1
|
Silva FHDS, Underwood A, Almeida CP, Ribeiro TS, Souza-Fagundes EM, Martins AS, Eliezeck M, Guatimosim S, Andrade LO, Rezende L, Gomes HW, Oliveira CA, Rodrigues RC, Borges IT, Cassali GD, Ferreira E, Del Puerto HL. Transcription factor SOX3 upregulated pro-apoptotic genes expression in human breast cancer. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:212. [PMID: 36175695 DOI: 10.1007/s12032-022-01758-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/20/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Sex-determining region Y-box 3 (SOX3) protein, a SOX transcriptions factors group, has been identified as a key regulator in several diseases, including cancer. Downregulation of transcriptions factors in invasive ductal carcinoma (IDC) can interfere in neoplasia development, increasing its aggressiveness. We investigated SOX3 protein expression and its correlation with apoptosis in the MDA-MB-231 cell line, as SOX3 and Pro-Caspase-3 immunoexpression in paraffin-embedded invasive ductal carcinoma tissue samples from patients (n = 27). Breast cancer cell line MDA-MD-231 transfected with pEF1-SOX3 + and pEF1-Empty vector followed by cytotoxicity assay (MTT), Annexin-V FITC PI for apoptosis percentage assessment by flow cytometry, qPCR for apoptotic-related gene expression, immunofluorescence, and immunohistochemistry to SOX3 immunolocalization in culture cells, and paraffin-embedded invasive ductal carcinoma tissue samples. RESULTS Apoptotic rate was higher in cells transfected with pEF1-SOX3 + (56%) than controls (10%). MDA-MB-231 transfected with pEF1-SOX3 + presented upregulation of pro-apoptotic mRNA from CASP3, CASP8, CASP9, and BAX genes, contrasting with downregulation antiapoptotic mRNA from BCL2, compared to non-transfected cells and cells transfected with pEF1-empty vector (p < 0.005). SOX3 protein nuclear expression was detected in 14% (4/27 cases) of ductal carcinoma cases, and pro-Caspase-3 expression was positive in 50% of the cases. CONCLUSION Data suggest that SOX3 transcription factor upregulates apoptosis in breast cancer cell line MDA-MB-231, and has a down nuclear expression in ductal carcinoma cases, and need to be investigated as a tumor suppressor protein, and its loss of expression and non-nuclear action turn the cells resistant to apoptosis. Further studies are necessary to understand how SOX3 protein regulates the promoter regions of genes involved in apoptosis.
Collapse
Affiliation(s)
- Felipe Henrique de Souza Silva
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627 - Campus UFMG, Belo Horizonte, MG, 31270-901, Brazil
| | - Adam Underwood
- Division of Mathematics and Sciences, Walsh University, North Canton, OH, USA
| | - Camila Pereira Almeida
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627 - Campus UFMG, Belo Horizonte, MG, 31270-901, Brazil
| | - Thais Salviana Ribeiro
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627 - Campus UFMG, Belo Horizonte, MG, 31270-901, Brazil
| | - Elaine M Souza-Fagundes
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Almir S Martins
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Marcos Eliezeck
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Silvia Guatimosim
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Luciana O Andrade
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Luisa Rezende
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Hipacia Werneck Gomes
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Cleida Aparecida Oliveira
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | | | - Isabella Terra Borges
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627 - Campus UFMG, Belo Horizonte, MG, 31270-901, Brazil
| | - Geovanni Dantas Cassali
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627 - Campus UFMG, Belo Horizonte, MG, 31270-901, Brazil
| | - Enio Ferreira
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627 - Campus UFMG, Belo Horizonte, MG, 31270-901, Brazil
| | - Helen Lima Del Puerto
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Presidente Antônio Carlos, 6627 - Campus UFMG, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
2
|
LINC00662 Promotes Proliferation and Invasion and Inhibits Apoptosis of Glioma Cells Through miR-483-3p/SOX3 Axis. Appl Biochem Biotechnol 2022; 194:2857-2871. [PMID: 35275355 DOI: 10.1007/s12010-022-03855-2] [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: 09/15/2021] [Accepted: 02/11/2022] [Indexed: 11/02/2022]
