1
|
Liu H, Chen L, Chen Y, Jin Y, Chen X, Ma N, Yang F, Bi H, Wen X, Xu S, Chen J, Lin Y, Yang Y, Wu Y, Chen Y. TCP1 promotes the progression of malignant tumours by stabilizing c-Myc through the AKT/GSK-3β and ERK signalling pathways. Commun Biol 2025; 8:563. [PMID: 40185866 PMCID: PMC11971430 DOI: 10.1038/s42003-025-07867-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 03/03/2025] [Indexed: 04/07/2025] Open
Abstract
The chaperonin tailless complex polypeptide 1 (TCP1) is a key subunit of chaperonin containing TCP1 (CCT) that regulates the folding and stability of proteins during cancer progression. Here, the prognostic significance of TCP1 was explored mainly in patients with hepatocellular carcinoma (HCC) and pancreatic ductal adenocarcinoma (PDAC). We showed that TCP1 expression was significantly greater in clinically malignant tumour tissues than in normal tissues and that high TCP1 expression was associated with poor prognosis. TCP1 suppression not only decreased the proliferation and invasion of cancer cells in vitro but also inhibited tumour growth and metastasis in vivo. The underlying mechanisms were determined by ubiquitination assays and Co-IP (Co-Immunoprecipitation) experiments, and it was found that TCP1 regulated the stability of c-Myc through the RAC-alpha serine/threonine-protein kinase (AKT) /Glycogen synthase kinase 3β (GSK-3β) and extracellular regulated protein kinases (ERK) signalling pathways. Moreover, TCP1 knock-in (TCP1-KI) dramatically promoted the occurrence of diethylnitrosamine (DEN) -induced primary HCC in mice. Our results highlight the critical role of TCP1 in HCC and PDAC and reveal a novel mechanism to suppress HCC and PDAC by targeting c-Myc via the TCP1-induced promotion of the AKT/GSK-3β and ERK signalling pathways. TCP1 is able to modulate the stability of target proteins by multiple pathways, thus promoting the progression of HCC and PDAC. Our study identifies TCP1 as a prognostic novel marker and therapeutic target of HCC and PDAC.
Collapse
Affiliation(s)
- Hekun Liu
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Linying Chen
- Department of Pathology, the First Affiliated Hospital of Fujian Medical University, No. 20, Chazhong Road, 350005, Fuzhou, Fujian, China
| | - Yuwen Chen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Yiyi Jin
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Xiance Chen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Nengjun Ma
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Fan Yang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Huixia Bi
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Xinxin Wen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Shenmin Xu
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Juan Chen
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Yanping Lin
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, 350122, Fuzhou, Fujian, China
| | - Yinghong Yang
- Department of Pathology, Fujian Medical University Union Hospital, No. 29, Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Yong Wu
- Fujian Institute of Haematology, Fujian Key Laboratory on Haematology, Fujian Medical University Union Hospital, No. 29, Xinquan Road, 350001, Fuzhou, Fujian, China.
| | - Yuanzhong Chen
- Fujian Institute of Haematology, Fujian Key Laboratory on Haematology, Fujian Medical University Union Hospital, No. 29, Xinquan Road, 350001, Fuzhou, Fujian, China.
| |
Collapse
|
2
|
Mehta RK, Tan M, Hassan MK, Zhao T, Markovitz DM, Lawrence TS, Nyati MK. Investigating NanoLuc-EGFR engineered cell lines for real-time monitoring of EGFR protein dynamics in live cells. Biochem Biophys Res Commun 2024; 733:150711. [PMID: 39312880 DOI: 10.1016/j.bbrc.2024.150711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/04/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024]
Abstract
Evaluating the steady-state protein level of the EGFR in live cells presents significant challenges compared to measuring its kinase activity. Traditional testing methods, such as immunoblotting, ELISA, and immunofluorescence assays, are generally restricted to fixed cells or cell lysates. Despite their utility, these methods are cumbersome and provide only intermittent snapshots of EGFR levels at specific time points. With emerging trends in drug development shifting toward engineering novel agents that promote protein degradation, rather than simply inhibiting kinase activity, a tool that enables real-time, quantitative detection of drug effects in live cells could catalyze advances in the field. Such an innovation would expedite the drug development process, enhancing the translation of research findings into effective, patient-centered therapies. The NanoLuc-EGFR cell line, created through CRISPR genome editing, allows for the continuous tracking and analysis of EGFR protein levels and their degradation within live cells. This approach provides quantitative monitoring of protein dynamics in real time, offering insights that go beyond absolute protein levels to include aspects such as protein stability and degradation rate. Using this cell line model, we observed that AT13387 and H84T BanLec induce EGFR degradation in A549-HiBiT cells, with the results confirmed by immunoblotting. In contrast, Erlotinib, Osimertinib, and Cetuximab inhibit EGFR phosphorylation without altering total EGFR levels, as validated by the HiBiT luciferase assay. The NanoLuc-EGFR cell line marks a significant advancement in understanding protein regulation and serves as an instrumental platform for investigating targeted therapies that modulate protein kinases, especially those that induce protein degradation.
