Figure - available from: Oncogene
This content is subject to copyright. Terms and conditions apply.
Enhancement of xenograft growth and metastasis by ectopic expression of IFIT1 or IFIT3 in OSCC cells. a Growth curves of SAS-Vector, SAS-IFIT1, and SAS-IFIT3 cells derived xenografts in nude mice. Aliquots of 5 × 10⁵ cells were subcutaneously implanted into nude mice. The results shown are the means ± SD from five mice. **P < 0.01 compared with the SAS-Vector group. P-values were determined by ANOVA test. b–d Tail vein administration of SAS-Vector, SAS-IFIT1, and SAS-IFIT3 cells. Kaplan-Meier survival curve of mice injected with SAS-IFIT1 or SAS-IFIT3 cells compared with SAS-Vector control cells (b). The average number of tumor lesions formed in the lungs (c); and the number of lymph nodes with tumor cell invasion (d). P-values were determined by Kruskal Wallis test and Fisher’s exact test compared to the SAS-Vector control. e–g Orthotopic xenograft tongue tumors of SAS-Vector, SAS-IFIT1 or SAS-IFIT3 cells. e Representative fluorescence images of the tongue and associated lymph node metastasis in neck tissues of orthotopic xenograft tongue tumors derived from SAS-Vector, SAS-IFIT1, and SAS-IFIT3 cells. f H&E-stained histopathology sections of the tongue and metastatic lymph nodes in neck tissues. The images of the areas within the black boxes were magnified to visualize the tumor cells in tongue and cervical lymph nodes (g) Percentage of cervical lymph node metastasis in orthotopic mice

Enhancement of xenograft growth and metastasis by ectopic expression of IFIT1 or IFIT3 in OSCC cells. a Growth curves of SAS-Vector, SAS-IFIT1, and SAS-IFIT3 cells derived xenografts in nude mice. Aliquots of 5 × 10⁵ cells were subcutaneously implanted into nude mice. The results shown are the means ± SD from five mice. **P < 0.01 compared with the SAS-Vector group. P-values were determined by ANOVA test. b–d Tail vein administration of SAS-Vector, SAS-IFIT1, and SAS-IFIT3 cells. Kaplan-Meier survival curve of mice injected with SAS-IFIT1 or SAS-IFIT3 cells compared with SAS-Vector control cells (b). The average number of tumor lesions formed in the lungs (c); and the number of lymph nodes with tumor cell invasion (d). P-values were determined by Kruskal Wallis test and Fisher’s exact test compared to the SAS-Vector control. e–g Orthotopic xenograft tongue tumors of SAS-Vector, SAS-IFIT1 or SAS-IFIT3 cells. e Representative fluorescence images of the tongue and associated lymph node metastasis in neck tissues of orthotopic xenograft tongue tumors derived from SAS-Vector, SAS-IFIT1, and SAS-IFIT3 cells. f H&E-stained histopathology sections of the tongue and metastatic lymph nodes in neck tissues. The images of the areas within the black boxes were magnified to visualize the tumor cells in tongue and cervical lymph nodes (g) Percentage of cervical lymph node metastasis in orthotopic mice

Source publication
Article
Full-text available
IFIT1 and IFIT3 are abundant products of interferon-stimulating genes. While the importance of IFIT1 and IFIT3 in the prognosis of cancer has been reported, the molecular basis of IFIT1 and IFIT3 in cancer progression remains unexplored. In the present study, we investigated the modes of action and the clinical significance of IFIT1 and IFIT3 in or...

Similar publications

Article
Full-text available
Background Oral squamous cell carcinoma (OSCC) is a common oral cancer. The current study aims to elucidate the potential roles of long noncoding RNA (lncRNA) LHFPL3-AS1 in OSCC development. Methods Gene expression was measured by qRT-PCR in tumor tissues and cell lines. Loss-of-function assays were performed to analyze the effects of LHFPL3-AS1 o...

Citations

... IFIT3 can enhance the anti-liver cancer effect of IFN-α by binding transcriptional activators STAT1 and STAT2 [53]. However, IFIT3 can also promote tumor progression and metastasis in squamous cell carcinoma of the head and neck [54]. The specific role of IFIT3 in NPC needs to be investigated. ...
