelini
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna Italy
| |
Collapse
|
17
|
Pantaleo MA, Ravegnini G, Astolfi A, Simeon V, Nannini M, Saponara M, Urbini M, Gatto L, Indio V, Sammarini G, Santini D, Ferracin M, Negrini M, Hrelia P, Biasco G, Angelini S. Integrating miRNA and gene expression profiling analysis revealed regulatory networks in gastrointestinal stromal tumors. Epigenomics 2016; 8:1347-1366. [PMID: 27625077 DOI: 10.2217/epi-2016-0030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
AIM Currently, little is known about differences in miRNA expression between KIT/PDGFRA mutant and KIT/PDGFRA wild-type (WT)-SDH deficient gastrointestinal stromal tumors (GIST). This prompted us to perform an integrated multiple expression profile of miRNA and mRNA, constructing an original miRNA-mRNA regulatory network in KIT/PDGFRA WT-SDH deficient GIST patients. PATIENTS & METHODS Analyses were carried out on KIT/PDGFRA mutant versus KIT/PDGFRA WT-SDH deficient GIST. Genome-wide miRNA and gene-expression analysis were performed using Agilent Human miRNA microarray and Affimetrix array, respectively. RESULTS Three potential regulatory networks (IGF1R → miR-139-5p/miR-455/let-7b, cyclin-dependent kinase 6 (CDK6) → miR-139-5p/let-7b and CD44 → miR-330-3p) were identified. CONCLUSION The miR-139-5p, 455-5p and let-7b signature, in particular, may represent an important therapeutic target in KIT/PDGFRA WT-SDH deficient GIST, usually characterized by IGF1R overexpression.
Collapse
Affiliation(s)
- Maria Abbondanza Pantaleo
- 'Giorgio Prodi' Cancer Research Center, University of Bologna, Bologna, Italy.,Department of Specialized, Experimental & Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Gloria Ravegnini
- Department of Pharmacy & Biotechnology, Via Irnerio 48, 40126 Bologna, Italy
| | - Annalisa Astolfi
- 'Giorgio Prodi' Cancer Research Center, University of Bologna, Bologna, Italy
| | - Vittorio Simeon
- Laboratory of Pre-Clinical & Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (PZ), Italy
| | - Margherita Nannini
- Department of Specialized, Experimental & Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Maristella Saponara
- Department of Specialized, Experimental & Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Milena Urbini
- Department of Specialized, Experimental & Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Lidia Gatto
- Department of Specialized, Experimental & Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Valentina Indio
- Department of Specialized, Experimental & Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Giulia Sammarini
- Department of Pharmacy & Biotechnology, Via Irnerio 48, 40126 Bologna, Italy
| | - Donatella Santini
- Pathology Unit, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Manuela Ferracin
- Department of Specialized, Experimental & Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Massimo Negrini
- Department of Morphology, Surgery & Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Patrizia Hrelia
- Department of Pharmacy & Biotechnology, Via Irnerio 48, 40126 Bologna, Italy
| | - Guido Biasco
- 'Giorgio Prodi' Cancer Research Center, University of Bologna, Bologna, Italy.,Department of Specialized, Experimental & Diagnostic Medicine, Sant'Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Sabrina Angelini
- Department of Pharmacy & Biotechnology, Via Irnerio 48, 40126 Bologna, Italy
| |
Collapse
|
18
|
Polymorphisms in DNA repair genes in gastrointestinal stromal tumours: susceptibility and correlation with tumour characteristics and clinical outcome. Tumour Biol 2016; 37:13413-13423. [PMID: 27460091 DOI: 10.1007/s13277-016-5276-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/15/2016] [Indexed: 12/13/2022] Open
Abstract
DNA repair pathways play an essential role in cancer susceptibility by maintaining genomic integrity. This led us to investigate the influence of polymorphisms in the genes coding repair pathway enzymes on gastrointestinal stromal tumours (GIST) susceptibility, tumour characteristics and clinical outcome. We investigated a panel of 20 polymorphisms in 11 genes in 81 cases and 147 controls. The XPD rs13181 wild-type allele and hOGG1 rs1052133 and XPF rs1800067 minor alleles were significantly associated with disease susceptibility. XPA rs1800975 and rs2808668 were associated with tumour size (P = 0.018), metastatic status at onset (P = 0.035) and mitotic index (P = 0.002). With regards to outcome treatment, the XPD rs50872 minor allele had a significant favourable impact on time to progression (TTP). Similarly, the XPC rs2228000 minor allele was correlated with a longer TTP (P = 0.03). On the contrary, the XPC rs2228001 and hOGG1 rs1052133 minor alleles were associated with a diminished TTP (P = 0.005 and P = 0.01, respectively). Regarding OS, we found the presence of at least one hOGG1 (rs1052133) minor allele that had a 60 % lower risk to die compared to the wild-type carriers (P = 0.04). Furthermore, the XRCC3 rs861539 variant allele is associated with a hazard of early death compared with the wild-type genotype (P = 0.04). To the best of our knowledge, this is the first study on polymorphisms in DNA repair genes, belonging to the different pathways, extensively evaluated in GIST patients. Through this multiple candidate gene approach, we report for the first time the significant associations between polymorphisms in DNA repair genes, susceptibility, clinical pathological features and clinical outcome in GIST.
Collapse
|
19
|
Ravegnini G, Sammarini G, Angelini S, Hrelia P. Pharmacogenetics of tyrosine kinase inhibitors in gastrointestinal stromal tumor and chronic myeloid leukemia. Expert Opin Drug Metab Toxicol 2016; 12:733-42. [DOI: 10.1080/17425255.2016.1184649] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Gloria Ravegnini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Giulia Sammarini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Sabrina Angelini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
20
|
Personalized Medicine in Gastrointestinal Stromal Tumor (GIST): Clinical Implications of the Somatic and Germline DNA Analysis. Int J Mol Sci 2015; 16:15592-608. [PMID: 26184165 PMCID: PMC4519915 DOI: 10.3390/ijms160715592] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/07/2015] [Accepted: 07/07/2015] [Indexed: 12/26/2022] Open
Abstract
Gastrointestinal stromal tumors (GIST) are the most common mesenchymal tumors of the gastrointestinal tract. They are characterized by gain of function mutations in KIT or PDGFRA tyrosine kinase receptors, with their consequent constitutive activation. The gold standard therapy is imatinib that offers a good and stable response for approximately 18–36 months. However, resistance is very common and it is vital to identify new biomarkers. Up until now, there have been two main approaches with focus to characterize novel targets. On the one hand, the focus is on the tumor genome, as the final clinical outcome depends mainly from the cancer specific mutations/alterations patterns. However, the germline DNA is important as well, and it is inconceivable to think the patients response to the drug is not related to it. Therefore the aim of this review is to outline the state of the art of the personalized medicine in GIST taking into account both the tumor DNA (somatic) and the patient DNA (germline).
Collapse
|