1
|
Odarenko KV, Sen’kova AV, Salomatina OV, Markov OV, Salakhutdinov NF, Zenkova MA, Markov AV. Soloxolone para-methylanilide effectively suppresses aggressive phenotype of glioblastoma cells including TGF-β1-induced glial-mesenchymal transition in vitro and inhibits growth of U87 glioblastoma xenografts in mice. Front Pharmacol 2024; 15:1428924. [PMID: 39135794 PMCID: PMC11317440 DOI: 10.3389/fphar.2024.1428924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024] Open
Abstract
Soloxolone amides are semisynthetic triterpenoids that can cross the blood-brain barrier and inhibit glioblastoma growth both in vitro and in vivo. Here we investigate the impact of these compounds on processes associated with glioblastoma invasiveness and therapy resistance. Screening of soloxolone amides against glioblastoma cells revealed the ability of compound 7 (soloxolone para-methylanilide) to inhibit transforming growth factor-beta 1 (TGF-β1)-induced glial-mesenchymal transition Compound 7 inhibited morphological changes, wound healing, transwell migration, and expression of mesenchymal markers (N-cadherin, fibronectin, Slug) in TGF-β1-induced U87 and U118 glioblastoma cells, while restoring their adhesiveness. Confocal microscopy and molecular docking showed that 7 reduced SMAD2/3 nuclear translocation probably by direct interaction with the TGF-β type I and type II receptors (TβRI/II). In addition, 7 suppressed stemness of glioblastoma cells as evidenced by inhibition of colony forming ability, spheroid growth, and aldehyde dehydrogenase (ALDH) activity. Furthermore, 7 exhibited a synergistic effect with temozolomide (TMZ) on glioblastoma cell viability. Using N-acetyl-L-cysteine (NAC) and flow cytometry analysis of Annexin V-FITC-, propidium iodide-, and DCFDA-stained cells, 7 was found to synergize the cytotoxicity of TMZ by inducing ROS-dependent apoptosis. Further in vivo studies showed that 7, alone or in combination with TMZ, effectively suppressed the growth of U87 xenograft tumors in mice. Thus, 7 demonstrated promising potential as a component of combination therapy for glioblastoma, reducing its invasiveness and increasing its sensitivity to chemotherapy.
Collapse
Affiliation(s)
- Kirill V. Odarenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Aleksandra V. Sen’kova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Oksana V. Salomatina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Oleg V. Markov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Nariman F. Salakhutdinov
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Andrey V. Markov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
2
|
Chen L, Gong J, Yong X, Li Y, Wang S. A review of typical biological activities of glycyrrhetinic acid and its derivatives. RSC Adv 2024; 14:6557-6597. [PMID: 38390501 PMCID: PMC10882267 DOI: 10.1039/d3ra08025k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/01/2024] [Indexed: 02/24/2024] Open
Abstract
Glycyrrhetinic acid, a triterpenoid compound primarily sourced from licorice root, exhibits noteworthy biological attributes, including anti-inflammatory, anti-tumor, antibacterial, antiviral, and antioxidant effects. Despite these commendable effects, its further advancement and application, especially in clinical use, have been hindered by its limited druggability, including challenges such as low solubility and bioavailability. To enhance its biological activity and pharmaceutical efficacy, numerous research studies focus on the structural modification, associated biological activity data, and underlying mechanisms of glycyrrhetinic acid and its derivatives. This review endeavors to systematically compile and organize glycyrrhetinic acid derivatives that have demonstrated outstanding biological activities over the preceding decade, delineating their molecular structures, biological effects, underlying mechanisms, and future prospects for assisting researchers in finding and designing novel glycyrrhetinic acid derivatives, foster the exploration of structure-activity relationships, and aid in the screening of potential candidate compounds.
