1
|
Liu Z, Liu Y, Kang X, Li L, Xiang Y. Subcellular Organelle Targeting as a Novel Approach to Combat Tumor Metastasis. Pharmaceutics 2025; 17:198. [PMID: 40006565 PMCID: PMC11859411 DOI: 10.3390/pharmaceutics17020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/28/2025] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor metastasis, the spread of cancer cells from the primary site to distant organs, remains a formidable challenge in oncology. Central to this process is the involvement of subcellular organelles, which undergo significant functional and structural changes during metastasis. Targeting these specific organelles offers a promising avenue for enhanced drug delivery and metastasis therapeutic efficacy. This precision increases the potency and reduces potential off-target effects. Moreover, by understanding the role of each organelle in metastasis, treatments can be designed to disrupt the metastatic process at multiple stages, from cell migration to the establishment of secondary tumors. This review delves deeply into tumor metastasis processes and their connection with subcellular organelles. In order to target these organelles, biomembranes, cell-penetrating peptides, localization signal peptides, aptamers, specific small molecules, and various other strategies have been developed. In this review, we will elucidate targeting delivery strategies for each subcellular organelle and look forward to prospects in this domain.
Collapse
Affiliation(s)
- Zefan Liu
- Department of General Surgery, First People‘s Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China; (Z.L.); (Y.L.)
| | - Yang Liu
- Department of General Surgery, First People‘s Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China; (Z.L.); (Y.L.)
| | - Xin Kang
- Department of General Surgery, First People‘s Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China; (Z.L.); (Y.L.)
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China;
| | - Yucheng Xiang
- School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| |
Collapse
|
2
|
Grit JL, McGee LE, Tovar EA, Essenburg CJ, Wolfrum E, Beddows I, Williams K, Sheridan RTC, Schipper JL, Adams M, Arumugam M, Vander Woude T, Gurunathan S, Field JM, Wulfkuhle J, Petricoin EF, Graveel CR, Steensma MR. p53 modulates kinase inhibitor resistance and lineage plasticity in NF1-related MPNSTs. Oncogene 2024; 43:1411-1430. [PMID: 38480916 PMCID: PMC11068581 DOI: 10.1038/s41388-024-03000-9] [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: 06/06/2023] [Revised: 02/16/2024] [Accepted: 03/01/2024] [Indexed: 05/05/2024]
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are chemotherapy resistant sarcomas that are a leading cause of death in neurofibromatosis type 1 (NF1). Although NF1-related MPNSTs derive from neural crest cell origin, they also exhibit intratumoral heterogeneity. TP53 mutations are associated with significantly decreased survival in MPNSTs, however the mechanisms underlying TP53-mediated therapy responses are unclear in the context of NF1-deficiency. We evaluated the role of two commonly altered genes, MET and TP53, in kinome reprograming and cellular differentiation in preclinical MPNST mouse models. We previously showed that MET amplification occurs early in human MPNST progression and that Trp53 loss abrogated MET-addiction resulting in MET inhibitor resistance. Here we demonstrate a novel mechanism of therapy resistance whereby p53 alters MET stability, localization, and downstream signaling leading to kinome reprogramming and lineage plasticity. Trp53 loss also resulted in a shift from RAS/ERK to AKT signaling and enhanced sensitivity to MEK and mTOR inhibition. In response to MET, MEK and mTOR inhibition, we observed broad and heterogeneous activation of key differentiation genes in Trp53-deficient lines suggesting Trp53 loss also impacts lineage plasticity in MPNSTs. These results demonstrate the mechanisms by which p53 loss alters MET dependency and therapy resistance in MPNSTS through kinome reprogramming and phenotypic flexibility.
Collapse
Affiliation(s)
- Jamie L Grit
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Lauren E McGee
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Elizabeth A Tovar
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Curt J Essenburg
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Emily Wolfrum
- Bioinformatics & Biostatistics Core, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Ian Beddows
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Kaitlin Williams
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | | | - Joshua L Schipper
- Flow Cytometry Core, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Marie Adams
- Genomics Core, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Menusha Arumugam
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Thomas Vander Woude
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Sharavana Gurunathan
- Department of Pharmacology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Jeffrey M Field
- Department of Pharmacology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Julia Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 20110, USA
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 20110, USA
| | - Carrie R Graveel
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Matthew R Steensma
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA.
- Helen DeVos Children's Hospital, Corewell Health System, Grand Rapids, MI, 49503, USA.
- Michigan State University College of Human Medicine, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
3
|
Choi D, Gonzalez‐Suarez AM, Dumbrava MG, Medlyn M, de Hoyos‐Vega JM, Cichocki F, Miller JS, Ding L, Zhu M, Stybayeva G, Gaspar‐Maia A, Billadeau DD, Ma WW, Revzin A. Microfluidic Organoid Cultures Derived from Pancreatic Cancer Biopsies for Personalized Testing of Chemotherapy and Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303088. [PMID: 38018486 PMCID: PMC10837378 DOI: 10.1002/advs.202303088] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 10/17/2023] [Indexed: 11/30/2023]
Abstract
Patient-derived cancer organoids (PDOs) hold considerable promise for personalizing therapy selection and improving patient outcomes. However, it is challenging to generate PDOs in sufficient numbers to test therapies in standard culture platforms. This challenge is particularly acute for pancreatic ductal adenocarcinoma (PDAC) where most patients are diagnosed at an advanced stage with non-resectable tumors and where patient tissue is in the form of needle biopsies. Here the development and characterization of microfluidic devices for testing therapies using a limited amount of tissue or PDOs available from PDAC biopsies is described. It is demonstrated that microfluidic PDOs are phenotypically and genotypically similar to the gold-standard Matrigel organoids with the advantages of 1) spheroid uniformity, 2) minimal cell number requirement, and 3) not relying on Matrigel. The utility of microfluidic PDOs is proven by testing PDO responses to several chemotherapies, including an inhibitor of glycogen synthase kinase (GSKI). In addition, microfluidic organoid cultures are used to test effectiveness of immunotherapy comprised of NK cells in combination with a novel biologic. In summary, our microfluidic device offers considerable benefits for personalizing oncology based on cancer biopsies and may, in the future, be developed into a companion diagnostic for chemotherapy or immunotherapy treatments.
Collapse
Affiliation(s)
- Daheui Choi
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMN55905USA
| | | | - Mihai G. Dumbrava
- Division of Experimental PathologyMayo ClinicRochesterMN55905USA
- Center for Individualized MedicineEpigenomics programMayo ClinicRochesterMN55905USA
| | - Michael Medlyn
- Division of Oncology ResearchCollege of MedicineMayo ClinicRochesterMN55905USA
| | | | - Frank Cichocki
- Department of MedicineUniversity of MinnesotaMinneapolisMN55455USA
| | | | - Li Ding
- Division of Oncology ResearchCollege of MedicineMayo ClinicRochesterMN55905USA
| | - Mojun Zhu
- Division of Medical OncologyMayo ClinicRochesterMN55905USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMN55905USA
| | - Alexandre Gaspar‐Maia
- Division of Experimental PathologyMayo ClinicRochesterMN55905USA
- Center for Individualized MedicineEpigenomics programMayo ClinicRochesterMN55905USA
| | - Daniel D. Billadeau
- Division of Oncology ResearchCollege of MedicineMayo ClinicRochesterMN55905USA
| | - Wen Wee Ma
- Division of Medical OncologyMayo ClinicRochesterMN55905USA
| | - Alexander Revzin
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMN55905USA
| |
Collapse
|
4
|
Zhang W, Chen T, Yang P, Li X, Zhu D, Su Z, Yang X, Jin R, Lan T, Guo H. Total flavonoids of Litchi chinensis Sonn. seed inhibit prostate cancer growth in bone by regulating the bone microenvironment via inactivation of the HGFR/NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117327. [PMID: 37871755 DOI: 10.1016/j.jep.2023.117327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/10/2023] [Accepted: 10/15/2023] [Indexed: 10/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Litchi chinensis Sonn. (Litchi) seed, a traditional Chinese medicine, is habitually used in the clinical treatment of prostate cancer (PCa)-induced bone pain. In our previous study, flavonoids have been identified as the active ingredient of litchi seed against PCa. However, its anti-tumor activities in bone and associated molecular mechanisms are still unclear. AIM OF THE STUDY To investigate the effects and underlying mechanisms of total flavonoids of litchi seed (TFLS) on the growth of PCa in bone. MATERIALS AND METHODS The effect of TFLS on the growth of PCa in bone was observed using a mouse model constructed with tibial injection of luciferase-expressing RM1-luc cells. Conditioned medium (CM) from bone marrow stromal cells OP9 and CM treated with TFLS (T-CM) was used to investigate the effect on the proliferation, colony formation, and apoptosis of PCa cells (LNCaP, PC3, RM1). An antibody microarray was performed to detect cytokine expression in the supernatant fraction of OP9 cell cultures treated with TFLS or left untreated. Western blot assay was employed to determine the expression and activity of HGFR and its key downstream proteins, Akt, mTOR, NF-κB, and Erk, in PCa cells. The potential target was further verified using immunofluorescence and immunohistochemistry assays. RESULTS Treatment with TFLS (80 mg/kg, 24 days) significantly suppressed the growth of RM1 cells in bone. CM from bone marrow stromal cells OP9 stimulated the proliferation and colony formation of the PCa cells as well as inhibited the apoptosis of PC3 cells, while T-CM reversed the effects mediated by OP9 cells in vitro. In an antibody array assay, TFLS regulated the majority of cytokines in OP9 cell culture supernatant, among which HGF, HGFR, IGF-1R, and PDGF-AA showed the greatest fold changes. Mechanistically, CM upregulated HGFR and promoted phosphorylation of NF-κB while T-CM induced reduction of HGFR and dephosphorylation of NF-κB in PC3 cells. Moreover, T-CM inhibited NF-κB entry into PC3 cell nuclei. Data from in vivo experiments further confirmed the inhibitory effects of TFLS on NF-κB. CONCLUSION TFLS suppresses the growth of PCa in bone through regulating bone microenvironment and the underlying mechanism potentially involves attenuation of the HGFR/NF-κB signaling axis.
