1
|
Salah RA, El-Derby AM, El-Gammal Z, Wadie B, Ahmed SM, Elshenawy SE, Magdy S, Salah A, Gabr M, Mohamed I, El-Badri N. Hepatocellular carcinoma patients serum modulates the regenerative capacities of adipose mesenchymal stromal cells. Heliyon 2024; 10:e24794. [PMID: 38333871 PMCID: PMC10850426 DOI: 10.1016/j.heliyon.2024.e24794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent cancers causing the highest mortality rate worldwide. Treatment options of surgery, radiation, cytotoxic drugs and liver transplantation suffer significant side effects and a high frequency of relapse. Stem cell therapy has been proposed as a new effective therapy, however, controversial reports are emerging on the role of mesenchymal stem cells in cancer. In this work, we aimed to assess the regenerative capacities of adipose mesenchymal stem cells when exposed to serum from HCC patients, by assessing the effect of the sera on modulating the regenerative capacities of h-AMSCs and the cancer properties in HCC cells. This will pave the way for maximizing the efficacy of MSCs in cancer therapy. Our data show that HCC serum-treated hA-MSCs suffered oncogene-induced senescence as shown by their altered morphology and ameliorated proliferation and differentiation. The cells were enlarged with small irregular nuclei, swollen rough endoplasmic reticulum cisternae, and aging lysosomes typified by dark residual bodies. HCC serum-treated Huh-7 cancer cells on the other hand displayed higher tumor aggressiveness as depicted by altered morphology, increased cellular proliferation and migration, and decreased percentage of early and late apoptotic cells. Our findings provide evidence that exposure of hA-MSCs to the serum of HCC patients decreases their regenerative capacities and should be considered when employed as a potential therapy in HCC patients.
Collapse
Affiliation(s)
- Radwa Ayman Salah
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Azza M. El-Derby
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Zaynab El-Gammal
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
- Stem Cells and Regenerative Medicine Department, Egypt Center for Research and Regenerative Medicine (ECRRM), Giza, 12578, Egypt
| | - Bishoy Wadie
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Sara M. Ahmed
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Shimaa E. Elshenawy
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
- Stem Cells and Regenerative Medicine Department, Egypt Center for Research and Regenerative Medicine (ECRRM), Giza, 12578, Egypt
| | - Shireen Magdy
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Ayman Salah
- Department of Hepatogastroenterology, Kasr El-Aini Cairo University, Cairo, Egypt
| | - Mahmoud Gabr
- Urology and Nephrology Center, Mansoura, 35516, Egypt
| | - Ihab Mohamed
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| |
Collapse
|
2
|
Liu Z, Tan X, Peng L, Gao W, Zeng P. Hederagenin Induces Apoptosis of Human Hepatoma HepG2 Cells via the Mitochondrial Pathway. Comb Chem High Throughput Screen 2024; 27:1495-1503. [PMID: 37817515 PMCID: PMC11327765 DOI: 10.2174/0113862073254353230925074944] [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: 04/04/2023] [Revised: 07/23/2023] [Accepted: 08/18/2023] [Indexed: 10/12/2023]
Abstract
OBJECTIVE The objective of this study is to assess the antitumor effects of hederagenin (HDG) in liver cancer (LC) cells and explore the related mechanisms. MATERIALS AND METHODS HepG2 cells were treated with HDG and cisplatin, respectively. The CCK8 assay was used to detect cell activity, DAPI staining was used to detect the proportion of living cells, TUNEL assay to detect the proportion of apoptotic cells, flow cytometry to detect the membrane potential, fluoroscopic electron microscopy to detect microstructural changes to the mitochondrial, and western blot analysis and high-content screening to detect apoptosisrelated proteins. RESULTS Treatment with HDG inhibited the growth of HepG2 cells, decreased the proportion of viable cells, increased the proportion of apoptotic cells, and significantly increased the proportion of cells in the G1 phase. Fluorescence staining showed that HDG damaged the mitochondria of HepG2 cells and significantly decreased the number of mitochondria. Flow cytometry showed that HDG decreased the mitochondrial membrane potential of HepG2 cells. Observations by electron microscopy showed that HDG caused swelling and vacuole formation of the mitochondria of HepG2 cells. HDG significantly reduced the average fluorescence intensity of Bcl-2 in HepG2 cells and significantly increased that of the pro-apoptosis proteins Bax, Cytochrome-c, and Caspase-3. CONCLUSION HDG induced apoptosis of HepG2 cells via the mitochondrial pathway.
