1
|
Lavareze L, Kimura TDC, Scarini JF, de Lima-Souza RA, Gonçalves MWA, Sales de Sá R, Aquino IG, Fernandes PM, Ribeiro FCP, Altemani A, Mariano FV, Fillmore GC, Egal ESA. Advances and current concepts on Eph receptors and ephrins in upper digestive tract cancers. Front Oncol 2025; 14:1520306. [PMID: 39839790 PMCID: PMC11746119 DOI: 10.3389/fonc.2024.1520306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Erythropoietin-producing hepatocellular (Eph) receptors comprise the largest group of surface receptors and are responsible for cellular signals. Eph/ephrin signaling has been identified to play a role in key cancer development and progression processes, especially in the upper gastrointestinal tract. The Eph/ephrin system has been described as a tumor suppressor in duodenal cancer, while in esophageal, gastric, hepatic, and pancreatic cancer, the system has been related to tumor progression. For their significant role in developing a wide range of malignancies, Eph receptors and their ligands have proven to be an important target for new anticancer therapies. In this review, we present an overview of the literature and highlight evidence supporting the role of the Eph/ephrin system in upper digestive tract cancers. In addition, we discuss molecular findings that represent promising therapeutic targets for these cancers.
Collapse
Affiliation(s)
- Luccas Lavareze
- Department of Pathology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Department of Oral Diagnosis, Piracicaba School of Dentistry, University of Campinas (FOP/UNICAMP), Piracicaba, São Paulo, Brazil
| | - Talita de Carvalho Kimura
- Department of Pathology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Department of Oral Diagnosis, Piracicaba School of Dentistry, University of Campinas (FOP/UNICAMP), Piracicaba, São Paulo, Brazil
| | - João Figueira Scarini
- Department of Pathology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Department of Oral Diagnosis, Piracicaba School of Dentistry, University of Campinas (FOP/UNICAMP), Piracicaba, São Paulo, Brazil
| | - Reydson Alcides de Lima-Souza
- Department of Pathology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Department of Oral Diagnosis, Piracicaba School of Dentistry, University of Campinas (FOP/UNICAMP), Piracicaba, São Paulo, Brazil
| | - Moisés Willian Aparecido Gonçalves
- Department of Pathology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Department of Oral Diagnosis, Piracicaba School of Dentistry, University of Campinas (FOP/UNICAMP), Piracicaba, São Paulo, Brazil
| | - Raisa Sales de Sá
- Department of Pathology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Department of Oral Diagnosis, Piracicaba School of Dentistry, University of Campinas (FOP/UNICAMP), Piracicaba, São Paulo, Brazil
| | - Iara Gonçalves Aquino
- Department of Pathology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Department of Oral Diagnosis, Piracicaba School of Dentistry, University of Campinas (FOP/UNICAMP), Piracicaba, São Paulo, Brazil
| | - Patricia Maria Fernandes
- Department of Pathology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | | | - Albina Altemani
- Department of Pathology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Fernanda Viviane Mariano
- Department of Pathology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Gary Chris Fillmore
- Biorepository and Molecular Pathology, Huntsman Cancer Institute, University of Utah (UU), Salt Lake City, UT, United States
| | - Erika Said Abu Egal
- Department of Pathology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Biorepository and Molecular Pathology, Huntsman Cancer Institute, University of Utah (UU), Salt Lake City, UT, United States
| |
Collapse
|
2
|
Kim Y, Miller WT. Contrasting Effects of Cancer-Associated Mutations in EphA3 and EphB2 Kinases. Biochemistry 2024:10.1021/acs.biochem.3c00674. [PMID: 38252844 PMCID: PMC11265570 DOI: 10.1021/acs.biochem.3c00674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Erythropoietin-producing hepatoma (Eph) receptors are a family of tyrosine kinases that can act as tumor promoters or tumor suppressors, depending on the receptor and cancer cell type. Cancer-associated somatic mutations have been identified in all Eph receptors, but in most cases, the functional effects of the mutations are unknown. In this study, we expressed and purified the kinase domains of wild-type (WT) EphA3 and EphB2 along with 16 cancer-associated mutants. We identified mutations that decrease EphA3 activity and both activating and inhibitory mutations in EphB2. To shed light on the mechanisms by which the mutations altered kinase activity, we measured the thermal stabilities of the enzymes and performed steady-state kinetic experiments. We also expressed the full-length receptors in HEK293T cells to determine the cellular effects. WT EphB2 promoted downstream ERK signaling, while a kinase-inactive mutant (S706F) was similar to the control cells. In contrast, WT EphA3 (but not loss-of-function mutants) inhibited ERK signaling. The reciprocal effects of EphB2 and EphA3 on ERK phosphorylation in HEK293T cells were also evident in Ras-GTP loading. Thus, consistent with the dual roles of Eph receptors as tumor promoters and tumor suppressors, somatic mutations have the potential to increase or decrease Eph function, resulting in changes in the downstream signaling transduction.