Collapse
Affiliation(s)
- Ranjit K Mehta
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mingjia Tan
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mohammed K Hassan
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Tengda Zhao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - David M Markovitz
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mukesh K Nyati
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
3
|
Zhang X, Hou J, Zhou G, Wang H, Wu Z. zDHHC3-mediated S-palmitoylation of SLC9A2 regulates apoptosis in kidney clear cell carcinoma. J Cancer Res Clin Oncol 2024; 150:194. [PMID: 38619631 PMCID: PMC11018659 DOI: 10.1007/s00432-024-05737-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 04/01/2024] [Indexed: 04/16/2024]
Abstract
PURPOSE Kidney clear cell carcinoma (KIRC) has a poor prognosis, high morbidity and mortality rates, and high invasion and metastasis rate, and effective therapeutic targets are lacking. zDHHC3 has been implicated in various cancers, but its specific role in KIRC remains unclear. METHODS In this study, we performed a pan-cancer analysis, bioinformatics analysis, and cell experiment to detect the role of zDHHC3 in KIRC. RESULTS zDHHC3 was significantly down-regulated in KIRC, and that its high expression was associated with favorable patient outcomes. We identified 202 hub genes that were most relevant to high zDHHC3 expression and KIRC, and found that they were involved mainly in ion transport and renal cell carcinoma. Among these hub genes, SLC9A2 was identified as a downstream gene of zDHHC3. zDHHC3 suppression led to decreased expression and S-palmitoylation of SLC9A2, which further inhibited the apoptosis of Caki-2 cells. CONCLUSION Our findings suggest that zDHHC3 plays an important role in KIRC, due partly to its regulation of SLC9A2 S-palmitoylation. The targeting of the zDHHC3-SLC9A2 axis may provide a new option for the clinical treatment of KIRC.
Collapse
Affiliation(s)
- Xiuyun Zhang
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, 430000, China
| | - Junpeng Hou
- Department of Orthopedic Surgery, Zhengzhou Central Hospital Affiliated With Zhengzhou University, Zhengzhou, 450000, China
| | - Guangyuan Zhou
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Haixia Wang
- School of Medicine, Shihezi University, Shihezi, 832003, Xinjiang Province, China
| | - Zeang Wu
- First Affiliated Hospital of Shihezi University, Dr. Zeang Wu, 107 North Second Road, Shihezi, 832003, Xinjiang Province, China.
| |
Collapse
|
4
|
Xu Y, Xing Z, Abdalla Ibrahim Suliman R, Liu Z, Tang F. Ferroptosis in liver cancer: a key role of post-translational modifications. Front Immunol 2024; 15:1375589. [PMID: 38650929 PMCID: PMC11033738 DOI: 10.3389/fimmu.2024.1375589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/26/2024] [Indexed: 04/25/2024] Open
Abstract
Ferroptosis is an emerging form of regulated cell death in an oxidative stress- and iron-dependent manner, primarily induced by the over-production of reactive oxygen species (ROS). Manipulation of ferroptosis has been considered a promising therapeutic approach to inhibit liver tumor growth. Nevertheless, the development of resistance to ferroptosis in liver cancer poses a significant challenge in cancer treatment. Post-translational modifications (PTMs) are crucial enzymatic catalytic reactions that covalently regulate protein conformation, stability and cellular activities. Additionally, PTMs play pivotal roles in various biological processes and divergent programmed cell death, including ferroptosis. Importantly, key PTMs regulators involved in ferroptosis have been identified as potential targets for cancer therapy. PTMs function of two proteins, SLC7A11, GPX4 involved in ferroptosis resistance have been extensively investigated in recent years. This review will summarize the roles of PTMs in ferroptosis-related proteins in hepatocellular carcinoma (HCC) treatment.