Collapse
Affiliation(s)
- Zhuo Liu
- Affiliated Hospital, Hunan Academy of Traditional Chinese Medicine, Changsha 410006, P.R. China
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, P.R. China
| | - Xiaoning Tan
- Affiliated Hospital, Hunan Academy of Traditional Chinese Medicine, Changsha 410006, P.R. China
| | - Lian Peng
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, P.R. China
| | - Wenhui Gao
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, P.R. China
| | - Puhua Zeng
- Affiliated Hospital, Hunan Academy of Traditional Chinese Medicine, Changsha 410006, P.R. China
| |
Collapse
|
3
|
Wu J, Deng R, Yan J, Zhu B, Wang J, Xu Y, Gui S, Jin X, Lu X. A cell transmembrane peptide chimeric M(27-39)-HTPP targeted therapy for hepatocellular carcinoma. iScience 2023; 26:106766. [PMID: 37234089 PMCID: PMC10205784 DOI: 10.1016/j.isci.2023.106766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/09/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent malignant tumor, with a growing incidence and death rate worldwide. The aims and challenges of treating HCC include targeting the tumor, entering the tumor tissue, inhibiting the spread and growth of tumor cells. M27-39 is a small peptide isolated from the antimicrobial peptide Musca domestica cecropin (MDC), whereas HTPP is a liver-targeting, cell-penetrating peptide obtained from the circumsporozoite protein (CSP) of Plasmodium parasites. In this study, M27-39 was modified by HTPP to form M(27-39)-HTPP, which targeted tumor penetration to treat HCC. Here, we revealed that M(27-39)-HTPP had a good ability to target and penetrate the tumor, effectively limit the proliferation, migration, and invasion, and induce the apoptosis in HCC. Notably, M(27-39)-HTPP demonstrated good biosecurity when administered at therapeutic doses. Accordingly, M(27-39)-HTPP could be used as a new, safe, and efficient therapeutic peptide for HCC.
Collapse
Affiliation(s)
- Jibin Wu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou Higher Education Mega Center, Guangzhou 510006, People’s Republic of China
| | - Rui Deng
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou Higher Education Mega Center, Guangzhou 510006, People’s Republic of China
| | - Jianling Yan
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou Higher Education Mega Center, Guangzhou 510006, People’s Republic of China
| | - Baokang Zhu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou Higher Education Mega Center, Guangzhou 510006, People’s Republic of China
| | - Jian Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou Higher Education Mega Center, Guangzhou 510006, People’s Republic of China
| | - Yinghua Xu
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing 102629, People’s Republic of China
| | - Shuiqing Gui
- Intensive Care Unit, Shenzhen Second People’s Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen 518031, People’s Republic of China
| | - Xiaobao Jin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou Higher Education Mega Center, Guangzhou 510006, People’s Republic of China
| | - Xuemei Lu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou Higher Education Mega Center, Guangzhou 510006, People’s Republic of China
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, People’s Republic of China
| |
Collapse
|
4
|
Shen L, Li H, Liu R, Zhou C, Bretches M, Gong X, Lu L, Zhang Y, Zhao K, Ning B, Yang SY, Zhang A. DEPDC1 as a crucial factor in the progression of human osteosarcoma. Cancer Med 2023; 12:5798-5808. [PMID: 36479633 PMCID: PMC10028160 DOI: 10.1002/cam4.5340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/22/2022] [Accepted: 07/03/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Novel therapeutic strategies are emerging with the increased understanding of the underlying mechanisms of human osteosarcoma. This current study tends to decipher the potentially critical role of DEP domain-containing 1 (DEPDC1), a tumor-related gene, during the progression of osteosarcoma. METHODS Bioinformatics analysis of 25,035 genes from the National Center for Biotechnology Information (NCBI) databases was performed to screen differentially expressed genes between osteosarcoma and normal control groups, complemented by the examination of 85 clinical osteosarcoma specimens. Furthermore, the manipulation of DEPDC1 expression levels by using silencing RNA (siRNA) or lentiviral vector intervention on human osteosarcoma cells was performed to reveal its role and interactions in in vitro and in vivo settings. RESULTS Gene expression profile analysis and immunohistochemical (IHC) examination suggested that DEPDC1 is highly expressed in human osteosarcoma cells and tumor tissue. The silencing of DEPDC1 arrested osteosarcoma cell proliferation, promoted apoptosis, and ceased tumor metastasis. Studies involving clinical human osteosarcoma cases exhibited a strong correlation of DEPDC1 over-expressed osteosarcoma specimens with a reduced patient survival rate. CONCLUSIONS Collectively, this study demonstrated that DEPDC1 is a critical driver in the promotion of osteosarcoma progression and results in poor patient prognosis. Genetically targeting or pharmacologically inhibiting DEPDC1 may serve as a promising strategy for treating human osteosarcoma.