Collapse
Affiliation(s)
- Weiquan Zhang
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & Pharmaceutical College, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China; Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Tao Chen
- Department of Orthopedics, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, 10 Huadong Road, Nanning, 530011, China
| | - Peilin Yang
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & Pharmaceutical College, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Xiaolan Li
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & Pharmaceutical College, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China; Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Dan Zhu
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & Pharmaceutical College, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Zhiheng Su
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & Pharmaceutical College, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Xin Yang
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China
| | - Ronghua Jin
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & Pharmaceutical College, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China.
| | - Taijin Lan
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, 179 Mingxiu Dong Road, Nanning, 530001, China.
| | - Hongwei Guo
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & Pharmaceutical College, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China; Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, 22 Shuangyong Road, Nanning, 530021, China.
| |
Collapse
|
5
|
Li Y, Chen L, Zheng Q, Liu G, Wang M, Wei S, Chen T. Lactate dehydrogenase A promotes nasopharyngeal carcinoma progression through the TAK1/NF-κB Axis. Mol Biol Rep 2024; 51:152. [PMID: 38236332 DOI: 10.1007/s11033-023-09130-9] [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: 05/11/2023] [Accepted: 12/07/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a malignant tumor that originates in the nasopharyngeal mucosa and is common in China and Southeast Asian countries. Cancer cells reprogram glycolytic metabolism to promote their growth, survival and metastasis. Glycolysis plays an important role in NPC development, but the underlying mechanisms remain incompletely elucidated. Lactate dehydrogenase A (LDHA) is a crucial glycolytic enzyme, catalyzing the last step of glycolysis. This study aims to investigate the exact role of LDHA, which catalyzes the conversion of pyruvate into lactate, in NPC development. METHODS AND RESULTS The western blot and immunohistochemical (IHC) results indicated that LDHA was significantly upregulated in NPC cells and clinical samples. LDHA knockdown by shRNA significantly inhibited NPC cell proliferation and invasion. Further knockdown of LDHA dramatically weakened the tumorigenicity of NPC cells in vivo. Mechanistic studies showed that LDHA activated TGF-β-activated kinase 1 (TAK1) and subsequent nuclear factor κB (NF-κB) signaling to promote NPC cell proliferation and invasion. Exogenous lactate supplementation restored NPC cell proliferation and invasion inhibited by LDHA knockdown, and this restorative effect was reversed by NF-κB inhibitor (BAY 11-7082) or TAK1 inhibitor (5Z-7-oxozeaenol) treatment. Moreover, clinical sample analyses showed that LDHA expression was positively correlated with TAK1 Thr187 phosphorylation and poor prognosis. CONCLUSIONS Our results suggest that LDHA and its major metabolite lactate drive NPC progression by regulating TAK1 and its downstream NF-κB signaling, which could become a therapeutic target in NPC.
Collapse
Affiliation(s)
- Yingzi Li
- State Key Laboratory of Respiratory Disease at People's Hospital of Yangjiang, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Yangjiang Key Laboratory of Respiratory Disease, Department of Respiratory Medicine, People's Hospital of Yangjiang, Yangjiang, 529500, Guangdong, China
| | - Lanfang Chen
- State Key Laboratory of Respiratory Disease at People's Hospital of Yangjiang, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Qiaochong Zheng
- Yangjiang Key Laboratory of Respiratory Disease, Department of Respiratory Medicine, People's Hospital of Yangjiang, Yangjiang, 529500, Guangdong, China
| | - Guanxin Liu
- State Key Laboratory of Respiratory Disease at People's Hospital of Yangjiang, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Mengjiao Wang
- Clinical Research Lab Center, Guizhou Provincial People's Hospital, Guizhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Shupei Wei
- State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, Guangdong, China.
| | - Tao Chen
- State Key Laboratory of Respiratory Disease at People's Hospital of Yangjiang, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
- Yangjiang Key Laboratory of Respiratory Disease, Department of Respiratory Medicine, People's Hospital of Yangjiang, Yangjiang, 529500, Guangdong, China.
| |
Collapse
|
6
|
Gupta R, Kadhim MM, Turki Jalil A, Obayes AM, Aminov Z, Alsaikhan F, Ramírez-Coronel AA, Ramaiah P, Tayyib NA, Luo X. Multifaceted role of NF-κB in hepatocellular carcinoma therapy: Molecular landscape, therapeutic compounds and nanomaterial approaches. ENVIRONMENTAL RESEARCH 2023; 228:115767. [PMID: 36966991 DOI: 10.1016/j.envres.2023.115767] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 05/16/2023]
Abstract
The predominant kind of liver cancer is hepatocellular carcinoma (HCC) that its treatment have been troublesome difficulties for physicians due to aggressive behavior of tumor cells in proliferation and metastasis. Moreover, stemness of HCC cells can result in tumor recurrence and angiogenesis occurs. Another problem is development of resistance to chemotherapy and radiotherapy in HCC cells. Genomic mutations participate in malignant behavior of HCC and nuclear factor-kappaB (NF-κB) has been one of the oncogenic factors in different human cancers that after nuclear translocation, it binds to promoter of genes in regulating their expression. Overexpression of NF-κB has been well-documented in increasing proliferation and invasion of tumor cells and notably, when its expression enhances, it induces chemoresistance and radio-resistance. Highlighting function of NF-κB in HCC can shed some light on the pathways regulating progression of tumor cells. The first aspect is proliferation acceleration and apoptosis inhibition in HCC cells mediated by enhancement in expression level of NF-κB. Moreover, NF-κB is able to enhance invasion of HCC cells via upregulation of MMPs and EMT, and it triggers angiogenesis as another step for increasing spread of tumor cells in tissues and organs. When NF-κB expression enhances, it stimulates chemoresistance and radio-resistance in HCC cells and by increasing stemness and population of cancer-stem cells, it can provide the way for recurrence of tumor. Overexpression of NF-κB mediates therapy resistance in HCC cells and it can be regulated by non-coding RNAs in HCC. Moreover, inhibition of NF-κB by anti-cancer and epigenetic drugs suppresses HCC tumorigenesis. More importantly, nanoparticles are considered for suppressing NF-κB axis in cancer and their prospectives and results can also be utilized for treatment of HCC. Nanomaterials are promising factors in treatment of HCC and by delivery of genes and drugs, they suppress HCC progression. Furthermore, nanomaterials provide phototherapy in HCC ablation.
Collapse
Affiliation(s)
- Reena Gupta
- Institute of Pharmaceutical Research, GLA University, District-Mathura, U. P., India
| | - Mustafa M Kadhim
- Department of Dentistry, Kut University College, Kut, Wasit, 52001, Iraq; Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad, 10022, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq.