Collapse
Affiliation(s)
- Liang Chen
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Pharmacy Hainan Medical University No. 3, XueYuan Road, LongHua District Haikou City Hainan Province 571199 China
| | - Jingwen Gong
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Pharmacy Hainan Medical University No. 3, XueYuan Road, LongHua District Haikou City Hainan Province 571199 China
| | - Xu Yong
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University Shanghai 200433 China
| | - Youbin Li
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Pharmacy Hainan Medical University No. 3, XueYuan Road, LongHua District Haikou City Hainan Province 571199 China
| | - Shuojin Wang
- Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Pharmacy Hainan Medical University No. 3, XueYuan Road, LongHua District Haikou City Hainan Province 571199 China
| |
Collapse
|
3
|
Done G, Ari F, Akgun O, Akgun H, Cevatemre B, Gençkal HM. The Mechanism for Anticancer and Apoptosis‐Inducing Properties of Cu(II) Complex with Quercetin and 1,10‐Phenanthroline. ChemistrySelect 2022. [DOI: 10.1002/slct.202203242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Gulseven Done
- Department of Biology Faculty of Science and Arts Bursa Uludag University 16059 Bursa Turkey
| | - Ferda Ari
- Department of Biology Faculty of Science and Arts Bursa Uludag University 16059 Bursa Turkey
| | - Oguzhan Akgun
- Department of Biology Faculty of Science and Arts Bursa Uludag University 16059 Bursa Turkey
| | - Halime Akgun
- Department of Biology Faculty of Science and Arts Bursa Uludag University 16059 Bursa Turkey
| | - Buse Cevatemre
- Koc University Research Center for Translational Medicine (KUTTAM) 34450 Istanbul Turkey
| | - Hasene Mutlu Gençkal
- Department of Chemistry Faculty of Science and Arts Bursa Uludag University 16059 Bursa Turkey
| |
Collapse
|
4
|
Markov AV, Ilyina AA, Salomatina OV, Sen’kova AV, Okhina AA, Rogachev AD, Salakhutdinov NF, Zenkova MA. Novel Soloxolone Amides as Potent Anti-Glioblastoma Candidates: Design, Synthesis, In Silico Analysis and Biological Activities In Vitro and In Vivo. Pharmaceuticals (Basel) 2022; 15:ph15050603. [PMID: 35631429 PMCID: PMC9145754 DOI: 10.3390/ph15050603] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 01/27/2023] Open
Abstract
The modification of natural or semisynthetic triterpenoids with amines can be explored as a promising strategy for improving their pharmacological properties. Here, we report the design and synthesis of 11 novel amide derivatives of soloxolone methyl (SM), a cyano enone-bearing derivative of 18βH-glycyrrhetinic acid. Analysis of their bioactivities in vitro and in silico revealed their high toxicity against a panel of tumor cells (average IC50(24 h) = 3.7 µM) and showed that the formation of amide moieties at the C-30 position of soloxolone did not enhance the cytotoxicity of derivatives toward tumor cells compared to SM, though it can impart an ability to pass across the blood–brain barrier. Further HPLC–MS/MS and mechanistic studies verified significant brain accumulation of hit compound 12 (soloxolone tryptamide) in a murine model and showed its high anti-glioblastoma potential. It was found that 12 induced ROS-dependent and autophagy-independent death of U87 and U118 glioblastoma cells via mitochondrial apoptosis and effectively blocked their clonogenicity, motility and capacity to form vessel-like structures. Further in vivo study demonstrated that intraperitoneal injection of 12 at a dosage of 20 mg/kg effectively inhibited the growth of U87 glioblastoma in a mouse xenograft model, reducing the proliferative potential of the tumor and leading to a depletion of collagen content and normalization of blood vessels in tumor tissue. The obtained results clearly demonstrate that 12 can be considered as a promising leading compound for drug development in glioblastoma treatment.
Collapse
Affiliation(s)
- Andrey V. Markov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.A.I.); (O.V.S.); (A.V.S.); (M.A.Z.)
- Correspondence: ; Tel.: +7-383-363-51-61
| | - Anna A. Ilyina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.A.I.); (O.V.S.); (A.V.S.); (M.A.Z.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia; (A.A.O.); (A.D.R.)
| | - Oksana V. Salomatina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.A.I.); (O.V.S.); (A.V.S.); (M.A.Z.)
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia;
| | - Aleksandra V. Sen’kova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.A.I.); (O.V.S.); (A.V.S.); (M.A.Z.)
| | - Alina A. Okhina
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia; (A.A.O.); (A.D.R.)
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia;
| | - Artem D. Rogachev
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia; (A.A.O.); (A.D.R.)
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia;
| | - Nariman F. Salakhutdinov
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia;
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.A.I.); (O.V.S.); (A.V.S.); (M.A.Z.)