Collapse
Affiliation(s)
- Lin Shen
- Department of Orthopaedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Han Li
- Department of Endocrinology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Ronghan Liu
- Department of Orthopaedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chendan Zhou
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China
| | - Morgan Bretches
- Department of Orthopaedic Surgery, University of Kansas School of Medicine-Wichita, Wichita, Kansas, USA
- Department of Biological Sciences, Wichita State University, Wichita, Kansas, USA
| | - Xuan Gong
- Department of Biological Sciences, Wichita State University, Wichita, Kansas, USA
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Laitong Lu
- Department of Orthopaedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ying Zhang
- Department of Orthopaedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Kai Zhao
- Department of Orthopaedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Bin Ning
- Department of Orthopaedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shang-You Yang
- Department of Orthopaedic Surgery, University of Kansas School of Medicine-Wichita, Wichita, Kansas, USA
- Department of Biological Sciences, Wichita State University, Wichita, Kansas, USA
| | - Aijun Zhang
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
5
|
Dahiya M, Dureja H. Sorafenib for hepatocellular carcinoma: potential molecular targets and resistance mechanisms. J Chemother 2021; 34:286-301. [PMID: 34291704 DOI: 10.1080/1120009x.2021.1955202] [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] [Indexed: 12/20/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most widespread typical therapy-resistant, unresectable type of malignant solid tumour with a high death rate constituting huge medical concern. Sorafenib is a small molecule oral multi-target kinase potent inhibitor that acts by suppressing/blocking the multiplication of the tumour cells, angiogenesis, and encouraging apoptosis of the tumour cells. Though, the precise mechanism of tumour cell death induction by sorafenib is yet under exploration. Furthermore, genetic heterogeneity plays a critical role in developing sorafenib resistance, which leads the way to identify the need for predictive biomarkers responsible for drug resistance. Therefore, it is essential to find out the fundamental resistance mechanisms to expand therapeutic plans. The authors summarize the molecular concepts of resistance, progression, potential molecular targets, HCC management therapies, and discussion on the advancements expected in the coming future, inclusive of biomarker-driven treatment strategies, which may provide the prospects to design innovative therapeutically targeted strategies for the HCC treatment and the clinical implementation of emerging targeted agents.
Collapse
Affiliation(s)
- Mandeep Dahiya
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| |
Collapse
|
6
|
Wang Y, Liu S, Chen Q, Ren Y, Li Z, Cao S. Novel small molecular inhibitor of Pit-Oct-Unc transcription factor 1 suppresses hepatocellular carcinoma cell proliferation. Life Sci 2021; 277:119521. [PMID: 33891940 DOI: 10.1016/j.lfs.2021.119521] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/08/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent fatal malignancies in the Chinese population, due to high rates of hepatitis virus infection. Molecular targeted drugs such as sorafenib are the anti-tumor agents of choice for HCC treatment, but their results are generally unsatisfactory. In the present study the use of Pit-Oct-Unc transcription factor 1 (OCT1/POU2F1) as a potential therapeutic target for HCC was investigated, and a novel small molecular inhibitor of OCT1 (SMIO-1) was designed and its therapeutic efficacy against HCC was assessed. OCT1 expression was higher in HCC specimens than in corresponding non-tumor tissues, and higher OCT1 was associated with poorer prognosis in advanced HCC patients undergoing sorafenib treatment. For the first time, the novel SMIO-1 was investigated in conjunction with OCT1 via molecular docking. Interaction between SMIO-1 and OCT1 was confirmed via OCT1 point mutation. Treatment with SMIO-1 repressed OCT1 transcription factor activation by disrupting the interaction between OCT1 and its cofactors. It also repressed the proliferation and metastasis of HCC cells, and inhibited proliferation-related and metastasis-related genes downstream of OCT1. Therefore, SMIO-1 is a promising strategy for HCC treatment.
