1
|
Wang C, Liang H, Li Y, Tang Z, Zhang Y. Chemokine (C-C motif) ligand 18/membrane-associated 3/forkhead box O1 axis promotes the proliferation, migration, and invasion of intrahepatic cholangiocarcinoma. Bioengineered 2022; 13:12738-12748. [PMID: 35609322 PMCID: PMC9276021 DOI: 10.1080/21655979.2022.2069383] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Phosphatidylinositol Transfer Protein, Membrane-Associated 3 (PITPNM3) often bind with chemokine (C-C motif) ligand 18 (CCL18) to promote tumor progression. However, the role of PITPNM3 in intrahepatic cholangiocarcinoma (ICC) is unclear. We first searched GEPIA database and detected the PITPNM3 expression using immunohistochemistry and real-time quantitative PCR. The results showed that PITPNM3 is high expression in ICC tissues and cells. Then we investigated the cell function of CLL18 and PITPNM3 through cell clone formation assay and transwell assay. The results indicated that CCL18 treatment promoted the proliferation, migration, and invasion of ICC cells. Silence of PITPNM3 reversed the effect of CCL18 on cell function. Simultaneously, we detected key protein expression of forkhead box O1 (FOXO1) and nuclear factor kappa B (NF-KB) through western blotting and found that CCL18 activated NF-KB pathway while inhibited FOXO1 pathway, the effect of which were attenuated by silence of PITPNM3. Finally, we confirmed which pathway affected the cell function using inhibitor of FOXO1 (AS1842856) and activator of NF-KB (Asatone). The results showed that AS1842856, not Asatone, relieved the inhibitory effect of si-PITPNM3 on the cell function of CCL18. In short, CCL18 treatment activated PITPNM3 to promote the proliferation, migration, and invasion of ICC via FOXO1 signaling pathway. These results provided a new insight for the diagnosis and therapy of ICC.
Collapse
Affiliation(s)
- Chusi Wang
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hao Liang
- Department of General Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanjie Li
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhaofeng Tang
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yingcai Zhang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Zhang C, Hu J, Li H, Ma H, Othmane B, Ren W, Yi Z, Qiu D, Ou Z, Chen J, Zu X. Emerging Biomarkers for Predicting Bladder Cancer Lymph Node Metastasis. Front Oncol 2021; 11:648968. [PMID: 33869048 PMCID: PMC8044933 DOI: 10.3389/fonc.2021.648968] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/05/2021] [Indexed: 12/24/2022] Open
Abstract
Bladder cancer is one of the leading causes of cancer deaths worldwide. Early detection of lymph node metastasis of bladder cancer is essential to improve patients' prognosis and overall survival. Current diagnostic methods are limited, so there is an urgent need for new specific biomarkers. Non-coding RNA and m6A have recently been reported to be abnormally expressed in bladder cancer related to lymph node metastasis. In this review, we tried to summarize the latest knowledge about biomarkers, which predict lymph node metastasis in bladder cancer and their mechanisms. In particular, we paid attention to the impact of non-coding RNA on lymphatic metastasis of bladder cancer and its specific molecular mechanisms, as well as some prediction models based on imaging, pathology, and biomolecules, in an effort to find more accurate diagnostic methods for future clinical application.