| |
Collapse
|
5
|
Uncovering the anti-angiogenic effect of semisynthetic triterpenoid CDDO-Im on HUVECs by an integrated network pharmacology approach. Comput Biol Med 2021; 141:105034. [PMID: 34802714 DOI: 10.1016/j.compbiomed.2021.105034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/03/2021] [Accepted: 11/11/2021] [Indexed: 01/01/2023]
Abstract
AIM To reveal the molecular mechanism of anti-angiogenic activity of semisynthetic triterpenoid CDDO-Im. MATERIALS AND METHODS Using re-analysis of cDNA microarray data of CDDO-Im-treated human vascular endothelial cells (HUVECs) (GSE71622), functional annotation of revealed differentially expressed genes (DEGs) and analysis of their co-expression, the key processes induced by CDDO-Im in HUVECs were identified. Venn diagram analysis was further performed to reveal the common DEGs, i.e. genes both susceptible to CDDO-Im and involved in the regulation of angiogenesis. A list of probable protein targets of CDDO-Im was prepared based on Connectivity Map/cheminformatics analysis and chemical proteomics data, among which the proteins that were most associated with the angiogenesis-related regulome were identified. Finally, identified targets were validated by molecular docking and text mining approaches. KEY FINDINGS The effect of CDDO-Im in HUVECs can be divided into two main phases: the short early phase (0.5-3 h) with an acute FOXD1/CEBPA/JUNB-regulated pro-angiogenic response induced by xenobiotic stress, and the second anti-angiogenic step (6-24 h) with massive suppression of various angiogenesis-related processes, accompanied by the activation of cytoprotective mechanisms. Our analysis showed that the anti-angiogenic activity of CDDO-Im is mediated by its inhibition of the expression of PLAT, ETS1, A2M, SPAG9, RASGRP3, FBXO32, GCNT1 and HDGFRP3 and its direct interactions with EGFR, mTOR, NOS2, HSP90AA1, MDM2, SYK, IRF3, ATR and KIF14. SIGNIFICANCE Our findings provide valuable insights into the understanding of the molecular mechanisms of the anti-angiogenic activity of cyano enone-bearing triterpenoids and revealed a range of novel promising therapeutic targets to control pathological neovascularization.
Collapse
|
6
|
Erkisa M, Ari F, Büyükköroğlu G, Şenel B, Yilmaz VT, Ulukaya E. Preparation and Characterization of Palladium Derivate-Loaded Micelle Formulation in Vitro as an Innovative Therapy Option against Non-Small Cell Lung Cancer Cells. Chem Biodivers 2021; 18:e2100402. [PMID: 34370383 DOI: 10.1002/cbdv.202100402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/09/2021] [Indexed: 01/05/2023]
Abstract
Nanoparticles have been used in cancer treatments to target tumor and reduce side effects. In this study, we aimed to increase the effectiveness of palladium(II) complex [PdCl(terpy)](sac) ⋅ 2H2 O, which previously showed anticancer potential, by preparing the nanoparticle formulation. An inhalable micellar dispersion containing a palladium(II) complex (PdNP) was prepared and its physicochemical characteristics were evaluated using in vitro tests. Morphology, size and surface charges of particle and loading/encapsulation efficiency of PdNP were analyzed by scanning electron microscopy, zeta sizer and inductively coupled plasma mass spectrometry while aerosol properties of PdNP were measured by the next generation impactor. A549 and H1299 non-small lung cancer cell types were used for cytotoxicity using SRB and ATP assays. Fluorescent staining and M30 antigen assay were carried out for cell death evaluation. Apoptosis was confirmed by flow cytometry analyses. SEM, particle size, and zeta potential results showed the particles have inhalable properties. The amount of the palladium(II) complex loaded into the particles was quantified which indicated high encapsulation efficiencies (97 %). The micellar dispersion expected to reach the alveolar region and the brachial region was determined 35 % and 47 %, respectively. PdNP showed an anti-growth effect by increasing reactive oxygen species that is followed by the induction of mitochondria-dependent apoptosis that is evidenced by pyknotic nuclei and M30 antigen level increments and disruption of polarization of membrane in mitochondria (Δψm). The results show that PdNP might be a promising inhalable novel complex to be used in non-small cell lung cancer, which warrants animal studies in further.
Collapse
Affiliation(s)
- Merve Erkisa
- Bursa Uludag University, Science and Art Faculty, Department of Biology, 16059, Bursa, Turkey.,Istinye University, Molecular Cancer Research Center (ISUMKAM), 34010, Istanbul, Turkey
| | - Ferda Ari
- Bursa Uludag University, Science and Art Faculty, Department of Biology, 16059, Bursa, Turkey
| | - Gülay Büyükköroğlu
- Anadolu University, Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, 26470, Eskisehir, Turkey
| | - Behiye Şenel
- Anadolu University, Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, 26470, Eskisehir, Turkey
| | - Veysel Turan Yilmaz
- Bursa Uludag University, Science and Art Faculty, Department of Chemistry, 16059, Bursa, Turkey
| | - Engin Ulukaya
- Istinye University, School of Medicine, Department of Clinical Biochemistry, 34010, Istanbul, Turkey
| |
Collapse
|
7
|
Fu X, Zhao W, Li K, Zhou J, Chen X. Cryptotanshinone Inhibits the Growth of HCT116 Colorectal Cancer Cells Through Endoplasmic Reticulum Stress-Mediated Autophagy. Front Pharmacol 2021; 12:653232. [PMID: 34220498 PMCID: PMC8248532 DOI: 10.3389/fphar.2021.653232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 04/20/2021] [Indexed: 12/19/2022] Open
Abstract
Among cancers, colorectal cancer (CRC) has one of the highest annual incidence and death rates. Considering severe adverse reactions associated with classical chemotherapy medications, traditional Chinese medicines have become potential drug candidates. In the current study, the effects of cryptotanshinone (CPT), a major component of Salvia miltiorrhiza Bunge (Danshen) on CRC and underlying mechanism were explored. First of all, data from in vitro experiments and in vivo zebrafish models indicated that CPT selectively inhibited the growth and proliferation of HCT116 and SW620 cells while had little effect on SW480 cells. Secondly, both ER stress and autophagy were associated with CRC viability regulation. Interestingly, ER stress inhibitor and autophagy inhibitor merely alleviated cytotoxic effects on HCT116 cells in response to CPT stimulation, while have little effect on SW620 cells. The significance of apoptosis, autophagy and ER stress were verified by clinical data from CRC patients. In summary, the current study has revealed the anti-cancer effects of CPT in CRC by activating autophagy signaling mediated by ER stress. CPT is a promising drug candidate for CRC treatment.