Collapse
Affiliation(s)
- Yue Wang
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Number 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, People's Republic of China.
| | - Shuo Liu
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Number 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, People's Republic of China
| | - Qin Chen
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Number 44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, People's Republic of China
| | - Yixin Ren
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, Hubei Province, People's Republic of China
| | - Zhongxiang Li
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001, Liaoning Province, People's Republic of China.
| | - Shuang Cao
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430072, Hubei Province, People's Republic of China.
| |
Collapse
|
7
|
Ni CJ, Qin XS, Huang ZS. Role of Wnt/β-catenin signaling pathway in occurrence and development of hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2021; 29:190-196. [DOI: 10.11569/wcjd.v29.i4.190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Studies have shown that the occurrence and development of liver cancer are related to a variety of signaling pathways. The Wnt/β-catenin signaling pathway is involved in all stages of liver disease progression, from initial liver damage to inflammation, fibrosis, and cirrhosis, as well as the occurrence and progression of tumors. Abnormal Wnt/β-catenin signaling promotes the development and progression of different liver diseases, including cancer. This review introduces the activation, biological function, and regulatory mechanism of the Wnt/β-catenin signaling pathway, discusses the role of ngthis pathway in the occurrence and progression of liver cancer, and describes factors that can inhibit the Wnt/β-catenin signaling pathway, such as small molecule inhibitors, traditional Chinese medicine extracts, and microRNAs, with an aim to provide reference for the basic and clinical research of liver cancer.
Collapse
Affiliation(s)
- Cai-Ju Ni
- Graduate School of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Xiao-Shan Qin
- Graduate School of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China,Department of Gastroenterology, The Affiliated Hospital of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Zan-Song Huang
- Department of Gastroenterology, The Affiliated Hospital of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China,Guangxi Clinical Research Center for Hepatobiliary Diseases, Baise 533000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
8
|
Wang H, Wang R, Huang D, Li S, Gao B, Kang Z, Tang B, Xie J, Yan F, Liang R, Li H, Yan J. Homoharringtonine Exerts Anti-tumor Effects in Hepatocellular Carcinoma Through Activation of the Hippo Pathway. Front Pharmacol 2021; 12:592071. [PMID: 33716735 PMCID: PMC7943857 DOI: 10.3389/fphar.2021.592071] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/07/2021] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent subtype of liver cancer with a mortality rate of approximately 3–6/100,000 and is the third leading cause of cancer-related death worldwide. Although several small-molecule drugs have been developed for the treatment of HCC, the choice of an agent for patients who require systemic chemotherapy at an advanced stage is still limited. The Hippo pathway is an evolutionarily conserved tumor suppressive pathway commonly dysregulated in HCC, which makes it a promising target for anti-HCC therapies. Homoharringtonine (HHT) is an FDA-approved anti-leukemia drug with proven strong anti-tumor activity in solid tumors. In this study, we found that HHT could significantly inhibit HCC cell growth by suppressing cell proliferation and colony formation. Moreover, HHT repressed cell invasion and migration remarkably. Additionally, HHT induced cell cycle arrest at S phase and promoted apoptosis. Most importantly, we showed that HHT-induced apoptosis was a consequence of the Hippo pathway activation. Consistently, the MST1/2 inhibitor, XMU-MP-1, could restore cell viability and reverse HHT-induced cell apoptosis. Furthermore, in vivo results confirmed the tumor inhibitory effect of HHT. Taken together, our findings suggest that HHT is a potential alternative therapeutic agent for the treatment of HCC.
Collapse
Affiliation(s)
- Haina Wang
- Department of Hematology, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Second Hospital of Dalian Medical University, Dalian, China.,Diamond Bay Institute of Hematology, Second Hospital of Dalian Medical University, Dalian, China
| | - Rui Wang
- Department of Hematology, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Second Hospital of Dalian Medical University, Dalian, China
| | - Dan Huang
- Department of Hematology, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Second Hospital of Dalian Medical University, Dalian, China
| | - Sihan Li
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Beibei Gao
- Department of Hematology, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Second Hospital of Dalian Medical University, Dalian, China
| | - Zhijie Kang
- Department of Hematology, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Second Hospital of Dalian Medical University, Dalian, China
| | - Bo Tang
- Department of Hematology, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Second Hospital of Dalian Medical University, Dalian, China
| | - Jiajun Xie
- Department of Hematology, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Second Hospital of Dalian Medical University, Dalian, China
| | - Fanzhi Yan
- Department of Hematology, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Second Hospital of Dalian Medical University, Dalian, China
| | - Rui Liang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, Second Hospital of Dalian Medical University, Dalian, China
| | - Hua Li
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Jinsong Yan
- Department of Hematology, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Second Hospital of Dalian Medical University, Dalian, China.,Diamond Bay Institute of Hematology, Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
9
|