Collapse
Affiliation(s)
- Chunyu Zhang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Jiao Hu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Huihuang Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Hongzhi Ma
- Department of Radiation Oncology, Hunan Cancer Hospital, Central South University, Changsha, China
| | - Belaydi Othmane
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Wenbiao Ren
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.,George Whipple Lab for Cancer Research, University of Rochester Medical Institute, Rochester, NY, United States
| | - Zhenglin Yi
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Dongxu Qiu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenyu Ou
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Jinbo Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
3
|
Li F, Xie W, Fang Y, Xie K, Liu W, Hou L, Tan W. HnRNP-F promotes the proliferation of bladder cancer cells mediated by PI3K/AKT/FOXO1. J Cancer 2021; 12:281-291. [PMID: 33391425 PMCID: PMC7738822 DOI: 10.7150/jca.50490] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/21/2020] [Indexed: 12/24/2022] Open
Abstract
Our previous study showed that heterogeneous nuclear ribonucleoprotein F (hnRNP-F) could induce epithelial-mesenchymal transition and metastasis in bladder cancer (BC), however, the role and mechanism of hnRNP-F in mediating the proliferative ability of BC cells remain unclear. HnRNP-F promoted the proliferation of BC cells by using BC cell lines and cell counting kit-8 (CCK8), colony formation and flow cytometry assays in vitro. Furthermore, the association of hnRNP-F with the phosphoinositide 3‑kinase (PI3K)/protein kinase B (AKT) signalling pathway was confirmed by western blotting after bioinformatic analysis. HnRNP-F expression was significantly decreased by treatment with the PI3K/AKT signalling pathway inhibitor LY294002, whereas hnRNP-F knockdown did not significantly affect PI3K or AKT expression, suggesting that hnRNP-F is likely a downstream target of the PI3K/AKT pathway. Forkhead box O1 (FOXO1) is a molecule downstream of PI3K/AKT and can be inhibited by phosphorylation. In addition, chromatin immunoprecipitation (ChIP) and luciferase reporter assays indicated that FOXO1 expression was negatively correlated with hnRNP-F expression as FOXO1 was found to bind to the promoter region of hnRNP-F mRNA and inhibit its transcription. To sum up, our findings suggest that hnRNP-F expression is regulated by the PI3K/AKT-mediated phosphorylation of FOXO1, with phosphorylation inhibiting FOXO1, which subsequently allows hnRNP-F to promote proliferation. This finding is a novel discovery in BC and could help reveal the mechanism of BC progression.
Collapse
Affiliation(s)
- Fei Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R.China
| | - Weiwei Xie
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R.China
| | - Yunze Fang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R.China
| | - Kunfeng Xie
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R.China
| | - Wendong Liu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R.China
| | - Lina Hou
- Department of Healthy Management, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Wanlong Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R.China
| |
Collapse
|
4
|
Wang Y, Lyu Z, Qin Y, Wang X, Sun L, Zhang Y, Gong L, Wu S, Han S, Tang Y, Jia Y, Kwong DLW, Kam N, Guan XY. FOXO1 promotes tumor progression by increased M2 macrophage infiltration in esophageal squamous cell carcinoma. Am J Cancer Res 2020; 10:11535-11548. [PMID: 33052231 PMCID: PMC7546008 DOI: 10.7150/thno.45261] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 08/28/2020] [Indexed: 12/11/2022] Open
Abstract
Objective: The transcription factor forkhead box protein O1 (FOXO1) is critical for regulating cytokine and chemokine secretion. However, its function in the tumor microenvironment (TME) remains largely unexplored. In this study, we characterized the prognostic value of FOXO1 and the interaction between tumor-derived FOXO1 and M2 macrophages in esophageal squamous cell carcinoma (ESCC). Methods: FOXO1 expression and macrophage infiltration in clinical samples and mouse models were quantified using quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry staining. Western blotting, qRT-PCR, and enzyme-linked immunosorbent assay were used to evaluate chemokine ligand 20 (CCL20) and colony stimulating factor 1 (CSF-1) expression in FOXO1(+) and FOXO1(-) tumor cells. Macrophage phenotypes were determined using qRT-PCR, flow cytometry, and RNA sequencing. Transcriptional activity was measured using chromatin immunoprecipitation (ChIP)-qPCR. Tumor viability was investigated using XTT proliferation and foci formation assays. Results: FOXO1 upregulation in tumor tissues was found to drive the polarization of M0 macrophages and infiltration of M2 macrophages into the TME, resulting in worse prognosis in ESCC patients. CSF-1, a vital factor inducing M0-to-M2 polarization, was upregulated via a FOXO1-mediated mechanism. RNA sequencing results corroborated that the FOXO1-induced macrophages exhibited similar molecular signatures to the IL4-stimulated M2 macrophages. The transwell assays showed that FOXO1 promoted the migration of M2 macrophages via CCL20 secretion, which could be inhibited using an anti-CCL20 antibody. FOXO1(+) tumor-induced M2 macrophages promoted tumor proliferation via the FAK-PI3K-AKT pathway and the PI3K inhibitor could effectively impede the oncogenical process. Conclusions: FOXO1 facilitated M0-to-M2 polarization and the recruitment of M2 macrophages in the TME via the transcriptional modulation of CCL20 and CSF-1. Our data deciphered the FOXO1-dependent mechanism in M2 macrophage infiltration in the TME of ESCC, which has implications for the development of novel prognostic and therapeutic targets to optimize the current treatment against ESCC.