Collapse
Affiliation(s)
- Yunyoung Kim
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York 11794, United States
| | - W Todd Miller
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York 11794, United States
- Department of Veterans Affairs Medical Center, Northport, New York 11768, United States
| |
Collapse
|
3
|
Wang L, Zhao QF, Yang BB, Liang HJ, Zhang XE, Zhang XY, Yang WJ, Guo ZY, Xu X, Tian F, Wu QH. STAT3-EphA7 axis contributes to the progression of esophageal squamous cell carcinoma. Acta Oncol 2023; 62:1757-1766. [PMID: 37738252 DOI: 10.1080/0284186x.2023.2259601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND Our previous study has revealed that EphA7 was upregulated in patient-derived esophageal squamous cell carcinoma (ESCC) xenografts with hyper-activated STAT3, but its mechanism was still unclear. MATERIALS AND METHODS To assess the association between EphA7 and STAT3, western blotting, immunofluorescence, ChIP assay, and qRT-PCR were conducted. Truncated mutation and luciferase assay were performed to examine the promoter activity of EphA7. CCK-8 assay and colony formation were performed to assess the proliferation of ESCC. Cell-derived xenograft models were established to evaluate the effects of EphA7 on ESCC tumor growth. RNA-seq analyses were used to assess the effects of EphA7 on related signals. RESULTS In this study, EphA7 was found upregulated in ESCC cell lines with high STAT3 activation, and immunofluorescence also showed that EphA7 was co-localized with phospho-STAT3 in ESCC cells. Interestingly, suppressing STAT3 activation by the STAT3 inhibitor Stattic markedly inhibited the protein expression of EphA7 in ESCC cells, in contrast, activation of STAT3 by IL-6 obviously upregulated the protein expression of EphA7. Moreover, the transcription of EphA7 was also mediated by the activation of STAT3 in ESCC cells, and the -2000∼-1500 region was identified as the key promoter of EphA7. Our results also indicated that EphA7 enhanced the cell proliferation of ESCC, and silence of EphA7 significantly suppressed ESCC tumor growth. Moreover, EphA7 silence markedly abolished STAT3 activation-derived cell proliferation of ESCC. Additionally, RNA-seq analyses indicated that several tumor-related signaling pathways were significantly changed after EphA7 downregulation in ESCC cells. CONCLUSION Our results showed that the transcriptional expression of EphA7 was increased by activated STAT3, and the STAT3 signaling may act through EphA7 to promote the development of ESCC.
Collapse
Affiliation(s)
- Li Wang
- Department of Obstetrics and Gynecology, Center of Genetics and Prenatal Diagnosis, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Qiao-Feng Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, P. R. China
| | - Bing-Bing Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, P. R. China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, P. R. China
| | - Hui-Jie Liang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, P. R. China
| | - Xian-E Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, P. R. China
- Second People's Hospital of Henan Province, Zhengzhou, P. R. China
| | - Xiao-Yan Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, P. R. China
| | - Wan-Jing Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, P. R. China
| | - Zhi-Yu Guo
- SanQuan College of XinXiang Medical University, Xinxiang, P. R. China
| | - Xin Xu
- College of Biotechnology, Center for Self-Propelled Nanotechnologies, Suzhou Industrial Park Institute of Services Outsourcing, Suzhou, P. R. China
- Translational Cancer Research Laboratory, Suzhou Acumen Medical Technology, Suzhou, P. R. China
| | - Fang Tian
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, P. R. China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, P. R. China
| | - Qing-Hua Wu
- Department of Obstetrics and Gynecology, Center of Genetics and Prenatal Diagnosis, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| |
Collapse
|
4
|
Saikia M, Bhattacharyya DK, Kalita JK. Identification of Potential Biomarkers Using Integrative Approach: A Case Study of ESCC. SN COMPUTER SCIENCE 2023; 4:114. [PMID: 36573207 PMCID: PMC9769493 DOI: 10.1007/s42979-022-01492-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 11/03/2022] [Indexed: 12/24/2022]
Abstract
This paper presents a consensus-based approach that incorporates three microarray and three RNA-Seq methods for unbiased and integrative identification of differentially expressed genes (DEGs) as potential biomarkers for critical disease(s). The proposed method performs satisfactorily on two microarray datasets (GSE20347 and GSE23400) and one RNA-Seq dataset (GSE130078) for esophageal squamous cell carcinoma (ESCC). Based on the input dataset, our framework employs specific DE methods to detect DEGs independently. A consensus based function that first considers DEGs common to all three methods for further downstream analysis has been introduced. The consensus function employs other parameters to overcome information loss. Differential co-expression (DCE) and preservation analysis of DEGs facilitates the study of behavioral changes in interactions among DEGs under normal and diseased circumstances. Considering hub genes in biologically relevant modules and most GO and pathway enriched DEGs as candidates for potential biomarkers of ESCC, we perform further validation through biological analysis as well as literature evidence. We have identified 25 DEGs that have strong biological relevance to their respective datasets and have previous literature establishing them as potential biomarkers for ESCC. We have further identified 8 additional DEGs as probable potential biomarkers for ESCC, but recommend further in-depth analysis.