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan; Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Ecuador; Epidemiology and Biostatistics Research Group, CES University, Colombia; Educational Statistics Research Group (GIEE), National University of Education, Ecuador
| | | | - Nahla A Tayyib
- Faculty of Nursing, Umm al- Qura University, Makkah, Saudi Arabia
| | - Xuanming Luo
- Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, 200031, China.
| |
Collapse
|
7
|
Xiong X, Song Q, Jing M, Yan W. Identification of PANoptosis-Based Prognostic Signature for Predicting Efficacy of Immunotherapy and Chemotherapy in Hepatocellular Carcinoma. Genet Res (Camb) 2023; 2023:6879022. [PMID: 37313428 PMCID: PMC10260314 DOI: 10.1155/2023/6879022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 06/15/2023] Open
Abstract
Background PANoptosis has been a research hotspot, but the role of PANoptosis in hepatocellular carcinoma (HCC) remains widely unknown. Drug resistance and low response rate are the main limitations of chemotherapy and immunotherapy in HCC. Thus, construction of a prognostic signature to predict prognosis and recognize ideal patients for corresponding chemotherapy and immunotherapy is necessary. Method The mRNA expression data of HCC patients was collected from TCGA database. Through LASSO and Cox regression, we developed a prognostic signature based on PANoptosis-related genes. KM analysis and ROC curve were implemented to evaluate the prognostic efficacy of this signature, and ICGC and GEO database were used as external validation cohorts. The immune cell infiltration, immune status, and IC50 of chemotherapeutic drugs were compared among different risk subgroups. The relationships between the signature and the efficacy of ICI therapy, sorafenib treatment, and transcatheter arterial chemoembolization (TACE) therapy were investigated. Result A 3-gene prognostic signature was constructed which divided the patients into low- and high-risk subgroups. Low-risk patients had better prognosis, and the risk score was proved to be an independent predictor of overall survival (OS), which had a well predictive effect. Patients in high-risk population had more immunosuppressive cells (Tregs, M0 macrophages, and MDSCs), higher TIDE score and TP53 mutation rate, and elevated activity of base excision repair (BER) pathways. Patients with low risk benefited more from ICI, TACE, and sorafenib therapy. The predictive value of the risk score was comparable with TIDE and MSI for OS under ICI therapy. The risk score could be a biomarker to predict the response to ICI, TACE, and sorafenib therapy. Conclusion The novel signature based on PANoptosis is a promising biomarker to distinguish the prognosis predict the benefit of ICI, TACE, and sorafenib therapy, and forecast the response to them.
Collapse
Affiliation(s)
- Xiaofeng Xiong
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianben Song
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengjia Jing
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yan
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Shang C, Sun Y, Wang Y, Shi H, Han X, Mo Y, Wang D, Ke Y, Zeng X. CXCL10 conditions alveolar macrophages within the premetastatic niche to promote metastasis. Cancer Lett 2022; 537:215667. [DOI: 10.1016/j.canlet.2022.215667] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 01/16/2023]
|
9
|
Tey SK, Wong SWK, Yeung CLS, Li JYK, Mao X, Chung CYS, Yam JWP. Liver cancer cells with nuclear MET overexpression release translation regulatory protein-enriched extracellular vesicles exhibit metastasis promoting activity. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e39. [PMID: 38939527 PMCID: PMC11080920 DOI: 10.1002/jex2.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 06/29/2024]
Abstract
MET receptor tyrosine kinase is a cell surface receptor that plays important role in embryonic development and tissue regeneration. Aberrant MET activation has been widely reported in different human cancers, making MET an attractive therapeutic target. The presence of truncated MET within the nucleus (nMET) with potential novel functions poses a great challenge to the current therapeutic strategies against MET surface receptor. Previous work has demonstrated the promoting effect of nMET in aggressive properties of hepatocellular carcinoma (HCC) cells by activating TAK1/NF-κB signalling pathway. Herein, we report the role of nMET in modulating tumour microenvironment and tumour metastasis mediated by extracellular vesicles (EVs). EVs released by nMET overexpressing cells enhanced cell motility and provoked metastasis. Proteomic profiling revealed the enrichment of translational regulatory proteins in EVs derived from nMET overexpressing cells. These proteins include eukaryotic initiation factor (EIF), ribosomal protein small subunit (RPS) and ribosomal protein larger subunit (RPL) gene families. Knockdown of EIF3I, RPS3A and RPL10 diminished the promoting effect of EVs in cell migration invasiveness and metastasis. In conclusion, the findings reveal an unrecognized capacity of nMET to augment HCC through the release of EVs with oncogenic effect. Targeting these translation-related proteins may serve as an alternative treatment for patients with nMET overexpression.
Collapse
Affiliation(s)
- Sze Keong Tey
- Department of PathologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
- School of Biological SciencesCollege of ScienceNanyang Technological UniversitySingapore
| | - Samuel Wan Ki Wong
- Department of PathologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Cherlie Lot Sum Yeung
- Department of PathologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Jason Ying Ki Li
- School of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Xiaowen Mao
- Department of PathologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Clive Yik Sham Chung
- School of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
| | - Judy Wai Ping Yam
- Department of PathologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong
- State Key Laboratory of Liver Research (The University of Hong Kong)Hong Kong
| |
Collapse
|
10
|
He T, Gao Y, Fang Y, Zhang Y, Zhang S, Nan F, Ding J, Chen Y. The HDAC inhibitor GCJ-490A suppresses c-Met expression through IKKα and overcomes gefitinib resistance in non-small cell lung cancer. Cancer Biol Med 2022; 19:j.issn.2095-3941.2021.0130. [PMID: 35188360 PMCID: PMC9425179 DOI: 10.20892/j.issn.2095-3941.2021.0130] [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] [Indexed: 11/14/2022] Open
Abstract
Objective: The novel compound GCJ-490A has been discovered as a pan-histone deacetylase (HDAC) inhibitor that exerts potent inhibitory activity against HDAC1, HDAC3, and HDAC6. Because of the important roles of HDACs in lung cancer development and the high distribution of GCJ-490A in lung tissue, we explored the anti-tumor potency of GCJ-490A against non-small cell lung cancer (NSCLC) in vitro and in vivo in this study. Methods: The in vitro effects of GCJ-490A alone or combined with the EGFR inhibitor gefitinib against NSCLC were measured with proliferation, apoptosis, and colony formation assays. NSCLC xenograft models were used to investigate the efficacy of GCJ-490A combined with gefitinib for the treatment of NSCLC in vivo. Western blot assays, luciferase reporter assays, chromatin immunoprecipitation assays, quantitative real time-PCR, immunohistochemistry, and transcription factor activity assays were used to elucidate possible mechanisms. Results: GCJ-490A effectively inhibited NSCLC cell proliferation and induced apoptosis in vitro and in vivo. Interestingly, inhibition of HDAC1 and HDAC6 by GCJ-490A increased histone acetylation at the IKKα promoter and enhanced IKKα transcription, thus decreasing c-Met. Moreover, this c-Met downregulation was found to be essential for the synergistic anti-tumor activity of GCJ-490A and gefitinib. Conclusions: These findings highlight the promising potential of HDAC inhibitors in NSCLC treatment and provide a rational basis for the application of HDAC inhibitors in combination with EGFR inhibitors in clinical trials.
Collapse
Affiliation(s)
- Ting He
- Division of Anti-Tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yinglei Gao
- Division of Anti-Tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yanfen Fang
- Division of Anti-Tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yangming Zhang
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shuwei Zhang
- University of Chinese Academy of Sciences, Beijing 100049, China.,State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Fajun Nan
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jian Ding
- Division of Anti-Tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Chen
- Division of Anti-Tumor Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
11
|
Wang W, Gao W, Zhu Q, Alasbahi A, Seki E, Yang L. TAK1: A Molecular Link Between Liver Inflammation, Fibrosis, Steatosis, and Carcinogenesis. Front Cell Dev Biol 2021; 9:734749. [PMID: 34722513 PMCID: PMC8551703 DOI: 10.3389/fcell.2021.734749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/22/2021] [Indexed: 12/22/2022] Open
Abstract
Chronic insult and persistent injury can cause liver inflammation, fibrosis, and carcinogenesis; it can also be associated with metabolic disorders. Identification of critical molecules that link the process of inflammation and carcinogenesis will provide prospective therapeutic targets for liver diseases. Rapid advancements in gene engineering technology have allowed the elucidation of the underlying mechanism of transformation, from inflammation and metabolic disorders to carcinogenesis. Transforming growth factor-β-activated kinase 1 (TAK1) is an upstream intracellular protein kinase of nuclear factor kappa-B (NF-κB) and c-Jun N-terminal kinases, which are activated by numerous cytokines, growth factors, and microbial products. In this study, we highlighted the functional roles of TAK1 and its interaction with transforming growth factor-β, WNT, AMP-activated protein kinase, and NF-κB signaling pathways in liver inflammation, steatosis, fibrosis, and carcinogenesis based on previously published articles.
