1
|
Qin H, Wang C, Hua Y. LINC01123 is associated with prognosis of oral squamous cell carcinoma and involved in tumor progression by sponging miR-34a-5p. Oral Surg Oral Med Oral Pathol Oral Radiol 2021; 133:50-59. [PMID: 34511356 DOI: 10.1016/j.oooo.2021.07.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/22/2021] [Accepted: 07/13/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Oral squamous cell carcinoma (OSCC) is a malignant tumor. This study aimed to investigate the role of a long noncoding RNA (lncRNA), LINC01123, in OSCC prognosis and progression and to explore the underlying mechanisms. STUDY DESIGN OSCC tissues were collected from 102 patients, and 4 OSCC cell lines were analyzed. The expression levels of LINC01123 and miR-34a-5p were estimated using quantitative real-time polymerase chain reaction (qRT-PCR). Cell counting kit-8 (CCK-8) and Transwell assays were used to assess the proliferation, migration, and invasion of OSCC cells. Kaplan-Meier survival analysis was used to analyze the prognostic value of LINC01123 in OSCC. RESULTS The analysis results showed that LINC01123 was overexpressed in OSCC tumor tissues; also, the prognosis of patients with OSCC with high LINC01123 expression levels was poor. The knockdown of LINC01123 inhibited the proliferation, migration, and invasion of OCSS cells. MiR-34a-5p was a target of LINC01123, and its inhibitor could reverse the effect of silenced LINC01123 on the progression of OSCC. CONCLUSIONS Highly expressed LINC01123 was associated with poor prognosis of OSCC and regulated OSCC cell proliferation, invasion, and migration by sponging miR-34a-5p. Therefore, the LINC01123/miR-34a-5p axis may provide new ideas for the prognosis and treatment of OSCC.
Collapse
Affiliation(s)
- Huan Qin
- Department of Stomatology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, 261031, China
| | - Changlei Wang
- Department of Stomatology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, 261031, China
| | - Yingjie Hua
- Department of Stomatology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, 261031, China..
| |
Collapse
|
2
|
Affiliation(s)
- Huiling Wang
- Guangxi Key Laboratory of Bio‐targeting Theranostics National Center for International Research of Bio‐targeting Theranostics Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy Guangxi Medical University Nanning China
| | - Yong Huang
- Guangxi Key Laboratory of Bio‐targeting Theranostics National Center for International Research of Bio‐targeting Theranostics Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy Guangxi Medical University Nanning China
| | - Jian He
- Guangxi Key Laboratory of Bio‐targeting Theranostics National Center for International Research of Bio‐targeting Theranostics Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy Guangxi Medical University Nanning China
| | - Liping Zhong
- Guangxi Key Laboratory of Bio‐targeting Theranostics National Center for International Research of Bio‐targeting Theranostics Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy Guangxi Medical University Nanning China
| | - Yongxiang Zhao
- Guangxi Key Laboratory of Bio‐targeting Theranostics National Center for International Research of Bio‐targeting Theranostics Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy Guangxi Medical University Nanning China
| |
Collapse
|
3
|
OSCC Exosomes Regulate miR-210-3p Targeting EFNA3 to Promote Oral Cancer Angiogenesis through the PI3K/AKT Pathway. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2125656. [PMID: 32695810 PMCID: PMC7368228 DOI: 10.1155/2020/2125656] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022]
Abstract