Article
Full-text available
Background Dermatomyositis (DM) is prone to nasopharyngeal carcinoma (NPC), but the mechanism is unclear. This study aimed to explore the potential pathogenesis of DM and NPC. Methods The datasets GSE46239, GSE142807, GSE12452, and GSE53819 were downloaded from the GEO dataset. The disease co-expression module was obtained by R-package WGCNA. We built PPI networks for the key modules. ClueGO was used to analyze functional enrichment for the key modules. DEG analysis was performed with the R-package "limma". R-package “pROC” was applied to assess the diagnostic performance of hub genes. MiRNA-mRNA networks were constructed using MiRTarBase and miRWalk databases. Results The key modules that positively correlated with NPC and DM were found. Its intersecting genes were enriched in the negative regulation of viral gene replication pathway. Similarly, overlapping down-regulated DEGs in DM and NPC were also enriched in negatively regulated viral gene replication. Finally, we identified 10 hub genes that primarily regulate viral biological processes and type I interferon responses. Four key genes (GBP1, IFIH1, IFIT3, BST2) showed strong diagnostic performance, with AUC>0.8. In both DM and NPC, the expression of key genes was correlated with macrophage infiltration level. Based on hub genes’ miRNA-mRNA network, hsa-miR-146a plays a vital role in DM-associated NPC. Conclusions Our research discovered pivot genes between DM and NPC. Viral gene replication and response to type I interferon may be the crucial bridge between DM and NPC. By regulating hub genes, MiR-146a will provide new strategies for diagnosis and treatment in DM complicated by NPC patients. For individuals with persistent viral replication in DM, screening for nasopharyngeal cancer is necessary.
... At last, we selected 6 DEPs (MX1, IFIT1, IFIH1, P4HB, ISG15 and SOD2) for qPCR assay, and the results were consistent with the iTRAQ data, which indicated the accuracy and reliability of iTRAQ. IFIH1 was reported to facilitate oral squamous cell carcinoma (OSCC) invasion [39] . Recent studies have implicated IFIH proteins as prognostic markers to determine the clinical outcome of many cancers, for they wildly regulated the cell functions of many cancers [40] . ...
Article
Full-text available
Background HIF1A-AS1 , an antisense transcript of HIF1α gene, is a 652-bp LncRNA that is globally expressed in multiple tissues of animals. Recent evidence indicated that HIF1A-AS1 was involved in tumorigenesis of several types of cancer. However, the role of lncRNA in PC has not been reported, and the molecular mechanism remains elusive. Results In order to investigate the role of HIF1A-AS1 in PC, it was overexpressed in some PC cell lines (PANC-1, PATU8988 and SW1990), and a series of experiments including cell viability detection, flow cytometry, transwell migration, clone formation and wound healing were performed. Functionally, the results indicated that overexpression of HIF1A-AS1 could greatly inhibit proliferation and migration and promote apoptosis of PC cells. Moreover, the isobaric tags for relative and absolute quantification (iTRAQ) quantitative proteomics analysis was implemented to explore the underlying mechanism and the results indicated that OE of HIF1A-AS1 globally affected the expression levels of multiple proteins associated with metabolism of cancer. At last, the network analysis revealed that most of these differentially expressed proteins (DEPs) were integrated and severed essential roles in regulatory function. In view of this, we guessed HIF1A-AS1 overexpression induced the dysfunction of metabolism and disordered proteins’ translation, which may account for its excellent tumour suppressor effect. Conclusions HIF1A-AS1 altered the cell function of PC cell lines via affecting the expression of numerous proteins. In summary, HIF1A-AS1 may exhibit a potential therapeutic effect on PC, and our study provided useful information in this filed.
... 4,[11][12][13][14][15][16] There are many factors that can contribute to the development of cancer and increase mortality rates in patients. 4,17,18 One of these influential factors is a gene mutation that can potentially lead to neoplastic changes in the oral cavity. 19 Multiple genetic and environmental factors trigger the inactivation of tumor suppressor genes and, or activation of oncogenes and, therefore, induce gene mutations. ...