Collapse
Affiliation(s)
- Weijun Wang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenkang Gao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingjing Zhu
- Department of Liver Diseases, Wuhan Jinyintan Hospital, Wuhan, China
| | - Afnan Alasbahi
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ekihiro Seki
- Department of Medicine, Cedars-Sinai, Los Angeles, CA, United States
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Ge QY, Chen J, Li GX, Tan XL, Song J, Ning D, Mo J, Du PC, Liu QM, Liang HF, Ding ZY, Zhang XW, Zhang BX. GRAMD4 inhibits tumour metastasis by recruiting the E3 ligase ITCH to target TAK1 for degradation in hepatocellular carcinoma. Clin Transl Med 2021; 11:e635. [PMID: 34841685 PMCID: PMC8597946 DOI: 10.1002/ctm2.635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 10/13/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Aberrant TAK1 (transforming growth factor β-activated kinase 1) activity is known to be involved in a variety of malignancies, but the regulatory mechanisms of TAK1 remain poorly understood. GRAMD4 (glucosyltransferase Rab-like GTPase activator and myotubularin domain containing 4) is a newly discovered p53-independent proapoptotic protein with an unclear role in HCC (hepatocellular carcinoma). RESULTS In this research, we found that GRAMD4 expression was lower in HCC samples, and its downregulation predicted worse prognosis for patients after surgical resection. Functionally, GRAMD4 inhibited HCC migration, invasion and metastasis. Mechanistically, GRAMD4 interacted with TAK1 to promote its protein degradation, thus, resulting in the inactivation of MAPK (Mitogen-activated protein kinase) and NF-κB pathways. Furthermore, GRAMD4 was proved to recruit ITCH (itchy E3 ubiquitin protein ligase) to promote the ubiquitination of TAK1. Moreover, high expression of TAK1 was correlated with low expression of GRAMD4 in HCC patients. CONCLUSIONS GRAMD4 inhibits the migration and metastasis of HCC, mainly by recruiting ITCH to promote the degradation of TAK1, which leads to the inactivation of MAPK and NF-κB signalling pathways.
Collapse
Affiliation(s)
- Qian yun Ge
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
- Clinical Medical Research Center of Hepatic SurgeryWuhanP. R. China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanP. R. China
| | - Jin Chen
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
- Clinical Medical Research Center of Hepatic SurgeryWuhanP. R. China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanP. R. China
| | - Gan xun Li
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
- Clinical Medical Research Center of Hepatic SurgeryWuhanP. R. China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanP. R. China
| | - Xiao long Tan
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
- Clinical Medical Research Center of Hepatic SurgeryWuhanP. R. China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanP. R. China
| | - Jia Song
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
- Clinical Medical Research Center of Hepatic SurgeryWuhanP. R. China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanP. R. China
| | - Deng Ning
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
- Clinical Medical Research Center of Hepatic SurgeryWuhanP. R. China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanP. R. China
| | - Jie Mo
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
- Clinical Medical Research Center of Hepatic SurgeryWuhanP. R. China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanP. R. China
| | - Peng cheng Du
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
- Clinical Medical Research Center of Hepatic SurgeryWuhanP. R. China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanP. R. China
| | - Qiu meng Liu
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
- Clinical Medical Research Center of Hepatic SurgeryWuhanP. R. China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanP. R. China
| | - Hui fang Liang
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
- Clinical Medical Research Center of Hepatic SurgeryWuhanP. R. China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanP. R. China
| | - Ze yang Ding
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
- Clinical Medical Research Center of Hepatic SurgeryWuhanP. R. China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanP. R. China
| | - Xue wu Zhang
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
- Clinical Medical Research Center of Hepatic SurgeryWuhanP. R. China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanP. R. China
| | - Bi xiang Zhang
- Hepatic Surgery CenterTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
- Clinical Medical Research Center of Hepatic SurgeryWuhanP. R. China
- Hubei Key Laboratory of Hepato‐Pancreato‐Biliary DiseasesWuhanP. R. China
- Key Laboratory of Organ TransplantationMinistry of EducationWuhanP. R. China
- Key Laboratory of Organ TransplantationNational Health CommissionWuhanP. R. China
- Key Laboratory of Organ TransplantationChinese Academy of Medical SciencesWuhanP. R. China
| |
Collapse
|
13
|
Intermediate between Idiopathic Hypereosinophilia and Chronic Eosinophilic Leukemia: A Report of Two Hypereosinophilic Cases with Possible Novel Molecular Mutations. Case Rep Hematol 2021; 2021:1142124. [PMID: 34513100 PMCID: PMC8426067 DOI: 10.1155/2021/1142124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/08/2021] [Accepted: 08/14/2021] [Indexed: 11/17/2022] Open
Abstract
To distinguish a reactive eosinophilia from its malignant counterpart is challenging. Establishing clonality of the eosinophils is crucial and considered the determining factor for establishing a diagnosis. Cases of hypereosinophilia without clear reactive etiologies, no evidence of end-organ damage, normal cytogenetics, and no molecular mutations are termed as “Idiopathic Hypereosinophilia (IHE).” For cases which lie between the spectrum of chronic eosinophilic leukemia (CEL) and IHE, identification of underlying molecular abnormalities might be helpful in better understanding the disease process and prognosis. Here, we report two cases of hypereosinophilia in which five possible novel molecular mutations were identified by targeted next-generation sequencing (NGS) analysis. They were FBXW7, KM2A, TCF3, ERBB4, and MET. With multiple genetic mutations, these cases could be classified as chronic eosinophilic leukemia. Both these young patients responded well to steroid therapy. While targeted NGS is a useful tool in identifying new molecular mutation associated with hypereosinophilia, our cases raise the question of further investigating this entity and if there is a possibility of an intermediate category lying between the spectrum of CEL and IHE. Defining hypereosinophilia with clonal molecular abnormality as a malignant process may need to be revisited. Even though attempts are being made to identify mutations in IHE, it might be more significant clinically to differentiate them based on response to steroid therapy and prognosis.
Collapse
|
14
|
LncRNA TUG1 exhibits pro-fibrosis activity in hypertrophic scar through TAK1/YAP/TAZ pathway via miR-27b-3p. Mol Cell Biochem 2021; 476:3009-3020. [PMID: 33791919 DOI: 10.1007/s11010-021-04142-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 03/19/2021] [Indexed: 01/19/2023]
Abstract
Hypertrophic Scar (HS) is a complicated fibrotic disease. In addition, its pathogenesis is still to be further explored. Long non-coding RNAs (lncRNAs) have been proved to be participated in multiple diseases, including HS. However, the role of lncRNA TUG1 in HS remains unclear. The expression level of RNA and protein in cells were detected by q-PCR and western blot, respectively. MTT assay was performed to test the cell proliferation. Cell migration was detected by transwell assay. Cell apoptosis was measured by flow cytometry. Dual luciferase report assay and RNA pull down were used to verify the relationship between TUG1, miR-27b-3p and TAK1.TUG1 and TAK1 were upregulated in HS, while miR-27b-3p was downregulated. Knockdown of TUG1 significantly suppressed the proliferation and migration and induced the apoptosis of HS fibroblasts (HSF). In addition, silencing of TUG1 notably inhibited the extracellular matrix (ECM) biosynthesis in HSF. Overexpression of miR-27b-3p has the same effect on HS as that of TUG1 knockdown. Meanwhile, TUG1 could sponge miR-27b-3p, and TAK1 was the direct target of miR-27b-3p. Furthermore, knockdown of TUG1 significantly suppressed the fibrosis in HS via miR-27b-3p/TAK1/YAP/TAZ axis mediation. LncRNA TUG1 promotes the fibrosis in HS via sponging miR-27b-3p and then activates TAK1/YAP/TAZ pathway, which may serve as a potential target for treatment of HS.