This study is aimed at determining how oral squamous cell carcinoma (OSCC) regulates the angiogenesis of HUVECs through miR-210-3p expression and exploring the relationship among miR-210-3p, its target protein, and the possible mechanism of angiogenesis regulation. miR-210-3p expression was detected in OSCC tissues and juxta cancerous tissues (JCT), and the relationship among miR-210-3p, microvessel density (MVD), and histopathologic features was analyzed. A conditioned medium (CM) of the OSCC cell line CAL27 was collected to stimulate human umbilical vein endothelial cells (HUVECs), and the miR-210-3p levels and tube formation capability of HUVECs were measured. The target protein level of miR-210-3p was altered; then, PI3K/AKT pathway activation in HUVECs was detected. miR-210-3p was tested in exosomes separated from CAL27 CM, and the transfer of miR-210-3p from OSCC exosomes to HUVECs was verified. Then, we found that the OSCC tissues had higher miR-210-3p levels than the JCT, and miR-210-3p level was positively correlated with MVD and tumor grade. CAL27 CM was able to elevate miR-210-3p levels in HUVECs and promoted tube formation. EFNA3 was the target gene of miR-210-3p, and ephrinA3 protein level was able to influence the migration and proliferation of HUVECs. The levels of phosphorylated AKT in the HUVECs increased when ephrinA3 was downregulated, and the upregulation of ephrinA3 resulted in the suppression of the PI3K/AKT pathway. miR-210-3p was detected in exosomes isolated from the CM of CAL27 cells, and miR-210-3p level in the HUVECs was elevated after absorbing the OSCC exosomes. In conclusion, miR-210-3p was more overexpressed in OSCC tissues than in the JCT. The exosomes secreted by OSCC cells were able to upregulate miR-210-3p expression and reduce ephrinA3 expression in HUVECs and promoted tube formation through the PI3K/AKT signaling pathway.
Collapse
|
4
|
Pang X, Liang S, Wang T, Yu S, Yang R, Hou T, Liu Y, He C, Zhang N. Engineering Thermo-pH Dual Responsive Hydrogel for Enhanced Tumor Accumulation, Penetration, and Chemo-Protein Combination Therapy. Int J Nanomedicine 2020; 15:4739-4752. [PMID: 32753862 PMCID: PMC7342477 DOI: 10.2147/ijn.s253990] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/01/2020] [Indexed: 01/20/2023] Open
Abstract
Purpose Combined chemotherapeutic drug and protein drug has been a widely employed strategy for tumor treatment. To realize both tumor accumulation and deep tumor penetration for drugs with different pharmacokinetics, we propose a structure-transformable, thermo-pH dual responsive co-delivery system to co-load granzyme B/docetaxel (GrB/DTX). Methods Thermo-sensitive hydrogels based on diblock copolymers (mPEG-b-PELG) were synthesized through ring opening polymerization. GrB/DTX mini micelles (GDM) was developed by co-loading these two drugs in pH-sensitive mini micelles, and the GDM-incorporated thermo-sensitive hydrogel (GDMH) was constructed. The thermo-induced gelation behavior of diblock copolymers and the physiochemical properties of GDMH were characterized. GDMH degradation and deep tumor penetration of released mini micelles were confirmed. The pH-sensitive disassembly and lysosomal escape abilities of released mini micelles were evaluated. In vitro cytotoxicity was studied using MTT assays and the in vivo antitumor efficacy study was evaluated in B16-bearing C57BL/6 mice. Results GDMH was gelatinized at body temperature and can be degraded by proteinase to release mini micelles. The mini micelles incorporated in GDMH can achieve deep tumor penetration and escape from lysosomes to release GrB and DTX. MTT results showed that maximum synergistic antitumor efficacy of GrB and DTX was observed at mass ratio of 1:100. Our in vivo antitumor efficacy study showed that GDMH inhibited tumor growth in the subcutaneous tumor model and in the post-surgical recurrence model. Conclusion The smart-designed transformable GDMH can facilitate tumor accumulation, deep tumor penetration, and rapid drug release to achieve synergistic chemo-protein therapy.