... 21 The relatively low survival rate of OSCC-afflicted patients is mainly attributed to its high propensity for lymphatic metastasis. 17,[22][23][24] Metastasis is a stage of tumor development that includes a variety of processes, such as the development of cellular motility and invasion capabilities, the epithelial-to-mesenchymal transition (EMT), and angiogenesis. Therefore, several steps are required for cancer cells to spread from their original site to the metastatic one, as shown in the invasion-metastasis cascade. ...
... Over expression inhibits metastasis and low expression induces cell migration by activating atypical PKC signaling. 17,59,60 TGFBI Transformed growth factor-beta-induced Overexpression of TGFBI promotes OSCC, and it is a key hub gene in a protein-protein interaction network. 4,61 LGALS3BP ...
Article
Full-text available
Background and Aims Oral squamous cell carcinoma is the most prevalent malignancy in the oral cavity, with a significant mortality rate. In oral squamous cell carcinoma patients, the survival rate could decrease because of delayed diagnosis. Thus, prevention, early diagnosis, and appropriate treatment can effectively increase the survival rate in patients. In this systematic review, we discussed the role of different genes in oral squamous cell carcinoma metastasis. Herein, we aimed to summarize clinical results, regarding the potential genes that promote oral squamous cell carcinoma metastasis. Methods This systematic review was carried out under the Preferred Reporting Items for Systematic Reviews and Meta‐Analysis guidelines. An electronic search for all relevant articles published in English between January 2018 and April 2022 was performed using Scopus, PubMed, and Google Scholar search engines. All original studies published in English were included, and we excluded studies that were in a non‐English language. Results A total of 4682 articles were found, of which 14 were relevant and detected significant genes in oral squamous cell carcinoma progression. These findings investigated the overexpression of interferon‐induced proteins with tetratricopeptide repeats 1 and 3 (IFIT1, IFT3), high‐mobility group A2 (HMGA2), transformed growth factor‐beta‐induced, lectin galactoside‐binding soluble 3 binding protein (LGALS3BP), bromodomain containing 4, COP9 signaling complex 6, heterogeneous nuclear ribonucleoproteins A2B1 (HNRNPA2B1), 5′−3′ exoribonuclease 2 (XRN2), cystatin‐A (CSTA), fibroblast growth factors 8 (FGF8), forkhead box P3, cadherin‐3, also known as P‐cadherin and Wnt family member 5A, ubiquitin‐specific‐processing protease 7, and retinoic acid receptor responder protein 2 genes lead to promote metastasis in oral squamous cell carcinoma. Overexpression of some genes (IFIT1, 3, LGALS3BP, HMGA2, HNRNPA2B1, XRN2, CSTA, and FGF8) was proven to be correlated with poor survival rates in oral squamous cell carcinoma patients. Conclusion Studies suggest that metastatic genes indicate a poor prognosis for oral squamous cell carcinoma patients. Detecting these metastatic genes in oral squamous cell carcinoma patients may be of predictive value and can also facilitate assessing oral squamous cell carcinoma development and its response to treatment.
... For example, overexpression of IFIT1 or IFIT3 increased oral squamous cell carcinoma (OSCC) resistance to multiple chemotherapeutic drugs, including 5FU, cisplatin, oxaliplatin, carboplatin [24,44]. On the contrary, the high expression of IFIT1 and IFIT3 in OSCC contributes to gefitinib's anti-tumor effect by enhancing p-EGFR recycling [45]. In our analysis, drug sensitivity test results showed that high expression of IFITs in AML was resistant to various drugs or small molecule, but sensitive to dasatinib. ...
Article
Full-text available
Background The Interferon-induced protein with tetratricopeptide repeat (IFIT) family, IFIT1/2/3/5, play an important role in different tumors progression. However, the prognosis significance and biological role of IFIT family members in acute myeloid leukemia (AML) remains unclear. Methods We obtained the gene expression data and clinical information of 173 AML patients from The Cancer Genome Atlas (TCGA) database. Several databases were used in our study, including GEPIA, MethSurv, STRING, GSCA and GeneMANIA database. Results The mRNA expression of IFIT1/2/3/5 was elevated in AML patients and had a high ability to distinguish AML from controls based on the receiver operating characteristic (ROC) curve (AUC > 0.9). Kaplan–Meier survival analysis showed that higher levels of IFIT2/3/5 expression predict poor prognosis in AML patients. Besides, the DNA methylation analysis suggested that 7 CpG sites of IFIT2, 4 CpG sites of IFIT3 and 10 CpG sites of IFIT5 were significantly associated with the prognosis of AML patients. In addition, IFIT2/3/5 expression was significantly positively associated with the immune cell infiltration and immune checkpoint expression, such as CTLA4, PDCD1, LAG3, and TIGIT. Finally, drug sensitivity analysis revealed that AML patients with high expression of IFIT2/3/5 were resistant to multiple drugs, but sensitive to dasatinib. Conclusion IFIT family genes might serve as biomarkers for diagnosis, prognosis and drug sensitivity in AML patients. The activation or blocking of IFIT-related signaling pathways may provide novel insights into immunotherapy for patients with AML.