Abstract
LINC00662 plays a prominent role in the carcinogenesis and progression of diverse cancers. However, its biological functions in glioma are still unclear. LINC00662 expression in glioma tissue samples and cell lines was examined by quantitative real-time polymerase chain reaction. The correlation between LINC00662 expression and the clinical characteristics of 50 patients with glioma was analyzed. LINC00662 knockdown and overexpression cell lines were constructed, and the effects of LINC00662 on the proliferation, invasion, and apoptosis of glioma cells were evaluated by cell counting kit-8, 5-ethynyl-2'-deoxyuridine, Transwell, and flow cytometry assays, respectively. Besides, the relationships among LINC00662, miR-483-3p, and sex-determining region Y-box 3 (SOX3) were assessed by dual-luciferase reporter assay and RNA immunoprecipitation assay. Western blot was used to detect the regulatory effects of LINC00662 and miR-483-3p on SOX3 expression in glioma cells. LINC00662 expression level was elevated in glioma tissues and cell lines compared to that in normal tissues and cell lines. LINC00662 high expression was associated with the adverse prognosis of patients with glioma. Knockdown of LINC00662 repressed the proliferation and invasion of glioma cells, and promoted apoptosis. Additionally, it was revealed that LINC00662 acted as the molecular sponge of miR-483-3p, and SOX3 was verified as a direct target of miR-483-3p. The inhibition of miR-483-3p expression and SOX3 overexpression reversed the biological effects of LINC00662 knockdown on glioma cells. This study reports the key regulatory role of LINC00662/miR-483-3p/SOX3 axis in the tumorigenesis and progression of glioma, bringing novel insights into the underlying mechanisms of glioma.
Collapse
|
3
|
Pluripotency Stemness and Cancer: More Questions than Answers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1376:77-100. [PMID: 34725790 DOI: 10.1007/5584_2021_663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Embryonic stem cells and induced pluripotent stem cells provided us with fascinating new knowledge in recent years. Mechanistic insight into intricate regulatory circuitry governing pluripotency stemness and disclosing parallels between pluripotency stemness and cancer instigated numerous studies focusing on roles of pluripotency transcription factors, including Oct4, Sox2, Klf4, Nanog, Sall4 and Tfcp2L1, in cancer. Although generally well substantiated as tumour-promoting factors, oncogenic roles of pluripotency transcription factors and their clinical impacts are revealing themselves as increasingly complex. In certain tumours, both Oct4 and Sox2 behave as genuine oncogenes, and reporter genes driven by composite regulatory elements jointly recognized by both the factors can identify stem-like cells in a proportion of tumours. On the other hand, cancer stem cells seem to be biologically very heterogeneous both among different tumour types and among and even within individual tumours. Pluripotency transcription factors are certainly implicated in cancer stemness, but do not seem to encompass its entire spectrum. Certain cancer stem cells maintain their stemness by biological mechanisms completely different from pluripotency stemness, sometimes even by engaging signalling pathways that promote differentiation of pluripotent stem cells. Moreover, while these signalling pathways may well be antithetical to stemness in pluripotent stem cells, they may cooperate with pluripotency factors in cancer stem cells - a paradigmatic example is provided by the MAPK-AP-1 pathway. Unexpectedly, forced expression of pluripotency transcription factors in cancer cells frequently results in loss of their tumour-initiating ability, their phenotypic reversion and partial epigenetic normalization. Besides the very different signalling contexts operating in pluripotent and cancer stem cells, respectively, the pronounced dose dependency of reprogramming pluripotency factors may also contribute to the frequent loss of tumorigenicity observed in induced pluripotent cancer cells. Finally, contradictory cell-autonomous and non-cell-autonomous effects of various signalling molecules operate during pluripotency (cancer) reprogramming. The effects of pluripotency transcription factors in cancer are thus best explained within the concept of cancer stem cell heterogeneity.
