1
|
Abd Alla J, Quitterer U. The RAF Kinase Inhibitor Protein (RKIP): Good as Tumour Suppressor, Bad for the Heart. Cells 2022; 11:cells11040654. [PMID: 35203304 PMCID: PMC8869954 DOI: 10.3390/cells11040654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 02/04/2023] Open
Abstract
The RAF kinase inhibitor protein, RKIP, is a dual inhibitor of the RAF1 kinase and the G protein-coupled receptor kinase 2, GRK2. By inhibition of the RAF1-MAPK (mitogen-activated protein kinase) pathway, RKIP acts as a beneficial tumour suppressor. By inhibition of GRK2, RKIP counteracts GRK2-mediated desensitisation of G protein-coupled receptor (GPCR) signalling. GRK2 inhibition is considered to be cardioprotective under conditions of exaggerated GRK2 activity such as heart failure. However, cardioprotective GRK2 inhibition and pro-survival RAF1-MAPK pathway inhibition counteract each other, because inhibition of the pro-survival RAF1-MAPK cascade is detrimental for the heart. Therefore, the question arises, what is the net effect of these apparently divergent functions of RKIP in vivo? The available data show that, on one hand, GRK2 inhibition promotes cardioprotective signalling in isolated cardiomyocytes. On the other hand, inhibition of the pro-survival RAF1-MAPK pathway by RKIP deteriorates cardiomyocyte viability. In agreement with cardiotoxic effects, endogenous RKIP promotes cardiac fibrosis under conditions of cardiac stress, and transgenic RKIP induces heart dysfunction. Supported by next-generation sequencing (NGS) data of the RKIP-induced cardiac transcriptome, this review provides an overview of different RKIP functions and explains how beneficial GRK2 inhibition can go awry by RAF1-MAPK pathway inhibition. Based on RKIP studies, requirements for the development of a cardioprotective GRK2 inhibitor are deduced.
Collapse
Affiliation(s)
- Joshua Abd Alla
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland;
| | - Ursula Quitterer
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland;
- Department of Medicine, Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Correspondence: ; Tel.: +41-44-632-9801
| |
Collapse
|
2
|
Giovanini G, Barros LRC, Gama LR, Tortelli TC, Ramos AF. A Stochastic Binary Model for the Regulation of Gene Expression to Investigate Responses to Gene Therapy. Cancers (Basel) 2022; 14:633. [PMID: 35158901 PMCID: PMC8833822 DOI: 10.3390/cancers14030633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/08/2021] [Accepted: 11/13/2021] [Indexed: 02/07/2023] Open
Abstract
In this manuscript, we use an exactly solvable stochastic binary model for the regulation of gene expression to analyze the dynamics of response to a treatment aiming to modulate the number of transcripts of a master regulatory switching gene. The challenge is to combine multiple processes with different time scales to control the treatment response by a switching gene in an unavoidable noisy environment. To establish biologically relevant timescales for the parameters of the model, we select the RKIP gene and two non-specific drugs already known for changing RKIP levels in cancer cells. We demonstrate the usefulness of our method simulating three treatment scenarios aiming to reestablish RKIP gene expression dynamics toward a pre-cancerous state: (1) to increase the promoter's ON state duration; (2) to increase the mRNAs' synthesis rate; and (3) to increase both rates. We show that the pre-treatment kinetic rates of ON and OFF promoter switching speeds and mRNA synthesis and degradation will affect the heterogeneity and time for treatment response. Hence, we present a strategy for reaching increased average mRNA levels with diminished heterogeneity while reducing drug dosage by simultaneously targeting multiple kinetic rates that effectively represent the chemical processes underlying the regulation of gene expression. The decrease in heterogeneity of treatment response by a target gene helps to lower the chances of emergence of resistance. Our approach may be useful for inferring kinetic constants related to the expression of antimetastatic genes or oncogenes and for the design of multi-drug therapeutic strategies targeting the processes underpinning the expression of master regulatory genes.