... [34] Mann-Whitney U test and Kaplan-Meier survival analysis showed that the expression of IFIT3 was correlated with T stage, lymph node metastasis, distant metastasis, OS and DFS in patients with ESCC, while Pidugu VK et al. also found in a clinical study that the increase of IFIT3 expression was signi cantly positively correlated with advanced T stage, lymph node metastasis, peripheral nerve invasion, lymphatic invasion and poor overall survival rate in patients with OSCC. [35] Zhao Y et al also found that the high expression of IFIT3 is independently related to the shortened survival period of pancreatic cancer patients, which can be used as a prognostic indicator. [36] Other studies have proved that IFIT3 silence can reduce the expression of IL-17 and IL-1 β, and decreases the migration ability of liver cancer cells. ...
Preprint
Full-text available
Objective This study aims to clarify the expression of IFIT3 in human esophageal squamous cell carcinoma(ESCC) and further explore the relationship between IFIT3 expression and clinical pathological characteristics and prognosis of ESCC patients. Methods The target gene IFIT3 was screened through differential expression gene analysis, cluster analysis, enrichment analysis, and construction of a protein protein interaction network (PPI network), and then validated through clinical patient tissue RNA extraction and reverse transcription quantitative PCR. The Mann Whitney U test and Kaplan Meier analysis were used to investigate the correlation between the relative expression of IFIT3 and the clinical pathological information and prognosis of ESCC patients. Results GEO2R detected 279 differentially expressed genes in ESCC and paracancerous tissues. Cluster analysis and enrichment analysis showed that Cluster 4 played an important role in immune-related functions. PPI network showed that IFIT3 was the hub gene in the Cluster 4. Clinical patient tissue samples confirm the differential expression of IFIT3 in ESCC and paracancerous tissues. Mann-Whitney U test showed that the relative expression of IFIT3 was significantly correlated with clinicopathological information in patients with ESCC. Kaplan-Meier survival analysis showed that the disease-free survival time and overall survival time of patients with low expression of IFIT3 were significantly longer than those of patients with high expression of IFIT3, and the correlations were more significant in some subgroups. Cox proportional hazards model showed that lymph node metastasis was an independent risk factor for the prognosis of ESCC patients. Conclusion IFIT3 is differentially expressed in the cancerous and paracancerous tissues of ESCC, and the relative expression level of IFIT3 is highly correlated with the clinical pathological characteristics and prognosis of ESCC patients.
... The results demonstrated that there was a negative correlation between KLF13 and IFIT1 in THCA samples (Additional file 1: Figure S2 and Additional file 2: Table S1). Previous studies have reported that IFIT1 acts as an oncogene in various tumors [20][21][22], whereas its significance, as well as its involvement in KLF13 suppression of tumor progression, in THCA remains to be determined. Thus, we further studied the regulation of KLF13 on the transcription activity of IFIT1. ...
... Another study showed the overexpression of IFIT1 in head and neck squamous cell carcinoma, and there was correlation with bad prognosis and tumorassociated macrophage markers [33]. IFIT1 expression has been shown to be positively related to the expression of p-EGFR Y1068 in Oral Squamous Cell Carcinoma [22]. Therefore, we intended to explore whether KLF13 suppressed THCA progression via regulating the expression of IFIT1. ...