Collapse
Affiliation(s)
- Xiaojing Fu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wenwen Zhao
- School of Basic Medicine, Qingdao University, Qingdao, China.,State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Kangkang Li
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jingyi Zhou
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xuehong Chen
- School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
8
|
张 梦, 杨 玉, 刘 敏, 梁 利, 罗 瑞, 尹 丹, 郭 风. [Estradiol activates ERK phosphorylation by binding to ERβ to inhibit proliferation and promote apoptosis of human chondrocytes]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:336-343. [PMID: 33849823 PMCID: PMC8075796 DOI: 10.12122/j.issn.1673-4254.2021.03.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To explore the effect of estradiol (E2) binding to its receptor ERβ on the proliferation and apoptosis of C28I2 cells. OBJECTIVE We cloned the sequence of ESR2 into a recombinant adenovirus plasmid (pAd-ESR2) and packaged the plasmid in HEK293 cells. Normal human chondrocyte C28I2 cells were transfected with Ad-ESR2 or small interfering RNA targeting ESR2-siRNA (ESR2-siRNA), and the effects of treatment with DMSO or E2 on the expression of the proteins associated with endoplasmic reticulum (ER) stress and cell apoptosis were determined using Western blotting. qRT-PCR was used to detect the expressions of proliferation-related marker genes, and an EdU kit and flow cytometry were used to assess cell proliferation and apoptosis. We also tested the effects of U0126 (an ERK pathway inhibitor) and E2, alone or in combination, on ER stress, apoptosis and the ERK signaling pathway in C28I2 cells infected with Ad-ESR2 using Western blotting. OBJECTIVE Overexpression of Ad-ESR2 in C28I2 cells significantly promoted the expressions of IRE1α, PERK, XBP1s, and cleaved caspase-12, inhibited proliferation related marker genes PCNA, cyclin B1, cyclin D1, and decreased the level of ERK phosphorylation following E2 treatment (all P < 0.05). Interference of ESR2 caused significant reduction in the expressions of ER stress-related proteins and apoptosis-related proteins, up-regulated the genes related to cell proliferation, and increased intracellular pERK/ERK ratio in C28I2 cells. The effect of E2 binding to ERβ, which promoted the expressions of ER stress associated proteins and apoptosis related proteins, was obviously antagonized by treatment of the cells with U0126. OBJECTIVE The binding of E2 to ERβ promotes ER stress and apoptosis in human chondrocytes by activating ERK pathway phosphorylation inhibit cell proliferation.
Collapse
Affiliation(s)
- 梦颖 张
- />重庆医科大学基础医学院细胞生物学与遗传学教研室,重庆 400016Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing 400016, China
| | - 玉有 杨
- />重庆医科大学基础医学院细胞生物学与遗传学教研室,重庆 400016Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing 400016, China
| | - 敏 刘
- />重庆医科大学基础医学院细胞生物学与遗传学教研室,重庆 400016Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing 400016, China
| | - 利 梁
- />重庆医科大学基础医学院细胞生物学与遗传学教研室,重庆 400016Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing 400016, China
| | - 瑞 罗
- />重庆医科大学基础医学院细胞生物学与遗传学教研室,重庆 400016Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing 400016, China
| | - 丹旸 尹
- />重庆医科大学基础医学院细胞生物学与遗传学教研室,重庆 400016Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing 400016, China
| | - 风劲 郭
- />重庆医科大学基础医学院细胞生物学与遗传学教研室,重庆 400016Department of Cell Biology and Genetics, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|