1
|
Han SR, Lee CH, Im JY, Kim JH, Kim JH, Kim SJ, Cho YW, Kim E, Kim Y, Ryu JH, Ju MH, Jeong JS, Lee SW. Targeted suicide gene therapy for liver cancer based on ribozyme-mediated RNA replacement through post-transcriptional regulation. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 23:154-168. [PMID: 33335800 PMCID: PMC7732968 DOI: 10.1016/j.omtn.2020.10.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/24/2020] [Indexed: 12/16/2022]
Abstract
Hepatocellular carcinoma (HCC) has high fatality rate and limited therapeutic options. Here, we propose a new anti-HCC approach with high cancer-selectivity and efficient anticancer effects, based on adenovirus-mediated Tetrahymena group I trans-splicing ribozymes specifically inducing targeted suicide gene activity through HCC-specific replacement of telomerase reverse transcriptase (TERT) RNA. To confer potent anti-HCC effects and minimize hepatotoxicity, we constructed post-transcriptionally enhanced ribozyme constructs coupled with splicing donor and acceptor site and woodchuck hepatitis virus post-transcriptional regulatory element under the control of microRNA-122a (miR-122a). Adenovirus encoding post-transcriptionally enhanced ribozyme improved trans-splicing reaction and decreased human TERT (hTERT) RNA level, efficiently and selectively retarding hTERT-positive liver cancers. Adenovirus encoding miR-122a-regulated ribozyme caused selective liver cancer cytotoxicity, the efficiency of which depended on ribozyme expression level relative to miR-122a level. Systemic administration of adenovirus encoding the post-transcriptionally enhanced and miR-regulated ribozyme caused efficient anti-cancer effects at a single dose of low titers and least hepatotoxicity in intrahepatic multifocal HCC mouse xenografts. Minimal liver toxicity, tissue distribution, and clearance pattern of the recombinant adenovirus were observed in normal animals administered either systemically or via the hepatic artery. Post-transcriptionally regulated RNA replacement strategy mediated by a cancer-specific ribozyme provides a clinically relevant, safe, and efficient strategy for HCC treatment.
Collapse
Affiliation(s)
- Seung Ryul Han
- R&D Center, Rznomics, Inc., Seongnam 13486, Republic of Korea
| | - Chang Ho Lee
- Department of Life Convergence, Research Institute of Advanced Omics, Dankook University, Yongin 16890, Republic of Korea
| | - Ji Young Im
- Department of Life Convergence, Research Institute of Advanced Omics, Dankook University, Yongin 16890, Republic of Korea
| | - Ju Hyun Kim
- Department of Life Convergence, Research Institute of Advanced Omics, Dankook University, Yongin 16890, Republic of Korea
| | - Ji Hyun Kim
- R&D Center, Rznomics, Inc., Seongnam 13486, Republic of Korea
| | - Sung Jin Kim
- Department of Life Convergence, Research Institute of Advanced Omics, Dankook University, Yongin 16890, Republic of Korea
| | - Young Woo Cho
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju 28160, Republic of Korea.,College of Pharmacy, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Eunkyung Kim
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju 28160, Republic of Korea.,College of Pharmacy, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Youngah Kim
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju 28160, Republic of Korea
| | - Ji-Ho Ryu
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju 28160, Republic of Korea.,College of Pharmacy, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Mi Ha Ju
- Department of Pathology and Immune-network Pioneer Research Center, Dong-A University College of Medicine, Busan 602-714, Republic of Korea
| | - Jin Sook Jeong
- Department of Pathology and Immune-network Pioneer Research Center, Dong-A University College of Medicine, Busan 602-714, Republic of Korea
| | - Seong-Wook Lee
- R&D Center, Rznomics, Inc., Seongnam 13486, Republic of Korea.,Department of Life Convergence, Research Institute of Advanced Omics, Dankook University, Yongin 16890, Republic of Korea
| |
Collapse
|
2
|
Lee CH, Han SR, Lee SW. Group I Intron-Based Therapeutics Through Trans-Splicing Reaction. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 159:79-100. [PMID: 30340790 DOI: 10.1016/bs.pmbts.2018.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In 1982, the Cech group discovered that an intron structure in an rRNA precursor of Tetrahymena thermophila is sufficient to complete splicing without assistance from proteins. This was the first moment that scientists recognized RNAs can have catalytic activities derived from their own unique three-dimensional structures and thus play more various roles in biological processes than thought before. Several additional catalytic RNAs, called ribozymes, were subsequently identified in nature followed by intense studies to reveal their mechanisms of action and to engineer them for use in fields such as molecular cell biology, therapeutics, imaging, etc. Naturally occurring RNA-targeting ribozymes can be broadly classified into two categories by their abilities: Self-cleavage and self-splicing. Since ribozymes use base-pairing to recognize cleavage sites, identification of the catalytic center of naturally occurring ribozymes enables to engineer from "self" to "trans" acting ones which has accelerated to design and use ribozyme as valuable tools in gene therapy fields. Especially, group I intron-based trans-splicing ribozyme has unique property to use as a gene therapeutic agent. It can destroy and simultaneously repair (and/or reprogram) target RNAs to yield the desired therapeutic RNAs, maintaining endogenous spatial and temporal gene regulation of target RNAs. There have been progressive improvements in trans-splicing ribozymes and successful applications of these elements in gene therapy and molecular imaging approaches for various pathogenic conditions. In this chapter, current status of trans-splicing ribozyme therapeutics, focusing on Tetrahymena group I intron-based ribozymes, and their future prospects will be discussed.