Han L, Lu Y, Wang X, Zhang S, Wang Y, Wu F, Zhang W, Wang X, Zhang L. Regulatory role and mechanism of the inhibition of the Mcl-1 pathway during apoptosis and polarization of H37Rv-infected macrophages. Medicine (Baltimore) 2020; 99:e22438. [PMID: 33080678 PMCID: PMC7572003 DOI: 10.1097/md.0000000000022438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Myeloid cell leukemia-1 (Mcl-1) plays an important role in the clearance of Mycobacterium tuberculosis (MTB) infection. It has the effect of anti-apoptosis, protecting macrophages that have engulfed pathogens and preventing pathogen clearance. Meanwhile, the MAPK signaling pathway plays a significant role in regulating Mcl-1 expression during tuberculosis infection. In the case of latent infection and active infection, the apoptosis and polarization of macrophages have a great influence during MTB infection, so we discussed the effect of Mcl-1 on apoptosis and polarization. Then, further discussed its mechanism. METHODS An infected RAW264.7 macrophage model was established to investigate the regulatory role and mechanism of the Mcl-1 pathway inhibition during apoptosis and polarization of H37Rv infection. First, Mcl-1 protein and mRNA was identified by western blotting and Real-Time Polymerase Chain Reaction (RT-PCR). RAW264.7 macrophage apoptosis was detected by flow cytometry. RT-PCR was utilized to detect Bax, Caspase-3, Cyt-c and Bcl-2 mRNA expression. Next, Then the expression levels of inflammation factors CD86, CD206, iNOS, Fizz1, IL-6, IL-10, TNF-α, and TGF-β was detected by ELISA. SEM was used to observe macrophages phenotype. Finally, Bax, Bcl-2 and Bcl-xl the expression was detected by western blotting. Confocal microscopy was used to analyze mitochondrial membrane potential using the JC-10 kit. RESULTS In this study, we found that inhibiting the Mcl-1 expression signaling pathway led to infection by different virulence Mycobacterium tuberculosis, as well as changes in Mcl-1 protein and mRNA expression. Concomitantly macrophage apoptosis rate also changed, While, two phenotypic states of M1 and M2 appeared in the infected cells. We also found that the mitochondrial pathway was activated, the expression of its related genes Bax, casepase3, and Cyt-c, increased, whereas that of Bcl-2 decreased, and the mitochondrial membrane depolarization function was changed. CONCLUSIONS We found that Mcl-1 affected the apoptosis and polarization of macrophages infected by Mycobacterium tuberculosis, mainly M1 in the early stage and M2 in the later stage. In addition, mitochondria played a crucial role in this process.
Collapse
Affiliation(s)
- Ling Han
- Department of Pathophysiology, the Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University
| | - Yang Lu
- Department of Pathophysiology, the Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University
| | - Xiaofang Wang
- Department of Pathophysiology, the Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University
| | - Shujun Zhang
- Department of Pathophysiology, the Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University
| | - Yingzi Wang
- Department of Pathophysiology, the Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University
| | - Fang Wu
- Department of Pathophysiology, the Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University
| | - Wanjiang Zhang
- Department of Pathophysiology, the Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University
| | - Xinmin Wang
- Department of Urinary Surgery, The First Affiliated Hospital, Medical College of Shihezi University, Shihezi, Xinjiang, China
| | - Le Zhang
- Department of Pathophysiology, the Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University
| |
Collapse
|
10
|
Salimi A, Saboji M, Seydi E. Synergistic Effects of Ellagic Acid and Sorafenib on Hepatocytes and Mitochondria Isolated from a Hepatocellular Carcinoma Rat Model. Nutr Cancer 2020; 73:2460-2468. [DOI: 10.1080/01635581.2020.1829653] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Ahmad Salimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mojdeh Saboji
- Department of Biology, Plant Physiology, Faculty of Science, Payame Noor University, Tehran, Iran
| | - Enayatollah Seydi
- Department of Occupational Health and Safety Engineering, School of Health, Alborz University of Medical Sciences, Karaj, Iran
- Research Center for Health, Safety and Environment, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
11
|
Carnosic acid increases sorafenib-induced inhibition of ERK1/2 and STAT3 signaling which contributes to reduced cell proliferation and survival of hepatocellular carcinoma cells. Oncotarget 2020; 11:3129-3143. [PMID: 32913557 PMCID: PMC7443370 DOI: 10.18632/oncotarget.27687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/07/2020] [Indexed: 11/25/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has increasing worldwide incidence but when unresectable lacks curative options. Treatment with a kinase inhibitor Sorafenib (Sf), while initially effective, results in only short increases in patient survival, thus there is a need for improved treatment regimens. Numerous treatment regimens have been explored wherein Sf is combined with other agents, such as non-toxic botanicals like Curcumin or Silibinin. Recently, we have shown that carnosic acid (CA), a component of the food preservative Rosemary Extract, can markedly enhance the cytotoxic actions of Sf in several cell lines derived from HCC, but not in the cell line Hu1545 derived from normal hepatocytes. CA has been shown to enhance Sf-induced cell death in the neoplastic cell lines, principally due to the composite of increased apoptosis and cytotoxic autophagy. In the present study we focused on the mechanisms that underlie the reduced proliferation and survival of HCC cells when CA is added to Sf and how this relates to the increase in Sf-induced DNA damage as well as to the elevation of cytoplasmic levels of reactive oxygen species (ROS). Importantly, the elevation of ROS levels induced by Sf was increased by adding CA. We found that CA enhanced Sf-induced prolongation of cell cycle, and the overall decrease in cell growth was associated with reduced activation of both STAT3 transcription factor (TF) and extracellular signal-regulated protein kinase (Erk)1/2. Our data suggest that a regimen incorporating CA, an inexpensive and non-toxic food additive, in the treatment of advanced HCC merits clinical evaluation.