Collapse
|
4
|
Jia X, Wei L, Zhang Z. NEAT1 Overexpression Indicates a Poor Prognosis and Induces Chemotherapy Resistance via the miR-491-5p/ SOX3 Signaling Pathway in Ovarian Cancer. Front Genet 2021; 12:616220. [PMID: 33995475 PMCID: PMC8118527 DOI: 10.3389/fgene.2021.616220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 01/27/2021] [Indexed: 12/15/2022] Open
Abstract
Background Accumulated studies have reported that dysregulated long non-coding RNAs (lncRNAs) are crucial in ovarian cancer (OC) initiation and development. However, detailed biological functions of lncRNA NEAT1 during the progression of OC remains to be uncovered. Purpose Our aim was to identify the role of NEAT1 in cisplatin resistance of ovarian cancer and the underlying mechanisms. Methods The expression patterns of NEAT1 in OC cell lines and tissue samples were identified by qRT-PCR. The cisplatin (DDP) sensitivity of OC cells was detected by MTT and CCK8 assay, while OC cell apoptosis and cell cycle were detected using flow cytometer assays. In addition, effects of NEAT1 on tumor growth were determined by xenograft tumor model. Luciferase reporter assay was conducted to prove the regulatory relation of miR-491-5p, NEAT1, and SOX3. Importantly, the expression of NEAT1 in exosomes from cisplatin-resistant patients was also determined by using qRT-PCR. Results In this study, upregulated NEAT1 was detected in OC cell lines and tissues. Meanwhile, NEAT1 was also increased in cisplatin-resistant OC cell lines and tissues. Upregulation of NEAT1 inhibited cisplatin-induced OC cell apoptosis and promoted cell proliferation, while knockdown of NEAT1 played the opposite role. These effects were also observed in vivo. Furthermore, direct interaction was observed between NEAT1 and miR-491-5p. NEAT1 led to the upregulation of miR-491-5p-targeted SOX3 mRNA. Importantly, this study also showed upregulated NEAT1 expression in serum exosomes derived from cisplatin-resistant patients. Conclusion NEAT1 is vital in the chemoresistance of ovarian cancer through regulating miR-491-5p/SOX3 pathway, showing that NEAT1 might be a potential target for OC resistance treatment.
Collapse
Affiliation(s)
- Xinzhuan Jia
- Department of Reproductive Medicine, The Fourth Hospital, Hebei Medical University, Shijiazhuang, China
| | - Lan Wei
- Department of Chest Surgery, Hebei Chest Hospital, Shijiazhuang, China
| | - Zhengmao Zhang
- Department of Gynecology, The Fourth Hospital, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
5
|
Lu S, Yu Z, Zhang X, Sui L. MiR-483 Targeted SOX3 to Suppress Glioma Cell Migration, Invasion and Promote Cell Apoptosis. Onco Targets Ther 2020; 13:2153-2161. [PMID: 32210581 PMCID: PMC7075338 DOI: 10.2147/ott.s240619] [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: 11/30/2019] [Accepted: 02/01/2020] [Indexed: 12/11/2022] Open
Abstract
Objective Glioma is the most common malignant brain tumor that has high aggressiveness. The aim of this study was to investigate the potential therapeutic targets for gliomas. Materials and Methods Real-time quantitative polymerase chain reaction (RT-qPCR) was employed to calculate the expression of miRNA and genes. The connection between the expression of miR-483 and patients' overall survival rate was evaluated using Kaplan-Meier analysis. In addition, the underlying mechanism was detected using luciferase assay. Results The expression level of miR-483 was significantly decreased in glioma tissue samples and cell lines, compared to the adjacent tissues and normal cell lines. Downregulation of miR-483 or upregulation of SOX3 was associated with overall survival of glioma patients. Additionally, overexpression of miR-483 promotes cell invasion and migration and inhibits apoptosis. In addition, miR-483 directly targeted to SOX3, and the expression of miR-483 has a negative correlation with SOX3 in glioma tissues. SOX3 reversed partial functions of miR-483 on cell migration, invasion, and promoted cell apoptosis in glioma. Conclusion MiR-483 inhibited glioma cell migration, invasion, and promoted glioma cell apoptosis by targeting SOX3. MiR-483 maybe acted as a potential target for the treatment of glioma.