Collapse
Affiliation(s)
- Ying Xu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Zhiyao Xing
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | | | - Zichuan Liu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, China
| | - Fengyuan Tang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
- Thinking Biomed (Beijing) Co., Ltd, Beijing Economic and Technological Development Zone, Beijing, China
| |
Collapse
|
5
|
Zhang L, Xu J, Li M, Chen X. The role of long noncoding RNAs in liquid-liquid phase separation. Cell Signal 2023; 111:110848. [PMID: 37557974 DOI: 10.1016/j.cellsig.2023.110848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/03/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023]
Abstract
Long noncoding RNAs (lncRNAs), which are among the most well-characterized noncoding RNAs, have attracted much attention due to their regulatory functions and potential therapeutic options in many types of disease. Liquid-liquid phase separation (LLPS), the formation of droplet condensates, is involved in various cellular processes, but the molecular interactions of lncRNAs in LLPS are unclear. In this review, we describe the research development on LLPS, including descriptions of various methods established to identify LLPS, summarize the physiological and pathological functions of LLPS, identify the molecular interactions of lncRNAs in LLPS, and present the potential applications of leveraging LLPS in the clinic. The aim of this review is to update the knowledge on the association between LLPS and lncRNAs, which might provide a new direction for the treatment of LLPS-mediated disease.
Collapse
Affiliation(s)
- Le Zhang
- Center for Reproductive Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia, China
| | - Jinjin Xu
- Department of Imaging Medicine, The People's Hospital of the Inner Mongolia Autonomous Region, Hohhot 010017, Inner Mongolia, China
| | - Muxuan Li
- The First Clinical Medical College of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia, China
| | - Xiujuan Chen
- Center for Reproductive Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia, China.
| |
Collapse
|
6
|
EAPB0503, an Imidazoquinoxaline Derivative Modulates SENP3/ARF Mediated SUMOylation, and Induces NPM1c Degradation in NPM1 Mutant AML. Int J Mol Sci 2022; 23:ijms23073421. [PMID: 35408798 PMCID: PMC8998649 DOI: 10.3390/ijms23073421] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/14/2022] [Accepted: 03/18/2022] [Indexed: 12/14/2022] Open
Abstract
Nucleophosmin-1 (NPM1) is a pleiotropic protein involved in numerous cellular processes. NPM1 shuttles between the nucleus and the cytoplasm, but exhibits a predominant nucleolar localization, where its fate and functions are exquisitely controlled by dynamic post-translational modifications (PTM). Sentrin/SUMO Specific Peptidase 3 (SENP3) and ARF are two nucleolar proteins involved in NPM1 PTMs. SENP3 antagonizes ARF-mediated NPM1 SUMOylation, to promote ribosomal biogenesis. In Acute Myeloid Leukemia (AML), NPM1 is frequently mutated, and exhibits an aberrant cytoplasmic localization (NPM1c). NPM1c mutations define a separate AML entity with good prognosis in some AML patients, rendering NPM1c as a potential therapeutic target. SENP3-mediated NPM1 de-SUMOylation induces resistance to therapy in NPM1c AML. Here, we demonstrate that the imidazoquinoxaline EAPB0503 prolongs the survival and results in selective reduction in the leukemia burden of NPM1c AML xenograft mice. Indeed, EAPB0503 selectively downregulates HDM2 expression and activates the p53 pathway in NPM1c expressing cells, resulting in apoptosis. Importantly, we unraveled that NPM1c expressing cells exhibit low basal levels of SUMOylation paralleled with high SENP3 and low ARF basal levels. EAPB0503 reverted these molecular players by inducing NPM1c SUMOylation and ubiquitylation, leading to its proteasomal degradation. EAPB0503-induced NPM1c SUMOylation is concurrent with SENP3 downregulation and ARF upregulation in NPM1c expressing cells. Collectively, these results provide a strong rationale for testing therapies modulating NPM1c post-translational modifications in the management of NPM1c AML.
Collapse
|
7
|
Post-translational modifications in tumor-associated carbonic anhydrases. Amino Acids 2021; 54:543-558. [PMID: 34436666 DOI: 10.1007/s00726-021-03063-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/05/2021] [Indexed: 12/31/2022]
Abstract
Human carbonic anhydrases IX (hCA IX) and XII (hCA XII) are two proteins associated with tumor formation and development. These enzymes have been largely investigated both from a biochemical and a functional point of view. However, limited data are currently available on the characterization of their post-translational modifications (PTMs) and the functional implication of these structural changes in the tumor environment. In this review, we summarize existing literature data on PTMs of hCA IX and hCA XII, such as disulphide bond formation, phosphorylation, O-/N-linked glycosylation, acetylation and ubiquitination, highlighting, when possible, their specific role in cancer pathological processes.