Collapse
Affiliation(s)
- Manaswita Saikia
- Department of Computer Science and Engineering, Tezpur University, Napaam, Tezpur, Assam 784028 India
| | - Dhruba K Bhattacharyya
- Department of Computer Science and Engineering, Tezpur University, Napaam, Tezpur, Assam 784028 India
| | - Jugal K Kalita
- Department of Computer Science, College of Engineering and Applied Science, University of Colorado, Colorado Springs, CO 80918 USA
| |
Collapse
|
5
|
Ming D, Ma J. EphA3 targeted by miR-3666 contributes to melanoma malignancy via activating ERK1/2 and p38 MAPK pathways. Open Med (Wars) 2022; 17:2098-2108. [PMID: 36578556 PMCID: PMC9758965 DOI: 10.1515/med-2022-0597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/18/2022] [Accepted: 10/11/2022] [Indexed: 12/23/2022] Open
Abstract
Melanoma is a rare, fatal type of skin tumor. Although EPH receptor A3 (EphA3) is deregulated in melanoma, its detailed role remained uncharacterized. Using real time quantitative PCR analysis and western blotting, EphA3 was identified to be upregulated in melanoma tissues and cells, while miR-3666 showed an opposite expression trend. Cell counting kit-8, scratch wound, and in vivo assays proved that EphA3 silence inhibited the melanoma cell proliferation and migration and retarded tumor growth in vivo. Furthermore, western blotting results displayed that EphA3 silence resulted in a low expression of p38-MAPK and p-ERK1/2. Mechanically, miR-3666 was proved to target EphA3 3'UTR by the luciferase reporter assay. Furthermore, miR-3666 mimic compromised the driven melanoma cell proliferation and migration by EphA3 overexpression. In addition, induction of ERK1/2 and p38 MAPK pathways offset the positive effect of EphA3 overexpression on melanoma cells. In conclusion, miR-3666 downregulated EphA3 expression and retarded melanoma malignancy via inactivating ERK1/2 and p38 MAPK pathways. Hence, miR-3666/EphA3 axis may represent a druggable target against melanoma progression.
Collapse
Affiliation(s)
- Di Ming
- Dermatological Department, Wuhan Asia General Hospital, Economic and Technological Development Zone, Wuhan 430056, Hubei, China
| | - Jingjing Ma
- Dermatological Department, Wuhan Asia General Hospital, No. 300 Taizihu North Road, Economic and Technological Development Zone, Wuhan 430056, Hubei, China
| |
Collapse
|
6
|
Xu QR, Du XH, Huang TT, Zheng YC, Li YL, Huang DY, Dai HQ, Li EM, Fang WK. Role of Cell-Cell Junctions in Oesophageal Squamous Cell Carcinoma. Biomolecules 2022; 12:biom12101378. [PMID: 36291586 PMCID: PMC9599896 DOI: 10.3390/biom12101378] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 02/05/2023] Open
Abstract
Cell-cell junctions comprise various structures, including adherens junctions, tight junctions, desmosomes, and gap junctions. They link cells to each other in tissues and regulate tissue homeostasis in critical cellular processes. Recent advances in cell-cell junction research have led to critical discoveries. Cell-cell adhesion components are important for the invasion and metastasis of tumour cells, which are not only related to cell-cell adhesion changes, but they are also involved in critical molecular signal pathways. They are of great significance, especially given that relevant molecular mechanisms are being discovered, there are an increasing number of emerging biomarkers, targeted therapies are becoming a future therapeutic concern, and there is an increased number of therapeutic agents undergoing clinical trials. Oesophageal squamous cell carcinoma (ESCC), the most common histological subtype of oesophageal cancer, is one of the most common cancers to affect epithelial tissue. ESCC progression is accompanied by the abnormal expression or localisation of components at cell-cell junctions. This review will discuss the recent scientific developments related to the molecules at cell-cell junctions and their role in ESCC to offer valuable insights for readers, provide a global view of the relationships between position, construction, and function, and give a reference for future mechanistic studies, diagnoses, and therapeutic developments.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - En-Min Li
- Correspondence: (E.-M.L.); (W.-K.F.)