Collapse
Affiliation(s)
- Guilherme Giovanini
- Escola de Artes, Ciências e Humanidades, Universidade de São Paulo, Av. Arlindo Béttio, 1000, São Paulo 03828-000, SP, Brazil;
| | - Luciana R. C. Barros
- Centro de Investigação Translacional em Oncologia, Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo, Instituto do Câncer do Estado de São Paulo, Av. Dr. Arnaldo, 251, São Paulo 01246-000, SP, Brazil; (L.R.C.B.); (L.R.G.); (T.C.T.J.)
| | - Leonardo R. Gama
- Centro de Investigação Translacional em Oncologia, Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo, Instituto do Câncer do Estado de São Paulo, Av. Dr. Arnaldo, 251, São Paulo 01246-000, SP, Brazil; (L.R.C.B.); (L.R.G.); (T.C.T.J.)
| | | | - Alexandre F. Ramos
- Escola de Artes, Ciências e Humanidades, Universidade de São Paulo, Av. Arlindo Béttio, 1000, São Paulo 03828-000, SP, Brazil;
- Centro de Investigação Translacional em Oncologia, Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo, Instituto do Câncer do Estado de São Paulo, Av. Dr. Arnaldo, 251, São Paulo 01246-000, SP, Brazil; (L.R.C.B.); (L.R.G.); (T.C.T.J.)
| |
Collapse
|
3
|
Huang Q, Wei J, Wei L, Zhang X, Bai F, Wen S, Wei Y, Tan S, Lu Z, Lin X. Retracted
: Role of RKIP in human hepatic stellate cell proliferation, invasion, and metastasis. J Cell Biochem 2018; 120:6168-6177. [DOI: 10.1002/jcb.27904] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 09/25/2018] [Indexed: 11/08/2022]
Affiliation(s)
- Quanfang Huang
- Department of Pharmacy, The First Affiliated Hospital of Guangxi University of Chinese Medicine Nanning Guangxi China
| | - Jinbin Wei
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University Nanning Guangxi China
| | - Ling Wei
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University Nanning Guangxi China
| | - Xiaolin Zhang
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University Nanning Guangxi China
| | - Facheng Bai
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University Nanning Guangxi China
| | - Shujuan Wen
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University Nanning Guangxi China
| | - Yuanyuan Wei
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University Nanning Guangxi China
| | - Shimei Tan
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University Nanning Guangxi China
| | - Zhongpeng Lu
- Department of Pharmacy, The First Affiliated Hospital of Guangxi University of Chinese Medicine Nanning Guangxi China
- Pharmaceutical College, University of Arkansas Medical School Little Rock Arkansas
| | - Xing Lin
- Life Sciences Institute and Pharmaceutical College, Guangxi Medical University Nanning Guangxi China
| |
Collapse
|
4
|
RKIP: A Key Regulator in Tumor Metastasis Initiation and Resistance to Apoptosis: Therapeutic Targeting and Impact. Cancers (Basel) 2018; 10:cancers10090287. [PMID: 30149591 PMCID: PMC6162400 DOI: 10.3390/cancers10090287] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/12/2018] [Accepted: 08/18/2018] [Indexed: 02/07/2023] Open
Abstract
RAF-kinase inhibitor protein (RKIP) is a well-established tumor suppressor that is frequently downregulated in a plethora of solid and hematological malignancies. RKIP exerts antimetastatic and pro-apoptotic properties in cancer cells, via modulation of signaling pathways and gene products involved in tumor survival and spread. Here we review the contribution of RKIP in the regulation of early metastatic steps such as epithelial–mesenchymal transition (EMT), migration, and invasion, as well as in tumor sensitivity to conventional therapeutics and immuno-mediated cytotoxicity. We further provide updated justification for targeting RKIP as a strategy to overcome tumor chemo/immuno-resistance and suppress metastasis, through the use of agents able to modulate RKIP expression in cancer cells.