Collapse
|
15
|
Jin X, Fu W, Li D, Wang N, Chen J, Zeng Z, Guo J, Liu H, Zhong X, Peng H, Yu X, Sun J, Zhang X, Wang X, Xu B, Lin Y, Liu J, Kutter C, Li Y. High Expression of LINC01268 is Positively Associated with Hepatocellular Carcinoma Progression via Regulating MAP3K7. Onco Targets Ther 2021; 14:1753-1769. [PMID: 33727826 PMCID: PMC7954037 DOI: 10.2147/ott.s295814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/22/2021] [Indexed: 11/29/2022] Open
Abstract
Objective As one of the most common neoplastic diseases, hepatocellular carcinoma (HCC) has a high morbidity and mortality, which seriously threatens human health and places a heavy burden on society and medical care. At present, effective early diagnosis, prognosis and treatment of HCC are limited. Altered gene expression patterns of lncRNA are associated with the occurrence, development and prognosis of various malignancies, including HCC. The aim of this study was to investigate the correlation between the expression of LINC01268 and HCC, and to elucidate the potential underlying molecular mechanism. Methods Expression level and localization of LINC01268 in human liver cancer cells and HCC tissues were investigated using RT-qPCR and fluorescent in situ hybridization (FISH), respectively. Correlation of expression levels of LINC01268 and MAP3K7 with differentiation and poor overall patient survival of HCC were analyzed using in house collected and publicly available HCC tissue data. RT-qPCR and Western blot were applied to inspect the effects of depletion and overexpression of LINC01268 on MAP3K7 expression. HCC cell proliferation and apoptosis were also investigated by simultaneous overexpression of LINC01268 and knockdown of MAP3K7, in order to delineate that MAP3K7 is a downstream effector of LINC01268. Results In this study, we identified that LINC01268 was highly expressed in HCC cell lines and tissues. High LINC01268 expression level was associated with lower HCC nodule number, moderate/poor differentiation and poor overall survival. Knockdown of LINC01268 inhibited the proliferation of HCC cells, which was enhanced by overexpression of LINC01268. Co-expression analysis implied an interaction between LINC01268 and MAP3K7. Similar to LINC01268, MAP3K7 was highly expressed in HCC cells, and positively correlated with moderate/poor differentiation as well as poor prognosis. Knockdown of LINC01268 in HCC cell lines led to reduction of MAP3K7 at both mRNA and protein levels. Phenotypic effects due to LINC01268 overexpression in HCC cells were reversed by knockdown of MAP3K7. Conclusion Taken together, the abnormal high expression of LINC01268 is associated with HCC progression via regulating MAP3K7, suggesting LINC01268 as a novel marker for HCC prognosis and potentially a new therapeutic target.
Collapse
Affiliation(s)
- Xiuli Jin
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Weixin Fu
- Science Experiment Center of China Medical University, Shenyang, 110122, People's Republic of China
| | - Dan Li
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Ningning Wang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Jiayu Chen
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Zilu Zeng
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Jiaqi Guo
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Hao Liu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Xinping Zhong
- Department of Hepatobiliary Surgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Hu Peng
- Emergency Department, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, People's Republic of China
| | - Xin Yu
- Department of Human Anatomy, School of Basic Medicine, Dali University, Dali, Yunnan, 671003, People's Republic of China
| | - Jing Sun
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Xinhe Zhang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Xue Wang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Beibei Xu
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Yingbo Lin
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, 17177, Sweden
| | - Jianping Liu
- Emergency Department, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, People's Republic of China
| | - Claudia Kutter
- Department of Microbiology, Tumor and Cell Biology, Science for Life Laboratory, Karolinska Institute, Stockholm, 17177, Sweden
| | - Yiling Li
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| |
Collapse
|
16
|
Srivastava S, Pang KM, Iida M, Nelson MS, Liu J, Nam A, Wang J, Mambetsariev I, Pillai R, Mohanty A, McDaniel N, Behal A, Kulkarni P, Wheeler DL, Salgia R. Activation of EPHA2-ROBO1 Heterodimer by SLIT2 Attenuates Non-canonical Signaling and Proliferation in Squamous Cell Carcinomas. iScience 2020; 23:101692. [PMID: 33196021 PMCID: PMC7644594 DOI: 10.1016/j.isci.2020.101692] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 08/21/2020] [Accepted: 10/13/2020] [Indexed: 12/25/2022] Open
Abstract
The tyrosine kinase receptor ephrin receptor A2 (EPHA2) is overexpressed in lung (LSCC) and head and neck (HNSCC) squamous cell carcinomas. Although EPHA2 can inhibit tumorigenesis in a ligand-dependent fashion via phosphorylation of Y588 and Y772, it can promote tumorigenesis in a ligand-independent manner via phosphorylation of S897. Here, we show that EPHA2 and Roundabout Guidance Receptor 1 (ROBO1) interact to form a functional heterodimer. Furthermore, we show that the ROBO1 ligand Slit Guidance Ligand 2 (SLIT2) and ensartinib, an inhibitor of EPHA2, can attenuate growth of HNSCC cells and act synergistically in LSCC cells. Our results suggest that patients with LSCC and HNSCC may be stratified and treated based on their EPHA2 and ROBO1 expression patterns. Although ~73% of patients with LSCC could benefit from SLIT2+ensartinib treatment, ~41% of patients with HNSCC could be treated with either SLIT2 or ensartinib. Thus, EPHA2 and ROBO1 represent potential LSCC and HNSCC theranostics.
Collapse
Affiliation(s)
- Saumya Srivastava
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Ka Ming Pang
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Mari Iida
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Wisconsin Institute for Medical Research, Madison, WI 53705-2275, USA
| | - Michael S. Nelson
- Light Microscopy Core, City of Hope National Medical Center, Duarte, CA, USA
| | - Jiayi Liu
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Arin Nam
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Jiale Wang
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Isa Mambetsariev
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Raju Pillai
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Atish Mohanty
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Nellie McDaniel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Wisconsin Institute for Medical Research, Madison, WI 53705-2275, USA
| | - Amita Behal
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Prakash Kulkarni
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Deric L. Wheeler
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Wisconsin Institute for Medical Research, Madison, WI 53705-2275, USA
| | - Ravi Salgia
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| |
Collapse
|
17
|
Chen MK, Hsu JL, Hung MC. Nuclear receptor tyrosine kinase transport and functions in cancer. Adv Cancer Res 2020; 147:59-107. [PMID: 32593407 DOI: 10.1016/bs.acr.2020.04.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Signaling functions of plasma membrane-localized receptor tyrosine kinases (RTKs) have been extensively studied after they were first described in the mid-1980s. Plasma membrane RTKs are activated by extracellular ligands and cellular stress stimuli, and regulate cellular responses by activating the downstream effector proteins to initiate a wide range of signaling cascades in the cells. However, increasing evidence indicates that RTKs can also be transported into the intracellular compartments where they phosphorylate traditional effector proteins and non-canonical substrate proteins. In general, internalization that retains the RTK's transmembrane domain begins with endocytosis, and endosomal RTK remains active before being recycled or degraded. Further RTK retrograde transport from endosome-Golgi-ER to the nucleus is primarily dependent on membranes vesicles and relies on the interaction with the COP-I vesicle complex, Sec61 translocon complex, and importin. Internalized RTKs have non-canonical substrates that include transcriptional co-factors and DNA damage response proteins, and many nuclear RTKs harbor oncogenic properties and can enhance cancer progression. Indeed, nuclear-localized RTKs have been shown to positively correlate with cancer recurrence, therapeutic resistance, and poor prognosis of cancer patients. Therefore, understanding the functions of nuclear RTKs and the mechanisms of nuclear RTK transport will further improve our knowledge to evaluate the potential of targeting nuclear RTKs or the proteins involved in their transport as new cancer therapeutic strategies.
Collapse
Affiliation(s)
- Mei-Kuang Chen
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Jennifer L Hsu
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
18
|
Xue H, Yu F, Zhang X, Liu L, Huang L. circ_0000638 inhibits neodymium oxide-induced bronchial epithelial cell inflammation through the miR-498-5p/NF-κB axis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 195:110455. [PMID: 32199215 DOI: 10.1016/j.ecoenv.2020.110455] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 03/02/2020] [Accepted: 03/06/2020] [Indexed: 06/10/2023]
Abstract
Inhalation of neodymium oxide, a type of rare earth metal, can induce various respiratory diseases, such as lung tissue inflammation, but the associated mechanisms underlying this induction are still unclear. In this study, we explored the role and mechanisms of circular RNA in neodymium oxide-induced airway inflammation. Our study showed that treating human bronchial epithelial (16HBE) cells with neodymium oxide caused an inflammatory response by upregulating the expression of interleukin-8 (IL-8) and interleukin-1 beta (IL-1β). Quantitative real-time polymerase chain reaction (qRT-PCR) analyses revealed significant downregulation of circRNA_0000638 in neodymium oxide-treated 16HBE cells. Data from functional analyses further showed that circ_0000638 inhibited inflammation induced by neodymium oxide in 16HBE cells. circ_0000638 targeted miR-498-5p and further increased the expression of NKRF (NF-κB repressing factor), which inhibited the activation of NF-κB (nuclear factor κB). Moreover, circ_0000638 reduced the expression of IL-8 and IL-1β by inhibiting NF-κB activation in neodymium oxide-treated 16HBE cells. These results suggest that circ_0000638 can inhibit NF-κB activation by competitively binding to miR-498-5p, further downregulating the expression of IL-8 and IL-1β in neodymium oxide-treated 16HBE cells.