Collapse
|
8
|
Godugu K, Sudha T, Davis PJ, Mousa SA. Nano Diaminopropane tetrac and integrin αvβ3 expression in different cancer types: Anti-cancer efficacy and Safety. Cancer Treat Res Commun 2021; 28:100395. [PMID: 34034044 DOI: 10.1016/j.ctarc.2021.100395] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 01/01/2023]
Abstract
Integrins are a family of heterodimeric plasma membrane glycoproteins, which regulate tumor growth, angiogenesis, migration, and metastasis. Integrin αvβ3 has been recognized as a putative target for the treatment of several cancers. Thus, the characterization of αvβ3 distribution in different human tumors is of substantial interest in tumor targeting and its suppression. In this study we evaluated the expression of integrin αvβ3 in different cancer types to define the expression pattern in cancer model. Furthermore, we investigated the effect of novel αvβ3 antagonist Diaminopropane Tetraiodothyroacetic acid conjugated to poly (lactic-co-glycolic acid) polymer and its nanoformulated form (NDAT), on different cancer cell lines both in vitro and in xenografts. In vitro, NDAT downregulated αv and β3 monomer expression. In vivo in tumor xenografts, similarly, NDAT downregulated αv and β3. Distinct reduction in tumor weight and viability was observed in glioblastoma xenografts treated with NDAT. Furthermore, NDAT was safe and tolerable in mice treated with high doses. In conclusion, NDAT is an effective and safe inhibitor of integrin αvβ3 expression in various cancer types, which indicates its impact on the targetability and suppression of αvβ3-associated tumor functions.
Collapse
Affiliation(s)
- Kavitha Godugu
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, , 1 Discovery Drive, Rensselaer, NY, USA
| | - Thangirala Sudha
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, , 1 Discovery Drive, Rensselaer, NY, USA
| | - Paul J Davis
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, , 1 Discovery Drive, Rensselaer, NY, USA
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, , 1 Discovery Drive, Rensselaer, NY, USA.
| |
Collapse
|
9
|
Lin TY, Hua WJ, Yeh H, Tseng AJ. Functional proteomic analysis reveals that fungal immunomodulatory protein reduced expressions of heat shock proteins correlates to apoptosis in lung cancer cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 80:153384. [PMID: 33113507 DOI: 10.1016/j.phymed.2020.153384] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Ling Zhi-8 (LZ-8) and GMI are two fungal immunomodulatory proteins (FIPs) with a similar structure and amino acid sequence and are respectively obtained from the medicinal mushroom Ganoderma lucidum and Ganoderma microsporum. They present the anti-cancer progression and metastasis. We previously demonstrated that LZ-8 reduces the tumor progression in lung cancer LLC1 cell-bearing mouse. However, it is unclear whether these FIPs induce changes in the protein expression profile in cancer cells and the mechanism for such a process is not defined. PURPOSE This study determines the changes in the proteomic profile for tumor lesions of LLC1 cell-bearing mouse received with LZ-8 and the potential mechanism for FIPs in anti-lung cancer cells. METHODS The proteomic profile of tumor lesions was determined using two-dimensional electrophoresis and a LTQ-OrbitrapXL mass spectrometer (LC-MS/MS). The biological processes and the signaling pathway enrichment analysis were performed using Ingenuity Pathway Analysis (IPA). The differentially expressed proteins were verified by Western blot. Cell viability was determined by MTT assay. Cell morphology was characterized using electron microscopy. Migration was detected using the Transwell assay. The apoptotic response was determined using Western blot and flow cytometry. RESULTS Obtained results showed that 21 proteins in the tumor lesions exhibited differential (2-fold change, p < 0.05) expression between PBS and LZ-8 treatment groups. LZ-8-induced changes in the proteomic profile that may relate to protein degradation pathways. Specifically, three heat shock proteins (HSPs), HSP60, 70 and 90, were significantly downregulated in tumor lesions of LLC1-bearing mouse received with LZ-8. Both LZ-8 and GMI reduced the protein levels for these HSPs in lung cancer cells. Functional studies showed that they inhibited cell migration but effectively induced apoptotic response in LLC1 cells in vitro. In addition, the inhibitors of HSP60 and HSP70 effectively inhibited cell migration and decreased cell viability of LLC1 cells. CONCLUSIONS LZ-8 induced changes in the proteomic profile of tumor lesions which may regulate the HSPs-related cell viability. Moreover, inhibition of HSPs may be related to the anti-lung cancer activity.