Collapse
Affiliation(s)
- Chang Ho Lee
- Department of Integrated Life Sciences, Dankook University, Yongin, Republic of Korea
| | | | - Seong-Wook Lee
- Department of Integrated Life Sciences, Dankook University, Yongin, Republic of Korea; Rznomics Inc., Gwangju, Republic of Korea.
| |
Collapse
|
3
|
Lee CH, Han SR, Lee SW. Therapeutic applications of group I intron-based trans-splicing ribozymes. WILEY INTERDISCIPLINARY REVIEWS-RNA 2018; 9:e1466. [PMID: 29383855 DOI: 10.1002/wrna.1466] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 12/10/2017] [Accepted: 12/14/2017] [Indexed: 12/21/2022]
Abstract
Since the breakthrough discovery of catalytic RNAs (ribozymes) in the early 1980s, valuable ribozyme-based gene therapies have been developed for incurable diseases ranging from genetic disorders to viral infections and cancers. Ribozymes can be engineered and used to downregulate or repair pathogenic genes via RNA cleavage mediated by trans-cleaving ribozymes or repair and reprograming mediated by trans-splicing ribozymes, respectively. Uniquely, trans-splicing ribozymes can edit target RNAs via simultaneous destruction and repair (and/or reprograming) to yield the desired therapeutic RNAs, thus selectively inducing therapeutic gene activity in cells expressing the target RNAs. In contrast to traditional gene therapy approaches, such as simple addition of therapeutic transgenes or inhibition of disease-causing genes, the selective repair and/or reprograming abilities of trans-splicing ribozymes in target RNA-expressing cells facilitates the maintenance of endogenous spatial and temporal gene regulation and reduction of disease-associated transcript expression. In molecular imaging technologies, trans-splicing ribozymes can be used to reprogram specific RNAs in living cells and organisms by the 3'-tagging of reporter RNAs. The past two decades have seen progressive improvements in trans-splicing ribozymes and the successful application of these elements in gene therapy and molecular imaging approaches for various pathogenic conditions, such as genetic, infectious, and malignant disease. This review provides an overview of the current status of trans-splicing ribozyme therapeutics, focusing on Tetrahymena group I intron-based ribozymes, and their future prospects. This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Chang Ho Lee
- Department of Integrated Life Sciences, Dankook University, Yongin, Republic of Korea
| | - Seung Ryul Han
- Department of Integrated Life Sciences, Dankook University, Yongin, Republic of Korea
| | - Seong-Wook Lee
- Department of Integrated Life Sciences, Dankook University, Yongin, Republic of Korea
| |
Collapse
|
4
|
Geisler A, Fechner H. MicroRNA-regulated viral vectors for gene therapy. World J Exp Med 2016; 6:37-54. [PMID: 27226955 PMCID: PMC4873559 DOI: 10.5493/wjem.v6.i2.37] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 03/02/2016] [Accepted: 03/17/2016] [Indexed: 02/06/2023] Open
Abstract
Safe and effective gene therapy approaches require targeted tissue-specific transfer of a therapeutic transgene. Besides traditional approaches, such as transcriptional and transductional targeting, microRNA-dependent post-transcriptional suppression of transgene expression has been emerging as powerful new technology to increase the specificity of vector-mediated transgene expression. MicroRNAs are small non-coding RNAs and often expressed in a tissue-, lineage-, activation- or differentiation-specific pattern. They typically regulate gene expression by binding to imperfectly complementary sequences in the 3' untranslated region (UTR) of the mRNA. To control exogenous transgene expression, tandem repeats of artificial microRNA target sites are usually incorporated into the 3' UTR of the transgene expression cassette, leading to subsequent degradation of transgene mRNA in cells expressing the corresponding microRNA. This targeting strategy, first shown for lentiviral vectors in antigen presenting cells, has now been used for tissue-specific expression of vector-encoded therapeutic transgenes, to reduce immune response against the transgene, to control virus tropism for oncolytic virotherapy, to increase safety of live attenuated virus vaccines and to identify and select cell subsets for pluripotent stem cell therapies, respectively. This review provides an introduction into the technical mechanism underlying microRNA-regulation, highlights new developments in this field and gives an overview of applications of microRNA-regulated viral vectors for cardiac, suicide gene cancer and hematopoietic stem cell therapy, as well as for treatment of neurological and eye diseases.