Collapse
|
5
|
Ide H, Goto T, Teramoto Y, Mizushima T, Jiang G, Nagata Y, Inoue S, Baras AS, Kashiwagi E, Miyamoto H. FOXO1 inactivation induces cisplatin resistance in bladder cancer. Cancer Sci 2020; 111:3397-3400. [PMID: 32678492 PMCID: PMC7469822 DOI: 10.1111/cas.14557] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/19/2020] [Accepted: 06/30/2020] [Indexed: 01/09/2023] Open
Abstract
We found that FOXO1-shRNA sublines or FOXO1-positive cells co-treated with a FOXO1 inhibitor were significantly more resistant to cisplatin treatment at pharmacological concentrations, compared with respective control sublines or those with mock treatment. Western blot demonstrated considerable increases in the expression levels of a phosphorylated inactive form of FOXO1 (p-FOXO1) in cisplatin-resistant sublines established by long-term culture with low/increasing doses of cisplatin, compared with respective controls. Immunohistochemistry in surgical specimens from patients with muscle-invasive bladder cancer undergoing cisplatin-based neoadjuvant therapy further showed a strong trend to associate between p-FOXO1 positivity and unfavorable response to chemotherapy.
Collapse
Affiliation(s)
- Hiroki Ide
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- James Buchanan Brady Urological InstituteJohns Hopkins University School of MedicineBaltimoreMDUSA
- Department of UrologyKeio University School of MedicineTokyoJapan
| | - Takuro Goto
- Department of Pathology and Laboratory MedicineUniversity of Rochester Medical CenterRochesterNYUSA
- James P. Wilmot Cancer InstituteUniversity of Rochester Medical CenterRochesterNYUSA
| | - Yuki Teramoto
- Department of Pathology and Laboratory MedicineUniversity of Rochester Medical CenterRochesterNYUSA
- James P. Wilmot Cancer InstituteUniversity of Rochester Medical CenterRochesterNYUSA
| | - Taichi Mizushima
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- James Buchanan Brady Urological InstituteJohns Hopkins University School of MedicineBaltimoreMDUSA
- Department of Pathology and Laboratory MedicineUniversity of Rochester Medical CenterRochesterNYUSA
- James P. Wilmot Cancer InstituteUniversity of Rochester Medical CenterRochesterNYUSA
| | - Guiyang Jiang
- Department of Pathology and Laboratory MedicineUniversity of Rochester Medical CenterRochesterNYUSA
- James P. Wilmot Cancer InstituteUniversity of Rochester Medical CenterRochesterNYUSA
| | - Yujiro Nagata
- Department of Pathology and Laboratory MedicineUniversity of Rochester Medical CenterRochesterNYUSA
- James P. Wilmot Cancer InstituteUniversity of Rochester Medical CenterRochesterNYUSA
| | - Satoshi Inoue
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- James Buchanan Brady Urological InstituteJohns Hopkins University School of MedicineBaltimoreMDUSA
- Department of Pathology and Laboratory MedicineUniversity of Rochester Medical CenterRochesterNYUSA
- James P. Wilmot Cancer InstituteUniversity of Rochester Medical CenterRochesterNYUSA
| | - Alexander S. Baras
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- James Buchanan Brady Urological InstituteJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Eiji Kashiwagi
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- James Buchanan Brady Urological InstituteJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Hiroshi Miyamoto
- Department of PathologyJohns Hopkins University School of MedicineBaltimoreMDUSA
- James Buchanan Brady Urological InstituteJohns Hopkins University School of MedicineBaltimoreMDUSA
- Department of Pathology and Laboratory MedicineUniversity of Rochester Medical CenterRochesterNYUSA
- James P. Wilmot Cancer InstituteUniversity of Rochester Medical CenterRochesterNYUSA
- Department of UrologyUniversity of Rochester Medical CenterRochesterNYUSA
| |
Collapse
|
6
|
Fan Z, Li L, Wang X, Miao G. Dysfunction of regulatory T cells mediated by AKT-FOXO1 signaling pathway occurs during the development of psoriasis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:799-809. [PMID: 32355530 PMCID: PMC7191153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/14/2019] [Indexed: 06/11/2023]
Abstract
Psoriasis is an immune-mediated skin disease with abnormal T cells. Regulatory T cells (Treg) are a kind of cell group with immunosuppressive effects. This study aimed to explore the role of Treg cells in the pathogenesis of psoriasis and its possible mechanism. Imiquimod induced psoriasis mice model was conducted. The skin lesions were evaluated according to the psoriasis area and severity index (PASI). Skin biopsies were taken for HE staining and immunohistochemical staining of IL-23, IL-17, IL-33 and TNF-α. CD4+CD25+ Treg cells were isolated. The proportions of Treg cells, cell proliferation, and immunosuppressive activity were analyzed by flow cytometry. The expression of AKT, Foxo1, pAKT, pFoxo1 protein, and the localization of Foxo1 protein in Treg cells were detected by western blot and immunofluorescence. The results showed that the psoriasis mice model was established successfully. There was no significant difference in the proportion of Treg cells between the two groups (P > 0.05). The cell proliferation abilities were decreased, and the immunosuppressive functions of Treg cells were weakened in the psoriatic group (P < 0.05). Western blot showed that pAKT and pFoxo1 levels of Treg cells were significantly increased in the psoriatic group (P < 0.05). Immunofluorescence showed that Foxo1 was mainly expressed in the nucleus of Treg cells in the control group, whereas expressed in the cytoplasm in the psoriasis group. Therefore, we concluded that the cell proliferation and immunosuppressive dysfunction of Treg cells mediated by AKT-FOXO1 signaling pathway may occurs during the development of psoriasis.