Collapse
Affiliation(s)
- Tung-Yi Lin
- Institute of Traditional Medicine, National Yang-Ming University, Taipei, Taiwan; Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.
| | - Wei-Jyun Hua
- Institute of Traditional Medicine, National Yang-Ming University, Taipei, Taiwan; Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Hsin Yeh
- Institute of Traditional Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ai-Jung Tseng
- Institute of Traditional Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
10
|
Zhang H, Han W. Protein Post-translational Modifications in Head and Neck Cancer. Front Oncol 2020; 10:571944. [PMID: 33117703 PMCID: PMC7561398 DOI: 10.3389/fonc.2020.571944] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/11/2020] [Indexed: 12/20/2022] Open
Abstract
Head and neck cancer (HNC) is one of the most common malignant tumors worldwide, and is prone to tumor recurrence and metastasis. At present, surgery combined with radiotherapy and chemotherapy is still the conventional treatment modality for patients with HNC. However, for patients with relapse or metastasis of HNC, the treatment outcome is not ideal, and the prognosis is poor. Thus, it is crucial to deepen the understand of tumor mechanisms. Post-translational modifications (PTMs) refer to covalent binding of small chemical molecular groups to amino-acid side-chain of proteins. Post-translational modification is an important regulator of protein function, and as such, a current research hotspot of epigenetics. In recent years, it has been found that tumor occurrence is often accompanied by the abnormality of PTMs. Indeed, the abnormality play an important role in tumor development, and can be used as a target for tumor diagnosis and treatment. To date, several types of protein PTMs involved in the development of HNC have been reported. This paper reviews the relationship between HNC and several major protein PTMs, including acetylation, methylation, and glycosylation, in order to provide clues for the future application about PTMs in diagnosis and treatment of HNC.
Collapse
Affiliation(s)
- Hongbo Zhang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wei Han
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
11
|
Dong G, Chen L, Zhang J, Liu T, Du L, Sheng C, Li M. Discovery of Turn-On Fluorescent Probes for Detecting PDEδ Protein in Living Cells and Tumor Slices. Anal Chem 2020; 92:9516-9522. [PMID: 32571022 DOI: 10.1021/acs.analchem.0c00335] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The first small-molecule fluorescent turn-on probes for detecting PDEδ protein were rationally designed, showing reasonable fluorescent properties and the fluorescent ability has been applied for visualization of the PDEδ protein in living cells and at tissue levels. The qPCR results showed that the mRNA expression of KRAS, PDEδ, AKT1, MAPK1, MEK7, RAF1, and mTOR were downregulated by probes 1-3 through PI3K/AKT/mTOR and MAPK signal pathways. The probes also can downregulate the protein level of pErk and tErk. Therefore, these small-molecule fluorescent probes are expected to be used in the screening of antipancreatic cancer drugs targeting the PDEδ protein, as well as in obtaining a better understanding of the pathological and physiological roles of PDEδ protein.
Collapse
Affiliation(s)
- Gaopan Dong
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Long Chen
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Jing Zhang
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Tingting Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Institute of Pharmacology, School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, China
| | - Lupei Du
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Chunquan Sheng
- Department of Medicinal Chemistry, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Minyong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
12
|
He H, Liu B, Luo H, Zhang T, Jiang J. Big data and artificial intelligence discover novel drugs targeting proteins without 3D structure and overcome the undruggable targets. Stroke Vasc Neurol 2020; 5:381-387. [PMID: 33376199 PMCID: PMC7804061 DOI: 10.1136/svn-2019-000323] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 12/27/2022] Open
Abstract
The discovery of targeted drugs heavily relies on three-dimensional (3D) structures of target proteins. When the 3D structure of a protein target is unknown, it is very difficult to design its corresponding targeted drugs. Although the 3D structures of some proteins (the so-called undruggable targets) are known, their targeted drugs are still absent. As increasing crystal/cryogenic
electron microscopy structures are deposited in Protein Data Bank, it is much more possible to discover the targeted drugs. Moreover, it is also highly probable to turn previous undruggable targets into druggable ones when we identify their hidden allosteric sites. In this review, we focus on the currently available advanced methods for the discovery of novel compounds targeting proteins without 3D structure and how to turn undruggable targets into druggable ones.