| | | |
Collapse
|
7
|
Subtype Classification and Prognosis Signature Construction of Osteosarcoma Based on Cellular Senescence-Related Genes. JOURNAL OF ONCOLOGY 2022; 2022:4421952. [PMID: 36106335 PMCID: PMC9467774 DOI: 10.1155/2022/4421952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/27/2022] [Accepted: 07/04/2022] [Indexed: 11/17/2022]
Abstract
Background. Cellular senescence (CS) is an alternative procedure that replaces or reinforces inadequate apoptotic responses and is used as an influencing factor for a variety of cancers. The value of CS gene in evaluating the immunotherapy response and clinical outcome of osteosarcoma (OS) has not been reported, and an accurate risk model based on CS gene has not been developed for OS patients. Methods. 279 CS genes were obtained from CellAge. Univariate Cox regression analysis was used to screen the CS gene which was significantly related to the prognosis of OS samples in TARGET data set. The prognosis, clinicopathological features, immune infiltration, gene expression at immune checkpoints, tumor immune dysfunction and exclusion (TIDE) score, and chemotherapy resistance of OS were analyzed among clusters. Least absolute shrinkage and selection operator (Lasso) Cox regression analysis to build cellular senescence-related gene signature (CSRS). Univariate and multivariate Cox regression analysis of CSRS and clinical parameters were carried out, and the parameters with independent prognostic value were used to construct nomogram. Results. Based on 30 CS genes related to OS prognosis, OS samples were divided into three clusters: C1, C2, and C3. C3 showed the lowest survival rate and metastasis rate and the highest immune score and stromal score and was more likely to respond to immune checkpoint blockade (ICB) treatment. A CSRS scoring system including four CS genes (MYC, DLX2, EPHA3, and LIMK1) was constructed, which could distinguish the survival outcome, tumor microenvironment (TME) status, and ICB treatment response of patients with different CSRS score. Nomogram constructed by CSRS score and metastatic has a high prognostic value for OS. Conclusions. Our study identified a molecular classification determined by CS-related genes and developed a new CSRS that has potential value in OS immunotherapy response and clinical outcome prediction.
Collapse
|
8
|
Indolium 1 Exerts Activity against Vemurafenib-Resistant Melanoma In Vivo. Antioxidants (Basel) 2022; 11:antiox11050798. [PMID: 35624662 PMCID: PMC9137681 DOI: 10.3390/antiox11050798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/06/2022] [Accepted: 04/13/2022] [Indexed: 02/05/2023] Open
Abstract
The development of targeted therapies (BRAF/MEK inhibitors) and immunotherapy have had a major impact on the treatment of melanoma. However, the majority of patients with advanced melanomas succumb to their disease. The mechanisms of resistance to both targeted therapies and immunotherapies are numerous and have been well-described. These include the alternative activation of BRAF/MEK signaling, novel compensating mutations in additional oncogenes, and loss of neoantigens. There has been limited development of small molecules that target alternative pathways in melanoma in the last two decades. We have previously identified triphenylmethanes as a class that shows activity against a wide variety of tumors. We have synthesized a novel triphenylmethane, indolium 1, and demonstrated its efficacy against an aggressive vemurafenib-resistant melanoma in vivo. Indolium 1 has a novel mechanism of action against melanoma, in that it results in induction of the tumor-suppressor EPHA3. We believe that pre-IND studies are warranted for this novel compound, given its mechanism of action and ability to inhibit the growth of vemurafenib resistant melanoma in vivo.