Article
Full-text available
Background Kruppel-like factor 13 (KLF13) is a transcription factor and plays an important role in carcinogenesis. However, the significance of KLF13 in thyroid carcinoma (THCA) is underdetermined. In this study, we aimed to explore the clinical relevance and function of KLF13 in the progress of THCA. Methods The expression of KLF13 in thyroid carcinoma and normal tissue was investigated by qPCR and IHC assay. The expression of KLF13 and IFIT1 in cell samples was investigated with Western blot assay. Cell proliferation ability was detected with CCK8 and colony formation assay. Cell growth in vivo with or without KLF13 overexpression was evaluated on a xenograft model. Cell migration ability was measured with Transwell assay. Cell cycle was detected with flow cytometer. The downstream genes of KLF13 were screened using RNA-seq assay. Luciferase activity was employed to assess the transcriptional regulation of KLF13 on IFIT1 promoter. Results KLF13 expression was downregulated in THCA samples. KLF13 knockdown and overexpression promoted and inhibited the proliferation and migration of THCA cells, respectively. The RNA-seq, RT-qPCR and immunoblotting data showed that KLF13 knockdown significantly potentiated IFIT1 expression at both mRNA and protein levels. Luciferase assays showed that KLF13 suppressed the transcription activity of IFIT1 promoter. Besides, IFIT1 upregulation was critical for the proliferation and migration of THCA cell lines. Lastly, silencing of IFIT1 greatly reversed the proliferation and migration induced by KLF13 knockdown. Conclusions In conclusion, KLF13 may function as an anti-tumor protein in THCA by regulating the expression of IFIT1 and offer a theoretical foundation for treating thyroid carcinoma.
... IFIT1 and IFIT3 has been shown to promote EGFR activation in OSCC cells and enhance the tumor-suppressive effect of gefitinib, an EGFR inhibitor. 35 We therefore investigated whether LOXL2 promotes EGFR activation in HNSCC cells. Figure 7A, ...
Article
Full-text available
Lysyl oxidase-like 2 (LOXL2) is a matrix-remodeling enzyme that has recently been identified as an important regulator of tumor progression and metastasis. This study discovered that LOXL2 expression in oral squamous cell carcinoma (OSCC) tissues was significantly associated with tumor clinical stage, lymph node metastasis and patients' overall survival time. LOXL2-overexpressing human buccal SCC TW2.6 (TW2.6/LOXL2) and hypopharyngeal SCC FaDu (FaDu/LOXL2) cells exhibited enhanced migration, invasion, epithelial-mesenchymal transition (EMT), and cancer stem cell (CSC) phenotypes, independently of its enzymatic activity. Moreover, TW2.6/LOXL2 significantly increased tumor-initiating frequency in SCID mice. We further demonstrated that LOXL2 increased the levels of interferon-induced protein with tetratricopeptide repeats 1 (IFIT1) and IFIT3 in TW2.6/LOXL2 and FaDu/LOXL2 cells. We also identified IFIT1 and IFIT3 as key downstream components of LOXL2 action in migration, invasion, EMT, and CSC phenotypes in TW2.6 and FaDu cells. Furthermore, a significant positive correlation between LOXL2 expression and IFIT1 and IFIT3 overexpression in human OSCC tissues was observed. In addition, TW2.6/LOXL2 and FaDu/LOXL2 cells were 3.3- to 3.6-fold more susceptible to the epidermal growth factor receptor (EGFR) inhibitor gefitinib than were their respective control cells. The antitumor effect of gefitinib on orthotopic TW2.6/LOXL2 xenograft tumor was fourfold higher than that on controls. Our results indicate that LOXL2 expression is a strong prognostic factor for OSCC and may be used as a marker to identify patients most likely to respond to EGFR-targeted therapy.
... Intriguingly, although IFITs share conserved structural motifs, their isoforms display differential expression and nonredundant functions [23,24]. Elevated levels of IFIT1, IFIT3 and IFIT5 have been shown to play significant roles in cancer progression, while the decreased expression of IFIT2 has been reported to enhance invasion, tumor progression, and drug resistance in various cancer types [25][26][27][28][29][30][31][32]. Furthermore, increased IFIT1 and IFIT3 expression but decreased IFIT2 expression is correlated with poor survival in OSCC patients [25,33]. ...