Collapse
Affiliation(s)
- Huiqin He
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Benquan Liu
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Hongyi Luo
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Tingting Zhang
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Jingwei Jiang
- Institute of Pharmacologic Science, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
13
|
Xanthohumol, a Prenylated Flavonoid from Hops, Induces Caspase-Dependent Degradation of Oncoprotein BCR-ABL in K562 Cells. Antioxidants (Basel) 2019; 8:antiox8090402. [PMID: 31527518 PMCID: PMC6769755 DOI: 10.3390/antiox8090402] [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: 08/05/2019] [Revised: 09/10/2019] [Accepted: 09/13/2019] [Indexed: 12/11/2022] Open
Abstract
BCR-ABL oncoprotein drives the initiation, promotion, and progression of chronic myelogenous leukemia (CML). Tyrosine kinase inhibitors are the first choice for CML therapy, however, BCR-ABL mediated drug resistance limits its clinical application and prognosis. A novel promising therapeutic strategy for CML therapy is to degrade BCR-ABL using small molecules. Antioxidant xanthohumol (XN) is a hop-derived prenylated flavonoid with multiple bioactivities. In this study, we showed XN could inhibit the proliferation, induce S phase cell cycle arrest, and stimulate apoptosis in K562 cells. XN degraded BCR-ABL in a concentration- and time-dependent manner, and the involved degradation pathway was caspase activation, while not autophagy induction or ubiquitin proteasome system (UPS) activation. Moreover, we revealed for the first time that XN could inhibit the UPS and autophagy in K562 cells, and the inhibitory effect of XN on autophagy could attenuate imatinib-induced autophagy and enhance the therapeutic efficiency of imatinib in K562 cells. Our present findings identified XN act as a degrader of BCR-ABL in K562 cells, and XN had potential to be developed as an alternate agent for CML therapy.
Collapse
|
14
|
Dokla EME, Fang CS, Abouzid KAM, Chen CS. 1,2,4-Oxadiazole derivatives targeting EGFR and c-Met degradation in TKI resistant NSCLC. Eur J Med Chem 2019; 182:111607. [PMID: 31446247 DOI: 10.1016/j.ejmech.2019.111607] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/02/2019] [Accepted: 08/07/2019] [Indexed: 11/15/2022]
Abstract
Development of small-molecule agents with the ability to facilitate oncoprotein degradation has emerged as a promising strategy for cancer therapy. Since EGFR and c-Met are both implicated in oncogenesis and tumor progression, we initiated a screening program by using an in-house library to identify agents capable of inducing the concomitant suppression of EGFR and c-Met expression, which led to the identification of compound 1, a 1,2,4-oxadiazole derivative. Based on the scaffold of 1, we developed a series of derivatives to assess their efficacies in facilitating the downregulation of EGFR and c-Met, among which compound 48 represented the optimal agent. 48 showed equipotent antiproliferative activity against a panel of five NSCLC cell lines with different EGFR mutational status (IC50 = 0.2-0.6 μM), while the same panel exhibited differential sensitivity to different EGFR kinase inhibitors tested. Cell cycle analysis indicated that the antiproliferative activity of 48 was associated with its ability to cause G2/M arrest and, to a lesser extent, apoptosis. Western blot and RT-PCR analyses revealed that 48 facilitated the downregulation of EGFR and c-Met at the protein level. In vivo data showed that oral administration of 48 was effective in suppressing gefitinib-resistant H1975 xenograft tumor growth in nude mice, and at a suboptimal dose, could sensitize H1975 tumors to gefitinib. Based on these findings, 48 represents a promising candidate for further development to target EGFR TKI-resistant NSCLC via dual inhibition of EGFR and c-Met oncoproteins.
Collapse
Affiliation(s)
- Eman M E Dokla
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Chun-Sheng Fang
- Institute of New Drug Development, China Medical University, Taichung, 40402, Taiwan
| | - Khaled A M Abouzid
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt; Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Sadat City, Menoufia, Egypt.
| | - Ching S Chen
- Institute of New Drug Development, China Medical University, Taichung, 40402, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 40447, Taiwan.