Collapse
|
9
|
Yang SH, Liu W, Peng J, Xu YJ, Liu YF, Li Y, Peng MY, Ou-Yang Z, Chen C, Liu EY. High Expression of RhoBTB3 Predicts Favorable Chemothrapy Outcomes in non-M3 Acute Myeloid Leukemia. J Cancer 2021; 12:4229-4239. [PMID: 34093823 PMCID: PMC8176412 DOI: 10.7150/jca.50472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 04/25/2021] [Indexed: 11/05/2022] Open
Abstract
Background: The expression patterns and prognostic significance of the Rho family GTPases in acute myeloid leukemia have not been systematically studied yet. Methods: In our study, we analyzed the expression patterns of 21 Rho family GTPases gene members in AML patients based on GEPIA database. 10 gene members with significant differential expression in AML tissue and healthy tissue were selected for subsequent research. Survival curve analysis in TCGA and GEO dataset preliminary showed that RhoBTB3 is related with the prognosis of non-M3 AML patients. The differential expression of RhoBTB3 on AML bone marrow and normal bone marrow was verified by RT-qPCR. We performed Kaplan-Meier survival analysis and Multivariate Cox analysis to assess the prognostic value of RhoBTB3 in non-M3 AML patients with different treatment regimens. Gene functional enrichment analysis of RhoBTB3 was performed using GO, KEGG and PPI network. Results: The AML patients from TCGA database were partitioned into 2 groups based on different treatment regimens: chemotherapy group and allo-HSCT group. In chemotherapy group, patients with higher expression level of RhoBTB3 showed relatively longer OS and EFS, multivariate Cox analysis revealed high RhoBTB3 mRNA expression as an independent favorable prognostic factor. However, in allo-HSCT group, no significant difference of OS and EFS were found between RhoBTB3 high and low subgroups. Meanwhile, allo-HSCT could circumvent the unfavorable prognosis that was associated with downregulation of RhoBTB3. Functional enrichment analysis showed the association of RhoBTB3 expression with several fundamental physiological components and pathways, including extracellular matrix components, extracellular structure organization, and cytokine-cytokine receptor interaction. Conclusions: Our study identified RhoBTB3 as a prognostic marker and may aid in the selection of the appropriate treatment options between chemotherapy and allo-HCST in non-M3 AML patients. Further researches are necessary to clarify the involvement of RhoBTB3 in the pathogenesis of AML.
Collapse
Affiliation(s)
- Shuang-Hui Yang
- Department of Hematology, XiangYa Hospital, Central South University, XiangYa Road No.87, Changsha 410008, China
| | - Wei Liu
- Department of Hematology, XiangYa Hospital, Central South University, XiangYa Road No.87, Changsha 410008, China
| | - Jie Peng
- Department of Hematology, XiangYa Hospital, Central South University, XiangYa Road No.87, Changsha 410008, China
| | - Ya-Jing Xu
- Department of Hematology, XiangYa Hospital, Central South University, XiangYa Road No.87, Changsha 410008, China
| | - Yan-Feng Liu
- Department of Hematology, XiangYa Hospital, Central South University, XiangYa Road No.87, Changsha 410008, China
| | - Yan Li
- Department of Hematology, XiangYa Hospital, Central South University, XiangYa Road No.87, Changsha 410008, China
| | - Min-Yuan Peng
- Department of Hematology, XiangYa Hospital, Central South University, XiangYa Road No.87, Changsha 410008, China
| | - Zhao Ou-Yang
- Department of Hematology, XiangYa Hospital, Central South University, XiangYa Road No.87, Changsha 410008, China
| | - Cong Chen
- Department of Hematology, XiangYa Hospital, Central South University, XiangYa Road No.87, Changsha 410008, China
| | - En-Yi Liu
- Department of Hematology, XiangYa Hospital, Central South University, XiangYa Road No.87, Changsha 410008, China
| |
Collapse
|
10
|
Markers of Angiogenesis, Lymphangiogenesis, and Epithelial-Mesenchymal Transition (Plasticity) in CIN and Early Invasive Carcinoma of the Cervix: Exploring Putative Molecular Mechanisms Involved in Early Tumor Invasion. Int J Mol Sci 2020; 21:ijms21186515. [PMID: 32899940 PMCID: PMC7554870 DOI: 10.3390/ijms21186515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 02/01/2023] Open
Abstract