Collapse
|
12
|
Zhou C, Huang Y, Chen Y, Xie Y, Wen H, Tan W, Wang C. miR-602 Mediates the RASSF1A/JNK Pathway, Thereby Promoting Postoperative Recurrence in Nude Mice with Liver Cancer. Onco Targets Ther 2020; 13:6767-6776. [PMID: 32764964 PMCID: PMC7368130 DOI: 10.2147/ott.s243651] [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: 12/24/2019] [Accepted: 06/19/2020] [Indexed: 12/25/2022] Open
Abstract
Purpose At present, there are few studies on the mechanisms underlying postoperative recurrence of liver cancer, and the mechanism of action of miR-602 in postoperative recurrence of liver tumors is not clear. Our goals were to investigate the effects of miR-602 on the expression of the Ras-associated domain family 1A (RASSF1A) gene and the regulation of primary and recurrent hepatic tumors to clarify the molecular mechanisms of miR-602 in postoperative hepatocellular carcinoma. Methods We constructed a mouse liver orthotopic tumor model and a mouse liver recurrent tumor model. We measured the expression levels of the RASSF1A gene and then analyzed the effects of miR-602 on the regulation of RASSF1A. We transiently transfected the miR-602 gene into cells that stably overexpressed RASSF1A and examined relevant indicators to elucidate the mechanisms by which miR-602 regulates the RASSF1A/c-Jun N-terminal kinase (JNK) pathway in recurrence and dormancy in liver cancer. Results RASSF1A expression was inversely related to that of JNK, activating transcription factor 2 (ATF-2), and c-Jun in SMMC7721 cells stably transfected with the RASSF1A gene and in recurrent mouse tumor tissues. After transient transfection of cells with miR-602 mimic or miR-602 inhibitor, the expression of miR-602 was inversely related to that of RASSF1A. Conclusion MiR-602 might inhibit the JNK signaling pathway by inhibiting the expression of RASSF1A, thereby promoting recurrence of liver cancer after surgery. The low expression levels of miR-602 in liver cancer tissues were closely related to postoperative recurrence; they could be used as a marker to judge the prognosis of patients with liver cancer.
Collapse
Affiliation(s)
- Cheng Zhou
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China.,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, People's Republic of China
| | - Yajing Huang
- Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Yongxu Chen
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China.,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, People's Republic of China
| | - Yingjie Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, People's Republic of China
| | - Huihong Wen
- Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Wei Tan
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, People's Republic of China
| | - Changjun Wang
- School of Medicine, South China University of Technology, Guangzhou, People's Republic of China.,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatric Institute, Guangzhou, People's Republic of China
| |
Collapse
|
13
|
Jakobi K, Beyer S, Koch A, Thomas D, Schwalm S, Zeuzem S, Pfeilschifter J, Grammatikos G. Sorafenib Treatment and Modulation of the Sphingolipid Pathway Affect Proliferation and Viability of Hepatocellular Carcinoma In Vitro. Int J Mol Sci 2020; 21:ijms21072409. [PMID: 32244391 PMCID: PMC7177910 DOI: 10.3390/ijms21072409] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) shows a remarkable heterogeneity and is recognized as a chemoresistant tumor with dismal prognosis. In previous studies, we observed significant alterations in the serum sphingolipids of patients with HCC. This study aimed to investigate the in vitro effects of sorafenib, which is the most widely used systemic HCC medication, on the sphingolipid pathway as well as the effects of inhibiting the sphingolipid pathway in HCC. Huh7.5 and HepG2 cells were stimulated with sorafenib, and inhibitors of the sphingolipid pathway and cell proliferation, viability, and concentrations of bioactive metabolites were assessed. We observed a significant downregulation of cell proliferation and viability and a simultaneous upregulation of dihydroceramides upon sorafenib stimulation. Interestingly, fumonisin B1 (FB1) and the general sphingosine kinase inhibitor SKI II were able to inhibit cell proliferation more prominently in HepG2 and Huh7.5 cells, whereas there were no consistent effects on the formation of dihydroceramides, thus implying an involvement of distinct metabolic pathways. In conclusion, our study demonstrates a significant downregulation of HCC proliferation upon sorafenib, FB1, and SKI II treatment, whereas it seems they exert antiproliferative effects independently from sphingolipids. Certainly, further data would be required to elucidate the potential of FB1 and SKI II as putative novel therapeutic targets in HCC.