| |
Collapse
|
15
|
Ray P, Tan YS, Somnay V, Mehta R, Sitto M, Ahsan A, Nyati S, Naughton JP, Bridges A, Zhao L, Rehemtulla A, Lawrence TS, Ray D, Nyati MK. Differential protein stability of EGFR mutants determines responsiveness to tyrosine kinase inhibitors. Oncotarget 2018; 7:68597-68613. [PMID: 27612423 PMCID: PMC5356576 DOI: 10.18632/oncotarget.11860] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 08/25/2016] [Indexed: 12/15/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) patients carrying specific EGFR kinase activating mutations (L858R, delE746-A750) respond well to tyrosine kinase inhibitors (TKIs). However, drug resistance develops within a year. In about 50% of such patients, acquired drug resistance is attributed to the enrichment of a constitutively active point mutation within the EGFR kinase domain (T790M). To date, differential drug-binding and altered ATP affinities by EGFR mutants have been shown to be responsible for differential TKI response. As it has been reported that EGFR stability plays a role in the survival of EGFR driven cancers, we hypothesized that differential TKI-induced receptor degradation between the sensitive L858R and delE746-A750 and the resistant T790M may also play a role in drug responsiveness. To explore this, we have utilized an EGFR-null CHO overexpression system as well as NSCLC cell lines expressing various EGFR mutants and determined the effects of erlotinib treatment. We found that erlotinib inhibits EGFR phosphorylation in both TKI sensitive and resistant cells, but the protein half-lives of L858R and delE746-A750 were significantly shorter than L858R/T790M. Third generation EGFR kinase inhibitor (AZD9291) inhibits the growth of L858R/T790M-EGFR driven cells and also induces EGFR degradation. Erlotinib treatment induced polyubiquitination and proteasomal degradation, primarily in a c-CBL-independent manner, in TKI sensitive L858R and delE746-A750 mutants when compared to the L858R/T790M mutant, which correlated with drug sensitivity. These data suggest an additional mechanism of TKI resistance, and we postulate that agents that degrade L858R/T790M-EGFR protein may overcome TKI resistance.
Collapse
Affiliation(s)
- Paramita Ray
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yee Sun Tan
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vishal Somnay
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ranjit Mehta
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Merna Sitto
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Aarif Ahsan
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA.,Current address: Oncology Research Unit East, Pfizer, Pearl River, NY 10965, USA
| | - Shyam Nyati
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - John P Naughton
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA.,Current address: Department of Otorhinolaryngology-Head and Neck Surgery, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10467, USA
| | - Alexander Bridges
- School of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lili Zhao
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alnawaz Rehemtulla
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dipankar Ray
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mukesh K Nyati
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
16
|
Wang J, Okkeri J, Pavic K, Wang Z, Kauko O, Halonen T, Sarek G, Ojala PM, Rao Z, Xu W, Westermarck J. Oncoprotein CIP2A is stabilized via interaction with tumor suppressor PP2A/B56. EMBO Rep 2017; 18:437-450. [PMID: 28174209 DOI: 10.15252/embr.201642788] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 12/20/2016] [Accepted: 01/09/2017] [Indexed: 01/20/2023] Open
Abstract
Protein phosphatase 2A (PP2A) is a critical human tumor suppressor. Cancerous inhibitor of PP2A (CIP2A) supports the activity of several critical cancer drivers (Akt, MYC, E2F1) and promotes malignancy in most cancer types via PP2A inhibition. However, the 3D structure of CIP2A has not been solved, and it remains enigmatic how it interacts with PP2A. Here, we show by yeast two-hybrid assays, and subsequent validation experiments, that CIP2A forms homodimers. The homodimerization of CIP2A is confirmed by solving the crystal structure of an N-terminal CIP2A fragment (amino acids 1-560) at 3.0 Å resolution, and by subsequent structure-based mutational analyses of the dimerization interface. We further describe that the CIP2A dimer interacts with the PP2A subunits B56α and B56γ. CIP2A binds to the B56 proteins via a conserved N-terminal region, and dimerization promotes B56 binding. Intriguingly, inhibition of either CIP2A dimerization or B56α/γ expression destabilizes CIP2A, indicating opportunities for controlled degradation. These results provide the first structure-function analysis of the interaction of CIP2A with PP2A/B56 and have direct implications for its targeting in cancer therapy.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Biological Structure, University of Washington, Seattle, WA, USA.,College of Life Sciences, Nankai University, Tianjin, China
| | - Juha Okkeri
- Turku Centre for Biotechnology, University of Turku, Turku, Finland.,Åbo Akademi University, Turku, Finland
| | - Karolina Pavic
