1
|
Miao N, Cai W, Ding S, Liu Y, Chen W, Sun T. Characterization of plasma exosomal microRNAs in responding to radiotherapy of human esophageal squamous cell carcinoma. Mol Med Rep 2022; 26:287. [PMID: 35894132 PMCID: PMC9366155 DOI: 10.3892/mmr.2022.12803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 07/11/2022] [Indexed: 11/30/2022] Open
Abstract
Radiotherapy is one of the main treatment methods for esophageal squamous cell carcinoma (ESCC). Previous research has shown that plasma exosomal microRNAs (miRNAs) can predict therapeutic outcome. In the present study, to identify potential exosomal miRNAs that respond to radiotherapy, plasma exosomal miRNAs from ESCC patients undergoing radiotherapy were isolated and sequenced. Upregulated and downregulated miRNAs were detected from patients pre- and post-radiotherapy, and it was found that they play distinct roles in DNA damage process and endosomal mediated transport. Based on wound healing and Cell Counting Kit-8 assays in TE-1 human esophageal cancer cells, it was identified that representative miRNA miR-652 and miR-30a alter migration but not proliferation. The present findings identified differentially expressed miRNAs in responding to radiotherapy, and added a reference to explore non-invasive plasma biomarkers to evaluate therapeutic effects in ESCC.
Collapse
Affiliation(s)
- Nan Miao
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian 361021, P.R. China
| | - Wenjie Cai
- Department of Radiation Oncology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Sijia Ding
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian 361021, P.R. China
| | - Yajuan Liu
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian 361021, P.R. China
| | - Wanhua Chen
- Department of Clinical Laboratory, First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, P.R. China
| | - Tao Sun
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, Fujian 361021, P.R. China
| |
Collapse
|
2
|
Muhuri M, Zhan W, Maeda Y, Li J, Lotun A, Chen J, Sylvia K, Dasgupta I, Arjomandnejad M, Nixon T, Keeler AM, Manokaran S, He R, Su Q, Tai PWL, Gao G. Novel Combinatorial MicroRNA-Binding Sites in AAV Vectors Synergistically Diminish Antigen Presentation and Transgene Immunity for Efficient and Stable Transduction. Front Immunol 2021; 12:674242. [PMID: 33995418 PMCID: PMC8113644 DOI: 10.3389/fimmu.2021.674242] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/07/2021] [Indexed: 12/26/2022] Open
Abstract
Recombinant adeno-associated virus (rAAV) platforms hold promise for in vivo gene therapy but are undermined by the undesirable transduction of antigen presenting cells (APCs), which in turn can trigger host immunity towards rAAV-expressed transgene products. In light of recent adverse events in patients receiving high systemic AAV vector doses that were speculated to be related to host immune responses, development of strategies to mute innate and adaptive immunity is imperative. The use of miRNA binding sites (miR-BSs) to confer endogenous miRNA-mediated regulation to detarget transgene expression from APCs has shown promise for reducing transgene immunity. Studies have shown that designing miR-142BSs into rAAV1 vectors were able to repress costimulatory signals in dendritic cells (DCs), blunt the cytotoxic T cell response, and attenuate clearance of transduced muscle cells in mice to allow sustained transgene expression in myofibers with negligible anti-transgene IgG production. In this study, we screened individual and combinatorial miR-BS designs against 26 miRNAs that are abundantly expressed in APCs, but not in skeletal muscle. The highly immunogenic ovalbumin (OVA) transgene was used as a proxy for foreign antigens. In vitro screening in myoblasts, mouse DCs, and macrophages revealed that the combination of miR-142BS and miR-652-5pBS strongly mutes transgene expression in APCs but maintains high myoblast and myocyte expression. Importantly, rAAV1 vectors carrying this novel miR-142/652-5pBS cassette achieve higher transgene levels following intramuscular injections in mice than previous detargeting designs. The cassette strongly inhibits cytotoxic CTL activation and suppresses the Th17 response in vivo. Our approach, thus, advances the efficiency of miRNA-mediated detargeting to achieve synergistic reduction of transgene-specific immune responses and the development of safe and efficient delivery vehicles for gene therapy.
Collapse
Affiliation(s)
- Manish Muhuri
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
| | - Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
| | - Yukiko Maeda
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - Jia Li
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Anoushka Lotun
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Jennifer Chen
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Katelyn Sylvia
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, United States
| | - Ishani Dasgupta
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, United States
| | - Motahareh Arjomandnejad
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, United States
| | - Thomas Nixon
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, United States
| | - Allison M. Keeler
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, United States
| | - Sangeetha Manokaran
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Ran He
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Qin Su
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Phillip W. L. Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
3
|
Zhao F, Yang G, Feng M, Cao Z, Liu Y, Qiu J, You L, Zheng L, Zhang T, Zhao Y. Expression, function and clinical application of stanniocalcin-1 in cancer. J Cell Mol Med 2020; 24:7686-7696. [PMID: 32468698 PMCID: PMC7348177 DOI: 10.1111/jcmm.15348] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/10/2019] [Accepted: 10/19/2019] [Indexed: 12/13/2022] Open
Abstract
The glycoprotein stanniocalcin-1 functions as a regulatory endocrine hormone that maintains the balance of calcium and phosphorus in bony fish and as a paracrine/autocrine factor involved in many physiological/pathological processes in humans, including carcinogenesis. In this review, we provide an overview of (a) the possible mechanisms through which STC1 affects the malignant properties of cancer, (b) transcriptional and post-transcriptional regulation pathways of STC1 and (c) the potential clinical relevance of STC1 as a cancer biomarker and even a therapeutic target in the future. Exploring the role of STC1 in cancer development may provide a better understanding of the tumorigenesis process in humans and may facilitate finding an effective therapeutic method against cancer.