Collapse
|
5
|
Xie SY, Li G, Han C, Yu YY, Li N. RKIP reduction enhances radioresistance by activating the Shh signaling pathway in non-small-cell lung cancer. Onco Targets Ther 2017; 10:5605-5619. [PMID: 29200875 PMCID: PMC5703172 DOI: 10.2147/ott.s149200] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) is exceptionally deadly because the tumors lack sensitive early-stage diagnostic biomarkers and are resistant to radiation and chemotherapy. Here, we investigated the role and mechanism of Raf kinase inhibitory protein (RKIP) in NSCLC radioresistance. The clinical data showed that the RKIP expression level was generally lower in radioresistant NSCLC tissues than in radiosensitive tissues. Reduced RKIP expression was related to NSCLC radioresistance and poor prognosis. In vitro experiments showed that RKIP knockdown increased radioresistance and metastatic ability in NSCLC cell lines. Mechanistically, RKIP reduction activated the Shh signaling pathway by derepressing Smoothened (Smo) and initiating glioma-associated oncogene-1 (Gli1)-mediated transcription in NSCLC. In addition, the inappropriately activated Shh–Gli1 signaling pathway then enhanced cancer stem cell (CSC) expression in the cell lines. The increasing quantity of CSCs in the tumor ultimately promotes the radiation resistance of NSCLC. Together, these results suggest that RKIP plays a vital role in radiation response and metastasis in NSCLC. RKIP reduction enhances radioresistance by activating the Shh signaling pathway and initiating functional CSCs. This role makes it a promising therapeutic target for improving the efficacy of NSCLC radiation treatment.
Collapse
Affiliation(s)
- Shi-Yang Xie
- Department of Radiation Oncology, The First Affiliated Hospital of Chi Medical University, Shenyang, China
| | - Guang Li
- Department of Radiation Oncology, The First Affiliated Hospital of Chi Medical University, Shenyang, China
| | - Chong Han
- Department of Radiation Oncology, The First Affiliated Hospital of Chi Medical University, Shenyang, China
| | - Yang-Yang Yu
- Department of Radiation Oncology, The First Affiliated Hospital of Chi Medical University, Shenyang, China
| | - Nan Li
- Department of Radiation Oncology, The First Affiliated Hospital of Chi Medical University, Shenyang, China
| |
Collapse
|
6
|
Lappas M. RKIP is decreased in laboring myometrium and modulates inflammation-induced pro-labor mediators. Reproduction 2017; 153:545-553. [PMID: 28280133 DOI: 10.1530/rep-17-0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/06/2017] [Accepted: 02/13/2017] [Indexed: 11/08/2022]
Abstract
Nuclear factor-kappa B (NF-κB)-induced inflammation plays a central role in the terminal process of human labor and delivery. Our previous studies show that IL1B induces NF-κB signaling through extracellular signal-regulated kinase (ERK; official gene symbol MAPK1), whereas TNF induces NF-κB-driven transcription of pro-labor mediators via an MAPK1-independent mechanism. Raf kinase inhibitor protein (RKIP) negatively regulates inflammation by inhibiting NF-κB activation directly or indirectly by inhibiting MAPK1. The role of RKIP in the processes of human labor and delivery is not known. The present study was performed to investigate the expression of RKIP in laboring and non-laboring human myometrium and determine the effect of siRNA knockdown of RKIP (siRKIP) on pro-labor mediators in human myometrial primary cells. Term labor was associated with a decrease in RKIP expression. Furthermore, RKIP expression was decreased in myometrial cells treated with IL1B and TNF, two likely factors contributing to preterm birth. The effect of siRKIP in primary myometrial cells was a significant augmentation of IL1B- and TNF-induced CXCL1 and CXCL8 mRNA abundance and secretion; PTGS2 mRNA levels and prostaglandin PGF2α release and MMP9 mRNA abundance and pro-MMP9 secretion. There was no effect of siRKIP on MAPK1 activation. On the other hand, RKIP knockdown was associated with increased activation of NF-κB RELA in the presence of IL1B and TNF. In conclusion, in human primary myometrial cells, RKIP negatively regulates IL1B- and TNF-induced expression and or secretion of pro-inflammatory and pro-labor mediators by inhibiting NF-κB RELA activation.