Collapse
|
5
|
Geng J, Liu Y, Jin Y, Tai J, Zhang J, Xiao X, Chu P, Yu Y, Wang SC, Lu J, Han S, Shi J, Guo Y, Ni X. MicroRNA-365a-3p promotes tumor growth and metastasis in laryngeal squamous cell carcinoma. Oncol Rep 2016; 35:2017-26. [PMID: 26883008 DOI: 10.3892/or.2016.4617] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/10/2015] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs (miRNAs) are increasingly recognized as oncogenes or tumor suppressors in laryngeal squamous cell carcinoma (LSCC). In this study, we analyzed the roles of miR-365a-3p, miR-143-5p, and miR-494-3p in LSCC using Annexin V/propidium iodide double staining and flow cyto-metry, along with a Transwell migration and invasion assay. The results showed that miR-365a-3p inhibitor significantly facilitated cell apoptosis and suppressed cell cycle progression, migration, and invasion in Hep-2 cells. However, miR-143-5p and miR-494-3p had no such influences. We then investigated the role of miR-365a-3p in LSCC in vivo and found that miR-365a-3p inhibitor suppressed LSCC xenograft tumor growth and metastasis in xenograft mouse models. Moreover, miR-365a-3p inhibitor significantly decreased the expression of p-AKT (Ser473), which indicated that miR-365a-3p can mediate PI3K/AKT signaling pathway transduction via p-AKT (Ser473) in LSCC. The data suggest that miR-365a-3p may act as an oncomiR and may promote growth and metastasis in LSCC via the PI3K/AKT signaling pathway, and thus miR‑365a-3p may be a potential therapeutic target for treatment of LSCC.
Collapse
Affiliation(s)
- Jiangqiao Geng
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Yuanhu Liu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Yaqiong Jin
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Jun Tai
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Jie Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Xiao Xiao
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Ping Chu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Yongbo Yu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Sheng Cai Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Jie Lu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Shujing Han
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Jin Shi
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Yongli Guo
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| | - Xin Ni
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 100045, P.R. China
| |
Collapse
|
6
|
Lee CH, Han SR, Lee SW. Therapeutic Applications of Aptamer-Based Riboswitches. Nucleic Acid Ther 2015; 26:44-51. [PMID: 26539634 DOI: 10.1089/nat.2015.0570] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aptamers bind to their targets with high affinity and specificity through structure-based complementarity, instead of sequence complementarity that is used by most of the oligonucleotide-based therapeutics. This property has been exploited in using aptamers as multifunctional therapeutic units, by attaching them to therapeutic drugs, nanoparticles, or imaging agents, or as direct molecular decoys for inducing loss-of-function or gain-of-function of targets. One of the most interesting fields of aptamer application is their development as molecular sensors to regulate artificial riboswitches. Naturally, the riboswitches sense small-molecule metabolites and respond by regulating the expression of the corresponding metabolic genes. Riboswitches are cis-acting RNA structures that consist of the sensing (aptamer) and the regulating (expression platform) domains. In principle, diverse riboswitches can be engineered and applied to control different steps of gene expression in bacterial species as well as eukaryotes, by simply replacing aptamers against various endogenous and/or exogenous targets. Although these engineered aptamer-based riboswitches are recently gaining attention, it is clear that aptamer-based riboswitches have a potential for next-generation therapeutics against various diseases because of their controllability, specificity, and modularity in regulating gene expression through various cellular processes, including transcription, splicing, stability, RNA interference, and translation. In this review, we provide a summary of the recently developed and engineered aptamer-based riboswitches focusing on their therapeutic availability and further discuss their clinical potential.