Collapse
Affiliation(s)
- Fangyu Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengyu Feng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yueze Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Xu J, Zhang Z, Chen Q, Yang L, Yin J. miR-146b Regulates Cell Proliferation and Apoptosis in Gastric Cancer by Targeting PTP1B. Dig Dis Sci 2020; 65:457-463. [PMID: 31441000 DOI: 10.1007/s10620-019-05771-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/30/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND/AIMS The purpose of this study is to explore the inhibition or activation effects of microRNA-146 B on the expression of PTP1B in gastric cancer cells. METHODS The expressions of PTP1B and miR-146b in gastric cancer were detected by RT-qPCR. The effects of miR-146b on cell apoptosis and proliferation of gastric cancer were detected. The methods used in the detection process included Annexin V/PI dying method, colony formation assay, and MTT assay. The downstream target gene miR-146b was predicted and screened by bioinformatics and luciferase reporter assay. The mRNA and protein expressions of the target gene PTP1B miR-146b were determined using RT-qPCR and western blot. The expression of miR-146 B in mice was detected by the cells transfected with microRNA-146 B in vivo. RESULTS Compared with normal tissues, PTP1B was higher and miR-146b was lower in cancer cells. Over-expression of miR-146b can inhibit cell viability and increase the apoptosis rate. According to the luciferase reporter assay, PTP1B was the downstream target gene of miR-146b. The re-introduction of PTP1B reversed the growth inhibition and apoptosis of gastric cancer cells induced by miR-146b. From the mouse xenograft model, the over-expression of miR-146b inhibited the tumor growth and reduced the expression level of PTP1B. CONCLUSION miR-146b directly inhibits the expression of PTP1B and suppressed the growth and development of gastric cancer.
Collapse
Affiliation(s)
- Jianguo Xu
- Department of Oncology Surgery, People's Hospital of Qinghai Province, Xining City, 810007, Qinghai Province, China
| | - Zilong Zhang
- Department of Oncology Surgery, People's Hospital of Qinghai Province, Xining City, 810007, Qinghai Province, China
| | - Qing Chen
- Department of Orthopedic, The 991 Hospital of PLA, Nanjing City, 441011, Jiangsu Province, China
| | - Lin Yang
- Department of Immunity, Medical College of Hubei University of Arts and Science, No. 296 Longzhong Road, Xiangyang City, 441053, Hubei Province, China
| | - Jiao Yin
- Department of Immunity, Medical College of Hubei University of Arts and Science, No. 296 Longzhong Road, Xiangyang City, 441053, Hubei Province, China.
| |
Collapse
|
5
|
Chang ZW, Jia YX, Zhang WJ, Song LJ, Gao M, Li MJ, Zhao RH, Li J, Zhong YL, Sun QZ, Qin YR. LncRNA-TUSC7/miR-224 affected chemotherapy resistance of esophageal squamous cell carcinoma by competitively regulating DESC1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018. [PMID: 29530057 PMCID: PMC5848549 DOI: 10.1186/s13046-018-0724-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND This study aims to clarify the underlying mechanism for the tumor suppressive function of lnc TUSC7 in chemotherapy resistance of esophageal squamous cell carcinoma (ESCC). METHODS TUSC7, miR-224 and DESC1 expressions in ESCC tissues and cells were detected by qRT-PCR. Protein level of DESC1, EGFR and p-AKT were observed by Western blot. Overall survival was calculated using the Kaplan-Meier method. Dual-luciferase reporter gene assay and RIP assay were used to comfirm TUSC7 binding to miR-224, and miR-224 binding to DESC1. Cell proliferation, apoptosis, and colony formation was detected by MTT, Flow Cytometry and Colony formation assays. RESULTS TUSC7 was downregulated in ESCC tissues and cells, and low TUSC7 indicated worse overall survival. The analysis of bioinformatics softwares showed that TUSC7 specifically bound to miR-224, and we proved miR-224 was upregulated in ESCC and negatively correlated with TUSC7 expression. Overexpression of TUSC7/inhibition of miR-224 suppressed cell proliferation, colony formation and chemotherapy resistance of ESCC cells, and promoted cell apoptosis. In addition, we confirmed that miR-224 specifically bound to DESC1, and negatively correlated with DESC1. TUSC7 suppressed the proliferation and chemotherapy resistance of ESCC cells by increasing DESC1 expression via inhibiting miR-224. We also confirmed DESC1 inhibited chemotherapy resistance of ESCC cells via EGFR/AKT. Finally, in vivo experiments demonstrated that overexpression of TUSC7 decreased tumor growth and chemotherapy resistance. CONCLUSION These findings suggested TUSC7 suppressed chemotherapy resistance of ESCC by downregulating miR-224 to modulate DESC1/EGFR/AKT pathway.
Collapse
Affiliation(s)
- Zhi-Wei Chang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Yong-Xu Jia
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Wei-Jie Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Li-Jie Song
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Ming Gao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Ming-Jun Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Rui-Hua Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Jing Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Ya-Li Zhong
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Qiao-Zhi Sun
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Yan-Ru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450052, Henan, People's Republic of China.
| |
Collapse
|