- Turku Centre for Biotechnology, University of Turku, Turku, Finland.,Åbo Akademi University, Turku, Finland
| | - Zhizhi Wang
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Otto Kauko
- Turku Centre for Biotechnology, University of Turku, Turku, Finland.,Åbo Akademi University, Turku, Finland.,Department of Pathology, University of Turku, Turku, Finland
| | - Tuuli Halonen
- Turku Centre for Biotechnology, University of Turku, Turku, Finland.,Åbo Akademi University, Turku, Finland
| | - Grzegorz Sarek
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland
| | - Päivi M Ojala
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland
| | - Zihe Rao
- College of Life Sciences, Nankai University, Tianjin, China
| | - Wenqing Xu
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Jukka Westermarck
- Turku Centre for Biotechnology, University of Turku, Turku, Finland .,Åbo Akademi University, Turku, Finland.,Department of Pathology, University of Turku, Turku, Finland
| |
Collapse
|
17
|
Huang H, Weng H, Dong B, Zhao P, Zhou H, Qu L. Oridonin Triggers Chaperon-mediated Proteasomal Degradation of BCR-ABL in Leukemia. Sci Rep 2017; 7:41525. [PMID: 28128329 PMCID: PMC5270248 DOI: 10.1038/srep41525] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/20/2016] [Indexed: 12/30/2022] Open
Abstract
Inducing degradation of oncoproteins by small molecule compounds has the potential to avoid drug resistance and therefore deserves to be exploited for new therapies. Oridonin is a natural compound with promising antitumor efficacy that can trigger the degradation of oncoproteins; however, the direct cellular targets and underlying mechanisms remain unclear. Here we report that oridonin depletes BCR-ABL through chaperon-mediated proteasomal degradation in leukemia. Mechanistically, oridonin poses oxidative stress in cancer cells and directly binds to cysteines of HSF1, leading to the activation of this master regulator of the chaperone system. The resulting induction of HSP70 and ubiquitin proteins and the enhanced binding to CHIP E3 ligase hence target BCR-ABL for ubiquitin-proteasome degradation. Both wild-type and mutant forms of BCR-ABL can be efficiently degraded by oridonin, supporting its efficacy observed in cultured cells as well as mouse tumor xenograft assays with either imatinib-sensitive or -resistant cells. Collectively, our results identify a novel mechanism by which oridonin induces rapid degradation of BCR-ABL as well as a novel pharmaceutical activator of HSF1 that represents a promising treatment for leukemia.
Collapse
Affiliation(s)
- Huilin Huang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou 510275, China
| | - Hengyou Weng
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou 510275, China
| | - Bowen Dong
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou 510275, China
| | - Panpan Zhao
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou 510275, China
| | - Hui Zhou
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou 510275, China
| | - Lianghu Qu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
18
|
Xu SW, Law BYK, Mok SWF, Leung ELH, Fan XX, Coghi PS, Zeng W, Leung CH, Ma DL, Liu L, Wong VKW. Autophagic degradation of epidermal growth factor receptor in gefitinib-resistant lung cancer by celastrol. Int J Oncol 2016; 49:1576-88. [PMID: 27498688 DOI: 10.3892/ijo.2016.3644] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 06/30/2016] [Indexed: 12/09/2022] Open
Abstract
Drug resistance of non-small cell lung cancer (NSCLC) is highly correlated to the mutation of the epidermal growth factor receptor (EGFR). Although EGFR tyrosine kinase inhibitors (TKIs) are available clinically, the molecular complexity of NSCLC has made it necessary to search for alternative therapeutic approaches to overcome the drug resistance of NSCLC. In the present study, we identified a triterpene molecule derived from the herbal plant Tripterygium wilfordii, celastrol, as a novel autophagy inducer. We demonstrate that celastrol exhibited selective cytotoxic effect towards EGFR mutant NSCLCs. In addition, celastrol also facilitated the autophagic degradation of Hsp90 client protein including EGFR and Akt on both EGFR wild-type and mutant NSCLCs via calcium-mediated autophagy. Blockage of celastrol-induced autophagic degradation of EGFR by autophagic inhibitor or calcium chelator decreased celastrol-mediated cell death in gefitinib-resistant NSCLCs. Overall, our findings suggest that celastrol may be developed as an effective anticancer agent for treatment of gefitinib-resistant NSCLC in the future.
Collapse
Affiliation(s)
- Su-Wei Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Betty Yuen Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Simon Wing Fai Mok
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Elaine Lai Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Xing Xing Fan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Paolo Saul Coghi
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Wu Zeng
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, P.R. China
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Hong Kong, P.R. China
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Vincent Kam Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| |
Collapse
|