Collapse
Affiliation(s)
- Zhixia Fan
- Department of Dermatology, Affiliated Hospital of Hebei University of Engineering Handan, Heibei, China
| | - Lingyu Li
- Department of Dermatology, Affiliated Hospital of Hebei University of Engineering Handan, Heibei, China
| | - Xin Wang
- Department of Dermatology, Affiliated Hospital of Hebei University of Engineering Handan, Heibei, China
| | - Guoying Miao
- Department of Dermatology, Affiliated Hospital of Hebei University of Engineering Handan, Heibei, China
| |
Collapse
|
7
|
Abstract
OBJECTIVES Pancreatic cancer (PC) is highly aggressive with multiple oncogenic mutations. The efficacy of current chemotherapy is poor, and new therapeutic targets are needed. The forkhead box (FOX) proteins are multidirectional transcriptional factors strongly implicated in malignancies. Their expression is consistently suppressed by several oncogenic pathways such as PI3K/AKT signaling activated in PC. A recent study showed that class IIa histone deacetylases (HDAC) can act as a transcriptional suppressor. In this study, we hypothesized that HDAC class IIa inhibition would upregulate FOXO3a expression, thereby inducing its transcription-dependent antitumor effects. METHODS We confirmed the change of FOXO3a expression and the effect of the cell growth inhibition by HDAC class IIa inhibition in AsPC-1 cells. Because FOXO3a is subject to ubiquitylation-mediated proteasome degradation, we examined the synergistic activation of FOXO3a by HDAC class IIa selective inhibitor TMP269 combined with proteasome inhibitor carfilzomib. RESULTS We observed that TMP269 induced FOXO3a expression in a dose-dependent manner and inhibited cell growth in AsPC-1 cells. G1/S arrest was observed. FOXO3a expression was further increased and cell growth inhibition was dramatically enhanced by TMP269 combined with carfilzomib. CONCLUSIONS Dual inhibition of class IIa HDACs and proteasome could be a promising new strategy for modifying FOXO3a activity against PC.