The establishment of a proangiogenic phenotype and epithelial-to-mesenchymal transition (EMT) are considered as critical events that promote the induction of invasive growth in epithelial tumors, and stimulation of lymphangiogenesis is believed to confer the capacity for early dissemination to cancer cells. Recent research has revealed substantial interdependence between these processes at the molecular level as they rely on common signaling networks. Of great interest are the molecular mechanisms of (lymph-)angiogenesis and EMT associated with the earliest stages of transition from intraepithelial development to invasive growth, as they could provide the source of potentially valuable tools for targeting tumor metastasis. However, in the case of early-stage cervical cancer, the players of (lymph-)angiogenesis and EMT processes still remain substantially uncharacterized. In this study, we used RNA sequencing to compare transcriptomes of HPV(+) preinvasive neoplastic lesions and early-stage invasive carcinoma of the cervix and to identify (lymph-)angiogenesis- and EMT-related genes and pathways that may underlie early acquisition of invasive phenotype and metastatic properties by cervical cancer cells. Second, we applied flow cytometric analysis to evaluate the expression of three key lymphangiogenesis/EMT markers (VEGFR3, MET, and SLUG) in epithelial cells derived from enzymatically treated tissue specimens. Overall, among 201 differentially expressed genes, a considerable number of (lymph-)angiogenesis and EMT regulatory factors were identified, including genes encoding cytokines, growth factor receptors, transcription factors, and adhesion molecules. Pathway analysis confirmed enrichment for angiogenesis, epithelial differentiation, and cell guidance pathways at transition from intraepithelial neoplasia to invasive carcinoma and suggested immune-regulatory/inflammatory pathways to be implicated in initiation of invasive growth of cervical cancer. Flow cytometry showed cell phenotype-specific expression pattern for VEGFR3, MET, and SLUG and revealed correlation with the amount of tumor-infiltrating lymphocytes at the early stages of cervical cancer progression. Taken together, these results extend our understanding of driving forces of angiogenesis and metastasis in HPV-associated cervical cancer and may be useful for developing new treatments.
Collapse
|
11
|
Tavares V, Pinto R, Assis J, Pereira D, Medeiros R. Dataset of GWAS-identified variants underlying venous thromboembolism susceptibility and linkage to cancer aggressiveness. Data Brief 2020; 30:105399. [PMID: 32258274 PMCID: PMC7114903 DOI: 10.1016/j.dib.2020.105399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/24/2020] [Accepted: 03/03/2020] [Indexed: 01/01/2023] Open
Abstract
Venous thromboembolism (VTE) is a common cardiovascular disease, for which several single nucleotide polymorphisms (SNPs) underlying susceptibility were identified. Apart from candidate gene approach, genome-wide association studies (GWAS) have contributed to the identification of novel VTE-associated SNPs, including some with no clear role in the haemostatic system. These genetic variants constitute potential cancer-related biomarkers, particularly predictive and prognostic biomarkers, as a two-way association between VTE and cancer is well established. The present dataset comprises the data obtained from GWAS performed to identify genetic variants associated with VTE risk. Furthermore, this dataset also comprises data regarding previously reported candidate gene and validation reports performed in adults of European ancestry that also analysed the VTE GWAS-identified variants. Lastly, to evaluate the impact of these genetic variants in carcinogenesis, a broad search was made, which has let us to establish putative links between several VTE-associated genes and cancer hallmarks in a review article entitled “Venous thromboembolism GWAS reported genetic makeup and the hallmarks of cancer: linkage to ovarian tumour behaviour”.
Collapse
Affiliation(s)
- Valéria Tavares
- Molecular Oncology and Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edificio Laboratórios, 1° piso, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.,ICBAS, Abel Salazar Institute for the Biomedical Sciences, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Ricardo Pinto
- Molecular Oncology and Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edificio Laboratórios, 1° piso, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
| | - Joana Assis
- Molecular Oncology and Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edificio Laboratórios, 1° piso, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.,FMUP, Faculty of Medicine, Porto University, Porto, Portugal
| | - Deolinda Pereira
- Molecular Oncology and Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edificio Laboratórios, 1° piso, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.,Oncology Department, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edificio Laboratórios, 1° piso, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.,ICBAS, Abel Salazar Institute for the Biomedical Sciences, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.,FMUP, Faculty of Medicine, Porto University, Porto, Portugal.,CEBIMED, Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
| |
Collapse
|