Collapse
Affiliation(s)
- Katja Jakobi
- Medizinische Klinik 1, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (K.J.); (S.Z.)
- Institut für Allgemeine Pharmakologie und Toxikologie, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (S.B.); (A.K.); (S.S.); (J.P.)
| | - Sandra Beyer
- Institut für Allgemeine Pharmakologie und Toxikologie, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (S.B.); (A.K.); (S.S.); (J.P.)
| | - Alexander Koch
- Institut für Allgemeine Pharmakologie und Toxikologie, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (S.B.); (A.K.); (S.S.); (J.P.)
| | - Dominique Thomas
- Institut für Klinische Pharmakologie und Toxikologie, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany;
| | - Stephanie Schwalm
- Institut für Allgemeine Pharmakologie und Toxikologie, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (S.B.); (A.K.); (S.S.); (J.P.)
| | - Stefan Zeuzem
- Medizinische Klinik 1, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (K.J.); (S.Z.)
| | - Josef Pfeilschifter
- Institut für Allgemeine Pharmakologie und Toxikologie, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (S.B.); (A.K.); (S.S.); (J.P.)
| | - Georgios Grammatikos
- Medizinische Klinik 1, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (K.J.); (S.Z.)
- Institut für Allgemeine Pharmakologie und Toxikologie, University Hospital, Goethe University Frankfurt am Main, 60590 Frankfurt, Germany; (S.B.); (A.K.); (S.S.); (J.P.)
- St Luke’s Hospital, 55236 Thessaloniki, Greece
- Correspondence: ; Tel.: +30-2316-014-910
| |
Collapse
|
14
|
Ding X, Sun W, Chen J. Retraction Note to: IL‑2 augments the sorafenib‑induced apoptosis in liver cancer by promoting mitochondrial fission and activating the JNK/TAZ pathway. Cancer Cell Int 2020; 19:358. [PMID: 31892857 PMCID: PMC6936145 DOI: 10.1186/s12935-019-1083-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Xiaoyan Ding
- Cancer Center, Beijing Ditan Hospital, Capital Medical University, No 8, Jingshundong Street Chaoyang District, Beijing, 100015 China
| | - Wei Sun
- Cancer Center, Beijing Ditan Hospital, Capital Medical University, No 8, Jingshundong Street Chaoyang District, Beijing, 100015 China
| | - Jinglong Chen
- Cancer Center, Beijing Ditan Hospital, Capital Medical University, No 8, Jingshundong Street Chaoyang District, Beijing, 100015 China
| |
Collapse
|
15
|
Faraj Shaglouf LH, Ranjpour M, Wajid S, Jain SK. Elevated expression of cellular SYNE1, MMP10, and GTPase1 and their regulatory role in hepatocellular carcinoma progression. PROTOPLASMA 2020; 257:157-167. [PMID: 31428857 DOI: 10.1007/s00709-019-01423-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/19/2019] [Indexed: 06/10/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver malignancy resulting in high mortality. HCC progression is associated with abnormal signal transduction that changes cell signaling pathways and ultimately leads to dysregulation of cell functions and uncontrolled cell proliferation. Present study was undertaken with the objective to identify differentially expressed proteins and quantify their transcript expression in the liver of HCC-bearing rats vis-à-vis controls and to decipher the network involving interaction of genes coding for the characterized proteins to an insight into mechanism of HCC tumorigenesis. 2D-Electrophoresis and MALDI-TOF-MS/MS were used to characterize differentially expressed proteins in DEN (diethylnitrosamine)-induced HCC tissue using the protocol reported by us earlier. Real-time PCR was performed to quantify the expression of transcripts for the identified proteins. GENEMANIA, an interacting network of genes coding for selected proteins, was deciphered that provided the functional role of these proteins in HCC progression. Upregulation of proteins SYNE1, MMP10, and MTG1 was observed. The mRNA quantification revealed elevated expression of their transcripts at HCC initiation, progression, and tumor stages. Network analysis showed the involvement of the genes coding for these proteins in dysregulation of signaling pathways during HCC development. The elevated expression of SYNE1, MMP10, and MTG1 suggests the role of these proteins as potential players in HCC progression and tumorigenesis.