Collapse
Affiliation(s)
- Xiuping Pang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Shuang Liang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Tianqi Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Shuangjiang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
| | - Rui Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Teng Hou
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Chaoliang He
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| |
Collapse
|
5
|
Chen S, Chen LH, Niu YH, Geng NB, Feng CJ. AEG-1 promotes angiogenesis and may be a novel treatment target for tongue squamous cell carcinoma. Oral Dis 2020; 26:876-884. [PMID: 32031319 DOI: 10.1111/odi.13300] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/15/2020] [Accepted: 01/27/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND The study explored the potential function of astrocyte elevated gene-1 (AEG-1) on angiogenesis in tongue squamous cell carcinoma (TSCC) in TSCC cell lines. METHODS The different degrees of angiogenesis were detected in TSCC cell lines expressing different levels of AEG-1 by chick chorioallantoic membrane (CAM) experimental model. Next, we established xenografts of different TSCC cell lines with different expression levels of AEG-1 in nude mice and conducted immunohistochemistry to evaluate the expression of the angiogenesis-associated factor, that is, vascular endothelial growth receptor factor 2 (VEGFR-2) and microvessel density (MVD). Vascular endothelial growth factor (VEGF) was detected by ELISA. RESULTS CAM assay showed that the number of vessels was significantly reduced in AEG-1-down um1 cell line (p < .05), whereas the number was significantly increased in AEG-1-over um2 cell line (p < .05). Moreover, up-regulated AEG-1 expression level was associated with higher tumor angiogenesis, which was reflected by augmented expression levels of VEGF (p < .01), VEGFR-2 (p < .05), and MVD counting (p < .01). CONCLUSIONS This study demonstrated that AEG-1 can promote tumor angiogenesis in TSCC and inhibition of tumor angiogenesis by repressing the expression of AEG-1 may be a novel potential treatment approach for TSCC.
Collapse
Affiliation(s)
- Shan Chen
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lei-Hui Chen
- Department of Stomatology, Guangdong No. 2 Provincial People's Hospital, Guangzhou, China
| | - Yan-Hong Niu
- Department of Oral and Maxillofacial Surgery, Huizhou Municipal Central Hospital, Huizhou, China
| | - Ning-Bo Geng
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chong-Jin Feng
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
6
|
Shao TR, Zheng ZN, Chen YC, Wu QQ, Huang GZ, Li F, Zeng WS, Lv XZ. LncRNA AC007271.3 promotes cell proliferation, invasion, migration and inhibits cell apoptosis of OSCC via the Wnt/β-catenin signaling pathway. Life Sci 2019; 239:117087. [PMID: 31759044 DOI: 10.1016/j.lfs.2019.117087] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/07/2019] [Accepted: 11/15/2019] [Indexed: 01/16/2023]
Abstract
AIMS Long noncoding RNA (lncRNA) AC007271.3 has been identified to be dysregulated in oral squamous cell carcinoma (OSCC) in our previous study. However, the precise role of AC007271.3 in OSCC remains unclear. In this study, we investigated the potential functions and the underlying mechanisms of AC007271.3 in OSCC. MATERIALS AND METHODS The expression levels of AC007271.3 in OSCC tissues and cell lines were examined using RT-qPCR. The relationship between AC007271.3 level and clinicopathological characteristics was analyzed, and its association with patient prognosis was assessed by the Kaplan-Meier method. The biological function of AC007271.3 and its role in the development of OSCC through Wnt/β-catenin signaling pathway were studied. KEY FINDINGS We identified that AC007271.3 was up-regulated and positively correlated with advanced clinical stage, lymph node metastasis, poor histological differentiation and unfavorable prognosis. We explored the expression, function, and molecular mechanism of AC007271.3 in OSCC cells. Overexpression of AC007271.3 remarkably promoted cell proliferation in vitro and in vivo, induced cell migration, invasion and inhibited apoptosis in vitro, while knockdown of AC007271.3 attenuated cell proliferation, migration, invasion and induced apoptosis. Mechanistically, AC007271.3 overexpression substantially increased the expression of β-catenin and the downstream target molecules CyclinD1, c-myc and Bcl-2, while silencing of AC007271.3 has the opposite effect. Rescued experiments showed that the ability to promote cell proliferation, migration, invasion and inhibiting apoptosis could be reversed when treated with the Wnt/β-catenin pathway inhibitor. SIGNIFICANCE Our data indicated that AC007271.3 could promote cell proliferation, invasion and inhibit cell apoptosis of OSCC via the Wnt/β-catenin signaling pathway, which might provide a novel therapeutic approach for OSCC.