Collapse
Affiliation(s)
- Chang Ho Lee
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, and Research Institute of Advanced Omics, Dankook University , Yongin, Republic of Korea
| | - Seung Ryul Han
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, and Research Institute of Advanced Omics, Dankook University , Yongin, Republic of Korea
| | - Seong-Wook Lee
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, and Research Institute of Advanced Omics, Dankook University , Yongin, Republic of Korea
| |
Collapse
|
7
|
Sang F, Yang Y, Yuan L, Ren J, Zhang Z. Development of a high-throughput real time PCR based on a hot-start alternative for Pfu mediated by quantum dots. NANOSCALE 2015; 7:15852-15862. [PMID: 26360777 DOI: 10.1039/c5nr03596a] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Hot start (HS) PCR is an excellent alternative for high-throughput real time PCR due to its ability to prevent nonspecific amplification at low temperature. Development of a cost-effective and simple HS PCR technique to guarantee high-throughput PCR specificity and consistency still remains a great challenge. In this study, we systematically investigated the HS characteristics of QDs triggered in real time PCR with EvaGreen and SYBR Green I dyes by the analysis of amplification curves, standard curves and melting curves. Two different kinds of DNA polymerases, Pfu and Taq, were employed. Here we showed that high specificity and efficiency of real time PCR were obtained in a plasmid DNA and an error-prone two-round PCR assay using QD-based HS PCR, even after an hour preincubation at 50 °C before real time PCR. Moreover, the results obtained by QD-based HS PCR were comparable to a commercial Taq antibody DNA polymerase. However, no obvious HS effect of QDs was found in real time PCR using Taq DNA polymerase. The findings of this study demonstrated that a cost-effective high-throughput real time PCR based on QD triggered HS PCR could be established with high consistency, sensitivity and accuracy.
Collapse
Affiliation(s)
- Fuming Sang
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai, 264209, People's Republic of China.
| | | | | | | | | |
Collapse
|
8
|
Li J, Lei H, Xu Y, Tao ZZ. miR-512-5p suppresses tumor growth by targeting hTERT in telomerase positive head and neck squamous cell carcinoma in vitro and in vivo. PLoS One 2015; 10:e0135265. [PMID: 26258591 PMCID: PMC4530866 DOI: 10.1371/journal.pone.0135265] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/20/2015] [Indexed: 12/14/2022] Open
Abstract
Telomerase activation has very important implications for head and neck squamous cell carcinoma (HNSCC), but the regulatory mechanisms of telomerase in HNSCC remain unclear. In our present study, we found that miR-512-5P was markedly downregulated in telomerase-positive HNSCC cell lines. Both in vitro and in vivo assays revealed that miR-512-5P mimic attenuated HNSCC cell proliferation, and tumor growth in nude mice, which exerts its tumor suppressor function through elevated apoptosis, inhibition of the telomerase activity, decrease of telomere-binding proteins and shortening of telomere length by human telomerase reverse transcriptase (hTERT) downregulation. Furthermore, the dual-luciferase reporter gene assay results demonstrated that hTERT was a direct target of miR-512-5P. We conclude that the frequently miR-512-5P overexpression can regulate hTERT and function as a tumor suppressor in HNSCC. Therefore, miR-512-5P may serve as a potential therapeutic agent for miR-based HNSCC therapy.
Collapse
Affiliation(s)
- Jun Li
- Department of Otolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Han Lei
- Hubei key laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yong Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ze-zhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- * E-mail:
| |
Collapse
|
9
|
Ma Z, Qiu X, Wang D, Li Y, Zhang B, Yuan T, Wei J, Zhao B, Zhao X, Lou J, Jin Y, Jin Y. MiR-181a-5p inhibits cell proliferation and migration by targeting Kras in non-small cell lung cancer A549 cells. Acta Biochim Biophys Sin (Shanghai) 2015; 47:630-8. [PMID: 26124189 DOI: 10.1093/abbs/gmv054] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 04/25/2015] [Indexed: 12/31/2022] Open
Abstract
MicroRNAs play important roles in carcinogenesis and tumor progress. Lung cancer is the leading cause of cancer mortality worldwide. In this study, the function of miR-181a-5p was investigated in non-small-cell lung cancer (NSCLC). Results showed that miR-181a-5p was significantly decreased in NSCLC tissues and cell lines. The proliferation and migration of A549 cells transfected with miR-181a-5p mimic was significantly inhibited. Luciferase activity assay results demonstrated that two binding sites of Kras could be directly targeted by miR-181a-5p. Furthermore, Kras was down-regulated by miR-181a-5p at both transcriptional and translational levels. SiRNA-mediated Kras down-regulation could mimic the effects of miR-181a-5p mimic in A549 cells. Our findings suggest that miR-181a-5p plays a potential role in tumor suppression by partially targeting Kras and has the potential therapeutic application in NSCLC patients.
Collapse
Affiliation(s)
- Zhongliang Ma
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Xiang Qiu
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Detao Wang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yanli Li
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Bingjie Zhang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Tianwei Yuan
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Jiali Wei
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Botao Zhao
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Xintai Zhao
- Shanghai Shines Pharmaceuticals Co., Ltd., Shanghai 200032, China
| | - Jiatao Lou
- Department of Laboratory Medicine, Shanghai Chest Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200030, China
| | - Yan Jin
- Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences, Shenzhen 518055, China
| | - Youxin Jin
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| |
Collapse
|