Collapse
Affiliation(s)
- Martha Lappas
- Mercy Perinatal Research CentreMercy Hospital for Women, Heidelberg, Victoria, Australia and Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne,Victoria, Australia
| |
Collapse
|
7
|
Tsao DA, Tseng WC, Chang HR. RKIP expression of liver and kidney after arsenic exposure. ENVIRONMENTAL TOXICOLOGY 2017; 32:1079-1082. [PMID: 27255443 DOI: 10.1002/tox.22291] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 04/28/2016] [Accepted: 05/06/2016] [Indexed: 06/05/2023]
Abstract
Arsenic is associated with cancers of kidney and liver. Raf kinase inhibitor protein (RKIP) has been identified as a member of a novel class of molecules that suppress the metastatic spread of tumors. In order to investigate the effect of arsenic to RKIP of liver and kidney, the expression of RKIP of liver and kidney with As (III) was explored in this study. Thirty male mice were chronically fed with 42.5 ppm, 85 ppm NaAsO2 and water for 180 days. The kidney and liver accumulation levels of As (III) in mice were determined by electro-thermal atomic absorption spectrometry. The method of RT-PCR, Western blotting analysis and immunohistochemistry were used to determine gene expression and protein expression of RKIP. The results showed that arsenic level was significantly increased in kidney and liver of As (III)-exposed mice as compared with control group. The gene expression and protein expression of RKIP was significantly decreased in kidney and liver of As (III)-exposed mice in comparison with these of control mice. These data suggested that RKIP decrease of liver and kidney with As (III) may be dangerous index in formation of cancer. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 1079-1082, 2017.
Collapse
Affiliation(s)
- Der-An Tsao
- Department of Medical Technology, Institute of Medical Technology, Fooyin University, Kaohsiung, Taiwan
| | - Wei-Chang Tseng
- Department of Medical Technology, Institute of Medical Technology, Fooyin University, Kaohsiung, Taiwan
| | - Huoy-Rou Chang
- Department of Biomedical Engineering, I-Shou University, Kaohsiung, Taiwan
| |
Collapse
|
8
|
Sánchez DI, González-Fernández B, San-Miguel B, de Urbina JO, Crespo I, González-Gallego J, Tuñón MJ. Melatonin prevents deregulation of the sphingosine kinase/sphingosine 1-phosphate signaling pathway in a mouse model of diethylnitrosamine-induced hepatocellular carcinoma. J Pineal Res 2017; 62. [PMID: 27696512 DOI: 10.1111/jpi.12369] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/27/2016] [Indexed: 02/06/2023]
Abstract
The sphingosine kinase (SphK)/sphingosine 1-phosphate (S1P) pathway is involved in multiple biological processes, including carcinogenesis. Melatonin shows beneficial effects in cell and animal models of hepatocellular carcinoma, but it is unknown if they are associated with the modulation of the SphK/S1P system, along with different downstream signaling pathways modified in cancer. We investigated the effects of melatonin in mice which received diethylnitrosamine (DEN) (35 mg/kg body weight i.p) once a week for 8 weeks. Melatonin was given at 5 or 10 mg/kg/day i.p. beginning 4 weeks after the onset of DEN administration and ending at the sacrifice time (10, 20, 30, or 40 weeks). Melatonin alleviated the distortion of normal hepatic architecture, lowered the incidence of preneoplastic/neoplastic lesions, and inhibited the expression of proliferative/cell cycle regulatory proteins (Ki67, PCNA, cyclin D1, cyclin E, CDK4, and CDK6). S1P levels and expression of SphK1, SphK2, and S1P receptors (S1PR1/S1PR3) were significantly elevated in DEN-treated mice. However, there was a decreased expression of S1P lyase. These effects were significantly abrogated in a time- and dose-dependent manner by melatonin, which also increased S1PR2 expression. Following DEN treatment, mice exhibited increased phosphorylation of PI3K, AKT, mTOR, STAT3, ERK, and p38, and a higher expression of NF-κB p50 and p65 subunits. Melatonin administration significantly inhibited those changes. Data obtained suggest a contribution of the SphK/S1P system and related signaling pathways to the protective effects of melatonin in hepatocarcinogenesis.