Collapse
Affiliation(s)
- Hainan Xue
- School of Public Health, Baotou Medical College, Baotou, 014030, Inner Mongolia, PR China
| | - Feng Yu
- School of Public Health, Baotou Medical College, Baotou, 014030, Inner Mongolia, PR China
| | - Xia Zhang
- School of Public Health, Baotou Medical College, Baotou, 014030, Inner Mongolia, PR China
| | - Ling Liu
- School of Public Health, Baotou Medical College, Baotou, 014030, Inner Mongolia, PR China
| | - Lihua Huang
- School of Public Health, Baotou Medical College, Baotou, 014030, Inner Mongolia, PR China.
| |
Collapse
|
19
|
Xiao H, Liu Z. Effects of microRNA‑217 on high glucose‑induced inflammation and apoptosis of human retinal pigment epithelial cells (ARPE‑19) and its underlying mechanism. Mol Med Rep 2019; 20:5125-5133. [PMID: 31702814 PMCID: PMC6854520 DOI: 10.3892/mmr.2019.10778] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/30/2019] [Indexed: 01/21/2023] Open
Abstract
Diabetic retinopathy is a major complication of diabetes. Increasing evidence has indicated that microRNAs (miRs) serves an important role in diabetic retinopathy. However, the expression and mechanism of miR-217 in high glucose-induced human retinal pigment epithelial cells ARPE-19 is still unclear. Therefore, the aim of this study was to investigate the role of miR-217 in high glucose-induced retinal epithelial cell damage, and further to explore the molecular mechanisms. In our study, we found that compared with control group, miR-217 was upregulated in high glucose-induced ARPE-19 cells. In addition, TargetScan and a dual-luciferase reporter gene assay showed that Sirtuin 1 (SIRT1) was a direct target of miR-217. Then, we performed reverse transcription-quantitative polymerase chain reaction assay and western blot assay to explore the expression of SIRT1 in high glucose-induced ARPE-19 cells. Our results demonstrated that SIRT1 was downregulated at the mRNA and protein levels in high glucose-induced ARPE-19 cells. Then, ARPE-19 cells were transfected with inhibitor control, miR-217 inhibitor or miR-217 inhibitor + SIRT1-small interfering RNA for 6 h, and then the cells were treated with 50 mM D-glucose for 24 h. We then investigated the effects of miR-217 inhibitor on ARPE-19 cell viability and apoptosis. An MTT assay revealed that miR-217 inhibitor significantly increased the viability and decreased the apoptosis of high glucose-induced ARPE-19 cells. ELISA indicated that miR-217 inhibitor significantly reduced the expression of inflammatory factors, such as interleukin (IL)-1β, tumor necrosis factor-α, and IL-6 in high glucose-treated ARPE-19 cells. Additionally, a western blot assay demonstrated that miR-217 inhibitor suppressed the expression of p-p65. The effects of miR-217 inhibitor on high glucose-treated ARPE-19 cells were significantly reversed by the silencing the SIRT1 gene. Therefore, our findings suggested that miR-217 inhibitor protected against retinal epithelial cell damage caused by high glucose via targeting SIRT1, thereby playing a protective role in diabetic retinopathy. Targeting miR-217 may have therapeutic potential in the treatment of diabetic retinopathy.
Collapse
Affiliation(s)
- Hongxia Xiao
- Department of Ophthalmology, Jing Men No. 2 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Zhen Liu
- Department of Ophthalmology, Chongqing Aier Eye Hospital, Chongqing 400020, P.R. China
| |
Collapse
|
20
|
Leung Z, Ko FCF, Tey SK, Kwong EML, Mao X, Liu BHM, Ma APY, Fung YME, Che CM, Wong DKH, Lai CL, Ng IOL, Yam JWP. Galectin-1 promotes hepatocellular carcinoma and the combined therapeutic effect of OTX008 galectin-1 inhibitor and sorafenib in tumor cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:423. [PMID: 31640796 PMCID: PMC6805403 DOI: 10.1186/s13046-019-1402-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 08/29/2019] [Indexed: 12/16/2022]
Abstract
Background Galectins are beta-galactose specific binding proteins. In human cancers, including hepatocellular carcinoma (HCC), galectin-1 (Gal-1) is often found to be overexpressed. In order to combat the dismal diagnosis and death rates of HCC, gene silencing and targeted inhibition of Gal-1 was investigated for its improved therapeutic potential. Methods Cellular and secretory Gal-1 levels were analyzed using HCC clinical samples. The study of Gal-1 was carried by both knockdown and overexpression approaches. The stable clones were tested by in vitro assays and in vivo experiments. Mass spectrometry was used to identify downstream targets of Gal-1. The upstream regulator of Gal-1, microRNA-22 (miR-22) was characterized by functional assays. The therapeutic effect of inhibiting Gal-1 was also analyzed. Results Gal-1 overexpression was observed in HCC and correlated with aggressive clinicopathological features and poorer survival. The loss of Gal-1 resulted in hindered cell migration, invasion and anchorage independent growth. This was also observed in the animal models, in that when Gal-1 was knocked down, there were fewer lung metastases. Proteomic profiling of control and Gal-1 knockdown cells identified that the level of retention in endoplasmic reticulum 1 (RER1) was suppressed when Gal-1 level was reduced. The cell motility of Gal-1 knockdown cells was enhanced upon the rescue of RER1 expression. In HCC tissues, Gal-1 and RER1 expressions displayed a significant positive correlation. The upstream regulator of Gal-1, miR-22 was observed to be underexpressed in HCC tissues and negatively correlated with Gal-1. Silencing of miR-22 resulted in the upregulation of Gal-1 and enhanced cell growth, migration and invasion. However, such enhancement was abolished in cells treated with OTX008, an inhibitor of Gal-1. Combinational treatment of OTX008 and sorafenib significantly reduced tumor growth and size. Conclusions Gal-1 overexpression was detected in HCC and this played a role in promoting tumorigenic processes and metastasis. The function of Gal-1 was found to be mediated through RER1. The correlations between miR-22, Gal-1 and RER1 expressions demonstrated the importance of miR-22 regulation on Gal-1/RER1 oncogenic activity. Lastly, the combinational treatment of OTX008 and sorafenib proved to be an improved therapeutic option compared to when administering sorafenib alone. Electronic supplementary material The online version of this article (10.1186/s13046-019-1402-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zoe Leung
- Department of Pathology, The University of Hong Kong, Hong Kong, China
| | | | - Sze Keong Tey
- Department of Pathology, The University of Hong Kong, Hong Kong, China
| | | | - Xiaowen Mao
- Department of Pathology, The University of Hong Kong, Hong Kong, China
| | | | - Angel Po Yee Ma
- Department of Pathology, The University of Hong Kong, Hong Kong, China
| | - Yi Man Eva Fung
- Department of Chemistry, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong, China
| | - Chi-Ming Che
- Department of Chemistry, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong, China
| | - Danny Ka Ho Wong
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Liver Research (The University of Hong Kong), Hong Kong, China
| | - Ching Lung Lai
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Liver Research (The University of Hong Kong), Hong Kong, China
| | - Irene Oi-Lin Ng
- Department of Pathology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Liver Research (The University of Hong Kong), Hong Kong, China
| | - Judy Wai Ping Yam
- Department of Pathology, The University of Hong Kong, Hong Kong, China. .,State Key Laboratory of Liver Research (The University of Hong Kong), Hong Kong, China. .,Department of Pathology, Block T, Queen Mary Hospital, Hong Kong, China.