Collapse
|
8
|
Han GH, Chay DB, Nam S, Cho H, Chung JY, Kim JH. Prognostic implications of forkhead box protein O1 (FOXO1) and paired box 3 (PAX3) in epithelial ovarian cancer. BMC Cancer 2019; 19:1202. [PMID: 31823759 PMCID: PMC6905044 DOI: 10.1186/s12885-019-6406-6] [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] [Received: 07/15/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Transcription factors forkhead box protein O1 (FOXO1) and paired box 3 (PAX3) have been reported to play important roles in various cancers. However, their role in epithelial ovarian cancer (EOC) has not been elucidated yet. Therefore, we evaluated the expression and clinical significance of FOXO1 and PAX3 in EOC. METHODS Immunohistochemical analyses of FOXO1 and PAX3 in 212 EOCs, 57 borderline ovarian tumors, 153 benign epithelial ovarian tumors, and 79 nonadjacent normal epithelial tissues were performed using tissue microarray. Various clinicopathological variables, including the survival of EOC patients, were compared. In addition, the effect of FOXO1 on cell growth was assessed in EOC cell lines. RESULTS FOXO1 and PAX3 protein expression levels were significantly higher in EOC tissues than in nonadjacent normal epithelial tissues, benign tissues, and borderline tumors (all p < 0.001). In EOC tissues, FOXO1 expression was positively correlated with PAX3 expression (Spearman's rho = 0.118, p = 0.149). Multivariate survival analysis revealed that high FOXO1 expression (hazard ratio = 2.77 [95% CI, 1.48-5.18], p = 0.001) could be an independent prognostic factor for overall survival. Most importantly, high expression of both FOXO1 and PAX3 showed a high hazard ratio (4.60 [95% CI, 2.00-10.55], p < 0.001) for overall survival. Also in vitro results demonstrated that knockdown of FOXO1 was associated with decreased cell viability, migration, and colony formation. CONCLUSIONS This study revealed that high expression of FOXO1/PAX3 is an indicator of poor prognosis in EOC. Our results suggest the promising potential of FOXO1 and PAX3 as prognostic and therapeutic markers. The possible link between biological functions of FOXO1 and PAX3 in EOC warrants further studies.
Collapse
Affiliation(s)
- Gwan Hee Han
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Doo Byung Chay
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sanghee Nam
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-Ro, Gangnam-Gu, Seoul, Seoul, 06273, South Korea
| | - Hanbyoul Cho
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, Republic of Korea. .,Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Joon-Yong Chung
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
9
|
Forkhead box protein O1 (FOXO1) and paired box gene 3 (PAX3) overexpression is associated with poor prognosis in patients with cervical cancer. Int J Clin Oncol 2019; 24:1429-1439. [PMID: 31302815 DOI: 10.1007/s10147-019-01507-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/16/2019] [Indexed: 01/04/2023]
Abstract
PURPOSE Forkhead box protein O1 (FOXO1) and paired box gene 3 (PAX3) have been reported to play an imported role in human cancers, but their role in cervical cancer has not yet been clarified. In this study, we evaluated the functional role of FOXO1 in cervical cancer cells and investigated the expression and clinical significance of FOXO1 and PAX3 in cervical lesions. METHODS In vitro assessment of cell function by cell viability, migration, and invasion assays were performed on FOXO1-knockdown cervical cancer cells. Immunohistochemical (IHC) staining analyses of FOXO1 and PAX3 were performed with a tissue microarray (TMA). The clinical significance was evaluated by comparing the data with various clinicopathologic characteristics, including survival of patients with cervical cancer. RESULTS In vitro results revealed that knockdown of FOXO1 is associated with decreased cell viability (p < 0.001), migration (p < 0.001), and invasion (p < 0.05), supporting the oncogenic role of FOXO1 in cervical cancer. FOXO1 and PAX3 expression was significantly higher in CIN (both p < 0.001) and cancer tissue (both p < 0.001) than in normal tissue. Multivariate analysis indicated that FOXO1 expression (hazard ratio 4.01 [95% CI 1.22-13.10], p = 0.021) and an advanced FIGO stage (hazard ratio 3.89 [95% CI 1.35-11.19], p = 0.012) were independent prognostic factors for overall survival. CONCLUSIONS This study reveals increased FOXO1 and PAX3 expression in cervical cancers and indicates an oncogenic role of FOXO1 in cervical cancer cells that correlates with poor patient survival.