Collapse
Affiliation(s)
- Diana I Sánchez
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
| | | | | | | | - Irene Crespo
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), León, Spain
| | - Javier González-Gallego
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), León, Spain
| | - María J Tuñón
- Institute of Biomedicine (IBIOMED), University of León, León, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), León, Spain
| |
Collapse
|
9
|
Nitta H, Shimose T, Emi Y, Imamura T, Ohnishi K, Kusumoto T, Yamamoto M, Fukuzawa K, Takahashi I, Higashi H, Tsuji A, Akagi Y, Oki E, Maehara Y, Baba H. Expression of the anaphylatoxin C5a receptor in gastric cancer: implications for vascular invasion and patient outcomes. Med Oncol 2016; 33:118. [DOI: 10.1007/s12032-016-0834-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/24/2016] [Indexed: 02/06/2023]
|
10
|
Jia H, Yang Q, Wang T, Cao Y, Jiang QY, Ma HD, Sun HW, Hou MX, Yang YP, Feng F. Rhamnetin induces sensitization of hepatocellular carcinoma cells to a small molecular kinase inhibitor or chemotherapeutic agents. Biochim Biophys Acta Gen Subj 2016; 1860:1417-30. [PMID: 27091611 DOI: 10.1016/j.bbagen.2016.04.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/23/2016] [Accepted: 04/13/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND The rapid development of multi-drug resistance (MDR) process has hindered the effectiveness of advanced hepatocellular carcinoma (HCC) treatments. Notch-1 pathway, which mediates the stress-response, promotes cell survival, EMT (epithelial-mesenchymal transition) process and induces anti-apoptosis in cancer cells, would be a potential target for overcoming MDR process. This study investigated the potential application of rhamnetin, a specific inhibitor of Notch-1 pathway, in anti-tumor drug sensitization of HCC treatment. METHODS The expression of miR-34a, proteins belonging to Notch-1 signaling pathway or MDR-related proteins was detected by quantitative polymerase chain reaction (qPCR) and western blot assay. To identify whether rhamnetin induces the chemotherapeutic sensitization in HCC cells, the MTT-assays, flow cytometry, soft agar, trans-well and nude mice assays were performed. RESULTS The endogenous expression of miR-34a was significantly increased and the expression of Notch-1 and Survivin was downregulated after rhamnetin treatment. Treatment of rhamnetin also reduced the expression of MDR related proteins P-GP (P-glycoprotein) and BCRP (breast cancer resistance protein). Rhamnetin increased the susceptibility of HCC cells and especially HepG2/ADR, a MDR HCC cell line, to a small molecular kinase inhibitor sorafenib or chemotherapeutic drugs etoposide and paclitaxel. The IC(50) value of those drugs correspondingly decreased. CONCLUSIONS Together, our findings suggest that rhamnetin treatment may attenuate the MDR process in HCC cells. These findings may contribute to more effective strategies for HCC therapy. GENERAL SIGNIFICANCE Rhamnetin acts as a promising sensitizer to chemotherapy and may be a novel approach to overcome the MDR process of HCC.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/enzymology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Cell Survival/drug effects
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Etoposide/pharmacology
- Gene Expression Regulation, Neoplastic
- Hep G2 Cells
- Humans
- Inhibitory Concentration 50
- Liver Neoplasms/drug therapy
- Liver Neoplasms/enzymology
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Male
- Mice, SCID
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Niacinamide/analogs & derivatives
- Niacinamide/pharmacology
- Paclitaxel/pharmacology
- Phenylurea Compounds/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Quercetin/analogs & derivatives
- Quercetin/pharmacology
- Receptor, Notch1/antagonists & inhibitors
- Receptor, Notch1/metabolism
- Signal Transduction/drug effects
- Sorafenib
- Transfection
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Hui Jia
- Department of Pharmacy, General Hospital of Shenyang Military Area Command, Shenyang 110016, PR China
| | - Qian Yang
- Center of Therapeutic Research for Liver Cancer, The 302nd Hospital, Beijing 100039, PR China
| | - Tao Wang
- Institute of Toxicology and Pharmacology, Chinese Academy of Military Medical Sciences, Beijing 100850, PR China
| | - Yu Cao
- Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Qi-Yu Jiang
- Research and Technology Service Center, The 302nd Hospital, Beijing 100039, PR China
| | - Hong-da Ma
- Department of Pharmacy, General Hospital of Shenyang Military Area Command, Shenyang 110016, PR China
| | - Hui-Wei Sun
- Center of Therapeutic Research for Liver Cancer, The 302nd Hospital, Beijing 100039, PR China
| | - Ming-Xiao Hou
- Department of Pharmacy, General Hospital of Shenyang Military Area Command, Shenyang 110016, PR China.
| | - Yong-Ping Yang
- Center of Therapeutic Research for Liver Cancer, The 302nd Hospital, Beijing 100039, PR China.
| | - Fan Feng
- Department of Pharmacy, General Hospital of Shenyang Military Area Command, Shenyang 110016, PR China.
| |
Collapse
|