... Elevated levels of IFIT1, IFIT3 and IFIT5 have been shown to play significant roles in cancer progression, while the decreased expression of IFIT2 has been reported to enhance invasion, tumor progression, and drug resistance in various cancer types [25][26][27][28][29][30][31][32]. Furthermore, increased IFIT1 and IFIT3 expression but decreased IFIT2 expression is correlated with poor survival in OSCC patients [25,33]. On the contrary, increased IFIT2 expression inhibits cell proliferation and triggers apoptosis in various cancer types [34][35][36][37]. ...
Article
Full-text available
(1) Background: Cancer stem cells (CSCs) are a small cell population associated with chemoresistance, metastasis and increased mortality rate in oral cancer. Interferon-induced proteins with tetratricopeptide repeats 2 (IFIT2) depletion results in epithelial to mesenchymal transition, invasion, metastasis, and chemoresistance in oral cancer. To date, no study has demonstrated the effect of IFIT2 depletion on the CSC-like phenotype in oral cancer cells. (2) Methods: Q-PCR, sphere formation, Hoechst 33,342 dye exclusion, immunofluorescence staining, and flow cytometry assays were performed to evaluate the expression of the CSC markers in IFIT2-depleted cells. A tumorigenicity assay was adopted to assess the tumor formation ability. Immunohistochemical staining was used to examine the protein levels of IFIT2 and CD24 in oral cancer patients. (3) Results: The cultured IFIT2 knockdown cells exhibited an overexpression of ABCG2 and CD44 and a downregulation of CD24 and gave rise to CSC-like phenotypes. Clinically, there was a positive correlation between IFIT2 and CD24 in the patients. IFIT2high/CD24high/CD44low expression profiles predicted a better prognosis in HNC, including oral cancer. The TNF-α blockade abolished the IFIT2 depletion-induced sphere formation, indicating that TNF-α may be involved in the CSC-like phenotypes in oral cancer. (4) Conclusions: The present study demonstrates that IFIT2 depletion promotes CSC-like phenotypes in oral cancer.
... Highlighting the diversity between IFIT proteins, IFIT1 and IFIT3 are distinguished from IFIT2 for their pro-metastatic action in the setting of OSCC [48,56]. Recent studies examining IFIT1 and IFIT3 expression in OSCC cell lines revealed that it was associated with EMT and increased cell invasion [48,56]. ...
... Highlighting the diversity between IFIT proteins, IFIT1 and IFIT3 are distinguished from IFIT2 for their pro-metastatic action in the setting of OSCC [48,56]. Recent studies examining IFIT1 and IFIT3 expression in OSCC cell lines revealed that it was associated with EMT and increased cell invasion [48,56]. Overexpression of IFIT1/3 results in unregulated recycling of phosphorylated epidermal growth factor (p-EGFR), which promotes increased expression of EMT-related markers, such as N-cadherin [56]. ...
... Recent studies examining IFIT1 and IFIT3 expression in OSCC cell lines revealed that it was associated with EMT and increased cell invasion [48,56]. Overexpression of IFIT1/3 results in unregulated recycling of phosphorylated epidermal growth factor (p-EGFR), which promotes increased expression of EMT-related markers, such as N-cadherin [56]. While the overexpression of IFIT1 and IFIT3 seems to contribute to metastatic disease, its effect on p-EGFR results in increased susceptibility to chemotherapeutic medications that inhibit tyrosine kinases, such as gefitinib [56]. ...
Article
Full-text available
The type-I interferon (IFN) system represents the first line of defense against viral pathogens. Recognition of the virus initiates complex signaling pathways that result in the transcriptional induction of IFNs, which are then secreted. Secreted IFNs stimulate nearby cells and result in the production of numerous proinflammatory cytokines and antiviral factors. Of particular note, IFN-induced tetratricopeptide repeat (IFIT) proteins have been thoroughly studied because of their antiviral activity against different viral pathogens. Although classically studied as an antiviral protein, IFIT expression has recently been investigated in the context of nonviral pathologies, such as cancer and sepsis. In oral squamous cell carcinoma (OSCC), IFIT1 and IFIT3 promote metastasis, while IFIT2 exhibits the opposite effect. The role of IFIT proteins during bacterial/fungal sepsis is still under investigation, with studies showing conflicting roles for IFIT2 in disease severity. In the setting of viral sepsis, IFIT proteins play a key role in clearing viral infection. As a result, many viral pathogens, such as SARS-CoV-2, employ mechanisms to inhibit the type-I IFN system and promote viral replication. In cancers that are characterized by upregulated IFIT proteins, medications that decrease IFIT expression may reduce metastasis and improve survival rates. Likewise, in cases of viral sepsis, therapeutics that increase IFIT expression may improve viral clearance and reduce the risk of septic shock. By understanding the effect of IFIT proteins in different pathologies, novel therapeutics can be developed to halt disease progression.