| |
Collapse
|
21
|
Yang Q, Xie B, Tang H, Meng W, Jia C, Zhang X, Zhang Y, Zhang J, Li H, Fu B. Minichromosome maintenance 3 promotes hepatocellular carcinoma radioresistance by activating the NF-κB pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:263. [PMID: 31208444 PMCID: PMC6580494 DOI: 10.1186/s13046-019-1241-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 05/22/2019] [Indexed: 12/16/2022]
Abstract
Background Hepatocellular carcinoma (HCC) is the most common tumors in the worldwide, it develops resistance to radiotherapy during treatment, understanding the regulatory mechanisms of radioresistance generation is the urgent need for HCC therapy. Methods qRT-PCR, western blot and immunohistochemistry were used to examine MCM3 expression. MTT assay, colony formation assay, terminal deoxynucleotidyl transferase nick end labeling assay and In vivo xenograft assay were used to determine the effect of MCM3 on radioresistance. Gene set enrichment analysis, luciferase reporter assay, western blot and qRT-PCR were used to examine the effect of MCM3 on NF-κB pathway. Results We found DNA replication initiation protein Minichromosome Maintenance 3 (MCM3) was upregulated in HCC tissues and cells, patients with high MCM3 expression had poor outcome, it was an independent prognostic factor for HCC. Cells with high MCM3 expression or MCM3 overexpression increased the radioresistance determined by MTT assay, colony formation assay, TUNEL assay and orthotopic transplantation mouse model, while cells with low MCM3 expression or MCM3 knockdown reduced the radioresistance. Mechanism analysis showed MCM3 activated NF-κB pathway, characterized by increasing the nuclear translocation of p65, the expression of the downstream genes NF-κB pathway and the phosphorylation of IKK-β and IκBα. Inhibition of NF-κB in MCM3 overexpressing cells using small molecular inhibitor reduced the radioresistance, suggesting MCM3 increased radioresistance through activating NF-κB pathway. Moreover, we found MCM3 expression positively correlated with NF-κB pathway in clinic. Conclusions Our findings revealed that MCM3 promoted radioresistance through activating NF-κB pathway, strengthening the role of MCM subunits in the tumor progression and providing a new target for HCC therapy. Electronic supplementary material The online version of this article (10.1186/s13046-019-1241-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qing Yang
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, 600# Tianhe Road, Guangzhou, 510630, China
| | - Binhui Xie
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Hui Tang
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, 600# Tianhe Road, Guangzhou, 510630, China
| | - Wei Meng
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, 600# Tianhe Road, Guangzhou, 510630, China
| | - Changchang Jia
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xiaomei Zhang
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yi Zhang
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, 600# Tianhe Road, Guangzhou, 510630, China
| | - Jianwen Zhang
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, 600# Tianhe Road, Guangzhou, 510630, China.
| | - Heping Li
- Department of Medical Oncology of the Eastern Hospital, The First Affiliated Hospital of Sun Yat-sen University, Zhongshan Er Road, Guangzhou, 510080, China.
| | - Binsheng Fu
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Organ Transplantation Research Center of Guangdong Province, 600# Tianhe Road, Guangzhou, 510630, China.
| |
Collapse
|
22
|
Cheng JS, Tsai WL, Liu PF, Goan YG, Lin CW, Tseng HH, Lee CH, Shu CW. The MAP3K7-mTOR Axis Promotes the Proliferation and Malignancy of Hepatocellular Carcinoma Cells. Front Oncol 2019; 9:474. [PMID: 31214512 PMCID: PMC6558008 DOI: 10.3389/fonc.2019.00474] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/17/2019] [Indexed: 12/28/2022] Open
Abstract
Targeted therapy is currently limited for patients with hepatocellular carcinoma (HCC) due to the lack of suitable targets. Kinases play pivotal roles in many cellular biological processes, whereas dysregulation of kinases may lead to various diseases, particularly cancer. However, the role of kinases in HCC malignancy remains unclear. In this study, we employed a kinome small interfering RNA (siRNA) library, comprising 710 kinase-related genes, to screen whether any kinases were essential for cell proliferation in various HCC cell lines. Through a kinome siRNA library screening, we found that MAP3K7 was a crucial gene for HCC cell proliferation. Pharmacological or genetic ablation of MAP3K7 diminished the growth, migration, and invasion of HCC cells, including primary HCC cells. Stable knockdown of MAP3K7 attenuated tumor formation in a spheroid cell culture model and tumor xenograft mouse model. In addition, silencing MAP3K7 reduced the phosphorylation and expression of mammalian target of rapamycin (mTOR) in HCC cells. MAP3K7 expression was positively correlated with mTOR expression in tumors of patients with HCC. Higher co-expression of MAP3K7 and mTOR was significantly associated with poor prognosis of HCC. Taken together, our results revealed that the MAP3K7-mTOR axis might promote tumorigenesis and malignancy, which provides a potential marker or therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Jin-Shiung Cheng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wei-Lun Tsai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Pei-Feng Liu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Yih-Gang Goan
- Division of Thoracic Surgery, Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chih-Wen Lin
- Division of Gastroenterology and Hepatology, E-Da Dachang Hospital, I-Shou University, Kaohsiung, Taiwan.,School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan
| | - Ho-Hsing Tseng
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Cheng-Hsin Lee
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chih-Wen Shu
- School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan
| |
Collapse
|
23
|
Frazier NM, Brand T, Gordan JD, Grandis J, Jura N. Overexpression-mediated activation of MET in the Golgi promotes HER3/ERBB3 phosphorylation. Oncogene 2019; 38:1936-1950. [PMID: 30390071 PMCID: PMC6417953 DOI: 10.1038/s41388-018-0537-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 09/07/2018] [Accepted: 09/25/2018] [Indexed: 12/16/2022]
Abstract
Ligand-dependent oligomerization of receptor tyrosine kinases (RTKs) results in their activation through highly specific conformational changes in the extracellular and intracellular receptor domains. These conformational changes are unique for each RTK subfamily, limiting cross-activation between unrelated RTKs. The proto-oncogene MET receptor tyrosine kinase overcomes these structural constraints and phosphorylates unrelated RTKs in numerous cancer cell lines. The molecular basis for these interactions is unknown. We investigated the mechanism by which MET phosphorylates the human epidermal growth factor receptor-3 (HER3 or ERBB3), a catalytically impaired RTK whose phosphorylation by MET has been described as an essential component of drug resistance to inhibitors targeting EGFR and HER2. We find that in untransformed cells, HER3 is not phosphorylated by MET in response to ligand stimulation, but rather to increasing levels of MET expression, which results in ligand-independent MET activation. Phosphorylation of HER3 by its canonical co-receptors, EGFR and HER2, is achieved by engaging an allosteric site on the HER3 kinase domain, but this site is not required when HER3 is phosphorylated by MET. We also observe that HER3 preferentially interacts with MET during its maturation along the secretory pathway, before MET is post translationally processed by cleavage within its extracellular domain. This results in accumulation of phosphorylated HER3 in the Golgi apparatus. We further show that in addition to HER3, MET phosphorylates other RTKs in the Golgi, suggesting that this mechanism is not limited to HER3 phosphorylation. These data demonstrate a link between MET overexpression and its aberrant activation in the Golgi endomembranes and suggest that non-canonical interactions between MET and other RTKs occur during maturation of receptors. Our study highlights a novel aspect of MET signaling in cancer that would not be accessible to inhibition by therapeutic antibodies.
Collapse
Affiliation(s)
- Nicole Michael Frazier
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Toni Brand
- Department of Otolaryngology - Head and Neck Surgery, University of California, San Francisco, CA, 94113, USA
| | - John D Gordan
- Division of Hematology and Oncology - University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Jennifer Grandis
- Department of Otolaryngology - Head and Neck Surgery, University of California, San Francisco, CA, 94113, USA
| | - Natalia Jura
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, 94158, USA.