Collapse
Affiliation(s)
- Shujing Lu
- Department of Critical Care Medicine, Liaocheng Third People's Hospital, Liaocheng 252000, Shandong, People's Republic of China
| | - Zhengyang Yu
- Department of Internal Neurology, Liaocheng Third People's Hospital, Liaocheng 252000, Shandong, People's Republic of China
| | - Xia Zhang
- Department of Internal Neurology, Liaocheng Third People's Hospital, Liaocheng 252000, Shandong, People's Republic of China
| | - Lingling Sui
- Department of Internal Neurology, Liaocheng Third People's Hospital, Liaocheng 252000, Shandong, People's Republic of China
| |
Collapse
|
6
|
Yang G, He F, Duan H, Shen J, Dong Q. lncRNA FLVCR-AS1 promotes osteosarcoma growth by targeting miR381-3p/CCND1. Onco Targets Ther 2020; 13:163-172. [PMID: 32021264 PMCID: PMC6966140 DOI: 10.2147/ott.s214813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/18/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose This article reports on FLVCR-AS1 effects on osteosarcoma (OS) growth. Methods Tumor tissue and adjacent normal tissue of 48 OS patients were collected. HOS and 143B cells were transfected. Gene expression was examined with qRT-PCR and Western blot. CCK8 assays and cell cloning was performed to measure cell proliferation. Cell cycle and apoptosis were assessed. Luciferase-reporter gene assays and RNA pull-down tests were used to detect targeting relationships between genes. Results Prominently higher FLVCR-AS1 expression was found in OS tissue and cells, and was associated with poor prognosis (P<0.05, P<0.01, or P<0.001). Compared with the siCtrl group, 143B and HOS cells of the siFLVCR-AS1 group had significantly lower OD450 values and clone numbers and obviously higher percentages of cells in the G1 phase and apoptosis (P<0.01 or P<0.001). miR381-3p expression was directly inhibited by FLVCR-AS1, and CCND1 expression was directly suppressed by miR381-3p. Compared with the FLVCR-AS1 group, 143B cells of the FLVCR-AS1+ miR381-3p mimic group and FLVCR-AS1+ siCCND1 group showed remarkably lower OD450 values and clone numbers obviously higher apoptosis and percentage of cells in the G1 phase (P<0.05, P<0.01, or P<0.001). Conclusion FLVCR-AS1 promoted OS growth by upregulating CCND1 expression via downregulation of miR381-3p.
Collapse
Affiliation(s)
- Guang Yang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.,Department of Orthopedics, First Affiliated Hospital of Kunming Medical University, Kunming 650200, China
| | - Fei He
- Department of Orthopedics, First Affiliated Hospital of Kunming Medical University, Kunming 650200, China
| | - Hao Duan
- Department of Orthopedics, First Affiliated Hospital of Kunming Medical University, Kunming 650200, China
| | - Jianlin Shen
- Department of Orthopedics, Affiliated Hospital of Putian University, Putian 351100, China
| | - Qirong Dong
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| |
Collapse
|
7
|
Bhatia S, Monkman J, Blick T, Duijf PH, Nagaraj SH, Thompson EW. Multi-Omics Characterization of the Spontaneous Mesenchymal-Epithelial Transition in the PMC42 Breast Cancer Cell Lines. J Clin Med 2019; 8:E1253. [PMID: 31430931 PMCID: PMC6723942 DOI: 10.3390/jcm8081253] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/15/2019] [Accepted: 08/15/2019] [Indexed: 12/16/2022] Open
Abstract
Epithelial-mesenchymal plasticity (EMP), encompassing epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET), are considered critical events for cancer metastasis. We investigated chromosomal heterogeneity and chromosomal instability (CIN) profiles of two sister PMC42 breast cancer (BC) cell lines to assess the relationship between their karyotypes and EMP phenotypic plasticity. Karyotyping by GTG banding and exome sequencing were aligned with SWATH quantitative proteomics and existing RNA-sequencing data from the two PMC42 cell lines; the mesenchymal, parental PMC42-ET cell line and the spontaneously epithelially shifted PMC42-LA daughter cell line. These morphologically distinct PMC42 cell lines were also compared with five other BC cell lines (MDA-MB-231, SUM-159, T47D, MCF-7 and MDA-MB-468) for their expression of EMP and cell surface markers, and stemness and metabolic profiles. The findings suggest that the epithelially shifted cell line has a significantly altered ploidy of chromosomes 3 and 13, which is reflected in their transcriptomic and proteomic expression profiles. Loss of the TGFβR2 gene from chromosome 3 in the epithelial daughter cell line inhibits its EMT induction by TGF-β stimulus. Thus, integrative 'omics' characterization established that the PMC42 system is a relevant MET model and provides insights into the regulation of phenotypic plasticity in breast cancer.
Collapse
Affiliation(s)
- Sugandha Bhatia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia.
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia.
- Translational Research Institute, Brisbane, QLD 4102, Australia.
| | - James Monkman
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Tony Blick
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Pascal Hg Duijf
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
- University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Shivashankar H Nagaraj
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Erik W Thompson
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia.
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia.
- Translational Research Institute, Brisbane, QLD 4102, Australia.
| |
Collapse
|