Collapse
Affiliation(s)
- Laila H Faraj Shaglouf
- Department of Biotechnology, School of Chemical and Life Science, Jamia Hamdard, New Delhi, 110062, India
| | - Maryam Ranjpour
- Department of Biotechnology, School of Chemical and Life Science, Jamia Hamdard, New Delhi, 110062, India
| | - Saima Wajid
- Department of Biotechnology, School of Chemical and Life Science, Jamia Hamdard, New Delhi, 110062, India
| | - Swatantra Kumar Jain
- Department of Biochemistry, Hamdard Institute of Medical Science and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
16
|
Wei L, Lun Y, Zhou X, He S, Gao L, Liu Y, He Z, Li B, Wang C. Novel urokinase-plasminogen activator inhibitor SPINK13 inhibits growth and metastasis of hepatocellular carcinoma in vivo. Pharmacol Res 2019; 143:73-85. [PMID: 30862605 DOI: 10.1016/j.phrs.2019.03.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/07/2019] [Accepted: 03/08/2019] [Indexed: 12/25/2022]
Abstract
Advanced hepatocellular carcinoma (HCC) is a highly aggressive malignancy that is a serious threat to the public health system of China. Urokinase-plasminogen activator (uPA) can promote the invasive growth and metastasis of HCC cells by activating matrix metalloproteinases (MMPs), leading to the breakage of the extra-cellular matrix. uPA is a promising target for advanced HCC treatment. In this stuy the expression of uPA was examined by quantitative polymerase chain reaction in hepatic cell lines. Protein interaction between uPA and SPINK13 was identified by immunoprecipitation. In vitro biochemical assay was used to examine the inhibitory effect of the SPINK13 on the direct cleaving of the recombinant pro-MMP9 by uPA. The antitumor effect of SPINK13 was examined by transwell assay or the nude mice tumor model.The expression of uPA was much higher in highly aggressive HCC cell lines than in lowly aggressive HCC cell lines or non-tumor hepatic cell lines. SPINK13 interacted with uPA in HCC cells and directly inhibited the cleaving of MMP9 by uPA. Treatment of the recombinant SPINK13 protein inhibited the invasion of HCC cells in several experiments, such as transwell experiments or the intrahepatic growth model. The results of the study indicated that SPINK13 could function as a promising therapeutic approach for patients with advanced HCC.
Collapse
Affiliation(s)
- Ling Wei
- Medical School of Chinese PLA & Department of Clinical Laboratory, Medical Laboratory Center, Chinese PLA General Hospital, Beijing 100853, PR China; Beijing Centre for Physical and Chemical Analysis, Beijing 100089, PR China
| | - Yongzhi Lun
- Department of Laboratory Medicine, School of Pharmacy and Medical Technology, Putian University, Putian 351100, Fujian Province, PR China
| | - Xiaoping Zhou
- Beijing Centre for Physical and Chemical Analysis, Beijing 100089, PR China
| | - Shang He
- Medical School of Chinese PLA & Department of Clinical Laboratory, Medical Laboratory Center, Chinese PLA General Hospital, Beijing 100853, PR China
| | - Lijuan Gao
- Beijing Centre for Physical and Chemical Analysis, Beijing 100089, PR China
| | - Yan Liu
- Beijing Centre for Physical and Chemical Analysis, Beijing 100089, PR China
| | - Zheng He
- Medical School of Chinese PLA & Department of Clinical Laboratory, Medical Laboratory Center, Chinese PLA General Hospital, Beijing 100853, PR China
| | - Baoming Li
- Beijing Centre for Physical and Chemical Analysis, Beijing 100089, PR China.
| | - Chengbin Wang
- Medical School of Chinese PLA & Department of Clinical Laboratory, Medical Laboratory Center, Chinese PLA General Hospital, Beijing 100853, PR China.
| |
Collapse
|