... However, more recent studies provide evidence that IFN can also mediate pro-tumorigenic effects [71]. Functional studies with IFIT proteins (IFIT1 and IFIT3) on various molecular signaling mechanisms implicates them in cancer progression and metastasis [92]. Interestingly, the pro-cancer growth effects of IFIT proteins involve Akt activation and were downregulated in miR193a-3p transfected LEC + MCF-7 spheroids. ...
... Interestingly, the pro-cancer growth effects of IFIT proteins involve Akt activation and were downregulated in miR193a-3p transfected LEC + MCF-7 spheroids. We observed similar effects on VECs exposed to secretome from MCF-7 cells transfected with miR193a-3p [29], suggesting that miR193a-3p inhibits spheroid growth by preventing MCF-7-LEC cross-talk by potentially downregulating IFIT proteins and inhibiting Akt [92]. Interestingly, in SUM149 inflammatory BC, the overexpression of IFITM1 enhances aggressive phenotype via signal transducer and activator of transcription 2 (STAT2) [93]. ...
Article
Full-text available
MicroRNA 193a-3p (miR193a-3p) is a short non-coding RNA with tumor suppressor properties. Breast cancer (BC) progression is governed by active interaction between breast cancer cells, vascular (V)/lymphatic (L) endothelial cells (ECs), and BC secretome. We have recently shown that miR193a-3p, a tumor suppressor miRNA, inhibits MCF-7 BC cell-driven growth of VECs via direct antimitogenic actions and alters MCF-7 secretome. Since LEC-BC cross-talk plays a key role in BC progression, we investigated the effects of miR193a-3p on MCF-7 secretome and estradiol-mediated growth effects in LECs and LEC + MCF-7 spheroids, and delineated the underlying mechanisms. Transfection of LECs with miR193a-3p, as well as secretome from MCF-7 transfected cells, inhibited LEC growth, and these effects were mimicked in LEC + MCF-7 spheroids. Moreover, miR193a-3p inhibited ERK1/2 and Akt phosphorylation in LECs and LEC + MCF-7 spheroids, which are importantly involved in promoting cancer development and metastasis. Treatment of LECs and LEC + MCF-7 spheroids with estradiol (E2)-induced growth, as well as ERK1/2 and Akt phosphorylation, and was abrogated by miR193a-3p and secretome from MCF-7 transfected cells. Gene expression analysis (GEA) in LEC + MCF-7 spheroids transfected with miR193a-3p showed significant upregulation of 54 genes and downregulation of 73 genes. Pathway enrichment analysis of regulated genes showed significant modulation of several pathways, including interferon, interleukin/cytokine-mediated signaling, innate immune system, ERK1/2 cascade, apoptosis, and estrogen receptor signaling. Transcriptomic analysis showed downregulation in interferon and anti-apoptotic and pro-growth molecules, such as IFI6, IFIT1, OSA1/2, IFITM1, HLA-A/B, PSMB8/9, and PARP9, which are known to regulate BC progression. The cytokine proteome array of miR193a-3p transfected MCF secretome and confirmed the upregulation of several growth inhibitory cytokines, including IFNγ, Il-1a, IL-1ra, IL-32, IL-33, IL-24, IL-27, cystatin, C-reactive protein, Fas ligand, MIG, and sTIM3. Moreover, miR193a-3p alters factors in MCF-7 secretome, which represses ERK1/2 and Akt phosphorylation, induces pro-apoptotic protein and apoptosis in LECs, and downregulates interferon-associated proteins known to promote cancer growth and metastasis. In conclusion, miR193a-3p can potentially modify the tumor microenvironment by altering pro-growth BC secretome and inhibiting LEC growth, and may represent a therapeutic molecule to target breast tumors/cancer.