Collapse
|
10
|
Ide H, Jiang G, Mizushima T, Fujita K, Inoue S, Yamaguchi S, Fushimi H, Nonomura N, Miyamoto H. Forkhead box O1 as an indicator of prognosis is inactivated in urothelial carcinoma of the upper urinary tract. Oncol Lett 2018; 17:482-487. [PMID: 30655790 DOI: 10.3892/ol.2018.9510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/23/2018] [Indexed: 12/14/2022] Open
Abstract
The transcription factor forkhead box O1 (FOXO1) can be inactivated via its phosphorylation, resulting in suppression of apoptosis. Using immunohistochemistry, the expression of a phosphorylated form of FOXO1 was assessed in upper urinary tract urothelial carcinoma (UUTUC) specimens. Overall, phospho-FOXO1 (p-FOXO1) was immunoreactive in all 99 UUTUC specimens [12 (12.1%) weak (1+), 46 (46.5%) moderate (2+) and 41 (41.4%) strong (3+)], which was significantly (P=0.018) increased, compared with benign urothelium specimens [77/82 (93.9%): 18 (22.0%) 1+, 41 (50.0%) 2+ and 18 (22.0%) 3+]. Muscle invasion (P=0.031) and lymphovascular invasion (P=0.025) were observed more frequently in p-FOXO1(2+/3+) tumor samples compared with p-FOXO1(1+) tumor samples. No statistically significant associations between p-FOXO1 expression and tumor grade or presence of concurrent carcinoma in situ, hydronephrosis or lymph node metastasis were observed. Furthermore, the levels of p-FOXO1 and estrogen receptor-β expression were significantly (P<0.05) correlated in UUTUC samples [correlation coefficient (CC)=0.244], particularly in tumor samples from male patients (CC=0.330). Additionally, patients with p-FOXO1(3+) tumors had a significantly increased risk of cancer-specific mortality (P=0.043), compared with those with p-FOXO1(1+/2+) tumors. Multivariate analysis further demonstrated a notable, albeit not significant, association between p-FOXO1 expression and cancer-specific survival (hazard ratio=2.204; P=0.053). These findings indicate that FOXO1 is inactivated in UUTUC specimens and p-FOXO1 overexpression may serve as a predictor of poor patient outcomes.
Collapse
Affiliation(s)
- Hiroki Ide
- Department of Pathology and James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Guiyang Jiang
- Department of Pathology and Laboratory Medicine and James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Taichi Mizushima
- Department of Pathology and James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pathology and Laboratory Medicine and James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Kazutoshi Fujita
- Department of Urology, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
| | - Satoshi Inoue
- Department of Pathology and James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pathology and Laboratory Medicine and James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Seiji Yamaguchi
- Department of Urology, Osaka General Medical Center, Osaka 558-8558, Japan
| | - Hiroaki Fushimi
- Department of Pathology, Osaka General Medical Center, Osaka 558-8558, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
| | - Hiroshi Miyamoto
- Department of Pathology and James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pathology and Laboratory Medicine and James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA.,Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
11
|
Zhang P, Gao H, Li Q, Chen X, Wu X. Downregulation of microRNA‑660 inhibits cell proliferation and invasion in osteosarcoma by directly targeting forkhead box O1. Mol Med Rep 2018; 18:2433-2440. [PMID: 29901128 DOI: 10.3892/mmr.2018.9165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/22/2018] [Indexed: 11/05/2022] Open
Abstract
The abnormal expression of microRNAs (miRNAs/miRs) has been observed in osteosarcoma (OS), and these differently expressed miRNAs contribute to the occurrence and development of OS by regulating various biological behaviours. Therefore, a comprehensive understanding of the detailed roles of aberrantly expressed miRNAs in OS progression may be favourable to the identification of promising therapeutic strategies for the treatment of patients with this malignancy. The present study demonstrated that miR‑660‑5p (miR‑660) expression was significantly upregulated in OS tissues and cell lines compared with that in normal adjacent tissues and normal human osteoblast hFOB1.19, respectively. miR‑660 downregulation led to a significant decrease in the proliferation and invasion of OS cells. Forkhead box O1 (FOXO1) was predicted as a potential target of miR‑660. The subsequent luciferase reporter assay indicated that miR‑660 directly binds to the 3'‑untranslated region of FOXO1. Furthermore, miR‑660 inhibition increased the FOXO1 expression in OS cells at mRNA and protein levels. Moreover, FOXO1 was downregulated in OS tissues and this downregulation was negatively correlated with miR‑660 levels. Besides, rescue experiments demonstrated that FOXO1 knockdown abolished the effects of miR‑660 knockdown on OS cell proliferation and invasion. These results suggest that miR‑660 may serve oncogenic roles in OS by directly targeting FOXO1. Targeting miR‑660 may be an effective candidate for the treatment of patients with OS.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Neurosurgery, Fujian Sanbo Funeng Brain Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Haifeng Gao
- Department of Oncology, Dongying People's Hospital, Dongying, Shandong 257000, P.R. China
| | - Qing Li
- Department of Pathology, Dongying People's Hospital, Dongying, Shandong 257000, P.R. China
| | - Xinlei Chen
- Department of Orthopedics, Central Hospital of Zibo, Zibo, Shandong 255022, P.R. China
| | - Xifa Wu
- Department of Orthopedics, Central Hospital of Zibo, Zibo, Shandong 255022, P.R. China
| |
Collapse
|