Collapse
Affiliation(s)
- Ting-Ru Shao
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, China
| | - Ze-Nan Zheng
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, China
| | - Yue-Chuan Chen
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, China
| | - Qing-Qing Wu
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, China
| | - Guang-Zhao Huang
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, China
| | - Fei Li
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, China
| | - Wei-Sen Zeng
- Department of Cell Biology, School of Basic Medicine, Southern Medical University, Guangzhou, China.
| | - Xiao-Zhi Lv
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Cheung LH, Zhao Y, Alvarez-Cienfuegos A, Mohamedali KA, Cao YJ, Hittelman WN, Rosenblum MG. Development of a human immuno-oncology therapeutic agent targeting HER2: targeted delivery of granzyme B. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:332. [PMID: 31362764 PMCID: PMC6668111 DOI: 10.1186/s13046-019-1333-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/21/2019] [Indexed: 01/24/2023]
Abstract
Background Immunotherapeutic approaches designed to augment T and B cell mediated killing of tumor cells has met with clinical success in recent years suggesting tremendous potential for treatment in a broad spectrum of tumor types. After complex recognition of target cells by T and B cells, delivery of the serine protease granzyme B (GrB) to tumor cells comprises the cytotoxic insult resulting in a well-characterized, multimodal apoptotic cascade. Methods We designed a recombinant fusion construct, GrB-Fc-4D5, composed of a humanized anti-HER2 scFv fused to active GrB for recognition of tumor cells and internal delivery of GrB, simulating T and B cell therapy. We assessed the construct’s antigen-binding specificity and GrB enzymatic activity, as well as in vitro cytotoxicity and internalization into target and control cells. We also assessed pharmacokinetic and toxicology parameters in vivo. Results GrB-Fc-4D5 was highly cytotoxic to Her2 positive cells such as SKBR3, MCF7 and MDA-MB-231 with IC50 values of 56, 99 and 27 nM, respectively, and against a panel of HER2+ cell lines regardless of endogenous expression levels of the PI-9 inhibitor. Contemporaneous studies with Kadcyla demonstrated similar levels of in vitro activity against virtually all cells tested. GrB-Fc-4D5 internalized rapidly into target SKOV3 cells within 1 h of exposure rapidly delivering GrB to the cytoplasmic compartment. In keeping with its relatively high molecular weight (160 kDa), the construct demonstrated a terminal-phase serum half-life in mice of 39.2 h. Toxicity studies conducted on BALB/c mice demonstrated no statistically significant changes in SGPT, SGOT or serum LDH. Histopathologic analysis of tissues from treated mice demonstrated no drug-related changes in any tissues examined. Conclusion GrB-Fc-4D5 shows excellent, specific cytotoxicity and demonstrates no significant toxicity in normal, antigen-negative murine models. This construct constitutes a novel approach against HER2-expressing tumors and is an excellent candidate for further development.
Collapse
Affiliation(s)
- Lawrence H Cheung
- Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Yunli Zhao
- Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Present address: Department of Pharmaceutical Analysis, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, China
| | - Ana Alvarez-Cienfuegos
- Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Khalid A Mohamedali
- Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Yu J Cao
- Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Present Address: Shenzhen Graduate School, School of Chemical Biology and Biotechnology, Peking University, Nanshan, Shenzhen, 518055, China
| | - Walter N Hittelman
- Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Michael G Rosenblum
- Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics, Unit 1950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| |
Collapse
|
8
|
Synergistic effect of granzyme B-azurin fusion protein on breast cancer cells. Mol Biol Rep 2019; 46:3129-3140. [DOI: 10.1007/s11033-019-04767-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 03/14/2019] [Indexed: 01/24/2023]
|