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
24
|
Xie Y, Nurkesh AA, Ibragimova N, Zhanzak Z, Meyerbekova A, Alexeyeva Z, Yesbolatova A, Satayeva M, Mustafa A, Manarbek L, Maipas A, Altaikyzy A, Keneskhanova Z, Akbay B, Chen Z. Systematic analysis of NLMP suggests nuclear localization of RTK/MET kinases resemble cancer cell clearance. J Exp Clin Cancer Res 2019; 38:43. [PMID: 30700325 PMCID: PMC6354337 DOI: 10.1186/s13046-018-1004-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/13/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Some membrane proteins can translocate into the nucleus, defined as nuclear localized membrane proteins (NLMPs), including receptor tyrosine kinases (RTKs). We previously showed that nuclear MET (nMET), a member of RTKs, mediates cancer stem-like cells self-renewal to promote cancer recurrence. However, it is unknown that nMET or mMET, which is the ancestor in the evolution of cancer cell survival and clearance. Here, we aim to study the NLMP functions in cell death, differentiation and survival. METHOD We applied the systematic reanalysis of functional NLMP and clinical investigations of nMET from databases. In addition, we used soft agar assay, immunoblotting, flow cytometry, and immunofluorescence confocal microscopy for examinations of nMET functions including stem-like cell formation, cell signaling, cell cycle regulation, and co-localization with regulators of cell signaling. ShRNA, antibody of recognizing surface membrane MET based treatment were used to downregulate endogenous nMET to uncover its function. RESULTS We predicted and demonstrated that nMET and nEGFR are most likely not ancestors. nMET overexpression induces both cell death and survival with drug resistance and stem cell-like characters. Moreover, the paradoxical function of nMET in both cell death and cell survival is explained by the fact that nMET induces stem cell-like cell growth, DNA damage repair, to evade the drug sensitization for survival of single cells while non-stem cell-like nMET expressing single cells may undergo clearance by cell death through cell cycle arrest induced by p21. CONCLUSION Taken together, our data suggest a link between nuclear RTK and cancer cell evolutionary clearance via cell death, and drug resistance for survival through stemness selection. Targeting evolved nuclear RTKs in cancer stem cells would be a novel avenue for precision cancer therapy.
Collapse
Affiliation(s)
- Yingqiu Xie
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Ayan A. Nurkesh
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Nazgul Ibragimova
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Zhuldyz Zhanzak
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Aizhan Meyerbekova
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Zhanna Alexeyeva
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Aiya Yesbolatova
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Madina Satayeva
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Aidana Mustafa
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Limara Manarbek
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Aisulu Maipas
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Akerke Altaikyzy
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Zhibek Keneskhanova
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Burkitkan Akbay
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Zhenbang Chen
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208 USA
| |
Collapse
|
25
|
Knockdown of PHF5A Inhibits Migration and Invasion of HCC Cells via Downregulating NF- κB Signaling. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1621854. [PMID: 30766880 PMCID: PMC6350539 DOI: 10.1155/2019/1621854] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/29/2018] [Accepted: 12/23/2018] [Indexed: 02/07/2023]
Abstract
Background Inflammation is the major risk factor for the progression of hepatocellular carcinoma (HCC), and the nuclear factor-κB (NF-κB) signaling plays the central role in the inflammation process. However, the activated mechanism of NF-κB signaling in HCC is unclear. Methods The expression of PHF5A is examined by qPCR, western blotting, and immunohistochemistry (IHC) assay. The potential of PHF5A (PHD-finger domain protein 5a) for migration and invasion is examined by wound healing and Transwell assay. Luciferase reporter assay, western blotting, and qPCR were applied to explore the mechanism by which PHF5A is involved in progression of HCC. Results PHF5A was significantly upregulated in HCC tissues and cells. Downregulation of PHF5A inhibits the migration and invasion of HCC cells. Further study demonstrated that PHF5A is implicated in HCC progression through NF-κB signaling. In addition, blocking the NF-κB signaling can weaken the stimulatory effect of PHF5A on migration and invasion of HCC cells. Conclusion PHF5A expression is upregulated in HCC tissues, and depletion of PHF5A inhibits the migration and invasion of HCC cells. Further experiments demonstrated that PHF5A is implicated in NF-κB signaling and knockdown of PHF5A downregulates the activity of NF-κB pathway to inhibit the tumor progression. The above results provide the evidence that PHF5A plays an indispensable role in progressive effect of NF-κB pathway in HCC and may be a novel therapeutic target of HCC.
Collapse
|
26
|
Grzeskowiak CL, Kundu ST, Mo X, Ivanov AA, Zagorodna O, Lu H, Chapple RH, Tsang YH, Moreno D, Mosqueda M, Eterovic K, Fradette JJ, Ahmad S, Chen F, Chong Z, Chen K, Creighton CJ, Fu H, Mills GB, Gibbons DL, Scott KL. In vivo screening identifies GATAD2B as a metastasis driver in KRAS-driven lung cancer. Nat Commun 2018; 9:2732. [PMID: 30013058 PMCID: PMC6048166 DOI: 10.1038/s41467-018-04572-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 05/02/2018] [Indexed: 12/26/2022] Open
Abstract
Genetic aberrations driving pro-oncogenic and pro-metastatic activity remain an elusive target in the quest of precision oncology. To identify such drivers, we use an animal model of KRAS-mutant lung adenocarcinoma to perform an in vivo functional screen of 217 genetic aberrations selected from lung cancer genomics datasets. We identify 28 genes whose expression promoted tumor metastasis to the lung in mice. We employ two tools for examining the KRAS-dependence of genes identified from our screen: 1) a human lung cell model containing a regulatable mutant KRAS allele and 2) a lentiviral system permitting co-expression of DNA-barcoded cDNAs with Cre recombinase to activate a mutant KRAS allele in the lungs of mice. Mechanistic evaluation of one gene, GATAD2B, illuminates its role as a dual activity gene, promoting both pro-tumorigenic and pro-metastatic activities in KRAS-mutant lung cancer through interaction with c-MYC and hyperactivation of the c-MYC pathway.
Collapse
Affiliation(s)
- Caitlin L Grzeskowiak
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Samrat T Kundu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiulei Mo
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Andrei A Ivanov
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Oksana Zagorodna
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Hengyu Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Richard H Chapple
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yiu Huen Tsang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Daniela Moreno
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Maribel Mosqueda
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Karina Eterovic
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jared J Fradette
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sumreen Ahmad
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Fengju Chen
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zechen Chong
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Chad J Creighton
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Haian Fu
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Gordon B Mills
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, 77030, USA.
| | - Kenneth L Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
27
|
Ji YX, Huang Z, Yang X, Wang X, Zhao LP, Wang PX, Zhang XJ, Alves-Bezerra M, Cai L, Zhang P, Lu YX, Bai L, Gao MM, Zhao H, Tian S, Wang Y, Huang ZX, Zhu XY, Zhang Y, Gong J, She ZG, Li F, Cohen DE, Li H. The deubiquitinating enzyme cylindromatosis mitigates nonalcoholic steatohepatitis. Nat Med 2018; 24:213-223. [PMID: 29291351 DOI: 10.1038/nm.4461] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/15/2017] [Indexed: 02/07/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a common clinical condition that can lead to advanced liver diseases. Lack of effective pharmacotherapies for NASH is largely attributable to an incomplete understanding of its pathogenesis. The deubiquitinase cylindromatosis (CYLD) plays key roles in inflammation and cancer. Here we identified CYLD as a suppressor of NASH in mice and in monkeys. CYLD is progressively degraded upon interaction with the E3 ligase TRIM47 in proportion to NASH severity. We observed that overexpression of Cyld in hepatocytes concomitantly inhibits lipid accumulation, insulin resistance, inflammation and fibrosis in mice with NASH induced in an experimental setting. Mechanistically, CYLD interacts directly with the kinase TAK1 and removes its K63-linked polyubiquitin chain, which blocks downstream activation of the JNK-p38 cascades. Notably, reconstitution of hepatic CYLD expression effectively reverses disease progression in mice with dietary or genetically induced NASH and in high-fat diet-fed monkeys predisposed to metabolic syndrome. Collectively, our findings demonstrate that CYLD mitigates NASH severity and identify the CYLD-TAK1 axis as a promising therapeutic target for management of the disease.
Collapse
Affiliation(s)
- Yan-Xiao Ji
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Zan Huang
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Xia Yang
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xiaozhan Wang
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ling-Ping Zhao
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Pi-Xiao Wang
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- Institute of Model Animal of Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Michele Alves-Bezerra
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, New York, USA
| | - Lin Cai
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
| | - Peng Zhang
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yue-Xin Lu
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Lan Bai
- Institute of Model Animal of Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mao-Mao Gao
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Huan Zhao
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Song Tian
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yong Wang
- Institute of Model Animal of Wuhan University, Wuhan, China
| | | | - Xue-Yong Zhu
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yan Zhang
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Jun Gong
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhi-Gang She
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Feng Li
- Basic Medical School, Wuhan University, Wuhan, China
| | - David E Cohen
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, New York, USA
| | - Hongliang Li
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
28
|
Affiliation(s)
- Paul E Hutchinson
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Paul K Wallace
- Department of Flow and Image Cytometry, Roswell Park Cancer Institute, Buffalo, New York 14263
| | - Vuong Nguyen
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|